Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Res Sq ; 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38699302

ABSTRACT

Transcribed Ultra-Conserved Regions (TUCRs) represent a severely understudied class of putative non-coding RNAs (ncRNAs) that are 100% conserved across multiple species. We performed the first-ever analysis of TUCRs in glioblastoma (GBM) and low-grade gliomas (LGG). We leveraged large human datasets to identify the genomic locations, chromatin accessibility, transcription, differential expression, correlation with survival, and predicted functions of all 481 TUCRs, and identified TUCRs that are relevant to glioma biology. Of these, we investigated the expression, function, and mechanism of action of the most highly upregulated intergenic TUCR, uc.110, identifying it as a new oncogene. Uc.110 was highly overexpressed in GBM and LGG, where it promoted malignancy and tumor growth. Uc.110 activated the WNT pathway by upregulating the expression of membrane frizzled-related protein (MFRP), by sponging the tumor suppressor microRNA miR-544. This pioneering study shows important roles for TUCRs in gliomas and provides an extensive database and novel methods for future TUCR research.

2.
bioRxiv ; 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38562826

ABSTRACT

Transcribed Ultra-Conserved Regions (TUCRs) represent a severely understudied class of putative non-coding RNAs (ncRNAs) that are 100% conserved across multiple species. We performed the first-ever analysis of TUCRs in glioblastoma (GBM) and low-grade gliomas (LGG). We leveraged large human datasets to identify the genomic locations, chromatin accessibility, transcription, differential expression, correlation with survival, and predicted functions of all 481 TUCRs, and identified TUCRs that are relevant to glioma biology. Of these, we investigated the expression, function, and mechanism of action of the most highly upregulated intergenic TUCR, uc.110, identifying it as a new oncogene. Uc.110 was highly overexpressed in GBM and LGG, where it promoted malignancy and tumor growth. Uc.110 activated the WNT pathway by upregulating the expression of membrane frizzled-related protein (MFRP), by sponging the tumor suppressor microRNA miR-544. This pioneering study shows important roles for TUCRs in gliomas and provides an extensive database and novel methods for future TUCR research.

3.
Int J Mol Sci ; 24(11)2023 May 26.
Article in English | MEDLINE | ID: mdl-37298284

ABSTRACT

microRNAs (miRNAs) play an important role in the pathology of glioblastoma (GBM), which is the most malignant and most common primary malignant brain tumor. miRNAs can target multiple genes simultaneously and are considered as potential therapeutic agents or targets. This study aimed to determine the role of miR-3174 in the pathobiology of GBM using both in vitro and in vivo approaches. This is the first study deciphering the role of miR-3174 in GBM. We studied the expression of miR-3174 and found it to be downregulated in a panel of GBM cell lines, GSCs and tissues relative to astrocytes and normal brain tissue. This finding led us to hypothesize that miR-3174 has a tumor-suppressive role in GBM. Exogenous expression of miR-3174 inhibited GBM cell growth and invasion, and hampered the neurosphere formation ability of GSCs. miR-3174 downregulated the expression of multiple tumor-promoting genes including CD44, MDM2, RHOA, PLAU and CDK6. Further, overexpression of miR-3174 reduced tumor volume in nude mice with intracranial xenografts. Immuno-histochemical study of brain sections with intracranial tumor xenografts revealed the pro-apoptotic and anti-proliferative activity of miR-3174. In conclusion, we demonstrated that miR-3174 has a tumor-suppressive role in GBM and could be exploited for therapeutic purposes.


Subject(s)
Brain Neoplasms , Glioblastoma , MicroRNAs , Animals , Mice , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Glioblastoma/metabolism , Mice, Nude , Genes, Tumor Suppressor , Brain/metabolism , Cell Proliferation/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic
4.
Cells ; 11(10)2022 05 19.
Article in English | MEDLINE | ID: mdl-35626721

ABSTRACT

Transcribed ultraconserved regions are putative lncRNA molecules that are transcribed from DNA that is 100% conserved in human, mouse, and rat genomes. This is notable, as lncRNAs are typically poorly conserved. TUCRs remain very understudied in many diseases, including cancer. In this review, we summarize the current literature on TUCRs in cancer with respect to expression deregulation, functional roles, mechanisms of action, and clinical perspectives.


Subject(s)
Neoplasms , RNA, Long Noncoding , Animals , Conserved Sequence/genetics , DNA , Genome , Mice , Neoplasms/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Rats
5.
PLoS One ; 9(5): e96239, 2014.
Article in English | MEDLINE | ID: mdl-24805821

ABSTRACT

Glioblastoma is the most common and lethal primary brain tumor. Tumor initiation and recurrence are likely caused by a sub-population of glioblastoma stem cells, which may derive from mutated neural stem and precursor cells. Since CD133 is a stem cell marker for both normal brain and glioblastoma, and to better understand glioblastoma formation and recurrence, we looked for dys-regulated microRNAs in human CD133+ glioblastoma stem cells as opposed to CD133+ neural stem cells isolated from normal human brain. Using FACS sorting of low-passage cell samples followed by microRNA microarray analysis, we found 43 microRNAs that were dys-regulated in common in three separate CD133+ human glioblastomas compared to CD133+ normal neural stem cells. Among these were several microRNAs not previously associated with cancer. We then verified the microRNAs dys-regulated in glioblastoma using quantitative real time PCR and Taqman analysis of the original samples, as well as human GBM stem cell and established cell lines and many human specimens. We show that two candidate oncogenic microRNAs, miR-363 and miR-582-5p, can positively influence glioblastoma survival, as shown by forced expression of the microRNAs and their inhibitors followed by cell number assay, Caspase 3/7 assay, Annexin V apoptosis/fluorescence activated cell sorting, siRNA rescue of microRNA inhibitor treatment, as well as 3'UTR mutagenesis to show luciferase reporter rescue of the most successful targets. miR-582-5p and miR-363 are shown to directly target Caspase 3, Caspase 9, and Bim.


Subject(s)
3' Untranslated Regions/genetics , Apoptosis Regulatory Proteins/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cell Survival/genetics , Glioblastoma/genetics , Membrane Proteins/metabolism , MicroRNAs/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins/metabolism , AC133 Antigen , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Caspase 3/genetics , Caspase 9/genetics , Cell Line, Tumor , Cell Survival/physiology , Cells, Cultured , Flow Cytometry , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Membrane Proteins/genetics , MicroRNAs/genetics , Peptides/genetics , Peptides/metabolism , Proto-Oncogene Proteins/genetics
6.
Cancer Res ; 74(5): 1541-53, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24425048

ABSTRACT

Great interest persists in useful prognostic and therapeutic targets in glioblastoma. In this study, we report the definition of miRNA (miR)-148a as a novel prognostic oncomiR in glioblastoma. miR-148a expression was elevated in human glioblastoma specimens, cell lines, and stem cells (GSC) compared with normal human brain and astrocytes. High levels were a risk indicator for glioblastoma patient survival. Functionally, miR-148a expression increased cell growth, survival, migration, and invasion in glioblastoma cells and GSCs and promoted GSC neurosphere formation. Two direct targets of miR-148a were identified, the EGF receptor (EGFR) regulator MIG6 and the apoptosis regulator BIM, which rescue experiments showed were essential to mediate the oncogenic activity of miR-148a. By inhibiting MIG6 expression, miR-148a reduced EGFR trafficking to Rab7-expressing compartments, which includes late endosomes and lysosomes. This process coincided with reduced degradation and elevated expression and activation of EGFR. Finally, inhibition of miR-148a strongly suppressed GSC and glioblastoma xenograft growth in vivo. Taken together, our findings provide a comprehensive analysis of the prognostic value and oncogenic function of miR-148a in glioblastoma, further defining it as a potential target for glioblastoma therapy.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , ErbB Receptors/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Membrane Proteins/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Bcl-2-Like Protein 11 , Cell Line, Tumor , Cell Movement/genetics , Humans , Mice , Prognosis , Up-Regulation/genetics , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins
7.
J Cancer Ther ; 4(8)2013 Oct.
Article in English | MEDLINE | ID: mdl-24273681

ABSTRACT

Our previous results indicated that both the secreted and the intracellular form of full length and 1-97 N-terminal fragment of IGFBP-3 induces apoptosis in PC-3 human prostate cancer cells in an IGF-dependent and independent manner. This study was undertaken to delineate possible down-stream signaling pathways that are involved in this process. Intact IGFBP-3 and its N-terminal 1-97 fragments with or without a signal pro-peptide was fused to YFP and expressed in PC-3 human prostate cancer cells. In some cases, the putative IGF-binding site present in full length IGFBP-3 and its N-terminal fragment was also mutated. Extent of apoptosis was quantified using FACS. Up-regulation of total Stat-1 and activation of phospho-Stat-1 was shown by western blot. TGF-ß signal was measured by luciferase reporter assay. Results from inhibitor studies indicated that both the Caspase 8 and caspase 9 pathways are involved in IGFBP-3 (non-secreted form) induced apoptosis in PC-3 cells. Exogenous addition of IGFBP-3 to PC-3 cells increased Stat-1 protein expression/tyrosine phosphorylation. Interestingly, results also showed that knockdown of Stat-1 by siRNA potentiated the IGFBP-3 induced apoptosis in PC-3 cells. In addition, both full-length IGFBP-3 and its 1-97 N-terminal fragments inhibited TGFß signaling in these cells. This is the first report that compares the signal transduction pathways involved in apoptotic pathways mediated by IGFBP-3 in PC-3 human prostate cancer cells. Non-secreted form of full length IGFBP-3 and its N-terminal fragments induced apoptosis in PC-3 cells via activation of caspase 8 and caspase 9. We noted that both secreted and non-secreted forms of IGFBP-3 are involved in modulating Stat-1 and TGF-ß pathways to induce apoptotic actions in PC-3 cells. Surprisingly, only non-secreted form of IGFBP-3 and its N-terminal fragments are involved in the induction of apoptosis in PC-3 cells via caspase 8 and caspase 9 activation. These studies clearly demonstrate that secreted and non-secreted FL and its 1-97 N-terminal fragments induce apoptosis in PC-3 cells by regulating different mechanistic pathways.

8.
Neuro Oncol ; 15(12): 1652-63, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24158111

ABSTRACT

BACKGROUND: Despite advances in the treatment of the most aggressive form of brain tumor, glioblastoma, patient prognosis remains disappointing. This failure in treatment has been attributed to dysregulated oncogenic pathways, as observed in other tumors. We and others have suggested the use of microRNAs (miRs) as therapeutic tools able to target multiple pathways in glioblastoma. METHODS: This work features PCR quantification of miRs and transient transfection of many glioblastoma cell lines with miRs, followed by cell number analysis, trypan blue staining, alamarBlue assay of cell viability, caspase-3/-7 activity assay, immunoblot of cleaved poly(ADP-ribose) polymerase and fluorescence activated cell sorting and imaging of apoptotic nuclei, cell invasion assays, MRIs of glioblastoma xenografts in mice using transiently transfected cells as well as posttumor treatment with lentiviral vector encoding miR-297, and analysis of miR-297 target diacylglycerol kinase (DGK)-α including immunoblot, 3'UTR luciferase activity, and rescue with DGK-α overexpression. Cell counts and DGK-α immunoblot were also analyzed in the context of hypoxia and with overexpression of heterogeneous ribonucleoprotein L (hnRNPL). RESULTS: We identified miR-297 as a highly cytotoxic microRNA in glioblastoma, with minimal cytotoxicity to normal astrocytes. miR-297 overexpression reduced in vitro invasiveness and in vivo tumor formation. DGK-α is shown to be a miR-297 target with a critical role in miR-297 toxicity. In addition, hypoxia and its mediator hnRNPL upregulated DGK-α and buffered the cytotoxic effects of miR-297. CONCLUSION: This work shows miR-297 as a novel and physiologic regulator of cancer cell survival, largely through targeting of DGK-α, and also indicates that hypoxia ameliorates miR-297 toxicity to cancer cells.


Subject(s)
Brain Neoplasms/mortality , Diacylglycerol Kinase/metabolism , Glioblastoma/mortality , Hypoxia/genetics , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Animals , Apoptosis , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Movement , Cell Proliferation , Diacylglycerol Kinase/genetics , Flow Cytometry , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, SCID , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Cancer Discov ; 3(7): 782-97, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23558954

ABSTRACT

Although diacylglycerol kinase α (DGKα) has been linked to several signaling pathways related to cancer cell biology, it has been neglected as a target for cancer therapy. The attenuation of DGKα activity via DGKα-targeting siRNA and small-molecule inhibitors R59022 and R59949 induced caspase-mediated apoptosis in glioblastoma cells and in other cancers, but lacked toxicity in noncancerous cells. We determined that mTOR and hypoxia-inducible factor-1α (HIF-1α) are key targets of DGKα inhibition, in addition to its regulation of other oncogenes. DGKα regulates mTOR transcription via a unique pathway involving cyclic AMP. Finally, we showed the efficacy of DGKα inhibition with short hairpin RNA or a small-molecule agent in glioblastoma and melanoma xenograft treatment models, with growth delay and decreased vascularity. This study establishes DGKα as a central signaling hub and a promising therapeutic target in the treatment of cancer.


Subject(s)
Brain Neoplasms/genetics , Diacylglycerol Kinase/genetics , Glioblastoma/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Apoptosis/drug effects , Brain Neoplasms/pathology , Cell Line, Tumor , Diacylglycerol Kinase/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Molecular Targeted Therapy , Piperidines/administration & dosage , Pyrimidinones/administration & dosage , Quinazolinones/administration & dosage , RNA, Small Interfering , Thiazoles/administration & dosage
10.
J Neurooncol ; 112(2): 153-63, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23307328

ABSTRACT

MicroRNAs and cancer stem cells have emerged as critical players in glioblastoma, one of the deadliest human cancers. In this study, we investigated the expression and function of microRNA-10b in glioblastoma cells and stem cells. An analysis of The Cancer Genome Atlas data revealed a correlation between high miR-10b levels and poor prognosis in glioblastoma patients. We measured the levels of miR-10b and found that it is upregulated in human glioblastoma tissues, glioblastoma cell and stem cell lines as compared to normal human tissues or astrocytes. Inhibition of miR-10b with a specific antagomir inhibited the proliferation of glioblastoma established and stem cell lines. Inhibition of miR-10b strongly reduced cell invasion and migration in glioblastoma cell and stem cell lines while overexpression of miR-10b induced cell migration and invasion. We also investigated several predicted targets of miR-10b but could not verify any of them experimentally. Additionally, miR-10b inhibition significantly decreased the in vivo growth of stem cell-derived orthotopic GBM xenografts. Altogether, our findings confirm the oncogenic effects of miR-10b in GBM cells and show for the first time a role of this microRNA in GBM stem cells. Targeting miR-10b might therefore inhibit glioblastoma stem cells, which are thought to be at the origin of glioblastoma and to contribute its recurrence and resistance to therapy.


Subject(s)
Apoptosis , Brain Neoplasms/pathology , Brain/metabolism , Cell Movement , Glioblastoma/pathology , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Animals , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Cell Adhesion , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/mortality , Humans , Mice , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Xenograft Model Antitumor Assays
11.
Neuro Oncol ; 14(10): 1215-26, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22962413

ABSTRACT

The Notch pathway is dysregulated and a potential target in glioblastoma multiforme (GBM). Currently available Notch inhibitors block γ-secretase, which is necessary for Notch processing. However, Notch is first cleaved by α-secretase outside the plasma membrane, via a disintegrin and metalloproteinase-10 and -17. In this work, we used a potent α-secretase inhibitor (ASI) to test inhibition of glioblastoma growth and inhibition of Notch and of both novel and known Notch targets. Featured in this study are luciferase reporter assays and immunoblot, microarray analysis, chromatin immunoprecipitation (ChIP), quantitative real-time PCR, cell number assay, bromodeoxyuridine incorporation, plasmid rescue, orthotopic xenograft model, and local delivery of treatment with convection-enhanced delivery using nanoparticles, as well as survival, MRI, and ex vivo luciferase assay. A CBF1-luciferase reporter assay as well as an immunoblot of endogenous Notch revealed Notch inhibition by the ASI. Microarray analysis, quantitative real-time PCR, and ChIP of ASI and γ-secretase inhibitor (GSI) treatment of GBM cells identified known Notch pathway targets, as well as novel Notch targets, including YKL-40 and leukemia inhibitory factor. Finally, we found that local nanoparticle delivery of ASIs but not GSIs increased survival time significantly in a GBM stem cell xenograft treatment model, and ASI treatment resulted in decreased tumor size and Notch activity. This work indicates α-secretase as an alternative to γ-secretase for inhibition of Notch in GBM and possibly other cancers as well, and it identifies novel Notch targets with biologic relevance and potential as biomarkers.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Brain Neoplasms/pathology , Cell Proliferation , Glioblastoma/pathology , Neoplastic Stem Cells/metabolism , Receptors, Notch/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Cycle , Chromatin Immunoprecipitation , Gene Expression Profiling , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , In Vitro Techniques , Luciferases/metabolism , Magnetics , Mice , Mice, Inbred BALB C , Nanoparticles , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Oligonucleotide Array Sequence Analysis , Piperidines/pharmacology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spiro Compounds/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
J Biol Chem ; 286(28): 25377-86, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21592959

ABSTRACT

Epidermal growth factor receptor (EGFR) is involved in development and progression of many human cancers. We have previously demonstrated that the ubiquitin-specific peptidase Usp18 (Ubp43) is a potent regulator of EGFR protein expression. Here we report that the 3'-untranslated region (3'-UTR) of the EGFR message modulates RNA translation following cell treatment with Usp18 siRNA, suggesting microRNA as a possible mediator. Given earlier evidence of EGFR regulation by the microRNA miR-7, we assessed whether miR-7 mediates Usp18 siRNA effects. We found that Usp18 depletion elevates miR-7 levels in several cancer cell lines because of a transcriptional activation and/or mRNA stabilization of miR-7 host genes and that miR-7 acts downstream of Usp18 to regulate EGFR mRNA translation via the 3'-UTR. Also, depletion of Usp18 led to a decrease in protein levels of other known oncogenic targets of miR-7, reduced cell proliferation and soft agar colony formation, and increased apoptosis. Notably, all of these phenotypes were reversed by a specific inhibitor of miR-7. Thus, our findings support a model in which Usp18 inhibition promotes up-regulation of miR-7, which in turn inhibits EGFR expression and the tumorigenic activity of cancer cells.


Subject(s)
3' Untranslated Regions , Endopeptidases/metabolism , ErbB Receptors/biosynthesis , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Models, Biological , Neoplasm Proteins/metabolism , Neoplasms/metabolism , RNA, Neoplasm/metabolism , Apoptosis/genetics , Cell Proliferation , Endopeptidases/genetics , ErbB Receptors/genetics , Gene Knockdown Techniques , HeLa Cells , Humans , MicroRNAs/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , RNA Stability/genetics , RNA, Neoplasm/genetics , Ubiquitin Thiolesterase
13.
Neuro Oncol ; 12(11): 1102-12, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20667897

ABSTRACT

Emerging studies have identified microRNAs (miRNAs) as possible therapeutic tools for the treatment of glioma, the most aggressive brain tumor. Their important targets in this tumor are not well understood. We recently found that the Notch pathway is a target of miRNA-326. Ectopic expression of miRNA-326 in glioma and glioma stem cells induced their apoptosis and reduced their metabolic activity. Computational target gene prediction revealed pyruvate kinase type M2 (PKM2) as another target of miRNA-326. PKM2 has recently been shown to play a key role in cancer cell metabolism. To investigate whether it might be a functionally important target of miR-326, we used RNA interference to knockdown PKM2 expression in glioma cells. Transfection of the established glioma and glioma stem cells with PKM2 siRNA reduced their growth, cellular invasion, metabolic activity, ATP and glutathione levels, and activated AMP-activated protein kinase. The cytotoxic effects exhibited by PKM2 knockdown in glioma and glioma stem cells were not observed in transformed human astrocytes. Western blot analysis of human glioblastoma specimens showed high levels of PKM2 protein, but none was observed in normal brain samples. Strikingly, cells with high levels of PKM2 expressed lower levels of miR-326, suggestive of endogenous regulation of PKM2 by miR-326. Our data suggest PKM2 inhibition as a therapy for glioblastoma, with the potential for minimal toxicity to the brain.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioma/genetics , MicroRNAs/genetics , Pyruvate Kinase/genetics , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/genetics , Gene Expression , Glioma/metabolism , Glioma/pathology , Humans , MicroRNAs/metabolism , Mutagenesis, Site-Directed , Pyruvate Kinase/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transfection
14.
J Neurosci ; 30(9): 3347-57, 2010 Mar 03.
Article in English | MEDLINE | ID: mdl-20203194

ABSTRACT

Maturation of the mammalian nervous system requires adequate provision of thyroid hormone and mechanisms that enhance tissue responses to the hormone. Here, we report that the development of cones, the photoreceptors for daylight and color vision, requires protection from thyroid hormone by type 3 deiodinase, a thyroid hormone-inactivating enzyme. Type 3 deiodinase, encoded by Dio3, is expressed in the immature mouse retina. In Dio3(-/-) mice, approximately 80% of cones are lost through neonatal cell death. Cones that express opsin photopigments for response to both short (S) and medium-long (M) wavelength light are lost. Rod photoreceptors, which mediate dim light vision, remain essentially intact. Excessive thyroid hormone in wild-type pups also eliminates cones. Cone loss is mediated by cone-specific thyroid hormone receptor beta2 (TRbeta2) as deletion of TRbeta2 rescues cones in Dio3(-/-) mice. However, rescued cones respond to short but not longer wavelength light because TRbeta2 under moderate hormonal stimulation normally induces M opsin and controls the patterning of M and S opsins over the retina. The results suggest that type 3 deiodinase limits hormonal exposure of the cone to levels that safeguard both cone survival and the patterning of opsins that is required for cone function.


Subject(s)
Iodide Peroxidase/genetics , Retina/enzymology , Retina/growth & development , Retinal Cone Photoreceptor Cells/enzymology , Thyroid Hormones/metabolism , Animals , Cell Death/genetics , Cell Differentiation/genetics , Cell Survival/genetics , Female , Gene Expression Regulation, Developmental/genetics , Light , Male , Mice , Mice, Knockout , Opsins/metabolism , Photic Stimulation , Retina/cytology , Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/radiation effects , Thyroid Hormone Receptors beta/metabolism , Vision, Ocular/genetics
15.
J Neurosci ; 29(48): 15161-8, 2009 Dec 02.
Article in English | MEDLINE | ID: mdl-19955368

ABSTRACT

Little is known of microRNA interactions with cellular pathways. Few reports have associated microRNAs with the Notch pathway, which plays key roles in nervous system development and in brain tumors. We previously implicated the Notch pathway in gliomas, the most common and aggressive brain tumors. While investigating Notch mediators, we noted microRNA-326 was upregulated following Notch-1 knockdown. This neuronally expressed microRNA was not only suppressed by Notch but also inhibited Notch proteins and activity, indicating a feedback loop. MicroRNA-326 was downregulated in gliomas via decreased expression of its host gene. Transfection of microRNA-326 into both established and stem cell-like glioma lines was cytotoxic, and rescue was obtained with Notch restoration. Furthermore, miR-326 transfection reduced glioma cell tumorigenicity in vivo. Additionally, we found microRNA-326 partially mediated the toxic effects of Notch knockdown. This work demonstrates a microRNA-326/Notch axis, shedding light on the biology of Notch and suggesting microRNA-326 delivery as a therapy.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Glioma/pathology , MicroRNAs/metabolism , Receptor, Notch1/metabolism , Signal Transduction/physiology , Apoptosis/genetics , Arrestins/genetics , Arrestins/metabolism , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Flow Cytometry/methods , Gene Expression Profiling , Humans , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oligonucleotide Array Sequence Analysis/methods , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Receptor, Notch1/genetics , Signal Transduction/genetics , Transfection/methods , Tumor Stem Cell Assay/methods , beta-Arrestins
16.
Cancer Res ; 69(19): 7569-76, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19773441

ABSTRACT

MicroRNA-34a (miR-34a) is a transcriptional target of p53 that is down-regulated in some cancer cell lines. We studied the expression, targets, and functional effects of miR-34a in brain tumor cells and human gliomas. Transfection of miR-34a down-regulated c-Met in human glioma and medulloblastoma cells and Notch-1, Notch-2, and CDK6 protein expressions in glioma cells. miR-34a expression inhibited c-Met reporter activities in glioma and medulloblastoma cells and Notch-1 and Notch-2 3'-untranslated region reporter activities in glioma cells and stem cells. Analysis of human specimens showed that miR-34a expression is down-regulated in glioblastoma tissues as compared with normal brain and in mutant p53 gliomas as compared with wild-type p53 gliomas. miR-34a levels in human gliomas inversely correlated to c-Met levels measured in the same tumors. Transient transfection of miR-34a into glioma and medulloblastoma cell lines strongly inhibited cell proliferation, cell cycle progression, cell survival, and cell invasion, but transfection of miR-34a into human astrocytes did not affect cell survival and cell cycle status. Forced expression of c-Met or Notch-1/Notch-2 transcripts lacking the 3'-untranslated region sequences partially reversed the effects of miR-34a on cell cycle arrest and cell death in glioma cells and stem cells, respectively. Also, transient expression of miR-34a in glioblastoma cells strongly inhibited in vivo glioma xenograft growth. Together, these findings represent the first comprehensive analysis of the role of miR-34a in gliomas. They show that miR-34a suppresses brain tumor growth by targeting c-Met and Notch. The results also suggest that miR-34a could serve as a potential therapeutic agent for brain tumors.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/therapy , Genetic Therapy/methods , Glioblastoma/genetics , Glioblastoma/therapy , MicroRNAs/genetics , Oncogenes , 3' Untranslated Regions , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Cycle/physiology , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/biosynthesis , Cyclin-Dependent Kinase 6/genetics , Down-Regulation , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , MicroRNAs/biosynthesis , MicroRNAs/metabolism , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/biosynthesis , Proto-Oncogene Proteins c-met/genetics , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/biosynthesis , Receptor, Notch1/genetics , Receptor, Notch2/antagonists & inhibitors , Receptor, Notch2/biosynthesis , Receptor, Notch2/genetics , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor alpha/biosynthesis , Receptor, Platelet-Derived Growth Factor alpha/genetics , Transfection
17.
Mol Cancer Ther ; 8(2): 376-85, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19190120

ABSTRACT

The tyrosine kinase receptor c-Met and its ligand hepatocyte growth factor (HGF) are frequently overexpressed and the tumor suppressor PTEN is often mutated in glioblastoma. Because PTEN can interact with c-Met-dependent signaling, we studied the effects of PTEN on c-Met-induced malignancy and associated molecular events and assessed the potential therapeutic value of combining PTEN restoration approaches with HGF/c-Met inhibition. We studied the effects of c-Met activation on cell proliferation, cell cycle progression, cell migration, cell invasion, and associated molecular events in the settings of restored or inhibited PTEN expression in glioblastoma cells. We also assessed the experimental therapeutic effects of combining anti-HGF/c-Met approaches with PTEN restoration or mTOR inhibition. PTEN significantly inhibited HGF-induced proliferation, cell cycle progression, migration, and invasion of glioblastoma cells. PTEN attenuated HGF-induced changes of signal transduction proteins Akt, GSK-3, JNK, and mTOR as well as cell cycle regulatory proteins p27, cyclin E, and E2F-1. Combining PTEN restoration to PTEN-null glioblastoma cells with c-Met and HGF inhibition additively inhibited tumor cell proliferation and cell cycle progression. Similarly, combining a monoclonal anti-HGF antibody (L2G7) with the mTOR inhibitor rapamycin had additive inhibitory effects on glioblastoma cell proliferation. Systemic in vivo delivery of L2G7 and PTEN restoration as well as systemic in vivo deliveries of L2G7 and rapamycin additively inhibited intracranial glioma xenograft growth. These preclinical studies show for the first time that PTEN loss amplifies c-Met-induced glioblastoma malignancy and suggest that combining anti-HGF/c-Met approaches with PTEN restoration or mTOR inhibition is worth testing in a clinical setting.


Subject(s)
Glioblastoma/enzymology , Glioblastoma/therapy , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction , Animals , Antibodies, Monoclonal/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Glioblastoma/pathology , Hepatocyte Growth Factor/antagonists & inhibitors , Humans , Mice , Neoplasm Invasiveness , Protein Kinases/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases , Xenograft Model Antitumor Assays
18.
Endocrinology ; 150(3): 1536-44, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18974269

ABSTRACT

Color vision is mediated by cone photoreceptors that express opsin photopigments with sensitivities to different light wavelengths. Most mammals, including mice, differentially express M and S opsins for response to medium-long and short wavelengths, respectively. Previous studies demonstrated that a thyroid hormone receptor (TRbeta2) is critical for opsin patterning: in TRbeta2-deficient mice, M opsin is lost and all cones instead express S opsin. Here, to investigate the requirement for thyroid hormone in cone development, we studied Tshr(-/-)mice as a model of congenital hypothyroidism. The onset of M opsin expression in Tshr(-/-)mice was severely delayed until after postnatal d 17 (P17), and M opsin expression failed to attain normal levels at older adult ages. S opsin showed a subtler change with an extended distribution pattern over the superior-inferior axis of the retina. Similar opsin abnormalities were detected in wild-type C57BL/6J mice made hypothyroid by methimazole treatment. In Tshr(-/-) mice, T(3) treatment from P8 recovered significant M opsin expression at P17. Tshr(-/-) mice produced normal numbers of cones, indicating that the major requirement for thyroid hormone is in opsin patterning rather than in cone generation. The phenotype is similar to, although milder than, that caused by loss of TRbeta2 and indicates the necessity for thyroid hormone for cone maturation.


Subject(s)
Body Patterning/genetics , Cone Opsins/genetics , Congenital Hypothyroidism/genetics , Gene Expression Regulation, Developmental , Retinal Cone Photoreceptor Cells/metabolism , Animals , Body Patterning/drug effects , Cone Opsins/metabolism , Congenital Hypothyroidism/chemically induced , Congenital Hypothyroidism/pathology , Female , Gene Expression Regulation, Developmental/drug effects , Methimazole , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Pregnancy , Receptors, Thyrotropin/genetics , Retina/drug effects , Retina/pathology , Retinal Cone Photoreceptor Cells/drug effects , Retinal Cone Photoreceptor Cells/pathology , Time Factors , Triiodothyronine/pharmacology
19.
Cancer Res ; 68(10): 3566-72, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18483236

ABSTRACT

microRNAs are noncoding RNAs inhibiting expression of numerous target genes, and a few have been shown to act as oncogenes or tumor suppressors. We show that microRNA-7 (miR-7) is a potential tumor suppressor in glioblastoma targeting critical cancer pathways. miR-7 potently suppressed epidermal growth factor receptor expression, and furthermore it independently inhibited the Akt pathway via targeting upstream regulators. miR-7 expression was down-regulated in glioblastoma versus surrounding brain, with a mechanism involving impaired processing. Importantly, transfection with miR-7 decreased viability and invasiveness of primary glioblastoma lines. This study establishes miR-7 as a regulator of major cancer pathways and suggests that it has therapeutic potential for glioblastoma.


Subject(s)
Brain Neoplasms/metabolism , Down-Regulation , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , MicroRNAs , Proto-Oncogene Proteins c-akt/metabolism , 3' Untranslated Regions , Cell Line, Tumor , Cell Proliferation , Cell Separation , Flow Cytometry , HeLa Cells , Humans , Transfection
20.
Carcinogenesis ; 29(5): 918-25, 2008 May.
Article in English | MEDLINE | ID: mdl-18359760

ABSTRACT

The Notch pathway plays a key role in the development and is increasingly recognized for its importance in cancer. We demonstrated previously the overexpression of Notch-1 and its ligands in gliomas and showed that their knockdown inhibits glioma cell proliferation and survival. To elucidate the mechanisms downstream of Notch-1 in glioma cells, we performed microarray profiling of glioma cells transfected with Notch-1 small interfering RNA. Notable among downregulated transcripts was the epidermal growth factor receptor (EGFR), known to be overexpressed or amplified in gliomas and prominent in other cancers as well. Further studies confirmed that Notch-1 inhibition decreased EGFR messenger RNA (mRNA) and EGFR protein in glioma and other cell lines. Transfection with Notch-1 increased EGFR expression. Additionally, we found a significant correlation in levels of EGFR and Notch-1 mRNA in primary high-grade human gliomas. Subsequent experiments showed that p53, an activator of the EGFR promoter, is regulated by Notch-1. Experiments with p53-positive and -null cell lines confirmed that p53 partially mediates the effects of Notch-1 on EGFR expression. These results show for the first time that Notch-1 upregulates EGFR expression and also demonstrate Notch-1 regulation of p53 in gliomas. These observations have significant implications for understanding the mechanisms of Notch in cancer and development.


Subject(s)
ErbB Receptors/genetics , Gene Expression Regulation , Glioma/genetics , Receptor, Notch1/physiology , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Biopsy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Genes, Reporter , Genes, p53 , Glioma/pathology , Humans , Luciferases/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptor, Notch1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...