Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
J Neurooncol ; 165(3): 449-458, 2023 Dec.
Article En | MEDLINE | ID: mdl-38015375

PURPOSE: There is a growing body of literature documenting glioma heterogeneity in terms of radiographic, histologic, molecular, and genetic characteristics. Incomplete spatial specification of intraoperative tumor samples may contribute to variability in the results of pathological and biological investigations. We have developed a system, termed geo-tagging, for routine intraoperative linkage of each tumor sample to its location via neuronavigation. METHODS: This is a single-institution, IRB approved, prospective database of undergoing clinically indicated surgery. We evaluated relevant factors affecting data collection by this registry, including tumor and surgical factors (e.g. tumor volume, location, grade and surgeon). RESULTS: Over a 2-year period, 487 patients underwent craniotomy for an intra-axial tumor. Of those, 214 underwent surgery for a newly diagnosed or recurrent glioma. There was significant variation in the average number of samples collected per registered case, with a range of samples from 2.53 to 4.75 per tumor type. Histology and grade impacted on sampling with a range of 2.0 samples per tumor in Grade four, IDH-WT gliomas to 4.5 samples in grade four, IDH-mutant gliomas. The range of cases with sampling per surgeon was 6 to 99 with a mean of 47.6 cases and there was a statistically significant differences between surgeons. Tumor grade did not have a statistically significant impact on number of samples per case. No significant correlation was found between the number of samples collected and enhancing tumor volume, EOR, or volume of tumor resected. CONCLUSION: We are using the results of this analysis to develop a prospective sample collection protocol.


Brain Neoplasms , Glioma , Humans , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Brain Neoplasms/surgery , Neoplasm Recurrence, Local , Glioma/diagnostic imaging , Glioma/genetics , Glioma/surgery , Magnetic Resonance Imaging/methods , Registries
2.
Int J Mol Sci ; 24(20)2023 Oct 14.
Article En | MEDLINE | ID: mdl-37894860

Temozolomide (TMZ) is an important first-line treatment for glioblastoma (GBM), but there are limitations to TMZ response in terms of durability and dependence on the promoter methylation status of the DNA repair gene O6-methylguanine DNA methyltransferase (MGMT). MGMT-promoter-hypermethylated (MGMT-M) GBMs are more sensitive to TMZ than MGMT-promoter-hypomethylated (MGMT-UM) GBMs. Moreover, TMZ resistance is inevitable even in TMZ-sensitive MGMT-M GBMs. Hence, epigenetic reprogramming strategies are desperately needed in order to enhance TMZ response in both MGMT-M and MGMT-UM GBMs. In this study, we present novel evidence that the epigenetic reactivation of Tumor Suppressor Candidate 3 (TUSC3) can reprogram sensitivity of GBM stem cells (GSCs) to TMZ irrespective of MGMT promoter methylation status. Interrogation of TCGA patient GBM datasets confirmed TUSC3 promoter regulation of TUSC3 expression and also revealed a strong positive correlation between TUSC3 expression and GBM patient survival. Using a combination of loss-of-function, gain-of-function and rescue studies, we demonstrate that TUSC3 reactivation is associated with enhanced TMZ response in both MGMT-M and MGMT-UM GSCs. Further, we provide novel evidence that the demethylating agent 5-Azacitidine (5-Aza) reactivates TUSC3 expression in MGMT-M GSCs, whereas the combination of 5-Aza and MGMT inhibitor Lomeguatrib is necessary for TUSC3 reactivation in MGMT-UM GSCs. Lastly, we propose a pharmacological epigenetic reactivation strategy involving TUSC3 that leads to significantly prolonged survival in MGMT-M and MGMT-UM orthotopic GSCs models. Collectively, our findings provide a framework and rationale to further explore TUSC3-mediated epigenetic reprogramming strategies that could enhance TMZ sensitivity and outcomes in GBM. Mechanistic and translational evidence gained from such studies could contribute towards optimal design of impactful trials for MGMT-UM GBMs that currently do not have good treatment options.


Brain Neoplasms , Glioblastoma , Humans , Temozolomide/pharmacology , Temozolomide/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Dacarbazine/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , DNA Methylation , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , O(6)-Methylguanine-DNA Methyltransferase/genetics , Epigenesis, Genetic , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
3.
Neuro Oncol ; 25(12): 2221-2236, 2023 12 08.
Article En | MEDLINE | ID: mdl-37436963

BACKGROUND: Schwannomas are common peripheral nerve sheath tumors that can cause severe morbidity given their stereotypic intracranial and paraspinal locations. Similar to many solid tumors, schwannomas and other nerve sheath tumors are primarily thought to arise due to aberrant hyperactivation of the RAS growth factor signaling pathway. Here, we sought to further define the molecular pathogenesis of schwannomas. METHODS: We performed comprehensive genomic profiling on a cohort of 96 human schwannomas, as well as DNA methylation profiling on a subset. Functional studies including RNA sequencing, chromatin immunoprecipitation-DNA sequencing, electrophoretic mobility shift assay, and luciferase reporter assays were performed in a fetal glial cell model following transduction with wildtype and tumor-derived mutant isoforms of SOX10. RESULTS: We identified that nearly one-third of sporadic schwannomas lack alterations in known nerve sheath tumor genes and instead harbor novel recurrent in-frame insertion/deletion mutations in SOX10, which encodes a transcription factor responsible for controlling Schwann cell differentiation and myelination. SOX10 indel mutations were highly enriched in schwannomas arising from nonvestibular cranial nerves (eg facial, trigeminal, vagus) and were absent from vestibular nerve schwannomas driven by NF2 mutation. Functional studies revealed these SOX10 indel mutations have retained DNA binding capacity but impaired transactivation of glial differentiation and myelination gene programs. CONCLUSIONS: We thus speculate that SOX10 indel mutations drive a unique subtype of schwannomas by impeding proper differentiation of immature Schwann cells.


Nerve Sheath Neoplasms , Neurilemmoma , Neuroma, Acoustic , Humans , INDEL Mutation , Transcriptional Activation , Neurilemmoma/genetics , Neurilemmoma/pathology , Neuroma, Acoustic/pathology , Mutation , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism
4.
Cancer Control ; 28: 10732748211040013, 2021.
Article En | MEDLINE | ID: mdl-34620004

INTRODUCTION: BRAF V600 E mutations have been identified in a subset of patients with primary brain tumors. Combination therapy with BRAF and Mitogen-activated protein kinase (MEK) inhibitors (BRAF/MEKi) targeting sequential steps in the MAPK pathway has replaced BRAFi monotherapy as the standard of care in multiple tumors with BRAF V600 E mutations, and clinical evidence for this strategy continues to grow in primary brain tumors. CASE SERIES: We describe four patients with BRAF V600 E mutated gliomas, including a 21-year-old woman with a ganglioglioma WHO grade I, a 19-year-old man with a pleomorphic xanthoastrocytoma WHO grade III, and 21-year-old and 33-year-old women with epithelioid GBM WHO grade IV, who achieved durable progression-free survival with combination BRAF/MEKi. CONCLUSION: Combination of BRAF/MEK inhibition can be a novel, promising approach as targeted therapy in gliomas with BRAF V600 E mutations, especially those that are resistant to standard therapy. Our cases, along with other early reports utilizing dabrafenib/trametinib, highlight the importance of somatic next-generation sequencing, particularly in younger patients. Interim results from clinical trials utilizing dabrafenib/trametinib have been promising thus far, and our case series suggests that durable clinical benefit is possible, even in the setting of glioblastoma, WHO grade IV.


Brain Neoplasms/drug therapy , Glioma/drug therapy , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Adult , Female , Humans , Male , Neoplasm Grading , Progression-Free Survival , Proto-Oncogene Proteins B-raf/genetics , Young Adult
5.
Clin Cancer Res ; 27(14): 4109-4125, 2021 07 15.
Article En | MEDLINE | ID: mdl-34035069

PURPOSE: Melanoma brain metastases (MBM) and leptomeningeal melanoma metastases (LMM) are two different manifestations of melanoma CNS metastasis. Here, we used single-cell RNA sequencing (scRNA-seq) to define the immune landscape of MBM, LMM, and melanoma skin metastases. EXPERIMENTAL DESIGN: scRNA-seq was undertaken on 43 patient specimens, including 8 skin metastases, 14 MBM, and 19 serial LMM specimens. Detailed cell type curation was performed, the immune landscapes were mapped, and key results were validated by IHC and flow cytometry. Association analyses were undertaken to identify immune cell subsets correlated with overall survival. RESULTS: The LMM microenvironment was characterized by an immune-suppressed T-cell landscape distinct from that of brain and skin metastases. An LMM patient with long-term survival demonstrated an immune repertoire distinct from that of poor survivors and more similar to normal cerebrospinal fluid (CSF). Upon response to PD-1 therapy, this extreme responder showed increased levels of T cells and dendritic cells in their CSF, whereas poor survivors showed little improvement in their T-cell responses. In MBM patients, therapy led to increased immune infiltrate, with similar T-cell transcriptional diversity noted between skin metastases and MBM. A correlation analysis across the entire immune landscape identified the presence of a rare population of dendritic cells (DC3) that was associated with increased overall survival and positively regulated the immune environment through modulation of activated T cells and MHC expression. CONCLUSIONS: Our study provides the first atlas of two distinct sites of melanoma CNS metastases and defines the immune cell landscape that underlies the biology of this devastating disease.


Brain Neoplasms/immunology , Brain Neoplasms/secondary , Melanoma/immunology , Melanoma/pathology , Melanoma/secondary , Meningeal Neoplasms/immunology , Meningeal Neoplasms/secondary , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Tumor Microenvironment/immunology , Humans
6.
Int J Mol Sci ; 23(1)2021 Dec 28.
Article En | MEDLINE | ID: mdl-35008722

Stemness reprogramming remains a largely unaddressed principal cause of lethality in glioblastoma (GBM). It is therefore of utmost importance to identify and target mechanisms that are essential for GBM stemness and self-renewal. Previously, we implicated BIRC3 as an essential mediator of therapeutic resistance and survival adaptation in GBM. In this study, we present novel evidence that BIRC3 has an essential noncanonical role in GBM self-renewal and stemness reprogramming. We demonstrate that BIRC3 drives stemness reprogramming of human GBM cell lines, mouse GBM cell lines and patient-derived GBM stem cells (GSCs) through regulation of BMP4 signaling axis. Specifically, BIRC3 induces stemness reprogramming in GBM through downstream inactivation of BMP4 signaling. RNA-Seq interrogation of the stemness reprogramming hypoxic (pseudopalisading necrosis and perinecrosis) niche in GBM patient tissues further validated the high BIRC3/low BMP4 expression correlation. BIRC3 knockout upregulated BMP4 expression and prevented stemness reprogramming of GBM models. Furthermore, siRNA silencing of BMP4 restored stemness reprogramming of BIRC3 knockout in GBM models. In vivo silencing of BIRC3 suppressed tumor initiation and progression in GBM orthotopic intracranial xenografts. The stemness reprograming of both GSCs and non-GSCs populations highlights the impact of BIRC3 on intra-tumoral cellular heterogeneity GBM. Our study has identified a novel function of BIRC3 that can be targeted to reverse stemness programming of GBM.


Baculoviral IAP Repeat-Containing 3 Protein/metabolism , Cellular Reprogramming , Glioblastoma/metabolism , Glioblastoma/pathology , Neoplastic Stem Cells/metabolism , Animals , Baculoviral IAP Repeat-Containing 3 Protein/genetics , Biomarkers, Tumor/metabolism , Bone Morphogenetic Protein 4/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Self Renewal/genetics , Cellular Reprogramming/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Humans , Mice , Neoplastic Stem Cells/pathology , Signal Transduction , Xenograft Model Antitumor Assays
7.
J Med Case Rep ; 14(1): 189, 2020 Oct 16.
Article En | MEDLINE | ID: mdl-33059729

BACKGROUND: Extramedullary disease in multiple myeloma often portends a worse diagnosis. In approximately 1% of cases, multiple myeloma may metastasize to the central nervous system as either leptomeningeal involvement or an intracranial, intraparenchymal lesion. Spinal cord metastases, however, are exceedingly rare. We present a case of spinal cord multiple myeloma as well as a literature review of reported cases. CASE PRESENTATION: A 66-year-old African American man with multiple myeloma presented with acute midthoracic pain and lower extremity paresis and paresthesia. Magnetic resonance imaging of the spine revealed two contrast-enhancing intramedullary enhancing lesions in the T1-T2 and T6-T7 cord. Resection with biopsy yielded a diagnosis of metastatic multiple myeloma. CONCLUSION: To date, only six cases of extramedullary disease to the spinal cord in patients with multiple myeloma have been reported, including our patient's case. In all cases, neurologic deficit was observed at presentation, and magnetic resonance imaging of the spine revealed an intramedullary, homogeneously enhancing lesion. Current evidence suggests worse prognosis in patients with extramedullary disease to the central nervous system, and treatment paradigms remain debatable.


Multiple Myeloma , Spinal Cord Neoplasms , Aged , Biopsy , Humans , Magnetic Resonance Imaging , Male , Multiple Myeloma/diagnosis , Multiple Myeloma/diagnostic imaging , Spinal Cord , Spinal Cord Neoplasms/diagnostic imaging , Spinal Cord Neoplasms/surgery
8.
World Neurosurg ; 143: 91-96, 2020 11.
Article En | MEDLINE | ID: mdl-32683005

BACKGROUND: Intracranial myxoid mesenchymal tumors (IMMTs) carrying an EWSR1-CREB gene family fusion are extremely rare and have only been identified in 10 previous reports. All but one has been found in children or young adults. Although there appear to be similarities to a myxoid variant of angiomatoid fibrous histiocytoma (AFH), clear histologic differences exist that appear to distinguish IMMTs as a distinct and novel entity. Previous reports have lacked detailed long-term follow-up and recommendations regarding treatment approach. CASE DESCRIPTION: This case describes a 48-year-old woman who presented with a left intraventricular mass that was identified on histology as an IMMT with an EWSR1-ATF1 gene fusion. After initial resection, the tumor demonstrated local recurrence. Repeat resection was performed followed by immediate demonstration of local and distant tumor recurrence. Histologic analysis of the tumor demonstrated a myxoid mesenchymal tumor distinct from AFH. Fractionated stereotactic radiation therapy was administered after the second resection, and tumor control was achieved at 1 year. CONCLUSIONS: Intracranial myxoid mesenchymal tumor is a novel and rare entity that has been previously identified in only 10 cases. This case is particularly remarkable because it is only the second IMMT case to occur in a middle-aged adult and shares striking similarities in clinical presentation to the previously reported case. Given the aggressive recurrence seen with the presented case, we recommend the treatment plan to be surgical resection followed by adjuvant radiation therapy.


Activating Transcription Factor 1/genetics , Cerebral Ventricle Neoplasms/genetics , Histiocytoma, Malignant Fibrous/genetics , RNA-Binding Protein EWS/genetics , Cerebral Ventricle Neoplasms/diagnostic imaging , Cerebral Ventricle Neoplasms/surgery , Female , Gene Fusion , Histiocytoma, Malignant Fibrous/diagnostic imaging , Histiocytoma, Malignant Fibrous/surgery , Humans , Magnetic Resonance Imaging , Middle Aged , Neoplasm Recurrence, Local , Neurosurgical Procedures/methods , Radiosurgery/methods , Radiotherapy, Adjuvant , Reoperation , Treatment Outcome
9.
Clin Cancer Res ; 26(9): 2163-2175, 2020 05 01.
Article En | MEDLINE | ID: mdl-31924735

PURPOSE: The development of leptomeningeal melanoma metastases (LMM) is a rare and devastating complication of the late-stage disease, for which no effective treatments exist. Here, we performed a multi-omics analysis of the cerebrospinal fluid (CSF) from patients with LMM to determine how the leptomeningeal microenvironment shapes the biology and therapeutic responses of melanoma cells. EXPERIMENTAL DESIGN: A total of 45 serial CSF samples were collected from 16 patients, 8 of these with confirmed LMM. Of those with LMM, 7 had poor survival (<4 months) and one was an extraordinary responder (still alive with survival >35 months). CSF samples were analyzed by mass spectrometry and incubated with melanoma cells that were subjected to RNA sequencing (RNA-seq) analysis. Functional assays were performed to validate the pathways identified. RESULTS: Mass spectrometry analyses showed the CSF of most patients with LMM to be enriched for pathways involved in innate immunity, protease-mediated damage, and IGF-related signaling. All of these were anticorrelated in the extraordinary responder. RNA-seq analysis showed CSF to induce PI3K/AKT, integrin, B-cell activation, S-phase entry, TNFR2, TGFß, and oxidative stress responses in the melanoma cells. ELISA assays confirmed that TGFß expression increased in the CSF of patients progressing with LMM. CSF from poorly responding patients conferred tolerance to BRAF inhibitor therapy in apoptosis assays. CONCLUSIONS: These analyses identified proteomic/transcriptional signatures in the CSF of patients who succumbed to LMM. We further showed that the CSF from patients with LMM has the potential to modulate BRAF inhibitor responses and may contribute to drug resistance.See related commentary by Glitza Oliva and Tawbi, p. 2083.


Melanoma , Proteomics , Disease Progression , Drug Resistance, Neoplasm , Gold , Humans , Melanoma/drug therapy , Melanoma/genetics , Phosphatidylinositol 3-Kinases , Tumor Microenvironment
10.
J Neurol ; 267(3): 783-790, 2020 Mar.
Article En | MEDLINE | ID: mdl-31748891

INTRODUCTION: A patient who was initially considered to have a glioblastoma (GBM) had molecular analysis, showing that it was a pleomorphic xanthoastrocytoma (PXA). Up to 78% of PXA tumors have BRAF V600E mutations. Primary brain tumors with BRAF mutations can have a good response to BRAF MEK inhibitors (BRAF MEKi), and there may be a synergistic response when combined with autophagy inhibitors. PRESENTATION OF THE CASE: A 20-year-old man found to have a large brain mass with midline shift underwent resection. He was diagnosed with "GBM" and treated with radiation and temozolomide with subsequent disease recurrence. Review of histology showed malignant PXA with BRAF V600E mutation. Treatment with Dabrafenib and Trametinib was started, and tumor size increased in size after 14 months of treatment. Given studies showing that resistance to BRAF inhibition can be overcome by autophagy inhibition, chloroquine was added. Patient has been on "triple" therapy for 15 months and has radiographically Stable Disease. At MCC, 3% of patients with gliomas have BRAF mutations who could potentially benefit from this combination therapy. CONCLUSION: This is the first report of a PXA patient receiving therapy with BRAF MEKi and an autophagy inhibitor with prolonged stable disease. This patient highlights the importance of a molecular interrogation in gliomas to provide an integrated diagnosis and effective treatment. This may be useful in up to 3% of glioma patients with BRAF mutations. Molecular testing in neuro-oncology is providing new avenues of diagnosis and treatment, and detailed molecular interrogation should be considered routine.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Astrocytoma/drug therapy , Brain Neoplasms/drug therapy , Proto-Oncogene Proteins B-raf/genetics , Astrocytoma/genetics , Brain Neoplasms/genetics , Chloroquine/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Imidazoles/therapeutic use , Male , Oximes/therapeutic use , Precision Medicine/methods , Pyridones/therapeutic use , Pyrimidinones/therapeutic use , Young Adult
11.
Sci Rep ; 9(1): 14072, 2019 Oct 01.
Article En | MEDLINE | ID: mdl-31575897

Epigenetic regulation of O6-alkylguanine DNA alkyltransferase (MGMT) is surrogate of intrinsic resistance to temozolomide (TMZ). However, mechanisms associated with adaptive resistance evolution of glioblastoma (GBM) relative to MGMT methylation remain unclear. We hereby report a paradoxical yet translational epigenetic regulation of plasticity towards adaptive resistance in GBM. Based on an adaptive resistance model of GBM cells with differential MGMT methylation profiles, MGMT-hypermethylation enhanced genetic and phenotypic plasticity towards adaptive resistance to TMZ while MGMT hypomethylation limited plasticity. The resulting model-associated adaptive resistance gene signature negatively correlated with GBM patient survival. XAF1, a tumor suppressor protein, paradoxically emerged as a mediator of differential plasticities towards adaptive resistance to TMZ through epigenetic regulation. XAF1 promoted resistance both in-vitro and in-vivo. Furthermore, XAF1 expression negatively correlated with XAF1 promoter methylation status, and negatively correlate with GBM patient survival. Collectively, XAF1 appears to have a pradoxical yet translational role in GBM.


Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents, Alkylating/therapeutic use , Apoptosis Regulatory Proteins/metabolism , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Epigenesis, Genetic , Glioblastoma/drug therapy , Temozolomide/therapeutic use , Tumor Suppressor Proteins/metabolism , Adaptation, Physiological , Cell Line, Tumor , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic/genetics , Evolution, Molecular , Glioblastoma/metabolism , Humans , Methylation , Tumor Suppressor Proteins/genetics
13.
Sci Rep ; 7(1): 9350, 2017 08 24.
Article En | MEDLINE | ID: mdl-28839258

Tumor hypoxia is an established facilitator of survival adaptation and mesenchymal transformation in glioblastoma (GBM). The underlying mechanisms that direct hypoxia-mediated survival in GBM habitats are unclear. We previously identified BIRC3 as a mediator of therapeutic resistance in GBM to standard temozolomide (TMZ) chemotherapy and radiotherapy (RT). Here we report that BIRC3 is a biomarker of the hypoxia-mediated adaptive mesenchymal phenotype of GBM. Specifically, in the TCGA dataset elevated BIRC3 gene expression was identified as a superior and selective biomarker of mesenchymal GBM versus neural, proneural and classical subtypes. Further, BIRC3 protein was highly expressed in the tumor cell niches compared to the perivascular niche across multiple regions in GBM patient tissue microarrays. Tumor hypoxia was found to mechanistically induce BIRC3 expression through HIF1-alpha signaling in GBM cells. Moreover, in human GBM xenografts robust BIRC3 expression was noted within hypoxic regions of the tumor. Importantly, selective inhibition of BIRC3 reversed therapeutic resistance of GBM cells to RT in hypoxic microenvironments through enhanced activation of caspases. Collectively, we have uncovered a novel role for BIRC3 as a targetable biomarker and mediator of hypoxia-driven habitats in GBM.


Baculoviral IAP Repeat-Containing 3 Protein/genetics , Glioblastoma/genetics , Hypoxia/genetics , Animals , Baculoviral IAP Repeat-Containing 3 Protein/metabolism , Biomarkers , Caspases/metabolism , Cell Line, Tumor , Disease Models, Animal , Endothelial Cells/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Silencing , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Prognosis , Radiation Tolerance/genetics , Tumor Microenvironment/genetics
14.
Cancer Control ; 22(2): 200-5, 2015 Apr.
Article En | MEDLINE | ID: mdl-26068765

BACKGROUND: Progress in molecular oncology during the last decade has enabled investigators to more precisely define and group gliomas. The impacts of isocitrate dehydrogenase (IDH) mutation (mut) status and other molecular markers on the classification, prognostication, and management of diffuse gliomas are likely to be far-reaching. METHODS: Clinical experience and the medical literature were used to assess the current status of glioma categorization and the likely impact of the pending revision of the classification scheme of the World Health Organization (WHO). RESULTS: IDH-mut is a defining event in most adult fibrillary astrocytomas (FAs) and nearly all oligodendrogliomas (ODs). The IDH-mut status of most gliomas can be established by immunohistochemistry for the most common mutant of IDH1 (R132H). IDH wild-type (wt) diffuse gliomas include several familiar entities -- in particular, glioblastoma (GBM) and most pediatric gliomas -- as well as an assortment of less well-defined entities. The codeletion of 1p/19q distinguishes OD from FA, which, by contrast, shows frequent loss of the α thalassemia/mental retardation syndrome X-linked protein. Mixed oligoastrocytomas are typically classifiable as either OD or FA using molecular testing. CONCLUSIONS: The current practice of designating IDH-mut WHO grade 4 astrocytoma as secondary GBM will likely be discouraged, and primary or de novo GBM, which is always IDH-wt, may lose this qualification. Histologically, low- or intermediate-grade IDH-wt gliomas with molecular changes characteristic of GBM might justify the designation of GBM WHO grade 3. Mixed oligoastrocytoma is losing popularity as a diagnostic term because most cases will fall into either the FA or OD category. Distinguishing IDH-mut from IDH-wt tumors in clinical trials is likely to clarify sensitivity rates or tumor resistance among subgroups, thus suggesting opportunities for targeted therapy.


Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioma/genetics , Glioma/pathology , Isocitrate Dehydrogenase/genetics , Astrocytoma/genetics , Biomarkers, Tumor , DNA Helicases/genetics , Genes, p53 , Glutarates/metabolism , Humans , Immunohistochemistry , Mutation , Nuclear Proteins/genetics , Oligodendroglioma/genetics , Prognosis , X-linked Nuclear Protein
16.
Chem Biol Interact ; 187(1-3): 425-31, 2010 Sep 06.
Article En | MEDLINE | ID: mdl-20122907

Butyrylcholinesterase (BuChE) is an enzyme capable of hydrolysing a wide variety of esters including acetylcholine, a molecule involved in neurotransmission and modulation of immune cell activity. In the brain, BuChE is expressed in white matter and certain populations of neurons and glia. Multiple sclerosis (MS) is an autoimmune disease affecting white matter characterized by neuroinflammation and neurodegeneration in the central nervous system. Here we demonstrate alterations in BuChE activity in MS white matter lesions, including diminished enzyme activity associated with myelin and an increased activity in cells with microglial morphology. Increased BuChE activity within MS lesions could contribute to the pro-inflammatory immune responses through hydrolysis of acetylcholine and to demyelination through hydrolytic deacylation of myelin proteins such as proteolipid protein. This suggests that BuChE could be a potential target for novel disease-modifying strategies for MS.


Brain/enzymology , Brain/pathology , Butyrylcholinesterase/metabolism , Multiple Sclerosis/enzymology , Multiple Sclerosis/pathology , Acetylcholinesterase/metabolism , Adult , Aged , Aged, 80 and over , Brain/metabolism , Case-Control Studies , Female , Humans , Immunohistochemistry , Inflammation/enzymology , Inflammation/metabolism , Inflammation/pathology , Male , Microglia/enzymology , Microglia/pathology , Middle Aged , Multiple Sclerosis/metabolism , Myelin Sheath/enzymology , Myelin Sheath/pathology
17.
Acta Neuropathol ; 116(3): 331-6, 2008 Sep.
Article En | MEDLINE | ID: mdl-18347805

Autosomal dominant sensory ataxia (ADSA), a rare hereditary ataxia, is characterized by progressive dysfunction of central sensory pathways. Its pathological features have not been previously documented. We report a case of a 61-year-old man with ADSA who died of congestive heart failure. Autopsy specimens of brain, thoracolumbar spinal cord, peripheral nerve and skeletal muscle were examined. There was no abnormality on gross examination. Microscopically, there were occasional swollen axons within the cerebral cortex and deep nuclei, particularly the subthalamic nucleus, with no neuronal loss, gliosis or microglial activation. There were many axonal spheroids within the medulla, particularly in the dorsal column nuclei. Axonal spheroids were also seen in the dorsal columns and ventral horns in the thoracolumbar spinal cord, but there was no Wallerian degeneration or demyelination. Amyloid precursor protein (APP) immunostaining of some of the spheroids suggested continuing dysfunction of axoplasmic flow in some regions. There was mild inflammation of peripheral nerve roots but no spheroid, and patchy chronic inflammation of skeletal muscle. In summary, the major pathological process in ADSA is a neuroaxonal dystrophy most prominent in the dorsal columns and dorsal column nuclei, consistent with the clinical pattern of central sensory pathway degeneration.


Ataxia/pathology , Neuroaxonal Dystrophies/pathology , Spinocerebellar Degenerations/pathology , Ataxia/genetics , Axons/pathology , Cerebral Cortex/pathology , Humans , Male , Middle Aged , Spheroids, Cellular/pathology
18.
Can J Neurol Sci ; 31(4): 558-64, 2004 Nov.
Article En | MEDLINE | ID: mdl-15595267

OBJECTIVES: Central neurocytoma is a tumour that typically occurs in young adults in close association with the lateral and third ventricles of the cerebrum. METHODS: We report the unusual case of a central neurocytoma that developed in the fourth ventricle of a 59-year-old woman and metastasized to the upper cervical canal. Subtotal excision and adjuvant radiotherapy were used to treat the lesion. Microscopic evaluation, discussion of the pathologic differential diagnosis and theories of the histogenesis of the tumour are presented. RESULTS AND CONCLUSIONS: Fourth ventricular neurocytoma is rare and has only been reported twice previously. It appears most likely that this tumour arises from subependymal progenitor cell lines.


Cerebral Ventricle Neoplasms/diagnosis , Epidural Neoplasms/diagnosis , Fourth Ventricle/pathology , Neoplasms, Second Primary/diagnosis , Neurocytoma/diagnosis , Cerebral Ventricle Neoplasms/radiotherapy , Cerebral Ventricle Neoplasms/surgery , Cervical Vertebrae , Epidural Neoplasms/radiotherapy , Epidural Neoplasms/surgery , Female , Fourth Ventricle/surgery , Humans , Magnetic Resonance Imaging , Middle Aged , Neoplasm Seeding , Neoplasms, Second Primary/radiotherapy , Neoplasms, Second Primary/surgery , Neurocytoma/radiotherapy , Neurocytoma/surgery , Treatment Outcome
19.
Can J Neurol Sci ; 30(4): 349-57, 2003 Nov.
Article En | MEDLINE | ID: mdl-14672267

BACKGROUND: 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase is a key rate-limiting enzyme in the mevalonate pathway, which generates precursors both for cholesterol biosynthesis and for the production of nonsteroidal mevalonate derivatives that are involved in a number of growth-regulatory processes. We have reported that lovastatin, a competitive inhibitor of HMG-CoA reductase, not only inhibits medulloblastoma proliferation in vitro, but also induces near-complete cell death via apoptosis. The mechanism of this phenomenon is unclear. Possible involvement of changes in expression of certain cell-cycle related genes led us to study some of them in more detail. METHODS: Medulloblastoma cell lines were exposed in vitro to lovastatin, and the effects of gene expression changes were studied using RT-PCR, antisense oligonucleotide, DNA electrophoresis and Western blotting analysis. RESULTS: 1) Levels of total Ras gene mRNA and individual Ras gene mRNA are stable in lovastatin treatment in all examined medulloblastoma cell lines. 2) Blocking c-myc gene over-expression does not enhance medulloblastoma cell sensitivity to lovastatin. 3) Following lovastatin treatment, p16 expression exhibits no change, but pronounced increases of p27KIP1 protein are observed in all examined cell lines. Lovastatin induces pronounced increases of p21WAF1 protein only in Daoy and UW228, but not in D283 Med and D341 Med. 4) Following lovastatin treatment, increased p53 protein is detected only in D341 Med, and bax protein is unchanged in all cell lines. CONCLUSIONS: Lovastatin-induced growth inhibition and apoptosis in medulloblastoma are not dependent on the regulation of Ras and c-myc gene expression, but may be mediated by p27KIP1 gene expression. Lovastatin-induced apoptosis in medulloblastoma is probably p53 independent, but p53 and p21WAF1 gene expression may also mediate anti-proliferative effects of lovastatin on specific medulloblastoma cell lines.


Apoptosis/drug effects , Cell Cycle/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Lovastatin/pharmacology , Medulloblastoma/metabolism , Apoptosis/physiology , Cell Cycle/physiology , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Gene Expression Regulation, Neoplastic/physiology , Genes, ras/drug effects , Genes, ras/physiology , Growth Inhibitors/pharmacology , Growth Inhibitors/therapeutic use , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Lovastatin/therapeutic use , Medulloblastoma/drug therapy
20.
J Neuropathol Exp Neurol ; 61(11): 947-55, 2002 Nov.
Article En | MEDLINE | ID: mdl-12430711

Oligodendroglioma represents a distinct type of diffuse glioma with a relatively favorable prognosis. Although an O2A-like glial progenitor cell of origin has been suggested, a neuronal-oligodendroglial progenitor cell is also of interest, particularly because variable degrees of neuronal marker expression have been reported in typical oligodendrogliomas. We present 2 female and 2 male patients (ages 34-54) with frontal lobe oligodendrogliomas containing a) morphologically distinct collections of small round cells with hyperchromatic nuclei, b) well-formed Homer Wright-like and perivascular rosettes, and c) demonstrable neuronal differentiation by immunohistochemistry and/or electron microscopy in the rosette-associated regions. Unlike extraventricular neurocytomas, these cases featured an infiltrative growth pattern and a classic oligodendroglioma immunophenotype in non-rosette bearing portions of each tumor. FISH analysis demonstrated chromosome 1p and 19q codeletions in 3 (75%) cases, both in regions with and without rosettes. Recurrences were common, although all patients are currently alive 4 months to 13 yr from initial diagnosis. Based on clinicopathologic and genetic features, we diagnosed these tumors as oligodendrogliomas with neurocytic differentiation. However, it is unclear whether they represent a) gliomas with divergent neuronal differentiation, b) a distinctive form of glioneuronal neoplasm, or c) a reflection of glioneuronal histogenesis in oligodendrogliomas in general. In any case, their occurrence suggests a histogenetic overlap between oligodendroglioma and extraventricular neurocytoma not previously recognized.


Brain Neoplasms/pathology , Cell Differentiation/genetics , Cell Transformation, Neoplastic/pathology , Neurons/pathology , Oligodendroglia/pathology , Oligodendroglioma/pathology , Stem Cells/pathology , Adult , Brain Neoplasms/metabolism , Brain Neoplasms/physiopathology , Cell Lineage/genetics , Cell Transformation, Neoplastic/metabolism , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 19/genetics , Female , Humans , Immunohistochemistry , Male , Microscopy, Electron , Middle Aged , Neurocytoma/metabolism , Neurocytoma/pathology , Neurons/metabolism , Neurons/ultrastructure , Oligodendroglia/metabolism , Oligodendroglia/ultrastructure , Oligodendroglioma/metabolism , Oligodendroglioma/physiopathology , Stem Cells/metabolism , Stem Cells/ultrastructure
...