Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Stem Cells ; 37(11): 1441-1454, 2019 11.
Article in English | MEDLINE | ID: mdl-31381815

ABSTRACT

In a previous study, we showed that folate receptor-α (FRα) translocates to the nucleus where it acts as a transcription factor and upregulates Hes1, Oct4, Sox2, and Klf4 genes responsible for pluripotency. Here, we show that acetylation and phosphorylation of FRα favor its nuclear translocation in the presence of folate and can cause a phenotypic switch from differentiated glial cells to dedifferentiated cells. shRNA-FRα mediated knockdown of FRα was used to confirm the role of FRα in dedifferentiation. Ocimum sanctum hydrophilic fraction-1 treatment not only blocks the folate mediated dedifferentiation of glial cells but also promotes redifferentiation of dedifferentiated glial cells, possibly by reducing the nuclear translocation of ~38 kDa FRα and subsequent interaction with chromatin assembly factor-1. Stem Cells 2019;37:1441-1454.


Subject(s)
Folate Receptor 1/metabolism , Neural Crest/drug effects , Neural Crest/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Cell Dedifferentiation/drug effects , Cell Dedifferentiation/genetics , Cell Line , Cell Line, Tumor , Folate Receptor 1/genetics , Folic Acid/analogs & derivatives , Folic Acid/pharmacology , Humans , Kruppel-Like Factor 4 , Nuclear Transfer Techniques , Ocimum sanctum/chemistry , Octamer Transcription Factor-3/metabolism , RNA, Small Interfering/genetics , SOXB1 Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Mol Cancer ; 16(1): 21, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28137267

ABSTRACT

BACKGROUND: Pilocytic astrocytomas (PAs) are the most common pediatric central nervous system neoplasms. In the majority of cases these tumors are benign and receive favorable prognosis following gross total surgical resection. In patients with progressive or symptomatic tumors, aggressive surgical resection is generally not feasible, thus radiation or chemotherapy are accepted initial or adjuvant interventions. Due to serious long-lasting side-effects, radiation is limited in young children; therefore, chemotherapy is widely practiced as an adjuvant treatment for these patients. However, chemotherapy can promote the emergence of multidrug resistant tumor cells that are more malignant than those of the original tumor. CD133, a putative stem cell marker in normal tissue and malignant brain tumors, enhances multidrug resistant gene 1 (MDR1) expression following chemotherapy in adult malignant glioblastomas. This study examines the relationship between CD133 and MDR1 in pediatric PAs exposed to chemotherapy, with the goal of identifying therapeutic targets that manifest as a result of chemotherapy. METHODS: Slides were obtained for 15 recurrent PAs, seven of which had received chemotherapy prior to surgical treatment for the recurrent tumor. These samples, as well as primary tumor tissue slides from the same patients were used to investigate CD133 and MDR1 expression via immunofluorescence. Archived frozen tissue samples from the same patients were used to examine CD133, MDR1 and PI3K-Akt-NF-κB signaling mediators, via western blot. Two drug resistant pediatric PA cell lines Res186 and Res199 were also used to evaluate the role of CD133 on cell response to cytotoxic therapy. RESULTS: CD133 and MDR1 were co-expressed and their expression was elevated in recurrent PAs from patients that had received chemotherapy, compared to patients that had not received chemotherapy. PI3K-Akt-NF-κB signaling mediator expression was also elevated in recurrent, chemotherapy-treated PA. Suppressing CD133 expression with siCD133 decreased levels of PI3K-Akt-NF-κB signaling mediators and MDR1, while increasing cell chemosensitivity, as indicated by quantification of apoptotic cells following chemotherapy. CONCLUSIONS: CD133 contributes to multidrug resistance by regulating MDR1 levels via the PI3K-Akt-NF-κB signal pathway not only in adult glioblastomas, but also in pediatric PAs. Targeting CD133, adjuvant to conventional chemotherapy may improve outcomes for children with recurrent PA.


Subject(s)
AC133 Antigen/metabolism , Astrocytoma/metabolism , Drug Resistance, Neoplasm , Neoplasm Recurrence, Local/metabolism , Up-Regulation , AC133 Antigen/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/metabolism , Adolescent , Astrocytoma/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemotherapy, Adjuvant , Child , Child, Preschool , Female , Gene Expression Regulation, Neoplastic , Humans , Infant , Male , NF-kappa B/genetics , Neoadjuvant Therapy , Neoplasm Recurrence, Local/drug therapy , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction
3.
Childs Nerv Syst ; 30(8): 1343-53, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24817375

ABSTRACT

PURPOSE: Maternal folate intake has reduced the incidence of human neural tube defects by 60-70 %. However, 30-40 % of cases remain nonresponsive to folate intake. The main purpose of this study was to understand the molecular mechanism of folate nonresponsiveness in a mouse model of neural tube defect. METHODS: We used a folate-nonresponsive Fkbp8 knockout mouse model to elucidate the molecular mechanism(s) of folate nonresponsiveness. Neurospheres were grown from neural stem cells isolated from the lumbar neural tube of E9.5 Fkbp8 (-/-) and wild-type embryos. Immunostaining was used to determine the protein levels of oligodendrocyte transcription factor 2 (Olig2), Nkx6.1, class III beta-tubulin (TuJ1), O4, glial fibrillary acidic protein (GFAP), histone H3 Lys27 trimethylation (H3K27me3), ubiquitously transcribed tetratricopeptide repeat (UTX), and Msx2, and quantitative real-time (RT)-PCR was used to determine the message levels of Olig2, Nkx6.1, Msx2, and noggin in neural stem cells differentiated in the presence and absence of folic acid. RESULTS: Fkbp8 (-/-)-derived neural stem cells showed (i) increased noggin expression; (ii) decreased Msx2 expression; (iii) premature differentiation--neurogenesis, oligodendrogenesis (Olig2 expression), and gliogenesis (GFAP expression); and (iv) increased UTX expression and decreased H3K27me3 polycomb modification. Exogenous folic acid did not reverse these markers. CONCLUSIONS: Folate nonresponsiveness could be attributed in part to increased noggin expression in Fkbp8 (-/-) embryos, resulting in decreased Msx2 expression. Folate treatment further increases Olig2 and noggin expression, thereby exacerbating ventralization.


Subject(s)
Carrier Proteins/metabolism , Folic Acid/adverse effects , Gene Expression Regulation, Developmental/genetics , Neural Tube Defects , Tacrolimus Binding Proteins/deficiency , Animals , Carrier Proteins/genetics , Cell Proliferation/drug effects , Cell Proliferation/physiology , Disease Models, Animal , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Tube Defects/chemically induced , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Pregnancy , Tacrolimus Binding Proteins/genetics
4.
Nanomedicine ; 10(2): 381-91, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23916888

ABSTRACT

This study examined a novel drug delivery system for treatment of malignant brain gliomas: DOX complexed with nanodiamonds (ND-Dox), and administered via convection-enhanced delivery (CED). Drug retention and toxicity were examined in glioma cell lines, and distribution, retention and toxicity were examined in normal rat parenchyma. Efficacy was assessed in a bioluminescence rodent tumor model. NDs markedly enhanced DOX uptake and retention in glioma cells. ND-Dox delivered via CED extended DOX retention and localized DOX toxicity in normal rodent parenchyma, and was significantly more efficient at killing tumor cells than uncomplexed DOX. Outcomes from this work suggest that CED of ND-Dox is a promising approach for brain tumor treatment. FROM THE CLINICAL EDITOR: In this paper, nanodiamonds were utilized to enhance delivery of DOX in a preclinical glioma model using a convection-enhanced delivery method, demonstrating remarkably enhanced efficacy.


Subject(s)
Brain Neoplasms/drug therapy , Drug Carriers , Glioma/drug therapy , Nanodiamonds , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Survival , Humans , Male , Nanomedicine , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Tissue Distribution
5.
Childs Nerv Syst ; 29(8): 1245-51, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23624780

ABSTRACT

OBJECTIVE: This study investigated epigenetic modifications in human central nervous system atypical teratoid rhabdoid tumors (AT/RTs), in response to inhibition of insulin-like growth factor receptor 1 (IGF-1R). MATERIALS AND METHODS: Tumor tissue was obtained from two pediatric patients, tissue was dissociated, and primary cultures were established. Cultured cells were treated with picropodophyllin (PPP; 0, 1, and 2 µM for 48 h), a selective IGF-1R inhibitor. Histone acetylation and methylation patterns (H3K9ac, H3K18ac, H3K4me3, H3K27me3) and levels of histone deacetylases (HDACs; HDAC1, HDAC3, and SirT1) and histone acetyl transferases (GCN5 and p300) were examined. H3K9ac and H3K18ac decreased in response to treatment with PPP. HDAC levels showed a biphasic response, increasing with 1 µM PPP, but then decreasing with 2 µM PPP. CONCLUSION: Inhibition of IGF-1R modified epigenetic status in AT/RT. Determining the mechanisms behind these modifications will guide the development of novel therapeutic targets for this malignant embryonal cancer.


Subject(s)
Central Nervous System Neoplasms/pathology , Epigenesis, Genetic/physiology , Epigenomics , Receptor, IGF Type 1/metabolism , Rhabdoid Tumor/pathology , Signal Transduction/drug effects , Teratoma/pathology , Acetylation/drug effects , Dose-Response Relationship, Drug , Epigenesis, Genetic/drug effects , Histone Acetyltransferases/metabolism , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Methylation/drug effects , Podophyllotoxin/analogs & derivatives , Podophyllotoxin/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/genetics , Tumor Cells, Cultured/drug effects
6.
Cancers (Basel) ; 15(13)2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37444539

ABSTRACT

Pediatric high-grade gliomas (pHGGs) are common malignant brain tumors without effective treatment and poor patient survival. Abnormal posttranslational modification at the histone H3 tail plays critical roles in tumor cell malignancy. We have previously shown that the trimethylation of lysine 4 at histone H3 (H3K4me3) plays a significant role in pediatric ependymoma malignancy and is associated with tumor therapeutic sensitivity. Here, we show that H3K4me3 and its methyltransferase WDR82 are elevated in pHGGs. A reduction in H3K4me3 by downregulating WDR82 decreases H3K4me3 promoter occupancy and the expression of genes associated with stem cell features, cell proliferation, the cell cycle, and DNA damage repair. A reduction in WDR82-mediated H3K4me3 increases the response of pediatric glioma cells to chemotherapy. These findings suggest that WDR82-mediated H3K4me3 is an important determinant of pediatric glioma malignancy and therapeutic response. This highlights the need for a more thorough understanding of the potential of WDR82 as an epigenetic target to increase therapeutic efficacy and improve the prognosis for children with malignant gliomas.

7.
Nat Cell Biol ; 25(3): 493-507, 2023 03.
Article in English | MEDLINE | ID: mdl-36849558

ABSTRACT

How abnormal neurodevelopment relates to the tumour aggressiveness of medulloblastoma (MB), the most common type of embryonal tumour, remains elusive. Here we uncover a neurodevelopmental epigenomic programme that is hijacked to induce MB metastatic dissemination. Unsupervised analyses of integrated publicly available datasets with our newly generated data reveal that SMARCD3 (also known as BAF60C) regulates Disabled 1 (DAB1)-mediated Reelin signalling in Purkinje cell migration and MB metastasis by orchestrating cis-regulatory elements at the DAB1 locus. We further identify that a core set of transcription factors, enhancer of zeste homologue 2 (EZH2) and nuclear factor I X (NFIX), coordinates with the cis-regulatory elements at the SMARCD3 locus to form a chromatin hub to control SMARCD3 expression in the developing cerebellum and in metastatic MB. Increased SMARCD3 expression activates Reelin-DAB1-mediated Src kinase signalling, which results in a MB response to Src inhibition. These data deepen our understanding of how neurodevelopmental programming influences disease progression and provide a potential therapeutic option for patients with MB.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Humans , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Medulloblastoma/genetics , Phosphorylation , Epigenomics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Adhesion Molecules, Neuronal/pharmacology , Cerebellar Neoplasms/genetics , Epigenesis, Genetic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism
8.
J Neurooncol ; 106(2): 261-70, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21842443

ABSTRACT

Interstitial chemotherapeutic drug infusion can bypass the blood-brain barrier, and provide high regional drug concentrations without systemic exposure. However, toxicity and efficacy for drugs administered via interstitial continuous (i.c.) infusion have not been characterized. In the current study, vincristine (VIN) was infused into the right frontal lobes of healthy Fisher 344 rats at 30, 45, 60, and 120 µg/ml over a period of 7 days at 1 µl/h, using an Alzet osmotic pump to evaluate toxicity. C6 rat glioblastoma cells transduced with a luciferase gene were inoculated into the right frontal lobe of a second group of rats. VIN was administered to tumor bearing rats via i.c. infusion 7 days later and tumor growth was monitored by bioluminescence intensity (BLI) to assess VIN efficacy, intravenous (i.v.) drug administration was used as a comparison drug delivery method. The results suggested that VIN toxicity is dose-dependent. Efficacy studies showed increased BLI, which correlates with histopathological tumor size, in saline-infused and i.v.-treated tumor-bearing rats. These rats survived an average of 28 ± 0.85 days and 33 ± 1.38 days, respectively. Both groups had large tumors at the time of death. Animals treated with VIN via i.c. infusion survived until day 90, the observation endpoint for this study. This was significantly longer than average survival times in the previous two groups. These results demonstrate that VIN via i.c. infusion is effective in reducing C6 glioblastoma tumors and prolonging rodent survival time compared to i.v. injection and suggest that chemotherapeutic drug administration via i.c. infusion may be a promising strategy for treating malignant brain tumors.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Vincristine/administration & dosage , Animals , Disease Models, Animal , Drug Delivery Systems , Infusions, Intraventricular , Luminescent Measurements , Male , Neoplasms, Experimental/drug therapy , Rats , Rats, Inbred F344
9.
Childs Nerv Syst ; 28(4): 565-74, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22282078

ABSTRACT

PURPOSE: Brain stem gliomas account for 20% of childhood brain tumors. Presently, there is no effective treatment for these tumors, and the prognosis remains poor. One reason for this is that chemotherapeutic drugs cannot cross the blood-brain barrier. In this study, we used a rodent brainstem tumor model, monitored both qualitatively and quantitatively, to examine the effectiveness of vincristine (VCR) administered via convection-enhanced delivery (CED). METHODS: C6 rat glioblastoma cells, transduced with an oncoretroviral plasmid containing a luciferase coding sequence, were inoculated into Fischer 344 rat brainstems. Tumor growth was monitored by bioluminescence intensity (BLI), and tumor volume was calculated from serial histopathologic sections. Therapeutic efficacy of VCR delivered via CED was assessed. Intravenous (I.V.) and intraperitoneal (I.P.) drug administration were used as a comparison for CED efficacy. RESULTS: BLI monitoring revealed progressive tumor growth in inoculated rats. Symptoms caused by tumor burden were evident 16-18 days after inoculation. BLI correlated quantitatively with tumor volume (r(2) = 0.9413), established by histopathological analysis of tumor growth within the pons. VCR administered through CED was more effective than I.V. or I.P. administration in reducing tumor size and increasing survival times. TUNEL assay results suggest that VCR induced glioblastoma cell apoptosis. CONCLUSIONS: VCR administered by CED was effective in reducing tumors and prolonging survival time.


Subject(s)
Brain Stem Neoplasms/drug therapy , Convection , Disease Models, Animal , Drug Delivery Systems/methods , Luminescent Measurements , Vincristine/administration & dosage , Animals , Brain Stem Neoplasms/diagnosis , Luminescent Measurements/methods , Male , Rats , Rats, Inbred F344 , Rodentia , Treatment Outcome , Xenograft Model Antitumor Assays/methods
10.
J Huazhong Univ Sci Technolog Med Sci ; 32(2): 227-232, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22528225

ABSTRACT

The effects of RNAi-mediated gene silencing of LRlG1 on proliferation and invasion of the human glioma cell line U251-MG and the possible mechanisms were explored in this study. The plasmids pGenesil2-LRIG1-shRNA1 and pGenesil2-LRIG1-shRNA2 were transfected into U251-MG glioma cells respectively by using Lipofectamine 2000 and the transfected cells in which the LRIG1 expression was stably suppressed were selected by G418. The cells transfected with negative shRNA served as control. The expression levels of LRIG1 mRNA and protein were measured by qRT-PCR and Western blotting, respectively. The cell cycle was analyzed by flow cytometry. The results showed that LRIG1 mRNA expression was reduced by 70% and 58% and LRIG1 protein expression by 58% and 26% in U251-MG cells transfected with pGenesil2-LRIG1-shRNAl and pGenesil2-LRIG1-shRNA2 relative to the negative shRNA-transfected U251-MG cells. The proliferative capacity of the LRIG1 specific siRNA-transfected cells was stronger than that of control cells. Cell cycle analysis showed that silencing LRIG1 significantly increased the percentage of S phase cells and the proliferation index (P<0.01). Moreover, silencing LRIG1 could promote the invasion of U251-MG cells (P<0.05). These findings suggested that LRIG1-targeting siRNA can exert a dramatically inhibitory effect on RNA transcription and protein expression of LRIG1, and LRIG1 down-regulation could promote the proliferation of U251-MG cells, arrest U251-MG cells in S phase, and enhance the invasion of U251-MG cells.


Subject(s)
Glioma/genetics , Glioma/pathology , Membrane Glycoproteins/metabolism , RNA Interference , Cell Line, Tumor , Cell Proliferation , Gene Silencing , Humans , Membrane Glycoproteins/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology
11.
J Immunother Cancer ; 10(9)2022 09.
Article in English | MEDLINE | ID: mdl-36096529

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most common malignant brain tumor with poor clinical outcomes. Immunotherapy has recently been an attractive and promising treatment of extracranial malignancies, however, most of clinical trials for GBM immunotherapy failed due to predominant accumulation of tumor-associated microglia/macrophages (TAMs). RESULTS: High level of LRIG2/soluble LRIG2 (sLRIG2) expression activates immune-related signaling pathways, which are associated with poor prognosis in GBM patients. LRIG2/sLRIGs promotes CD47 expression and facilitates TAM recruitment. Blockade of CD47-SIRPα interactions and inhibition of sLRIG2 secretion synergistically suppress GBM progression in an orthotropic murine GBM model. CONCLUSIONS: GBM cells with high level LRIG2 escape the phagocytosis by TAM via the CD47-SIRPα axis, highlighting a necessity for an early stage of clinical trial targeting LRIG2 and CD47-SIRPα as a novel treatment for patients with GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Brain Neoplasms/pathology , CD47 Antigen/metabolism , Humans , Immunity, Innate , Macrophages , Membrane Glycoproteins/metabolism , Mice
12.
Cancers (Basel) ; 13(5)2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33807733

ABSTRACT

Diffuse intrinsic pontine gliomas (DIPGs) account for ~15% of pediatric brain tumors, which invariably present with poor survival regardless of treatment mode. Several seminal studies have revealed that 80% of DIPGs harbor H3K27M mutation coded by HIST1H3B, HIST1H3C and H3F3A genes. The H3K27M mutation has broad effects on gene expression and is considered a tumor driver. Determination of the effects of H3K27M on posttranslational histone modifications and gene regulations in DIPG is critical for identifying effective therapeutic targets. Advanced animal models play critical roles in translating these cutting-edge findings into clinical trial development. Here, we review current molecular research progress associated with DIPG. We also summarize DIPG animal models, highlighting novel genomic engineered mouse models (GEMMs) and innovative humanized DIPG mouse models. These models will pave the way towards personalized precision medicine for the treatment of DIPGs.

13.
Neurooncol Adv ; 3(1): vdab085, 2021.
Article in English | MEDLINE | ID: mdl-34345820

ABSTRACT

BACKGROUND: Meningiomas are the most common tumor arising within the cranium of adults. Despite surgical resection and radiotherapy, many meningiomas invade the brain, and many recur, often repeatedly. To date, no chemotherapy has proven effective against such tumors. Thus, there is an urgent need for chemotherapeutic options for treating meningiomas, especially those that enhance radiotherapy. Palbociclib is an inhibitor of cyclin-dependent kinases 4 and 6 that has been shown to enhance radiotherapy in preclinical models of other cancers, is well-tolerated in patients, and is used to treat malignancies elsewhere in the body. We, therefore, sought to determine its therapeutic potential in preclinical models of meningioma. METHODS: Patient-derived meningioma cells were tested in vitro and in vivo with combinations of palbociclib and radiation. Outputs included cell viability, apoptosis, clonogenicity, engrafted mouse survival, and analysis of engrafted tumor tissues after therapy. RESULTS: We found that palbociclib was highly potent against p16-deficient, Rb-intact CH157 and IOMM-Lee meningioma cells in vitro, but was ineffective against p16-intact, Rb-deficient SF8295 meningioma cells. Palbociclib also enhanced the in vitro efficacy of radiotherapy when used against p16-deficient meningioma, as indicated by cell viability and clonogenic assays. In vivo, the combination of palbociclib and radiation extended the survival of mice bearing orthotopic p16 deficient meningioma xenografts, relative to each as a monotherapy. CONCLUSIONS: These data suggest that palbociclib could be repurposed to treat patients with p16-deficient, Rb-intact meningiomas, and that a clinical trial in combination with radiation therapy merits consideration.

14.
Cancers (Basel) ; 12(9)2020 Sep 14.
Article in English | MEDLINE | ID: mdl-32937871

ABSTRACT

Suprasellar germ cell tumors (S-GCTs) are rare, presenting in either solitary or multifocal fashion. In this study, we retrospectively examine 22 solitary S-GCTs and 20 bifocal germ cell tumors (GCTs) over a 30-year period and demonstrate clinical, radiographic, and prognostic differences between the two groups with therapeutic implications. Compared to S-GCTs, bifocal tumors were almost exclusively male, exhibited higher rate of metastasis, and had worse rates of progression free and overall survival trending toward significance. We also introduce a novel magnetic resonance (MR) imaging classification of suprasellar GCT into five types: a IIIrd ventricle floor tumor extending dorsally with or without an identifiable pituitary stalk (Type Ia, Ib), ventrally (Type III), in both directions (Type II), small lesions at the IIIrd ventricle floor extending to the stalk (Type IV), and tumor localized in the stalk (Type V). S-GCTs almost uniformly presented as Type I-III, while most bifocal GCTs were Type IV with a larger pineal mass. These differences are significant as bifocal GCTs representing concurrent primaries or subependymal extension may be treated with whole ventricle radiation, while cerebrospinal fluid (CSF)-borne metastases warrant craniospinal irradiation (CSI). Although further study is necessary, we recommend CSI for bifocal GCTs exhibiting high-risk features such as metastasis or non-germinomatous germ cell tumor histology.

15.
Cancers (Basel) ; 12(2)2020 Feb 24.
Article in English | MEDLINE | ID: mdl-32102285

ABSTRACT

Malignant gliomas are heterogeneous neoplasms. Glioma stem-like cells (GSCs) are undifferentiated and self-renewing cells that develop and maintain these tumors. These cells are the main population that resist current therapies. Genomic and epigenomic analyses has identified various molecular subtypes. Bone morphogenetic protein 4 (BMP4) reduces the number of GSCs through differentiation and induction of apoptosis, thus increasing therapeutic sensitivity. However, the short half-life of BMP4 impedes its clinical application. We previously reviewed BMP4 signaling in central nervous system development and glioma tumorigenesis and its potential as a treatment target in human gliomas. Recent advances in understanding both adult and pediatric malignant gliomas highlight critical roles of BMP4 signaling pathways in the regulation of tumor biology, and indicates its potential as a therapeutic molecule. Furthermore, significant progress has been made on synthesizing BMP4 biocompatible delivery materials, which can bind to and markedly extend BMP4 half-life. Here, we review current research associated with BMP4 in brain tumors, with an emphasis on pediatric malignant gliomas. We also summarize BMP4 delivery strategies, highlighting biocompatible BMP4 binding peptide amphiphile nanostructures as promising novel delivery platforms for treatment of these devastating tumors.

16.
Neoplasia ; 21(6): 505-515, 2019 06.
Article in English | MEDLINE | ID: mdl-31005631

ABSTRACT

BACKGROUND: Ependymomas (EPNs) are the third most common brain tumor in children. These tumors are resistant to available chemotherapeutic treatments, therefore new effective targeted therapeutics must be identified. Increasing evidence shows epigenetic alterations including histone posttranslational modifications (PTMs), are associated with malignancy, chemotherapeutic resistance and prognosis for pediatric EPNs. In this study we examined histone PTMs in EPNs and identified potential targets to improve chemotherapeutic efficacy. METHODS: Global histone H3 lysine 4 trimethylation (H3K4me3) levels were detected in pediatric EPN tumor samples with immunohistochemistry and immunoblots. Candidate genes conferring therapeutic resistance were profiled in pediatric EPN tumor samples with micro-array. Promoter H3K4me3 was examined for two candidate genes, CCND1 and ERBB2, with chromatin-immunoprecipitation coupled with real-time PCR (ChIP-PCR). These methods and MTS assay were used to verify a relationship between H3K4me3 levels and CCND1 and ERBB2, and to investigate cell viability in response to chemotherapeutic drugs in primary cultured pediatric EPN cells. RESULTS: H3K4me3 levels positively correlate with WHO grade malignancy in pediatric EPNs and are associated with progression free survival in patients with posterior fossa group A EPNs (PF-EPN-A). Reduction of H3K4me3 by silencing its methyltransferase SETD1A, in primary cultured EPN cells increased cell response to chemotherapy. CONCLUSIONS: Our results support the development of a novel treatment that targets H3K4me3 to increase chemotherapeutic efficacy in pediatric PF-EPN-A tumors.


Subject(s)
Cyclin D1/genetics , Ependymoma/drug therapy , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Receptor, ErbB-2/genetics , Carboplatin/pharmacology , Cell Survival/drug effects , Child, Preschool , Drug Resistance, Neoplasm/drug effects , Ependymoma/genetics , Ependymoma/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Pediatrics , Primary Cell Culture , Promoter Regions, Genetic/drug effects , Vincristine/pharmacology
17.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 37(5): 444-50, 2008 09.
Article in Zh | MEDLINE | ID: mdl-18925709

ABSTRACT

OBJECTIVE: To investigate the effects of leucine-rich repeats and immunoglobulin-like domains 3 (LRIG3) on the biological activity of glioblastoma cell line GL15. METHODS: Glioblastoma GL15 cells were cultured and transfected with LRIG3-EGFP plasmid. The location of LRIG3 in GL15 cells was observed with confocal microscopy. The proliferation and invasiveness of GL15 cells were detected with methyl thiazolyl tetrazolium (MTT) and Transwell methods respectively; the expression of epidermal growth factor receptor (EGFR) and LRIG3 mRNA and protein were detected with reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blot respectively. RESULT: After transfection with the plasmid LRIG-EGFP, LRIG3 fusion protein was found in cytoplasm of GL15 cells and cell proliferative and invasiveness were reduced. The expression of EGFR and LRIG3 varied with the duration of EGF treatment (100 ng/ml): the expression of EGFR decreased while the expression of LRIG3 increased as time prolonged. CONCLUSION: LRIG3 can inhibit the proliferation and invasiveness of glioblastoma cells and may be used as a target gene in gene therapy of glioblastoma.


Subject(s)
Brain Neoplasms/pathology , ErbB Receptors/metabolism , Glioblastoma/pathology , Membrane Proteins/metabolism , Cell Proliferation , Epidermal Growth Factor/genetics , ErbB Receptors/genetics , Humans , Membrane Proteins/genetics , Neoplasm Invasiveness , Plasmids/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Tumor Cells, Cultured
18.
Neoplasia ; 19(4): 261-270, 2017 04.
Article in English | MEDLINE | ID: mdl-28278424

ABSTRACT

Human glioma, in particular, malignant forms such as glioblastoma exhibit dismal survival rates despite advances in treatment strategies. A population of glioma cells with stem-like features, glioma cancer stem-like cells (GCSCs), contribute to renewal and maintenance of the tumor cell population and appear responsible for chemotherapeutic and radiation resistance. Bone morphogenetic protein 4 (BMP4), drives differentiation of GCSCs and thus improves therapeutic efficacy. Based on this observation it is imperative that the clinical merits of BMP4 in treating human gliomas should be addressed. This article reviews BMP4 signaling in central nervous system development and in glioma tumorigenesis, and the potential of this molecule as a treatment target in human gliomas. Further work needs to be done to determine if distinct lineages of GCSCs, associated with different glioma sub-classifications, proneural, neural, classical and mesenchymal, differ in responsiveness to BMP4 treatment. Additionally, interaction among BMP4 and cell matrix, tumor-vascular molecules and microglial immune cells also needs to be investigated, as this will enhance our knowledge about the role of BMP4 in human glioma and lead to the identification and/or development of novel therapeutic approaches that improve treatment outcomes of these devastating tumors.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Brain Neoplasms/metabolism , Glioma/metabolism , Bone Morphogenetic Protein 4/genetics , Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Carrier Proteins , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Central Nervous System/embryology , Central Nervous System/metabolism , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Glioma/diagnosis , Glioma/therapy , Humans , Phenotype , Protein Binding , Radiation Tolerance/genetics , Signal Transduction/drug effects , Tumor Microenvironment
19.
Article in English | MEDLINE | ID: mdl-17219981

ABSTRACT

The effects of epidermal growth factor (EGF) on proliferation of G15 glioma cells and the possible mechanisms were investigated. GFAP and EGFR expression was detected by immunohistochemical method. After the cells were treated with EGF at different concentrations, cell count method was used to determine the proliferation of glioma cells, cell cycle and apoptosis were analyzed by flow cytometry (FCM), and laser scan confocal microscope (LSCM) was used to measure the cytoplasmic free calcium. The results showed that GFAP was diffusedly expressed in GL15 cells and EGFR was over-expressed. EGF at doses of < or =1 ng/mL could significantly stimulate cell proliferation, cells in phase G0/G1 decreased, and those in phase S increased. EGF at doses of 10 and 100 ng/ml could inhibit the cell proliferation significantly, and the apoptosis ratio in high dose of EGF group was higher than in control group. EGF could significantly induce a quick rise of intracellular free calcium, but the peak value of intracellular free calcium activated by high dose of EGF was higher than by low dose of EGF. It was suggested that EGF had a dual effect on gliomas: low dose of EGF could stimulate the cell proliferation of gliomas, but high dose of EGF could induce the cell apoptosis and inhibit the proliferation of gliomas, which might be contributed to the difference of intracellular free calcium.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Epidermal Growth Factor/pharmacology , Glioma/pathology , Brain Neoplasms/pathology , Calcium/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , ErbB Receptors/metabolism , Glial Fibrillary Acidic Protein/metabolism , Humans
20.
Gene ; 534(2): 400-7, 2014 Jan 25.
Article in English | MEDLINE | ID: mdl-23954875

ABSTRACT

In vitro neural differentiation of human embryonic stem cells (hESCs) is an advantageous system for studying early neural development. The process of early neural differentiation in hESCs begins by initiation of primitive neuroectoderm, which is manifested by rosette formation, with consecutive differentiation into neural progenitors and early glial-like cells. In this study, we examined the involvement of early neural markers - OTX2, PAX6, Sox1, Nestin, NR2F1, NR2F2, and IRX2 - in the onset of rosette formation, during spontaneous neural differentiation of hESC and human induced pluripotent stem cell (hiPSC) colonies. This is in contrast to the conventional way of studying rosette formation, which involves induction of neuronal differentiation and the utilization of embryoid bodies. Here we show that OTX2 is highly expressed at the onset of rosette formation, when rosettes comprise no more than 3-5 cells, and that its expression precedes that of established markers of early neuronal differentiation. Importantly, the rise of OTX2 expression in these cells coincides with the down-regulation of the pluripotency marker OCT4. Lastly, we show that cells derived from rosettes that emerge during spontaneous differentiation of hESCs or hiPSCs are capable of differentiating into dopaminergic neurons in vitro, and into mature-appearing pyramidal and serotonergic neurons weeks after being injected into the motor cortex of NOD-SCID mice.


Subject(s)
Cell Differentiation/genetics , Dopaminergic Neurons/physiology , Embryonic Stem Cells/physiology , Pluripotent Stem Cells/physiology , Rosette Formation/methods , Serotonergic Neurons/physiology , Animals , Biomarkers/metabolism , Cell Line , Dopaminergic Neurons/metabolism , Down-Regulation/genetics , Embryonic Stem Cells/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Motor Cortex/metabolism , Motor Cortex/physiology , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Otx Transcription Factors/genetics , Otx Transcription Factors/metabolism , Pluripotent Stem Cells/metabolism , Serotonergic Neurons/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL