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










Publication year range
1.
Int J Mol Sci ; 22(16)2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34445128

ABSTRACT

The WNT (Wingless/Integrated) signaling pathway is implicated in various stages of glioblastoma, which is an aggressive brain tumor for which therapeutic options are limited. WNT has been recognized as a hallmark of therapeutic challenge due to its context-dependent role and critical function in healthy tissue homeostasis. In this review, we deeply scrutinize the WNT signaling pathway and its involvement in the genesis of glioblastoma as well as its acquired therapy resistance. We also provide an analysis of the WNT pathway in terms of its therapeutic importance in addition to an overview of the current targeted therapies under clinical investigation.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , Wnt Signaling Pathway/genetics , Animals , Humans
2.
Med Image Anal ; 70: 101995, 2021 05.
Article in English | MEDLINE | ID: mdl-33640720

ABSTRACT

In this paper, we propose a novel microscopy image translation method for transforming a bright-field microscopy image into three different fluorescence images to observe the apoptosis, nuclei, and cytoplasm of cells, which visualize dead cells, nuclei of cells, and cytoplasm of cells, respectively. These biomarkers are commonly used in high-content drug screening to analyze drug response. The main contribution of the proposed work is the automatic generation of three fluorescence images from a conventional bright-field image; this can greatly reduce the time-consuming and laborious tissue preparation process and improve throughput of the screening process. Our proposed method uses only a single bright-field image and the corresponding fluorescence images as a set of image pairs for training an end-to-end deep convolutional neural network. By leveraging deep convolutional neural networks with a set of image pairs of bright-field and corresponding fluorescence images, our proposed method can produce synthetic fluorescence images comparable to real fluorescence microscopy images with high accuracy. Our proposed model uses multi-task learning with adversarial losses to generate more accurate and realistic microscopy images. We assess the efficacy of the proposed method using real bright-field and fluorescence microscopy image datasets from patient-driven samples of a glioblastoma, and validate the method's accuracy with various quality metrics including cell number correlation (CNC), peak signal-to-noise ratio (PSNR), structural similarity index measure (SSIM), cell viability correlation (CVC), error maps, and R2 correlation.


Subject(s)
Image Processing, Computer-Assisted , Neural Networks, Computer , Humans , Microscopy, Fluorescence , Signal-To-Noise Ratio
3.
Cancers (Basel) ; 13(3)2021 Jan 20.
Article in English | MEDLINE | ID: mdl-33498427

ABSTRACT

(1) Background: Recent advances in precision oncology research rely on indicating specific genetic alterations associated with treatment sensitivity. Developing ex vivo systems to identify cancer patients who will respond to a specific drug remains important. (2) Methods: cells from 12 patients with glioblastoma were isolated, cultured, and subjected to high-content screening. Multi-parameter analyses assessed the c-Met level, cell viability, apoptosis, cell motility, and migration. A drug repurposing screen and large-scale drug sensitivity screening data across 59 cancer cell lines and patient-derived cells were obtained from 125 glioblastoma samples. (3) Results: High-content analysis of patient-derived cells provided robust and accurate drug responses to c-Met-targeted agents. Only the cells of one glioblastoma patient (PDC6) showed elevated c-Met level and high susceptibility to the c-Met inhibitors. Multi-parameter image analysis also reflected a decreased c-Met expression and reduced cell growth and motility by a c-Met-targeting antibody. In addition, a drug repurposing screen identified Abemaciclib as a distinct CDK4/6 inhibitor with a potent c-Met-inhibitory function. Consistent with this, we present large-scale drug sensitivity screening data showing that the Abemaciclib response correlates with the response to c-Met inhibitors. (4) Conclusions: Our study provides a new insight into high-content screening platforms supporting drug sensitivity prediction and novel therapeutics screening.

4.
ACS Omega ; 5(40): 25798-25809, 2020 Oct 13.
Article in English | MEDLINE | ID: mdl-33073104

ABSTRACT

c-Met, as a receptor expressed on the cell membrane, contributes to the growth and metastasis of tumors, as well as angiogenesis, mainly through the hepatocyte growth factor (HGF)/c-Met axis during tumor progression. Although several c-Met inhibitors, including small molecules and monoclonal antibody inhibitors, are currently being investigated, their clinical outcomes have not been promising. Development of an antibody-drug conjugate (ADC) against c-Met could be an attractive therapeutic strategy that would provide superior antitumor efficacy with broad-spectrum c-Met expression levels. In the present study, site-specific drug-conjugate technology was applied to develop an ADC using the human-mouse cross-reactive c-Met antibody and a prodrug pyrrolobenzodiazepine (PBD). The toxin payload was uniformly conjugated to the light-chain C-terminus of the native cIRCR201 antibody (drug-to-antibody ratio = 2), as confirmed using LC-MS. Using a high-throughput screening system, we found that cIRCR201-dPBD exhibited varying sensitivities depending on the expression levels of c-Met, and it induced receptor-mediated endocytosis and toxin-mediated apoptosis in 47 different cancer cell lines. cIRCR201-dPBD also showed significant antitumor activity on the MET-amplified cancer cells using in vivo xenograft models. Therefore, cIRCR201-dPBD could be a promising therapeutic strategy for tumors with c-Met expression.

5.
Genome Biol ; 21(1): 216, 2020 08 26.
Article in English | MEDLINE | ID: mdl-32847614

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is a complex disease with extensive molecular and transcriptional heterogeneity. GBM can be subcategorized into four distinct subtypes; tumors that shift towards the mesenchymal phenotype upon recurrence are generally associated with treatment resistance, unfavorable prognosis, and the infiltration of pro-tumorigenic macrophages. RESULTS: We explore the transcriptional regulatory networks of mesenchymal-associated tumor-associated macrophages (MA-TAMs), which drive the malignant phenotypic state of GBM, and identify macrophage receptor with collagenous structure (MARCO) as the most highly differentially expressed gene. MARCOhigh TAMs induce a phenotypic shift towards mesenchymal cellular state of glioma stem cells, promoting both invasive and proliferative activities, as well as therapeutic resistance to irradiation. MARCOhigh TAMs also significantly accelerate tumor engraftment and growth in vivo. Moreover, both MA-TAM master regulators and their target genes are significantly correlated with poor clinical outcomes and are often associated with genomic aberrations in neurofibromin 1 (NF1) and phosphoinositide 3-kinases/mammalian target of rapamycin/Akt pathway (PI3K-mTOR-AKT)-related genes. We further demonstrate the origination of MA-TAMs from peripheral blood, as well as their potential association with tumor-induced polarization states and immunosuppressive environments. CONCLUSIONS: Collectively, our study characterizes the global transcriptional profile of TAMs driving mesenchymal GBM pathogenesis, providing potential therapeutic targets for improving the effectiveness of GBM immunotherapy.


Subject(s)
Gene Regulatory Networks , Glioblastoma/genetics , Tumor-Associated Macrophages , Animals , Carcinogenesis , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Glioblastoma/pathology , Glioma/genetics , Humans , Immunotherapy , Macrophages/metabolism , Mice , Neurofibromin 1/genetics , Phenotype , Prognosis , Stem Cells , Transcriptome , Tumor Microenvironment
6.
Nat Commun ; 11(1): 3288, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32620753

ABSTRACT

The prognostic and therapeutic relevance of molecular subtypes for the most aggressive isocitrate dehydrogenase 1/2 (IDH) wild-type glioblastoma (GBM) is currently limited due to high molecular heterogeneity of the tumors that impedes patient stratification. Here, we describe a distinct binary classification of IDH wild-type GBM tumors derived from a quantitative proteomic analysis of 39 IDH wild-type GBMs as well as IDH mutant and low-grade glioma controls. Specifically, GBM proteomic cluster 1 (GPC1) tumors exhibit Warburg-like features, neural stem-cell markers, immune checkpoint ligands, and a poor prognostic biomarker, FKBP prolyl isomerase 9 (FKBP9). Meanwhile, GPC2 tumors show elevated oxidative phosphorylation-related proteins, differentiated oligodendrocyte and astrocyte markers, and a favorable prognostic biomarker, phosphoglycerate dehydrogenase (PHGDH). Integrating these proteomic features with the pharmacological profiles of matched patient-derived cells (PDCs) reveals that the mTORC1/2 dual inhibitor AZD2014 is cytotoxic to the poor prognostic PDCs. Our analyses will guide GBM prognosis and precision treatment strategies.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Isocitrate Dehydrogenase/genetics , Proteogenomics/methods , Proteomics/methods , Benzamides/pharmacology , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Glioblastoma/genetics , Glioblastoma/therapy , Humans , Isocitrate Dehydrogenase/classification , Isocitrate Dehydrogenase/metabolism , Kaplan-Meier Estimate , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/metabolism , Morpholines/pharmacology , Mutation , Prognosis , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology
7.
Cell Cycle ; 19(13): 1590-1601, 2020 07.
Article in English | MEDLINE | ID: mdl-32423265

ABSTRACT

p97 has recently emerged as a therapeutic target for cancer due to its essential functions in protein homeostasis. CB-5083 is a first-in-class, potent and selective ATP-competitive p97 inhibitor that induces proteotoxic stress in cancer cells. Potential mechanisms regulating the sensitivity of cells to p97 inhibition remain poorly studied. Here, we demonstrate that Thrombospondin-1 (THBS1) is a CB-5083-upregulated gene that helps confer resistance of HCT116 cells to CB-5083. Our immunoblotting and immunofluorescence data showed that CB-5083 significantly increases the steady-state abundance of THBS1. Blockade of THBS1 induction sensitized cells to CB-5083-mediated growth inhibition. Suppression of THBS1 caused an increase of CB-5083-induced sub-G1 population and caspase 3/7 activity suggesting that its function is linked to the survival of cancer cells in response to p97 inhibition. Altogether our data provide new evidence that THBS1 is important for the susceptibility of cells to p97 inhibition.


Subject(s)
Colonic Neoplasms/metabolism , Indoles/pharmacology , Pyrimidines/pharmacology , Thrombospondin 1/metabolism , Valosin Containing Protein/antagonists & inhibitors , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Thrombospondin 1/genetics , Up-Regulation/drug effects , Up-Regulation/genetics , Valosin Containing Protein/metabolism
8.
Sci Rep ; 10(1): 8096, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32415084

ABSTRACT

Abnormal regulation of ß-catenin initiates an oncogenic program that serves as a main driver of many cancers. Albeit challenging, ß-catenin is an attractive drug target due to its role in maintenance of cancer stem cells and potential to eliminate cancer relapse. We have identified C2, a novel ß-catenin inhibitor, which is a small molecule that binds to a novel allosteric site on the surface of ß-catenin. C2 selectively inhibits ß-catenin, lowers its cellular load and significantly reduces viability of ß-catenin-driven cancer cells. Through direct binding to ß-catenin, C2 renders the target inactive that eventually activates proteasome system for its removal. Here we report a novel pharmacologic approach for selective inhibition of ß-catenin via targeting a cryptic allosteric modulation site. Our findings may provide a new perspective for therapeutic targeting of ß-catenin.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Small Molecule Libraries/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/antagonists & inhibitors , Allosteric Regulation , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Apoptosis , Cell Proliferation , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Small Molecule Libraries/chemistry , Small Molecule Libraries/isolation & purification , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Genome Med ; 12(1): 17, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32070411

ABSTRACT

BACKGROUND: Gastric cancer is among the most lethal human malignancies. Previous studies have identified molecular aberrations that constitute dynamic biological networks and genomic complexities of gastric tumors. However, the clinical translation of molecular-guided targeted therapy is hampered by challenges. Notably, solid tumors often harbor multiple genetic alterations, complicating the development of effective treatments. METHODS: To address such challenges, we established a comprehensive dataset of molecularly annotated patient derivatives coupled with pharmacological profiles for 60 targeted agents to explore dynamic pharmacogenomic interactions in gastric cancers. RESULTS: We identified lineage-specific drug sensitivities based on histopathological and molecular subclassification, including substantial sensitivities toward VEGFR and EGFR inhibition therapies in diffuse- and signet ring-type gastric tumors, respectively. We identified potential therapeutic opportunities for WNT pathway inhibitors in ALK-mutant tumors, a significant association between PIK3CA-E542K mutation and AZD5363 response, and transcriptome expression of RNF11 as a potential predictor of response to gefitinib. CONCLUSIONS: Collectively, our results demonstrate the feasibility of drug screening combined with tumor molecular characterization to facilitate personalized therapeutic regimens for gastric tumors.


Subject(s)
Drug Resistance, Neoplasm , Pharmacogenomic Variants , Stomach Neoplasms/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Gefitinib/pharmacology , Gefitinib/therapeutic use , Humans , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrroles/pharmacology , Pyrroles/therapeutic use , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/genetics , Stomach Neoplasms/drug therapy , Transcriptome , Tumor Cells, Cultured , Wnt Signaling Pathway/drug effects
10.
Biotechniques ; 68(4): 180-184, 2020 04.
Article in English | MEDLINE | ID: mdl-32040335

ABSTRACT

CRISPR-Cas9 has proven to be a versatile tool for the discovery of essential genetic elements involved in various disease states. CRISPR-assisted dense mutagenesis focused on therapeutically challenging protein complexes allows us to systematically perturb protein-coding sequences in situ and correlate them with functional readouts. Such perturbations can mimic targeting by therapeutics and serve as a foundation for the discovery of highly specific modulators. However, translation of such genomics data has been challenging due to the missing link for proteomics under the physiological state of the cell. We present a method based on cellular thermal shift assays to easily interrogate proteomic shifts generated by CRISPR-assisted dense mutagenesis, as well as a case focused on NuRD epigenetic complex.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing/methods , Mutagenesis, Insertional/genetics , Proteome/genetics , Proteomics/methods , Cell Line , Humans , Proteome/analysis
11.
Cancers (Basel) ; 11(12)2019 Nov 22.
Article in English | MEDLINE | ID: mdl-31771104

ABSTRACT

Glioblastoma is a highly aggressive and lethal brain tumor, with limited treatment options. Abnormal activation of the neddylation pathway is observed in glioblastoma, and the NEDD8-activating enzyme (NAE) inhibitor, MLN4924, was previously shown to be effective in glioblastoma cell line models. However, its effect has not been tested in patient-derived glioblastoma stem cells. We first analyzed public data to determine whether NEDD8 pathway proteins are important in glioblastoma development and patient survival. NAE1 and UBA3 levels increased in glioblastoma patients; high NEDD8 levels were associated with poor clinical outcomes. Immunohistochemistry results also supported this result. The effects of MLN4924 were evaluated in 4 glioblastoma cell lines and 15 patient-derived glioblastoma stem cells using high content analysis. Glioblastoma cell lines and patient-derived stem cells were highly susceptible to MLN4924, while normal human astrocytes were resistant. In addition, there were various responses in 15 patient-derived glioblastoma stem cells upon MLN4924 treatment. Genomic analyses indicated that MLN4924 sensitive cells exhibited enrichment of Extracellular Signal Regulated Kinase (ERK) and Protein kinase B (AKT, also known as PKB) signaling. We verified that MLN4924 inhibits ERK and AKT phosphorylation in MLN4924 sensitive cells. Our findings suggest that patient-derived glioblastoma stem cells in the context of ERK and AKT activation are sensitive and highly regulated by neddylation inhibition.

12.
Genome Biol ; 20(1): 253, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31771620

ABSTRACT

BACKGROUND: Gynecologic malignancy is one of the leading causes of mortality in female adults worldwide. Comprehensive genomic analysis has revealed a list of molecular aberrations that are essential to tumorigenesis, progression, and metastasis of gynecologic tumors. However, targeting such alterations has frequently led to treatment failures due to underlying genomic complexity and simultaneous activation of various tumor cell survival pathway molecules. A compilation of molecular characterization of tumors with pharmacological drug response is the next step toward clinical application of patient-tailored treatment regimens. RESULTS: Toward this goal, we establish a library of 139 gynecologic tumors including epithelial ovarian cancers (EOCs), cervical, endometrial tumors, and uterine sarcomas that are genomically and/or pharmacologically annotated and explore dynamic pharmacogenomic associations against 37 molecularly targeted drugs. We discover lineage-specific drug sensitivities based on subcategorization of gynecologic tumors and identify TP53 mutation as a molecular determinant that elicits therapeutic response to poly (ADP-Ribose) polymerase (PARP) inhibitor. We further identify transcriptome expression of inhibitor of DNA biding 2 (ID2) as a potential predictive biomarker for treatment response to olaparib. CONCLUSIONS: Together, our results demonstrate the potential utility of rapid drug screening combined with genomic profiling for precision treatment of gynecologic cancers.


Subject(s)
Genital Neoplasms, Female/genetics , Pharmacogenomic Testing , Precision Medicine , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor , Female , Genital Neoplasms, Female/drug therapy , Humans
13.
Micron ; 126: 102718, 2019 11.
Article in English | MEDLINE | ID: mdl-31473399

ABSTRACT

The morphology of tumor cells is highly related to their phenotype and activity. To verify the drug response of a brain tumor patient, fluorescence microscope images of drug-treated patient-derived cells in each well are analyzed. Due to the limitation of the field of view (FOV), a large number of small FOVs are acquired to compose one complete microscope well. Here, we propose an automated method for accurately stitching tile-scanned fluorescence microscope images, even with noise and a narrow overlapping region between adjacent fields. The proposed method is based on intensity-based normalized cross-correlation (NCC) and a triangular method-based threshold. The proposed method's quantitative accuracy and the sensitivity of the input was compared to other existing stitching tools, MIST and FijiIS, setting manually stitched images as the ground truth. The test images were 20 samples of 3 × 3 grid images in three versions of the fluorescence channel. The distance between the location of each field and number of cells was determined for different input field overlap ranges (1%, 3%, 5%, and 10%), while the actual value was about 1.15%. The proposed method had a distance error of 1.5 pixels at an input overlap of 1%, showing the lowest minimum error at all channels. Regarding the difference in cell numbers, although the number of overlapping cells was always small because of the narrow overlapping range, the proposed method was able to generate the resultant image with the smallest difference. In addition, to confirm the size limitation of the proposed algorithm, the accuracy of stitching images of grid structures 3 × 3, 5 × 5, 10 × 10-20 × 20 was tested, showing consistent results. In conclusion, quantitative evaluation of the performance of the method proved its improved accuracy compared to other current state-of-art techniques, and it showed robust performance even with noise and a narrow overlapping region between adjacent fields.


Subject(s)
Automation , Brain/cytology , Image Processing, Computer-Assisted/methods , Microscopy, Fluorescence , Tumor Cells, Cultured/ultrastructure , Adult , Aged , Brain/pathology , Brain/surgery , Female , Glioblastoma , High-Throughput Screening Assays , Humans , Male , Middle Aged
14.
Cell ; 175(6): 1665-1678.e18, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30343896

ABSTRACT

Low-grade gliomas almost invariably progress into secondary glioblastoma (sGBM) with limited therapeutic option and poorly understood mechanism. By studying the mutational landscape of 188 sGBMs, we find significant enrichment of TP53 mutations, somatic hypermutation, MET-exon-14-skipping (METex14), PTPRZ1-MET (ZM) fusions, and MET amplification. Strikingly, METex14 frequently co-occurs with ZM fusion and is present in ∼14% of cases with significantly worse prognosis. Subsequent studies show that METex14 promotes glioma progression by prolonging MET activity. Furthermore, we describe a MET kinase inhibitor, PLB-1001, that demonstrates remarkable potency in selectively inhibiting MET-altered tumor cells in preclinical models. Importantly, this compound also shows blood-brain barrier permeability and is subsequently applied in a phase I clinical trial that enrolls MET-altered chemo-resistant glioma patients. Encouragingly, PLB-1001 achieves partial response in at least two advanced sGBM patients with rarely significant side effects, underscoring the clinical potential for precisely treating gliomas using this therapy.


Subject(s)
Brain Neoplasms , Exons , Glioblastoma , Mutation , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-met , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Drug Delivery Systems , Female , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
15.
Cell Death Dis ; 9(8): 792, 2018 07 18.
Article in English | MEDLINE | ID: mdl-30022047

ABSTRACT

Testing new ways to identify untapped opportunities for glioblastoma therapies remains highly significant. Amplification and overexpression of MDM2 gene is frequent in glioblastoma and disrupting the MDM2-p53 interaction is a promising strategy to treat the cancer. RG7112 is the first-in class inhibitor and recently discovered AMG232 is the most potent MDM2 inhibitor known to date. Here, we compared the effects of these two clinical MDM2 inhibitors in six glioblastoma cell lines and ten patient-derived glioblastoma stem cells. Targeted sequencing of the TP53, MDM2 genes and whole transcriptome analysis were conducted to verify genetic status associated with sensitivity and resistance to the drugs. Although TP53 wild-type glioblastoma cell lines are similarly sensitive to AMG232 and RG7112, we found that four TP53 wild-type out of ten patient-derived glioblastoma cells are much more sensitive to AMG232 than RG7112 (average IC50 of 76 nM vs. 720 nM). Among these, 464T stem cells containing MDM2 gene amplification were most sensitive to AMG232 with IC50 of 5.3 nM. Moreover, AMG232 exhibited higher selectivity against p53 wild-type cells over p53 mutant stem cells compared to RG7112 (average selectivity of 512-fold vs. 16.5-fold). Importantly, we also found that AMG232 is highly efficacious in three-dimensional (3D) tumor spheroids growth and effectively inhibits the stemness-related factors, Nestin and ZEB1. Our data provide new evidence that glioblastoma stem cells have high susceptibility to AMG232 suggesting the potential clinical implications of MDM2 inhibition for glioblastoma treatment. These will facilitate additional preclinical and clinical studies evaluating MDM2 inhibitors in glioblastoma and direct further efforts towards developing better MDM2-targeted therapeutics.


Subject(s)
Acetates/pharmacology , Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Piperidones/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Acetates/chemistry , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Imidazolines/chemistry , Imidazolines/pharmacology , Mutation , Nestin/metabolism , Piperidones/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism
16.
Cell Death Dis ; 8(3): e2693, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28333152

ABSTRACT

Radiotherapy represents the most effective non-surgical modality in cancer treatment. MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression, and are involved in many biological processes and diseases. To identify miRNAs that influence the radiation response, we performed miRNA array analysis using MCF7 cells at 2, 8, and 24 h post irradiation. We demonstrated that miR-770-5p is a novel radiation-inducible miRNA. When miR-770-5p was overexpressed, relative cell number was reduced due to increased apoptosis in MCF7 and A549 cells. Transcriptomic and bioinformatic analyses revealed that PDZ-binding kinase (PBK) might be a possible target of miR-770-5p for regulation of radiosensitivity. PBK regulation mediated by direct targeting of miR-770-5p was demonstrated using luciferase reporter assays along with wild-type and mutant PBK-3'untranslated region constructs. Radiation sensitivity increased and decreased in miR-770-5p- and anti-miR-770-5p-transfected cells, respectively. Consistent with this result, transfection of short interfering RNA against PBK inhibited cell proliferation, while ectopic expression of PBK restored cell survival from miR-770-5p-induced cell death. In addition, miR-770-5p suppressed tumor growth, and miR-770-5p and PBK levels were inversely correlated in xenograft model mice. Altogether, these data demonstrated that miR-770-5p might be a useful therapeutic target miRNA that sensitizes tumors to radiation via negative regulation of PBK.


Subject(s)
MicroRNAs/genetics , Mitogen-Activated Protein Kinase Kinases/genetics , Radiation Tolerance/genetics , 3' Untranslated Regions/genetics , A549 Cells , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Gene Expression Regulation, Neoplastic/genetics , HCT116 Cells , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Processing, Post-Translational/genetics , Transcriptional Activation/genetics
17.
Cell Chem Biol ; 23(4): 517-28, 2016 04 21.
Article in English | MEDLINE | ID: mdl-27105284

ABSTRACT

The AAA ATPase p97/VCP regulates protein homeostasis using a diverse repertoire of cofactors to fulfill its biological functions. Here we use the allosteric p97 inhibitor NMS-873 to analyze its effects on enzyme composition and the ability of cells to adapt to its cytotoxicity. We found that p97 inhibition changes steady state cofactor-p97 composition, leading to the enrichment of a subset of its cofactors and polyubiquitin bound to p97. We isolated cells specifically insensitive to NMS-873 and identified a new mutation (A530T) in p97. A530T is sufficient to overcome the cytotoxicity of NMS-873 and alleviates p97 composition changes caused by the molecule but not other p97 inhibitors. This mutation does not affect NMS-873 binding but increases p97 catalytic efficiency through altered ATP and ADP binding. Collectively, these findings identify cofactor-p97 interactions sensitive to p97 inhibition and reveal a new on-target mechanism to suppress the cytotoxicity of NMS-873.


Subject(s)
Acetanilides/pharmacology , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Benzothiazoles/pharmacology , Enzyme Inhibitors/pharmacology , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Small Molecule Libraries/pharmacology , Acetanilides/chemistry , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/genetics , Allosteric Regulation/drug effects , Benzothiazoles/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , HCT116 Cells , Humans , Models, Molecular , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Small Molecule Libraries/chemistry , Structure-Activity Relationship
18.
Proc Natl Acad Sci U S A ; 111(43): 15532-7, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25313037

ABSTRACT

X-linked inhibitor of apoptosis (XIAP)-associated factor 1 (XAF1) is a tumor suppressor that is frequently inactivated in many human cancers. However, the molecular mechanism underlying its growth-inhibitory function remains largely unknown. Here, we report that XAF1 forms a positive feedback loop with p53 and acts as a molecular switch in p53-mediated cell-fate decisions favoring apoptosis over cell-cycle arrest. XAF1 binds directly to the N-terminal proline-rich domain of p53 and thus interferes with E3 ubiquitin ligase MDM2 binding and ubiquitination of p53. XAF1 stimulates homeodomain-interacting protein kinase 2 (HIPK2)-mediated Ser-46 phosphorylation of p53 by blocking E3 ubiquitin ligase Siah2 interaction with and ubiquitination of HIPK2. XAF1 also steps up the termination of p53-mediated cell-cycle arrest by activating zinc finger protein 313 (ZNF313), a p21(WAF1)-targeting ubiquitin E3 ligase. XAF1 interacts with p53, Siah2, and ZNF313 through the zinc finger domains 5, 6, and 7, respectively, and truncated XAF1 isoforms preferentially expressed in cancer cells fail to form a feedback loop with p53. Together, this study uncovers a novel role for XAF1 in p53 stress response, adding a new layer of complexity to the mechanisms by which p53 determines cell-fate decisions.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Adaptor Proteins, Signal Transducing , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Enzyme Activation/drug effects , Feedback, Physiological/drug effects , HCT116 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Models, Biological , Neoplasm Proteins/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , Protein Isoforms/metabolism , Protein Stability/drug effects , Protein Structure, Tertiary , Proteolysis/drug effects , Proto-Oncogene Proteins c-mdm2/metabolism , Remission Induction , Signal Transduction/drug effects , Tumor Suppressor Protein p53/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects
19.
Cell Cycle ; 12(10): 1521-35, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23598720

ABSTRACT

TGF-ß1 plays biphasic functions in prostate tumorigenesis, inhibiting cell growth at early stages but promoting malignant progression at later stages. However, the molecular basis for the oncogenic conversion of TGF-ß1 function remains largely undefined. Here, we demonstrate that PPARδ is a direct transcription target of TGF-ß1 and plays a critical role in oncogenic redirection of TGF-ß1 signaling. Blockade of PPARδ induction enhances tumor cell response to TGF-ß1-mediated growth inhibition, while its activation promotes TGF-ß1-induced tumor growth, migration and invasion. PPARδ-mediated switch of TGF-ß1 function is associated with down- and upregulation of Smad and ERK signaling, respectively, and tightly linked to its function to activate ABCA1 cholesterol transporter followed by caveolin-1 (Cav1) induction. Intriguingly, TGF-ß1 activation of the PPARδ-ABCA1-Cav1 pathway facilitates degradation of TGF-ß receptors (TßRs) and attenuates Smad but enhances ERK response to TGF-ß1. Expression of PPARδ and Cav1 is tightly correlated in both prostate tissues and cell lines and significantly higher in cancer vs. normal tissues. Collectively, our study shows that PPARδ is a transcription target of TGF-ß1 and contributes to the oncogenic conversion of TGF-ß1 function through activation of the ABCA1-Cav1-TßR signaling axis.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Caveolin 1/metabolism , PPAR delta/metabolism , Transforming Growth Factor beta1/metabolism , ATP Binding Cassette Transporter 1/antagonists & inhibitors , ATP Binding Cassette Transporter 1/genetics , Animals , Caveolin 1/antagonists & inhibitors , Caveolin 1/genetics , Cell Line, Tumor , Cell Movement , Humans , Male , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , PPAR delta/antagonists & inhibitors , PPAR delta/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Smad Proteins/antagonists & inhibitors , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta1/pharmacology , Transplantation, Heterologous
20.
Cancer Res ; 72(16): 4097-109, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22706202

ABSTRACT

Caveolin-1 (CAV1) acts as a growth suppressor in various human malignancies, but its expression is elevated in many advanced cancers, suggesting the oncogenic switch of its role during tumor progression. To understand the molecular basis for the growth-promoting function of CAV1, we characterized its expression status, differential roles for tumor growth, and effect on glucose metabolism in colorectal cancers. Abnormal elevation of CAV1 was detected in a substantial fraction of primary tumors and cell lines and tightly correlated with promoter CpG sites hypomethylation. Depletion of elevated CAV1 led to AMPK activation followed by a p53-dependent G1 cell-cycle arrest and autophagy, suggesting that elevated CAV1 may contribute to ATP generation. Furthermore, CAV1 depletion downregulated glucose uptake, lactate accumulation, and intracellular ATP level, supporting that aerobic glycolysis is enhanced by CAV1. Consistently, CAV1 was shown to stimulate GLUT3 transcription via an HMGA1-binding site within the GLUT3 promoter. HMGA1 was found to interact with and activate the GLUT3 promoter and CAV1 increased the HMGA1 activity by enhancing its nuclear localization. Ectopic expression of HMGA1 increased glucose uptake, whereas its knockdown caused AMPK activation. In addition, GLUT3 expression was strongly induced by cotransfection of CAV1 and HMGA1, and its overexpression was observed predominantly in tumors harboring high levels of CAV1 and HMGA1. Together, these data show that elevated CAV1 upregulates glucose uptake and ATP production through HMGA1-mediated GLUT3 transcription, suggesting that CAV1 may render tumor cells growth advantages by enhancing aerobic glycolysis.


Subject(s)
Caveolin 1/biosynthesis , Colorectal Neoplasms/metabolism , Glucose Transporter Type 3/biosynthesis , HMGA1a Protein/metabolism , Adenosine Triphosphate/metabolism , Adenylate Kinase/metabolism , Aerobiosis , Animals , Autophagy/physiology , Caveolin 1/deficiency , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Line, Tumor , Colorectal Neoplasms/genetics , DNA Methylation , Female , Glucose/metabolism , Glucose Transporter Type 3/genetics , Glycolysis , HCT116 Cells , Humans , Immunoblotting , Mice , Mice, Nude , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...