Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(19)2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36232951

ABSTRACT

Brain-tumor-initiating cells (BTICs) of proneural and mesenchymal origin contribute to the highly malignant phenotype of glioblastoma (GB) and resistance to current therapies. BTICs of different subtypes were challenged with oxidative phosphorylation (OXPHOS) inhibition with metformin to assess the differential effects of metabolic intervention on key resistance features. Whereas mesenchymal BTICs varied according to their invasiveness, they were in general more glycolytic and less responsive to metformin. Proneural BTICs were less invasive, catabolized glucose more via the pentose phosphate pathway, and responded better to metformin. Targeting glycolysis may be a promising approach to inhibit tumor cells of mesenchymal origin, whereas proneural cells are more responsive to OXPHOS inhibition. Future clinical trials exploring metabolic interventions should account for metabolic heterogeneity of brain tumors.


Subject(s)
Brain Neoplasms , Glioblastoma , Metformin , Brain/metabolism , Brain Neoplasms/metabolism , Cell Line, Tumor , Glioblastoma/metabolism , Glucose/metabolism , Humans , Metformin/metabolism , Metformin/pharmacology , Metformin/therapeutic use , Neoplastic Stem Cells/metabolism
2.
J Neuropathol Exp Neurol ; 81(11): 885-899, 2022 10 18.
Article in English | MEDLINE | ID: mdl-35980299

ABSTRACT

von Hippel-Lindau (VHL) disease is an autosomal dominant hereditary cancer disorder caused by a germline mutation in the VHL tumor suppressor gene. Loss of the wild-type allele results in VHL deficiency and the potential formation of cerebellar hemangioblastomas, which resemble embryonic hemangioblast proliferation and differentiation processes. Multiple, microscopic, VHL-deficient precursors, termed developmentally arrested structural elements (DASEs), consistently involve the cerebellar molecular layer in VHL patients, indicating the tumor site of origin. Unlike hemangioblastomas, however, cerebellar DASEs do not express brachyury, a mesodermal marker for hemangioblasts. In this study, neuronal progenitors occupying the molecular layer were investigated as tumor cells of origin. By immunohistochemistry, cerebellar DASEs and hemangioblastomas lacked immunoreactivity with antibody ZIC1 (Zic family member 1), a granule cell progenitor marker with concordance from oligonucleotide RNA expression array analyses. Rather, cerebellar DASEs and hemangioblastomas were immunoreactive with antibody PAX2 (paired box 2), a marker of basket/stellate cell progenitors. VHL cerebellar cortices also revealed PAX2-positive cells in Purkinje and molecular layers, resembling the histological and molecular development of basket/stellate cells in postnatal non-VHL mouse and human cerebella. These data suggest that VHL deficiency can result in the developmental arrest of basket/stellate cells in the human cerebellum and that these PAX2-positive, initiated cells await another insult or signal to form DASEs and eventually, tumors.


Subject(s)
Cerebellar Neoplasms , Hemangioblastoma , von Hippel-Lindau Disease , Animals , Mice , Infant, Newborn , Humans , Hemangioblastoma/genetics , Hemangioblastoma/metabolism , Hemangioblastoma/pathology , von Hippel-Lindau Disease/complications , von Hippel-Lindau Disease/genetics , von Hippel-Lindau Disease/metabolism , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Cerebellum/pathology , Oligonucleotides/metabolism , RNA/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
3.
Mol Cell Endocrinol ; 470: 105-114, 2018 07 15.
Article in English | MEDLINE | ID: mdl-28986303

ABSTRACT

BACKGROUND: Pre-operative detection of corticotropin (ACTH) secreting microadenomas causing Cushing's disease (CD) improves surgical outcomes. Current best magnetic resonance imaging fails to detect up to 40% of these microadenomas. 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET) is specific, but not sensitive in detecting corticotropinomas. Theoretically, secretagogue stimulation with corticotropin releasing hormone (CRH) could improve detection of adenomas with 18F-FDG PET. Previous attempts with simultaneous CRH stimulation have failed to demonstrate increased 18F-FDG uptake in corticotropinomas. We hypothesized that CRH stimulation leads to a delayed elevation in glucose uptake in corticotropinomas. METHODS: Clinical data was analyzed for efficacy of CRH in improving 18FDG-PET detection of corticotropinomas in CD. Glucose transporter 1 (GLUT1) immunoreactivity was performed on surgical specimens. Ex-vivo, viable cells from these tumors were tested for secretagogue effects (colorimetric glucose uptake), and for fate of intracellular glucose (glycolysis stress analysis). Validation of ex-vivo findings was performed with AtT-20 cells. RESULTS: CRH increased glucose uptake in human-derived corticotroph tumor cells and AtT-20, but not in normal murine or human corticotrophs (p < 0.0001). Continuous and intermittent (1 h) CRH exposure increased glucose uptake in AtT-20 with maximal effect at 4 h (p = 0.001). Similarly, CRH and 8-Br-cAMP led to robust GLUT1 upregulation and increased membrane translocation at 2 h, while fasentin suppressed baseline (p < 0.0001) and CRH-mediated glucose uptake. Expectedly, intra-operatively collected corticotropinomas demonstrated GLUT1 overexpression. Lastly, human derived corticotroph tumor cells demonstrated increased glycolysis and low glucose oxidation. CONCLUSION: Increased and delayed CRH-mediated glucose uptake differentially occurs in adenomatous corticotrophs. Delayed secretagogue-stimulated 18F-FDG PET could improve microadenoma detection.


Subject(s)
ACTH-Secreting Pituitary Adenoma/metabolism , Corticotropin-Releasing Hormone/pharmacology , Glucose Transporter Type 1/metabolism , Glucose/metabolism , Animals , Arginine Vasopressin/pharmacology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Corticotrophs/metabolism , Dexamethasone/pharmacology , Glucose Transporter Type 1/genetics , Glycolysis/drug effects , Humans , Mice , Protein Transport/drug effects , Sheep , Transcription, Genetic/drug effects
4.
Sci Rep ; 7: 40822, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28094316

ABSTRACT

Von Hippel-Lindau (VHL)-associated hemangioblastomas (VHL-HB) arise in the central nervous system (CNS), and are a leading cause of morbidity and mortality in VHL disease. Currently, surgical resection is the most effective way to manage symptomatic VHL-HBs. Surgically unresectable VHL-HBs or those in frail patients are challenging problems. Therapies targeting oncologic and vascular endothelial growth factor (VEGF) pathways have failed to demonstrate tumor control. Our experience and previous reports on VHL-HB avidity to somatostatin analogues suggested somatostatin receptor (SSTR) expression in VHL-HBs, offering an alternative therapeutic strategy. We explored this possibility by demonstrating consistent histologic expression of SSTR1, 2a, 4, and 5 in VHL-HBs. We found that somatostatin analogue octreotide induces apoptosis in VHL-HB stromal cells in a dose-dependent fashion by BAX - caspase-3 pathway unrelated to canonical VHL pathway. When administered to a patient with unresectable symptomatic suprasellar hemangioblastoma, octreotide resulted in tumor volume reduction, symptom stabilization, and tumor cytopenia on repeat 68Ga-DOTA-TATE positron emission tomography (PET) within 6 months, suggesting tumor infarction. We conclude that VHL-HBs harbor multiple SSTR subtypes that offer actionable chemo-therapeutic strategy for management of symptomatic, unresectable tumors by somatostatin analogue therapy.


Subject(s)
Brain Neoplasms/drug therapy , Hemangioblastoma/drug therapy , Octreotide/pharmacology , Receptors, Somatostatin/genetics , von Hippel-Lindau Disease/drug therapy , Apoptosis/drug effects , Brain Neoplasms/pathology , Cells, Cultured , Female , Hemangioblastoma/pathology , Humans , Middle Aged , Octreotide/analogs & derivatives , Octreotide/therapeutic use , Organometallic Compounds/therapeutic use , Radiopharmaceuticals/therapeutic use , Receptors, Somatostatin/agonists , Receptors, Somatostatin/metabolism , von Hippel-Lindau Disease/pathology
5.
J Neurosurg ; 127(4): 905-916, 2017 10.
Article in English | MEDLINE | ID: mdl-28059647

ABSTRACT

OBJECTIVE To study peritumoral brain edema (PTBE), it is necessary to create a model that accurately simulates vasogenic brain edema (VBE) without introducing a complicated tumor environment. PTBE associated with brain tumors is predominantly a result of vascular endothelial growth factor (VEGF) secreted by brain tumors, and VEGF infusion alone can lead to histological blood-brain barrier (BBB) breakdown in the absence of tumor. VBE is intimately linked to BBB breakdown. The authors sought to establish a model for VBE with chronic infusion of VEGF that can be validated by serial in-vivo MRI and histological findings. METHODS Male Fischer rats (n = 182) underwent stereotactic striatal implantation of MRI-safe brain cannulas for chronic infusion of VEGF (2-20 µg/ml). Following a preinfusion phase (4-6 days), the rats were exposed to VEGF or control rat serum albumin (1.5 µl/hr) for as long as 144 hours. Serial MRI was performed during infusion on a high-field (9.4-T) machine at 12-24, 24-36, 48-72, and 120-144 hours. Rat brains were then collected and histological analysis was performed. RESULTS Control animals and animals infused with 2 µg/ml of VEGF experienced no neurological deficits, seizure activity, or abnormal behavior. Animals treated with VEGF demonstrated a significantly larger volume (42.90 ± 3.842 mm3) of T2 hyper-attenuation at 144 hours when compared with the volume (8.585 ± 1.664 mm3) in control animals (mean difference 34.31 ± 4.187 mm3, p < 0.0001, 95% CI 25.74-42.89 mm3). Postcontrast T1 enhancement in the juxtacanalicular region indicating BBB breakdown was observed in rats undergoing infusion with VEGF. At the later time periods (120-144 hrs) the volume of T1 enhancement (34.97 ± 8.99 mm3) was significantly less compared with the region of edema (p < 0.0001). Histologically, no evidence of necrosis or inflammation was observed with VEGF or control infusion. Immunohistochemical analysis demonstrated astrocyte activation, vascular remodeling, and increased claudin-5 expression in juxtacanalicular regions. Aquaporin-4 expression was increased in both control and VEGF animals in the juxtacanalicular regions. CONCLUSIONS The results of this study show that chronic brain infusion of VEGF creates a reliable model of VBE. This model lacks necrosis and inflammation that are characteristic of previous models of VBE. The model allows for a precise investigation into the mechanism of VBE formation. The authors also anticipate that this model will allow for investigation into the mechanism of glucocorticoid action in abrogating VBE, and to test novel therapeutic strategies targeting PTBE.


Subject(s)
Brain Edema , Vascular Endothelial Growth Factor A/administration & dosage , Animals , Brain Edema/diagnosis , Brain Edema/etiology , Brain Neoplasms/complications , Disease Models, Animal , Magnetic Resonance Imaging , Male , Rats, Inbred F344
6.
Sci Rep ; 6: 35486, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27748427

ABSTRACT

von Hippel-Lindau (VHL) patients develop multiple central nervous system hemangioblastomas (HB). Some HBs become symptomatic with exponential growth or cyst formation following long periods of quiescence. Understanding the factors underlying growth in hemangioblastoma may lead to better strategies to arrest or prevent tumor growth. In 5 VHL patients, we resected quiescent hemangioblastomas (Q-HB) that were en-route during surgical access to symptomatic hemangioblastomas (S-HB), for matched tumor analysis. Quantitative reverse transcriptase analysis demonstrated a 2-fold increase in EPO expression in all S-HB, while 4/5 showed either Hypoxia Inducible Factor-1α or 2α upregulation. Additionally, all S-HB had increased phosphorylated erythropoietin (EPO) receptor and phosphorylated STAT-5 relative to matched Q-HB, with increased phosphorylated JAK-2 largely confined to the stromal cells in clusters within the tumors. These findings suggest that Q-HB to S-HB conversion may be associated with an erythropoietin-signaling loop. Furthermore, we found that EPO is detectable in cyst fluid from S-HB (n = 14), while absent in CSF (n = 1). Additionally, S-HB presentation or S-HB resection does not result in discernible change in serum EPO or hemoglobin (n = 60). These observations suggest that the altered erythropoietin signaling is focal and suggests that studying modulation of erythropoietin receptor pathway may lead to strategies in preventing HB growth.


Subject(s)
Erythropoietin/metabolism , Hemangioblastoma/etiology , Hemangioblastoma/metabolism , Signal Transduction , von Hippel-Lindau Disease/etiology , von Hippel-Lindau Disease/metabolism , Adult , Biomarkers , Female , Hemangioblastoma/diagnosis , Humans , Male , Middle Aged , Radiography , Severity of Illness Index , von Hippel-Lindau Disease/diagnosis
7.
J Neurooncol ; 130(1): 111-122, 2016 10.
Article in English | MEDLINE | ID: mdl-27568035

ABSTRACT

Transcription factors that induce epithelial-mesenchymal transition (EMT) promote invasion, chemoresistance and a stem-cell phenotype in epithelial tumors, but their roles in central nervous system tumors are not well-understood. We hypothesized these transcription factors have a functional impact in grades II-III gliomas. Using the National Cancer Institute (NCI) Repository for Molecular Brain Neoplasia Data (REMBRANDT) and the Cancer Genome Atlas (TCGA) Lower-Grade Glioma (LGG) data, we determined the impact of EMT-promoting transcription factors (EMT-TFs) on overall survival in grades II-III gliomas, compared their expression across common genetic subtypes and subsequently validated these findings in a set of 31 tumors using quantitative real-time polymerase chain reaction (PCR) and immunohistochemistry. Increased expression of the gene coding for the transcriptional repressor Zinc Finger E box-binding Homeobox 1 (ZEB1) was associated with a significant increase in overall survival (OS) on Kaplan-Meier analysis. Genetic subtype analysis revealed that ZEB1 expression was relatively increased in IDH1/2-mutant gliomas, and IDH1/2-mutant gliomas expressed significantly lower levels of many ZEB1 transcriptional targets. Similarly, IDH1/2-mutant tumors expressed significantly higher levels of targets of microRNA 200C (MIR200C), a key regulator of ZEB1. In a validation study, ZEB1 mRNA was significantly increased in IDH1-mutant grades II-III gliomas, and ZEB1 protein expression was more pronounced in these tumors. Our findings demonstrate a novel relationship between IDH1/2 mutations and expression of ZEB1 and its transcriptional targets. Therapy targeting ZEB1-associated pathways may represent a novel therapeutic avenue for this class of tumors.


Subject(s)
Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/genetics , Glioma/metabolism , Isocitrate Dehydrogenase/genetics , Mutation/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Databases, Factual/statistics & numerical data , Female , Glioma/genetics , Glioma/mortality , Humans , Isocitrate Dehydrogenase/metabolism , Kaplan-Meier Estimate , Male , RNA, Messenger/metabolism , Statistics as Topic
8.
J Neurosurg ; 122(6): 1444-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25859802

ABSTRACT

OBJECT: Pituitary adenomas occasionally undergo infarction, apoplexy, which often destroys much of the tumor. It is well known that apoplexy can be precipitated by several acute factors, including cardiac surgery, other types of surgery, trauma, insulin infusion, and stimulation with administration of hypothalamic releasing factors. METHODS: The prior focus on mechanisms underlying pituitary apoplexy has been on these acute events. Less attention has been given to the endogenous features of pituitary tumors that make them susceptible to spontaneous infarction, despite that most pituitary apoplexy occurs in the absence of a recognized precipitating event. The authors examine intrinsic features of pituitary adenomas that render them vulnerable to apoplexy-features such as high metabolic demand, paucity of angiogenesis, and sparse vascularity, qualities that have previously not been linked with apoplexy-and argue that it is these features of adenomas that underlie their susceptibility to spontaneous infarction. The sensitivity of freshly cultured pituitary adenomas to hypoglycemia is assessed. RESULTS: Adenomas have high metabolic demand, limited angiogenesis, and reduced vessel density compared with the normal gland. Pituitary adenoma cells do not survive in the presence of reduced or absent concentrations of glucose. CONCLUSIONS: The authors propose that the frequent ischemic infarction of pituitary adenomas is the product of intrinsic features of these tumors. These endogenous qualities create a tenuous balance between high metabolic demand and marginal tissue perfusion. Thus, the tumor is vulnerable to spontaneous infarction or to acute ischemia by any event that acutely alters the balance between tumor perfusion and tumor metabolism, events such as acute systemic hypotension, abruptly decreased supply of nutrients, hypoglycemia with insulin administration, or increase in the tumor's metabolic demand due to administration of hypothalamic releasing factors. It may be possible to take advantage of these intrinsic features of pituitary adenomas by using aspects of this vulnerability for development of new approaches for treatment.


Subject(s)
Adenoma/pathology , Pituitary Apoplexy/pathology , Pituitary Neoplasms/pathology , Adenoma/metabolism , Adenoma/surgery , Energy Metabolism/physiology , Glucose/metabolism , Humans , Male , Middle Aged , Pituitary Apoplexy/metabolism , Pituitary Apoplexy/surgery , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/surgery
9.
Sci Rep ; 4: 4102, 2014 Feb 17.
Article in English | MEDLINE | ID: mdl-24531117

ABSTRACT

von Hippel-Lindau disease (VHL) patients develop highly vascular tumors, including central nervous system hemangioblastomas. It has been hypothesized that the vascular nature of these tumors is the product of reactive angiogenesis. However, recent data indicate that VHL-associated hemangioblastoma neoplastic cells originate from embryologically-arrested hemangioblasts capable of blood and endothelial cell differentiation. To determine the origin of tumor vasculature in VHL-associated hemangioblastomas, we analyzed the vascular elements in tumors from VHL patients. We demonstrate that isolated vascular structures and blood vessels within VHL-associated hemangioblastomas are a result of tumor-derived vasculogenesis. Further, similar to hemangioblastomas, we demonstrate that other VHL-associated lesions possess vascular tissue of tumor origin and that tumor-derived endothelial cells emerge within implanted VHL deficient UMRC6 RCC murine xenografts. These findings further establish the embryologic, developmentally arrested, hemangioblast as the tumor cell of origin for VHL-associated hemangioblastomas and indicate that it is also the progenitor cell for other VHL-associated tumors.


Subject(s)
Cerebellar Neoplasms/pathology , Hemangioblastoma/pathology , von Hippel-Lindau Disease/diagnosis , Animals , Cell Line, Tumor , Cerebellar Neoplasms/blood supply , Cerebellar Neoplasms/etiology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Factor VIII/metabolism , Hemangioblastoma/blood supply , Hemangioblastoma/etiology , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Loss of Heterozygosity , Male , Mice , Mice, Inbred NOD , Neovascularization, Pathologic , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Transplantation, Heterologous , von Hippel-Lindau Disease/complications , von Hippel-Lindau Disease/pathology
10.
J Neurosurg ; 119(2): 427-33, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23621605

ABSTRACT

OBJECT: The astrocytic contribution to the blood-brain barrier (BBB) in metastatic and primary malignant brain tumors is not well understood. To better understand the vascular properties associated with metastatic and primary malignant brain tumors, the authors systematically analyzed the astrocytic component of the BBB in brain neoplasms. METHODS: Twelve patients who underwent resection of metastatic or primary brain neoplasms (4 metastatic lesions, 2 low-grade astrocytomas, 2 anaplastic astrocytomas, and 4 glioblastoma multiforme) were included. Clinical, MRI, operative, histopathological and immunohistochemical (glial fibrillary acidic protein [GFAP], CD31, and aquaporin 4 [AQ4]) findings were analyzed. RESULTS: Intratumoral regions of MRI enhancement corresponded with breakdown of the normal astrocyte-endothelial cell relationship in the BBB in metastatic deposits and malignant gliomas. Metastases demonstrated lack of perivascular GFAP and AQ4 on CD31-positive intratumoral vessels. At the metastasis-brain interface, normalization of GFAP and AQ4 staining associated with intraparenchymal vessels was observed. Intratumoral vasculature in enhancing regions of high-grade gliomas revealed gaps in GFAP and AQ4 staining consistent with disintegration of the normal astrocyte-endothelial cell association in the BBB. Intratumoral vasculature in low-grade and nonenhancing regions of high-grade gliomas maintained the normal astrocyte-endothelial cell relationship seen in an intact BBB, with GFAP- and AQ4-positive glial processes that were uniformly associated with the CD31-positive vasculature. CONCLUSIONS: Regions of MRI enhancement in metastatic and primary malignancies correspond to areas of breakdown of the physiological astrocyte-endothelial cell relationship of the BBB, including loss of normal perivascular astrocytic architecture on GFAP and AQ4 immunohistochemistry. Nonenhancing areas are associated with preservation of the normal astrocyte-endothelial cell relationship of the intact BBB.


Subject(s)
Astrocytes/pathology , Astrocytoma/pathology , Blood-Brain Barrier/pathology , Brain Neoplasms/pathology , Glioblastoma/pathology , Adult , Aged , Astrocytes/metabolism , Astrocytoma/metabolism , Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Female , Glial Fibrillary Acidic Protein/metabolism , Glioblastoma/metabolism , Humans , Magnetic Resonance Imaging , Male , Middle Aged
12.
Neuro Oncol ; 14(11): 1357-66, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23074198

ABSTRACT

Carbonic anhydrase (CA) IX is over-expressed in glioblastoma; however, its functions in this context are unknown. Metabolically, glioblastomas are highly glycolytic, leading to a significant lactic acid load. Paradoxically, the intracellular pH is alkaline. We hypothesized that CAIX contributes to the extrusion of hydrogen ions into the extracellular space, thereby moderating intra- and extracellular pH and creating an environment conductive to enhanced invasion. We investigated the role of CAIX as a prognostic marker in patients with glioblastoma and its biological function in vitro. CAIX expression was analyzed in 59 patients with glioblastoma by immunohistochemistry. The expression levels were correlated to overall survival. In vitro, U251 and Ln 18 glioblastoma cells were incubated under hypoxia to induce CAIX expression, and RNA interference (RNAi) was used to examine the function of CAIX on cell attachment, invasion, intracellular energy transfer, and susceptibility to adjuvant treatment. High CAIX expression was identified as an independent factor for poor survival in patients with glioblastoma. In vitro, cell attachment and invasion were strongly reduced after knockdown of CAIX. Finally, the effects of radiation and chemotherapy were strongly augmented after CAIX interference and were accompanied by a higher rate of apoptotic cell death. CAIX is an independent prognostic factor for poor outcome in patients with glioblastoma. Cell attachment, invasion, and survival during adjuvant treatment are significantly influenced by high CAIX expression. These results indicate that inhibition of CAIX is a potential metabolic target for the treatment of patients with glioblastoma.


Subject(s)
Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Brain Neoplasms/enzymology , Carbonic Anhydrase IV/metabolism , Glioblastoma/enzymology , Blotting, Western , Brain Neoplasms/mortality , Cell Adhesion/physiology , Cell Movement/physiology , Female , Glioblastoma/mortality , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Invasiveness , Prognosis , RNA Interference
13.
J Neurosurg ; 115(4): 796-801, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21761968

ABSTRACT

OBJECT: In craniopharyngiomas, cystic growth causes pressure on vital structures of the adjacent brain, leading to significant morbidity. However, the molecular pathogenesis of this cyst formation remains unknown. Carbonic anhydrase IX (CA IX) is a tumor-associated, hypoxia-inducible enzyme, which can cause fluid production and development of cysts. The authors investigated CA IX expression in craniopharyngiomas and its correlation with the extent of cyst formation. In addition, the major pathways of CA IX regulation, hypoxia and p53 mutation, were analyzed. METHODS: Expression of CA IX was analyzed in 20 craniopharyngioma patients by means of in situ hybridization and immunohistochemistry. Preoperative imaging was used to quantify cyst volume. To analyze putative hypoxic induction of CA IX, immunohistochemical staining for HIF-1α and VEGF was performed. Since p53 negatively regulates CA IX expression, we also analyzed the tumors for p53 mutation by direct sequencing. RESULTS: Significant CA IX was found in 85% of the 20 cases. The extent of CA IX expression was significantly correlated with cyst volume. HIF-1α expression was largely absent in all tissue samples, whereas moderate VEGF expression was present in a subset of cases but without correlation to cyst volume. No p53 mutation was found in any of the analyzed tumors. CONCLUSIONS: Carbonic anhydrase IX, which is virtually absent in normal brain, is significantly upregulated in craniopharyngiomas and shows a significant association with cyst size. The mechanisms of regulation remain unknown, since neither hypoxia nor p53 appears to play a role. These results indicate that inhibition of CA IX may be a potential target for the adjuvant treatment in patients with cystic craniopharyngiomas.


Subject(s)
Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Carbonic Anhydrases/metabolism , Craniopharyngioma/metabolism , Pituitary Neoplasms/metabolism , Adolescent , Adult , Aged , Antigens, Neoplasm/genetics , Biomarkers, Tumor/genetics , Carbonic Anhydrase IX , Carbonic Anhydrases/genetics , Child , Child, Preschool , Craniopharyngioma/genetics , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Middle Aged , Pituitary Neoplasms/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
14.
J Neurosurg ; 115(3): 512-7, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21663414

ABSTRACT

OBJECT: Central nervous system hemangioblastomas are the most common manifestation of von Hippel-Lindau (VHL) disease, an autosomal dominant tumor suppressor syndrome that results in loss of VHL protein function and continuous upregulation of hypoxia-inducible factors. These tumors are composed of neoplastic stromal cells and abundant vasculature. Stromal cells express markers consistent with multipotent embryonically arrested hemangioblasts, which are precursors for hematopoietic and vascular lineages. Notch receptors are transmembrane signaling molecules that regulate multiple developmental processes including hematopoiesis and vasculogenesis. To investigate the importance of notch signaling in the development of VHL disease-associated CNS hemangioblastomas, the authors examined the presence of the four notch receptors and downstream notch effectors in this setting. METHODS: The authors used surgical specimens obtained from confirmed VHL-associated hemangioblastomas. Immunohistochemical analysis for the four notch receptors and the downstream effectors was performed on formalin-fixed paraffin-embedded sections. Western blot analysis for HES1 was performed on frozen specimens. RESULTS: All four notch receptors are present in hemangioblastomas. NOTCH1 and NOTCH4 receptors were widely and prominently expressed in both the stromal and vascular cells, NOTCH2 receptor expression was limited to primarily stromal cells, and NOTCH3 receptor expression was limited to vascular cells. All 4 receptors displayed a nuclear presence. Immunohistochemical analysis also demonstrated that downstream notch effectors, HES1 and HES5, were uniformly expressed in tumor stromal and vascular cells, but HES3, HEY1, and HEY2 were not. Strong HES1 expression was confirmed by Western blot analysis. CONCLUSIONS: The presence of all four notch receptors and downstream effector molecules suggests that the notch signaling pathway plays a critical role in the maintenance of the undifferentiated pluripotent phenotype of these tumors and in the associated vascular response. Moreover, the prominent expression of notch receptors in VHL-associated CNS hemangioblastomas reveals a new and possibly potent therapeutic target.


Subject(s)
Central Nervous System Neoplasms/metabolism , Hemangioblastoma/metabolism , Receptors, Notch/metabolism , von Hippel-Lindau Disease/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/metabolism , Central Nervous System Neoplasms/pathology , DNA-Binding Proteins/metabolism , Hemangioblastoma/pathology , Homeodomain Proteins/metabolism , Humans , Repressor Proteins , Transcription Factor HES-1 , Transcription Factors/metabolism , von Hippel-Lindau Disease/pathology
15.
Neoplasia ; 10(10): 1146-53, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18813354

ABSTRACT

Inactivation of the von Hippel-Lindau (VHL) gene and activation of the hypoxia-inducible factor (HIF) in susceptible cells precedes formation of tumorlets and frank tumor in the epididymis of male VHL patients. We performed detailed histologic and molecular pathologic analysis of tumor-free epididymal tissues from VHL patients to obtain further insight into early epididymal tumorigenesis. Four epididymides from two VHL patients were serially sectioned to allow for three-dimensional visualization of morphologic changes. Areas of interest were genetically analyzed by tissue microdissection, immunohistochemistry for HIF and markers for mesonephric differentiation, and in situ hybridization for HIF downstream target vascular endothelial growth factor. Structural analysis of the epididymides revealed marked deviations from the regular anatomic structure resulting from impaired organogenesis. Selected efferent ductules were represented by disorganized mesonephric cells, and the maldeveloped mesonephric material was VHL-deficient by allelic deletion analysis. Furthermore, we observed maldeveloped mesonephric material near cystic structures, which were also VHL-deficient and were apparent derivatives of maldeveloped material. Finally, a subset of VHL-deficient cells was structurally integrated in regular efferent ductules; proliferation of intraductular VHL-deficient cells manifests itself as papillary growth into the ductular lumen. Furthermore, we clarify that that there is a pathogenetic continuum between microscopic tumorlets and formation of tumor. In multiple locations, three-dimensional reconstruction revealed papillary growth to extend deeply into ductular lumina, indicative of progression into early hamartoma-like neoplasia. We conclude epididymal tumorigenesis in VHL disease to occur in two distinct sequential steps: maldevelopment of VHL-deficient mesonephric cells, followed by neoplastic papillary proliferation.


Subject(s)
Epididymis/abnormalities , Genital Diseases, Male/complications , Genital Diseases, Male/pathology , Genital Neoplasms, Male/etiology , Hamartoma/etiology , von Hippel-Lindau Disease/complications , Adult , Autopsy , Cell Proliferation , Cysts/etiology , Cysts/metabolism , Cysts/pathology , Disease Progression , Epididymis/growth & development , Epididymis/metabolism , Epididymis/pathology , Genital Diseases, Male/congenital , Genital Diseases, Male/metabolism , Genital Neoplasms, Male/pathology , Hamartoma/pathology , Humans , Male , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Wolffian Ducts/abnormalities , Wolffian Ducts/metabolism , Wolffian Ducts/pathology , von Hippel-Lindau Disease/metabolism , von Hippel-Lindau Disease/pathology
16.
J Cereb Blood Flow Metab ; 28(3): 482-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17684515

ABSTRACT

An adjuvant therapy to prolong the therapeutic window for stroke patients is urgently needed. This randomized, blinded, placebo-controlled study investigated adjuvant intravenous sodium nitrite with recombinant tissue plasminogen activator (rtPA) in middle cerebral artery occlusion (MCAO) with 6 and 2 h of ischemia followed by reperfusion in Sprague-Dawley rats (n=59). Quantitative diffusion, T(1)-, T(2)-weighted, and semiquantitative perfusion imaging were performed before and after reperfusion and at 48 h after ischemia to determine the spatiotemporal evolution of stroke. After 48 h animals were killed and examined to evaluate infarct size and evidence of hemorrhagic transformation. Factor VIII immunostaining was performed to assess vessel morphology. Nitrite treatment (6 h group: 37.5 micromol for more than 90 mins; 2 h group: 26.25 and 1.75 micromol for more than 60 mins) did not reduce infarct volume 48 h after MCAO compared with saline-treated placebo groups after 6 or 2 h of MCAO. Stroke progression from baseline to 48 h, based on the apparent diffusion coefficient and relative cerebral blood flow deficits before and after reperfusion and T(2)-weighted hyperintensity at 48 h, did not differ between treated and control animals. These results suggest that nitrite is not a protective adjuvant therapy to delayed rtPA administration after ischemic stroke in rats.


Subject(s)
Nitrites/pharmacology , Reperfusion Injury , Stroke/drug therapy , Thrombolytic Therapy/methods , Animals , Disease Models, Animal , Magnetic Resonance Imaging , Male , Nitrites/therapeutic use , Rats , Rats, Sprague-Dawley , Survival Rate , Treatment Outcome
17.
Neoplasia ; 9(9): 777-87, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17898873

ABSTRACT

Glioblastoma multiformes (GBMs) express increased aquaporin (AQP) 1 compared to normal brain. AQPs may contribute to edema, cell motility, and shuttling of H(2)O and H(+) from intracellular to extracellular space. We sought to gain insight into AQP1 function in GBM. In cultured 9L gliosarcoma cells, AQP1 expression was induced by dexamethasone, platelet-derived growth factor, NaCl, hypoxia, D-glucose (but not L-glucose), and fructose. Induction of AQP1 expression correlated with the level of glycolysis, maximized by increasing medium D-glucose or fructose and decreasing O(2), and was quantified by measuring lactate dehydrogenase (LDH) activity and medium lactate concentration. Upregulation of the protease cathepsin B was also observed in 9L cells cultured under glycolytic conditions. Immunohistochemical staining of human GBM specimens revealed increased coincident expression of AQP1, LDH, and cathepsin B in glioma cells associated with blood vessels at the tumor periphery. GBMs are known to exhibit aerobic glycolysis. Increased glucose metabolism at the tumor periphery may provide a scenario by which upregulation of AQP1, LDH, and cathepsin B contributes to acidification of the extracellular milieu and to invasive potential of glioma cells in perivascular space. The specific upregulation and metabolic consequences of increased AQP1 in gliomas may provide a therapeutic target, both as a cell surface marker and as a functional intervention.


Subject(s)
Aquaporin 1/physiology , Brain Neoplasms/pathology , Glioma/pathology , Gliosarcoma/pathology , Neoplasm Proteins/physiology , Aerobiosis , Animals , Aquaporin 1/biosynthesis , Aquaporin 1/genetics , Brain Neoplasms/metabolism , Cathepsin B/biosynthesis , Cathepsin B/genetics , Cell Hypoxia , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Culture Media, Serum-Free/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glioma/metabolism , Gliosarcoma/metabolism , Glycolysis , Hexoses/pharmacology , Humans , Hydrogen-Ion Concentration , Intracellular Fluid/chemistry , L-Lactate Dehydrogenase/biosynthesis , L-Lactate Dehydrogenase/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Rats , Response Elements
18.
J Neurosurg ; 107(1): 190-7, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17639894

ABSTRACT

Recent preclinical studies have demonstrated that convection-enhanced delivery (CED) can be used to perfuse the brain and brainstem with therapeutic agents while simultaneously tracking their distribution using coinfusion of a surrogate magnetic resonance (MR) imaging tracer. The authors describe a technique for the successful clinical application of this drug delivery and monitoring paradigm to the brainstem. Two patients with progressive intrinsic brainstem lesions (one with Type 2 Gaucher disease and one with a diffuse pontine glioma) were treated with CED of putative therapeutic agents mixed with Gd-diethylenetriamene pentaacetic acid (DTPA). Both patients underwent frameless stereotactic placement of MR imaging-compatible outer guide-inner infusion cannulae. Using intraoperative MR imaging, accurate cannula placement was confirmed and real-time imaging during infusion clearly demonstrated progressive filling of the targeted region with the drug and Gd-DTPA infusate. Neither patient had clinical or imaging evidence of short- or long-term infusate-related toxicity. Using this technique, CED can be used to safely perfuse targeted regions of diseased brainstem with therapeutic agents. Coinfused imaging surrogate tracers can be used to monitor and control the distribution of therapeutic agents in vivo. Patients with a variety of intrinsic brainstem and other central nervous system disorders may benefit from a similar treatment paradigm.


Subject(s)
Brain Stem , Cerebellar Neoplasms/pathology , Cerebellar Neoplasms/surgery , Gaucher Disease/pathology , Gaucher Disease/surgery , Glioma , Perfusion/methods , Pons , Surgery, Computer-Assisted/instrumentation , Brain Stem/blood supply , Brain Stem/pathology , Brain Stem/surgery , Catheters, Indwelling , Cerebrovascular Circulation/physiology , Contrast Media , Facial Paralysis/complications , Facial Paralysis/physiopathology , Gadolinium DTPA , Gaucher Disease/complications , Glioma/blood supply , Glioma/pathology , Glioma/surgery , Humans , Infant, Newborn , Intraoperative Care , Magnetic Resonance Imaging , Male , Neurosurgical Procedures , Pons/blood supply , Pons/pathology , Pons/surgery
19.
J Neurosurg ; 103(5): 853-68, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16304990

ABSTRACT

Overexpression of vascular endothelial growth factor (VEGF) is associated with several central nervous system (CNS) diseases and abnormalities, and is often postulated as a causative factor and promising therapeutic target in these settings. The authors' goal was to reassess the contribution of VEGF to the biology and pathology of the CNS. The authors review the literature relating to the following aspects of VEGF: 1) the biology of VEGF in normal brain; 2) the involvement of VEGF in CNS disorders other than tumors (traumatic and ischemic injuries arteriovenous malformations, inflammation); and 3) the role of VEGF in brain tumor biology (gliomas and the associated vasogenic edema, and hemangioblastomas). The authors conclude the following: first, that VEGF overexpression contributes to the phenotype associated with many CNS disorders, but VEGF is a reactive rather than a causative factor in many cases; and second, that use of VEGF as a therapeutic agent or target is complicated by the effects of VEGF not only on the cerebral vasculature, but also on astrocytes, neurons, and inflammatory cells. In many cases, therapeutic interventions targeting the VEGF/VEGF receptor axis are likely to be ineffective or even detrimental. Clinical manipulation of VEGF levels in the CNS must be approached with caution.


Subject(s)
Brain Diseases/pathology , Brain Diseases/physiopathology , Vascular Endothelial Growth Factor A/physiology , Animals , Humans
20.
Neuro Oncol ; 7(4): 465-75, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16212811

ABSTRACT

Malignant brain tumors exhibit distinct metabolic characteristics. Despite high levels of lactate, the intracellular pH of brain tumors is more alkaline than normal brain. Additionally, with increasing malignancy, brain tumors display intratumoral hypoxia. Carbonic anhydrase (CA) IX and XII are transmembrane isoenzymes that are induced by tissue hypoxia. They participate in regulation of pH homeostasis by catalyzing the reversible hydration of carbon dioxide. The aim of our study was to investigate whether brain tumors of different histology and grade of malignancy express elevated levels of CA IX and XII as compared to normal brain. We analyzed 120 tissue specimens from brain tumors (primary and metastatic) and normal brain for CA IX and XII expression by immunohistochemistry, Western blot, and in situ hybridization. Whereas normal brain tissue showed minimal levels of CA IX and XII expression, expression in tumors was found to be upregulated with increased level of malignancy. Hemangioblastomas, from patients with von Hippel-Lindau disease, also displayed high levels of CA IX and XII expression. Comparison of CA IX and XII staining with HIF-1alpha staining revealed a similar microanatomical distribution, indicating hypoxia as a major, but not the only, induction factor. The extent of CA IX and XII staining correlated with cell proliferation, as indicated by Ki67 labeling. The results demonstrate that CA IX and XII are upregulated in intrinsic and metastatic brain tumors as compared to normal brain tissue. This may contribute to the management of tumor-specific acid load and provide a therapeutic target.


Subject(s)
Brain Neoplasms/enzymology , Carbonic Anhydrases/biosynthesis , Hemangioblastoma/etiology , Blotting, Western , Hemangioblastoma/enzymology , Humans , Immunohistochemistry , In Situ Hybridization , von Hippel-Lindau Disease/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...