Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Stem Cells ; 34(6): 1702-7, 2016 06.
Article in English | MEDLINE | ID: mdl-26991945

ABSTRACT

Brain tumor-initiating cells (BTICs) are self-renewing multipotent cells critical for tumor maintenance and growth. Using single-cell microfluidic profiling, we identified multiple subpopulations of BTICs coexisting in human glioblastoma, characterized by distinct surface marker expression and single-cell molecular profiles relating to divergent bulk tissue molecular subtypes. These data suggest BTIC subpopulation heterogeneity as an underlying source of intra-tumoral bulk tissue molecular heterogeneity, and will support future studies into BTIC subpopulation-specific therapies. Stem Cells 2016;34:1702-1707.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Biomarkers, Tumor/metabolism , Brain Neoplasms/genetics , Cell Line, Tumor , Glioblastoma/genetics , Humans , Phenotype , Single-Cell Analysis , Transcription, Genetic
2.
BMC Cancer ; 15: 225, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25884624

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most malignant primary brain tumor in adults, with a median survival time of one and a half years. Traditional treatments, including radiation, chemotherapy, and surgery, are not curative, making it imperative to find more effective treatments for this lethal disease. γ-Glutamyl transferase (GGT) is a family of enzymes that was shown to control crucial redox-sensitive functions and to regulate the balance between proliferation and apoptosis. GGT7 is a novel GGT family member that is highly expressed in brain and was previously shown to have decreased expression in gliomas. Since other members of the GGT family were found to be altered in a variety of cancers, we hypothesized that GGT7 could regulate GBM growth and formation. METHODS: To determine if GGT7 is involved in GBM tumorigenesis, we modulated GGT7 expression in two GBM cell lines (U87-MG and U138) and monitored changes in tumorigenicity in vitro and in vivo. RESULTS: We demonstrated for the first time that GBM patients with low GGT7 expression had a worse prognosis and that 87% (7/8) of primary GBM tissue samples showed a 2-fold decrease in GGT7 expression compared to normal brain samples. Exogenous expression of GGT7 resulted in a 2- to 3-fold reduction in proliferation and anchorage-independent growth under minimal growth conditions (1% serum). Decreasing GGT7 expression using either short interfering RNA or short hairpin RNA consistently increased proliferation 1.5- to 2-fold. In addition, intracranial injections of U87-MG cells with reduced GGT7 expression increased tumor growth in mice approximately 2-fold, and decreased mouse survival. To elucidate the mechanism by which GGT7 regulates GBM growth, we analyzed reactive oxygen species (ROS) levels in GBM cells with modulated GGT7 expression. We found that enhanced GGT7 expression reduced ROS levels by 11-33%. CONCLUSION: Our study demonstrates that GGT7 is a novel player in GBM growth and that GGT7 can play a critical role in tumorigenesis by regulating anti-oxidative damage. Loss of GGT7 may increase the cellular ROS levels, inducing GBM occurrence and growth. Our findings suggest that GGT7 can be a promising biomarker and a potential therapeutic target for GBM.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , gamma-Glutamyltransferase/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/mortality , Glioblastoma/pathology , Heterografts , Humans , Prognosis , Reactive Oxygen Species/metabolism , gamma-Glutamyltransferase/genetics
3.
J Neurochem ; 116(2): 281-90, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21070240

ABSTRACT

D-serine is a co-agonist of NMDA receptor (NMDAR) and plays important roles in synaptic plasticity mechanisms. Serine racemase (SR) is a brain-enriched enzyme that converts L-serine to D-serine. SR interacts with the protein interacting with C-kinase 1 (PICK1), which is known to direct protein kinase C (PKC) to its targets in cells. Here, we investigated whether PKC activity regulates SR activity and D-serine availability in the brain. In vitro, PKC phosphorylated SR and decreased its activity. PKC activation increased SR phosphorylation in serine residues and reduced D-serine levels in astrocyte and neuronal cultures. Conversely, PKC inhibition decreased basal SR phosphorylation and increased cellular D-serine levels. In vivo modulation of PKC activity regulated both SR phosphorylation and D-serine levels in rat frontal cortex. Finally, rats that completed an object recognition task showed decreased SR phosphorylation and increased D-serine/total serine ratios, which was markedly correlated with decreased PKC activity in both cortex and hippocampus. Results indicate that PKC phosphorylates SR in serine residues and regulates D-serine availability in the brain. This interaction may be relevant for the regulation of physiological and pathological mechanisms linked to NMDAR function.


Subject(s)
Brain/metabolism , Protein Kinase C/physiology , Serine/metabolism , Animals , Animals, Newborn , Brain/physiology , Cells, Cultured , Male , Neurons/enzymology , Neurons/metabolism , Neurons/physiology , Phosphorylation/physiology , Protein Kinase C/metabolism , Racemases and Epimerases/metabolism , Racemases and Epimerases/physiology , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/physiology , Recognition, Psychology/physiology , Serine/chemistry
4.
Nat Commun ; 9(1): 4121, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30297829

ABSTRACT

Medulloblastoma is the most common malignant brain tumor of childhood. Group 3 medulloblastoma, the most aggressive molecular subtype, frequently disseminates through the leptomeningeal cerebral spinal fluid (CSF) spaces in the brain and spinal cord. The mechanism of dissemination through the CSF remains poorly understood, and the molecular pathways involved in medulloblastoma metastasis and self-renewal are largely unknown. Here we show that NOTCH1 signaling pathway regulates both the initiation of metastasis and the self-renewal of medulloblastoma. We identify a mechanism in which NOTCH1 activates BMI1 through the activation of TWIST1. NOTCH1 expression and activity are directly related to medulloblastoma metastasis and decreased survival rate of tumor-bearing mice. Finally, medulloblastoma-bearing mice intrathecally treated with anti-NRR1, a NOTCH1 blocking antibody, present lower frequency of spinal metastasis and higher survival rate. These findings identify NOTCH1 as a pivotal driver of Group 3 medulloblastoma metastasis and self-renewal, supporting the development of therapies targeting this pathway.


Subject(s)
Cell Proliferation/genetics , Cerebellar Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Medulloblastoma/genetics , Receptor, Notch1/genetics , Animals , Antibodies, Blocking/immunology , Antibodies, Blocking/pharmacology , Cell Line, Tumor , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/metabolism , Humans , Medulloblastoma/drug therapy , Medulloblastoma/metabolism , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasm Metastasis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Receptor, Notch1/immunology , Receptor, Notch1/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Xenograft Model Antitumor Assays/methods
5.
Nat Commun ; 9(1): 4651, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30389946

ABSTRACT

The original version of this Article omitted Suzana A. Kahn, Siddhartha S. Mitra & Samuel H. Cheshier as jointly supervising authors. This has now been corrected in both the PDF and HTML versions of the Article.

6.
Sci Transl Med ; 9(381)2017 03 15.
Article in English | MEDLINE | ID: mdl-28298418

ABSTRACT

Morbidity and mortality associated with pediatric malignant primary brain tumors remain high in the absence of effective therapies. Macrophage-mediated phagocytosis of tumor cells via blockade of the anti-phagocytic CD47-SIRPα interaction using anti-CD47 antibodies has shown promise in preclinical xenografts of various human malignancies. We demonstrate the effect of a humanized anti-CD47 antibody, Hu5F9-G4, on five aggressive and etiologically distinct pediatric brain tumors: group 3 medulloblastoma (primary and metastatic), atypical teratoid rhabdoid tumor, primitive neuroectodermal tumor, pediatric glioblastoma, and diffuse intrinsic pontine glioma. Hu5F9-G4 demonstrated therapeutic efficacy in vitro and in vivo in patient-derived orthotopic xenograft models. Intraventricular administration of Hu5F9-G4 further enhanced its activity against disseminated medulloblastoma leptomeningeal disease. Notably, Hu5F9-G4 showed minimal activity against normal human neural cells in vitro and in vivo, a phenomenon reiterated in an immunocompetent allograft glioma model. Thus, Hu5F9-G4 is a potentially safe and effective therapeutic agent for managing multiple pediatric central nervous system malignancies.


Subject(s)
Antibodies/therapeutic use , Antigens, Differentiation/metabolism , Brain Neoplasms/drug therapy , CD47 Antigen/immunology , Phagocytosis , Receptors, Immunologic/metabolism , Animals , Antibodies/pharmacology , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Child , Disease Models, Animal , Humans , Immunocompetence , Injections, Intraventricular , Medulloblastoma/drug therapy , Medulloblastoma/pathology , Meningeal Neoplasms/pathology , Meningeal Neoplasms/secondary , Mice, Inbred C57BL , Models, Biological , Neoplasm Metastasis , Phagocytosis/drug effects , Survival Analysis , Xenograft Model Antitumor Assays
7.
PLoS One ; 11(4): e0153550, 2016.
Article in English | MEDLINE | ID: mdl-27092773

ABSTRACT

Tumor-associated macrophages (TAMs) represent an important cellular subset within the glioblastoma (WHO grade IV) microenvironment and are a potential therapeutic target. TAMs display a continuum of different polarization states between antitumorigenic M1 and protumorigenic M2 phenotypes, with a lower M1/M2 ratio correlating with worse prognosis. Here, we investigated the effect of macrophage polarization on anti-CD47 antibody-mediated phagocytosis of human glioblastoma cells in vitro, as well as the effect of anti-CD47 on the distribution of M1 versus M2 macrophages within human glioblastoma cells grown in mouse xenografts. Bone marrow-derived mouse macrophages and peripheral blood-derived human macrophages were polarized in vitro toward M1 or M2 phenotypes and verified by flow cytometry. Primary human glioblastoma cell lines were offered as targets to mouse and human M1 or M2 polarized macrophages in vitro. The addition of an anti-CD47 monoclonal antibody led to enhanced tumor-cell phagocytosis by mouse and human M1 and M2 macrophages. In both cases, the anti-CD47-induced phagocytosis by M1 was more prominent than that for M2. Dissected tumors from human glioblastoma xenografted within NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice and treated with anti-CD47 showed a significant increase of M1 macrophages within the tumor. These data show that anti-CD47 treatment leads to enhanced tumor cell phagocytosis by both M1 and M2 macrophage subtypes with a higher phagocytosis rate by M1 macrophages. Furthermore, these data demonstrate that anti-CD47 treatment alone can shift the phenotype of macrophages toward the M1 subtype in vivo.


Subject(s)
Antibodies/pharmacology , CD47 Antigen/metabolism , Glioblastoma/drug therapy , Glioblastoma/pathology , Macrophages/drug effects , Macrophages/pathology , Phagocytosis/drug effects , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Glioblastoma/metabolism , Humans , Macrophages/metabolism , Mice , Mice, Inbred NOD , Phenotype
8.
PLoS One ; 10(8): e0133689, 2015.
Article in English | MEDLINE | ID: mdl-26241738

ABSTRACT

Connective-tissue growth factor (CTGF) is a modular secreted protein implicated in multiple cellular events such as chondrogenesis, skeletogenesis, angiogenesis and wound healing. CTGF contains four different structural modules. This modular organization is characteristic of members of the CCN family. The acronym was derived from the first three members discovered, cysteine-rich 61 (CYR61), CTGF and nephroblastoma overexpressed (NOV). CTGF is implicated as a mediator of important cell processes such as adhesion, migration, proliferation and differentiation. Extensive data have shown that CTGF interacts particularly with the TGFß, WNT and MAPK signaling pathways. The capacity of CTGF to interact with different growth factors lends it an important role during early and late development, especially in the anterior region of the embryo. ctgf knockout mice have several cranio-facial defects, and the skeletal system is also greatly affected due to an impairment of the vascular-system development during chondrogenesis. This study, for the first time, indicated that CTGF is a potent inductor of gliogenesis during development. Our results showed that in vitro addition of recombinant CTGF protein to an embryonic mouse neural precursor cell culture increased the number of GFAP- and GFAP/Nestin-positive cells. Surprisingly, CTGF also increased the number of Sox2-positive cells. Moreover, this induction seemed not to involve cell proliferation. In addition, exogenous CTGF activated p44/42 but not p38 or JNK MAPK signaling, and increased the expression and deposition of the fibronectin extracellular matrix protein. Finally, CTGF was also able to induce GFAP as well as Nestin expression in a human malignant glioma stem cell line, suggesting a possible role in the differentiation process of gliomas. These results implicate ctgf as a key gene for astrogenesis during development, and suggest that its mechanism may involve activation of p44/42 MAPK signaling. Additionally, CTGF-induced differentiation of glioblastoma stem cells into a less-tumorigenic state could increase the chances of successful intervention, since differentiated cells are more vulnerable to cancer treatments.


Subject(s)
Astrocytes/drug effects , Connective Tissue Growth Factor/pharmacology , Fibronectins/biosynthesis , Animals , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Fibronectins/genetics , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Glioblastoma/pathology , Humans , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nestin/analysis , Nestin/biosynthesis , Nestin/genetics , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Recombinant Proteins/pharmacology , SOXB1 Transcription Factors/analysis , Xenopus Proteins/pharmacology
9.
Neurosurgery ; 77(5): 794-802; discussion 802, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26225855

ABSTRACT

BACKGROUND: The presence, characteristics, and potential clinical relevance of neural progenitor populations within the neural placodes of myelomeningocele patients remain to be studied. Neural stem cells are known to reside adjacent to ependyma-lined surfaces along the central nervous system axis. OBJECTIVE: Given such neuroanatomic correlation and regenerative capacity in fetal development, we assessed myelomeningocele-derived neural placode tissue as a potentially novel source of neural stem and progenitor cells. METHODS: Nonfunctional neural placode tissue was harvested from infants during the surgical repair of myelomeningocele and subsequently further analyzed by in vitro studies, flow cytometry, and immunofluorescence. To assess lineage potential, neural placode-derived neurospheres were subjected to differential media conditions. Through assessment of platelet-derived growth factor receptor α (PDGFRα) and CD15 cell marker expression, Sox2+Olig2+ putative oligodendrocyte progenitor cells were successfully isolated. RESULTS: PDGFRαCD15 cell populations demonstrated the highest rate of self-renewal capacity and multipotency of cell progeny. Immunofluorescence of neural placode-derived neurospheres demonstrated preferential expression of the oligodendrocyte progenitor marker, CNPase, whereas differentiation to neurons and astrocytes was also noted, albeit to a limited degree. CONCLUSION: Neural placode tissue contains multipotent progenitors that are preferentially biased toward oligodendrocyte progenitor cell differentiation and presents a novel source of such cells for use in the treatment of a variety of pediatric and adult neurological disease, including spinal cord injury, multiple sclerosis, and metabolic leukoencephalopathies.


Subject(s)
Meningomyelocele/pathology , Neural Stem Cells/cytology , Neurons/cytology , Neurons/pathology , Oligodendroglia/cytology , Cell Differentiation/physiology , Cells, Cultured , Female , Humans , Infant, Newborn , Male , Neural Stem Cells/physiology , Neurons/physiology , Oligodendroglia/physiology
10.
Cureus ; 6(9)2014.
Article in English | MEDLINE | ID: mdl-28729960

ABSTRACT

Tumors of the central nervous system are challenging to treat due to the limited effectiveness and associated toxicities of chemotherapy and radiation therapy. For tumors that can be removed surgically, extent of malignant tissue resection has been shown to correlate with disease progression, recurrence, and survival. Thus, improved technologies for real-time brain tumor imaging are critically needed as tools for guided surgical resection. We previously engineered a novel peptide that binds with high affinity and unique specificity to αVß3, αVß5, and α5ß1 integrins, which are present on tumor cells, and the vasculature of many cancers, including brain tumors. In the current study, we conjugated this engineered peptide to a near infrared fluorescent dye (Alexa Fluor 680), and used the resulting molecular probe for non-invasive whole body imaging of patient-derived medulloblastoma xenograft tumors implanted in the cerebellum of mice. The engineered peptide exhibited robust targeting and illumination of intracranial medulloblastoma following both intravenous and intraperitoneal injection routes. In contrast, a variant of the engineered peptide containing a scrambled integrin-binding sequence did not localize to brain tumors, demonstrating that tumor-targeting is driven by specific integrin interactions. Ex vivo imaging was used to confirm the presence of tumor and molecular probe localization to the cerebellar region. These results warrant further clinical development of the engineered peptide as a tool for image-guided resection of central nervous system tumors.

11.
J Cereb Blood Flow Metab ; 29(6): 1109-20, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19337276

ABSTRACT

Intracerebral hemorrhage (ICH) is a major cause of disability in adults worldwide. The pathophysiology of this syndrome is complex, involving both inflammatory and redox components triggered by the extravasation of blood into the cerebral parenchyma. Hemoglobin, heme, and iron released therein seem be important in the brain damage observed in ICH. However, there is a lack of information concerning hemoglobin traffic and metabolism in brain cells. Here, we investigated the fate of hemoglobin and heme in cultured neurons and astrocytes, as well as in the cortex of adult rats. Hemoglobin was made traceable by conjugation to Alexa 488, whereas a fluorescent heme analogue (tin-protoporphyrin IX) was prepared to allow heme tracking. Using fluorescence microscopy we observed that neurons were more efficient in uptake hemoglobin and heme than astrocytes. Exposure of cortical neurons to hemoglobin or heme resulted in an oxidative stress condition. Viability assays showed that neurons were more susceptible to both hemoglobin and heme toxicity than astrocytes. Together, these results show that neurons, rather than astrocytes, preferentially take up hemoglobin-derived products, indicating that these cells are actively involved in the ICH-associated brain damage.


Subject(s)
Extracellular Space/metabolism , Heme/metabolism , Hemoglobins/metabolism , Animals , Astrocytes/metabolism , Cells, Cultured , Metalloporphyrins/metabolism , Neurons/metabolism , Oxidative Stress , Protoporphyrins/metabolism , Rats , Rats, Wistar
12.
Glia ; 55(16): 1690-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17886292

ABSTRACT

Gliomas are tumors derived from glia or their precursors within the central nervous system. Clinically, gliomas are divided into four grades and the glioblastoma multiforme (GBM), also referred as grade IV astrocytoma, is the most aggressive and the most common glioma in humans. The prognosis for patients with GBM remains dismal, with a median survival of 9-12 months. Despite their striking heterogeneity, common alterations in specific cellular signal transduction pathways occur within most GBMs. Previous work from our group identified the co-chaperone stress-inducible protein 1 (STI1) as a cell surface ligand for cellular prion (PrP(C)), which leads to the activation of several signal transduction pathways, some of which modulate cell survival. In the present work, we used thymidine incorporation assays to investigate the effect of STI1 upon proliferation of the human glioblastoma-derived cell line A172. Here we report that STI1 is secreted by and induces proliferation in tumor cells, an effect that is modulated by the Erk and PI3K pathways, and that, in contrast to glioma cells, STI1 does not induce proliferation of normal glia. In addition, our data suggest the involvement of PrP(C) in STI1-induced proliferation of A172 cells. These results provide initial evidence of a new functional role for STI1 on the physiology of human gliomas, and may lead to the identification of new therapeutic targets in these tumors.


Subject(s)
Glioma/metabolism , Glioma/pathology , Heat-Shock Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Astrocytes/cytology , Astrocytes/metabolism , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Heat-Shock Proteins/pharmacology , Humans , PrPC Proteins/metabolism , Recombinant Proteins/pharmacology , Signal Transduction , Thymidine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL