Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Neuro Oncol ; 19(4): 524-534, 2017 04 01.
Article in English | MEDLINE | ID: mdl-27994064

ABSTRACT

Background: The nuclear factor I-A (NFIA) transcription factor promotes glioma growth and inhibits apoptosis in glioblastoma (GBM) cells. Here we report that the NFIA pro-survival effect in GBM is mediated in part via a novel NFIA-nuclear factor-kappaB (NFκB) p65 feed-forward loop. Methods: We examined effects of gain- and loss-of-function manipulations of NFIA and NFκB p65 on each other's transcription, cell growth, apoptosis and sensitivity to chemotherapy in patient-derived GBM cells and established GBM cell lines. Results: NFIA enhanced apoptosis evasion by activating NFκB p65 and its downstream anti-apoptotic factors tumor necrosis factor receptor-associated factor 1 (TRAF1) and cellular inhibitor of apoptosis proteins (cIAPs). Induction of NFκB by NFIA was required to protect cells from apoptosis, and inhibition of NFκB effectively reversed the NFIA anti-apoptotic effect. Conversely, NFIA knockdown decreased expression of NFκB and anti-apoptotic genes TRAF1 and cIAPs, and increased baseline apoptosis. NFIA positively regulated NFκB transcription and NFκB protein level. Interestingly, NFκB also activated the NFIA promoter and increased NFIA level, and knockdown of NFIA was sufficient to attenuate the NFκB pro-survival effect, suggesting a reciprocal regulation between NFIA and NFκB in governing GBM cell survival. Supporting this, NFIA and NFκB expression levels were highly correlated in human GBM and patient-derived GBM cells. Conclusions: These data define a previously unknown NFIA-NFκB feed-forward regulation that may contribute to GBM cell survival.


Subject(s)
Apoptosis , Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , NFI Transcription Factors/metabolism , Transcription Factor RelA/metabolism , Cell Survival , Drug Resistance, Neoplasm , Humans , Tumor Cells, Cultured
2.
PLoS One ; 9(12): e116114, 2014.
Article in English | MEDLINE | ID: mdl-25541984

ABSTRACT

Glioblastoma multiforme (GBM) is a deadly primary brain malignancy. Glioblastoma stem cells (GSC), which have the ability to self-renew and differentiate into tumor lineages, are believed to cause tumor recurrence due to their resistance to current therapies. A subset of GSCs is marked by cell surface expression of CD133, a glycosylated pentaspan transmembrane protein. The study of CD133-expressing GSCs has been limited by the relative paucity of genetic tools that specifically target them. Here, we present CD133-LV, a lentiviral vector presenting a single chain antibody against CD133 on its envelope, as a vehicle for the selective transduction of CD133-expressing GSCs. We show that CD133-LV selectively transduces CD133+ human GSCs in dose-dependent manner and that transduced cells maintain their stem-like properties. The transduction efficiency of CD133-LV is reduced by an antibody that recognizes the same epitope on CD133 as the viral envelope and by shRNA-mediated knockdown of CD133. Conversely, the rate of transduction by CD133-LV is augmented by overexpression of CD133 in primary human GBM cultures. CD133-LV selectively transduces CD133-expressing cells in intracranial human GBM xenografts in NOD.SCID mice, but spares normal mouse brain tissue, neurons derived from human embryonic stem cells and primary human astrocytes. Our findings indicate that CD133-LV represents a novel tool for the selective genetic manipulation of CD133-expressing GSCs, and can be used to answer important questions about how these cells contribute to tumor biology and therapy resistance.


Subject(s)
Antigens, CD/immunology , Brain Neoplasms/pathology , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Glioblastoma/pathology , Glycoproteins/immunology , Neoplastic Stem Cells/pathology , Peptides/immunology , Transduction, Genetic , AC133 Antigen , Animals , Antigens, CD/analysis , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Genetic Vectors/genetics , Genetic Vectors/immunology , Glioblastoma/genetics , Glioblastoma/immunology , Glycoproteins/analysis , Humans , Lentivirus/genetics , Lentivirus/immunology , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Peptides/analysis , Tumor Cells, Cultured
3.
Neuro Oncol ; 16(2): 191-203, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24305710

ABSTRACT

Background Nuclear factor IA (NFIA), a transcription factor and essential regulator in embryonic glial development, is highly expressed in human glioblastoma (GBM) compared with normal brain, but its contribution to GBM and cancer pathogenesis is unknown. Here we demonstrate a novel role for NFIA in promoting growth and migration of GBM and establish the molecular mechanisms mediating these functions. Methods To determine the role of NFIA in glioma, we examined the effects of NFIA in growth, proliferation, apoptosis, and migration. We used gain-of-function (overexpression) and loss-of-function (shRNA knockdown) of NFIA in primary patient-derived GBM cells and established glioma cell lines in culture and in intracranial xenografts in mouse brains. Results Knockdown of native NFIA blocked tumor growth and induced cell death and apoptosis. Complementing this, NFIA overexpression accelerated growth, proliferation, and migration of GBM in cell culture and in mouse brains. These NFIA tumor-promoting effects were mediated via transcriptional repression of p53, p21, and plasminogen activator inhibitor 1 (PAI1) through specific NFIA-recognition sequences in their promoters. Importantly, the effects of NFIA on proliferation and apoptosis were independent of TP53 mutation status, a finding especially relevant for GBM, in which TP53 is frequently mutated. Conclusion NFIA is a modulator of GBM growth and migration, and functions by distinct regulation of critical oncogenic pathways that govern the malignant behavior of GBM.


Subject(s)
Brain Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , NFI Transcription Factors/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Cycle , Cell Movement , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Immunoenzyme Techniques , Mice , Mice, Nude , NFI Transcription Factors/antagonists & inhibitors , NFI Transcription Factors/genetics , Plasminogen Activator Inhibitor 1/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
4.
Neuro Oncol ; 12(2): 122-32, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20150379

ABSTRACT

Nuclear factor IA (NFIA) is a transcription factor that specifies glial cell identity and promotes astrocyte differentiation during embryonic development. Its expression and function in gliomas are not known. Here, we examined NFIA protein expression in gliomas and its association with clinical outcome in pediatric malignant astrocytomas. We analyzed expression of NFIA by immunohistochemistry in 88 existing glioma specimens from Childrens Hospital Los Angeles and the University of Southern California. Association between NFIA expression and progression-free survival (PFS) was examined in high-grade astrocytomas for which clinical data were available (n = 23, all children). NFIA was highly expressed in astrocytomas of all grades, but only in a minority of cells in oligodendroglial tumors. NFIA was expressed on a higher percentage of tumor cells in low-grade astrocytomas (91 +/- 5% and 77 +/- 14% in World Health Organization [WHO] I and II, respectively) compared with high-grade astrocytomas (48 +/- 18% and 37 +/- 16% in WHO III and IV, respectively; P < .001, low- vs high-grade astrocytomas). There was a significant association between NFIA expression and PFS in children with astrocytoma WHO grade III or IV (Cox regression P = .019; logrank trend test for NFIA tertiles P = .0040 and NFIA quartiles P = .014). The association was not consistently significant in this small series of patients after adjustment was made for WHO grade III or IV. This is the first study to demonstrate expression of NFIA protein in astrocytomas and its association with grades of astrocytoma and PFS, suggesting that NFIA may play a role in astrocytoma biology.


Subject(s)
Astrocytoma/metabolism , Brain Neoplasms/metabolism , NFI Transcription Factors/biosynthesis , Adolescent , Astrocytoma/mortality , Brain Neoplasms/mortality , Child , Child, Preschool , Disease-Free Survival , Female , Humans , Immunohistochemistry , Infant , Kaplan-Meier Estimate , Male , Retrospective Studies
5.
J Pediatr ; 146(6): 843-4, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15973330

ABSTRACT

Frasier syndrome is a relatively rare disorder associated with XY gonadal dysgenesis, gonadoblastoma, and kidney failure. In this report, we identify a classic mutation in the Wilms' tumor 1 gene in one of the original cases of Frasier syndrome reported in this Journal in 1964.


Subject(s)
Denys-Drash Syndrome/genetics , Genes, Wilms Tumor , Gonadal Dysgenesis, 46,XY/genetics , Point Mutation , Adenine , DNA Mutational Analysis , Female , Guanine , Humans , Introns , Middle Aged , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...