Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 61(4): 520-534, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26853146

ABSTRACT

Altered energy metabolism is a cancer hallmark as malignant cells tailor their metabolic pathways to meet their energy requirements. Glucose and glutamine are the major nutrients that fuel cellular metabolism, and the pathways utilizing these nutrients are often altered in cancer. Here, we show that the long ncRNA CCAT2, located at the 8q24 amplicon on cancer risk-associated rs6983267 SNP, regulates cancer metabolism in vitro and in vivo in an allele-specific manner by binding the Cleavage Factor I (CFIm) complex with distinct affinities for the two subunits (CFIm25 and CFIm68). The CCAT2 interaction with the CFIm complex fine-tunes the alternative splicing of Glutaminase (GLS) by selecting the poly(A) site in intron 14 of the precursor mRNA. These findings uncover a complex, allele-specific regulatory mechanism of cancer metabolism orchestrated by the two alleles of a long ncRNA.


Subject(s)
Glutaminase/genetics , Neoplasms/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Alleles , Alternative Splicing , Energy Metabolism , HCT116 Cells , Humans , Neoplasms/genetics , RNA Precursors/chemistry , RNA Precursors/metabolism , RNA, Messenger/metabolism
2.
Breast Cancer Res Treat ; 189(2): 333-345, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34241740

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype that lacks targeted therapies. Patients with TNBC have a very poor prognosis because the disease often metastasizes. New treatment approaches addressing drivers of metastasis and tumor growth are crucial to improving patient outcomes. Developing targeted gene therapy is thus a high priority for TNBC patients. PEA15 (phosphoprotein enriched in astrocytes, 15 kDa) is known to bind to ERK, preventing ERK from being translocated to the nucleus and hence blocking its activity. The biological function of PEA15 is tightly regulated by its phosphorylation at Ser104 and Ser116. However, the function and impact of phosphorylation status of PEA15 in the regulation of TNBC metastasis and in epithelial-to-mesenchymal transition (EMT) are not well understood. METHODS: We established stable cell lines overexpressing nonphosphorylatable (PEA15-AA) and phospho-mimetic (PEA15-DD) mutants. To dissect specific cellular mechanisms regulated by PEA15 phosphorylation status, we performed RT-PCR immune and metastasis arrays. In vivo mouse models were used to determine the effects of PEA15 phosphorylation on tumor growth and metastasis. RESULTS: We found that the nonphosphorylatable mutant PEA15-AA prevented formation of mammospheres and expression of EMT markers in vitro and decreased tumor growth and lung metastasis in in vivo experiments when compared to control, PEA15-WT and phosphomimetic PEA15-DD. However, phosphomimetic mutant PEA15-DD promoted migration, mesenchymal marker expression, tumorigenesis, and lung metastasis in the mouse model. PEA15-AA-mediated inhibition of breast cancer cell migratory capacity and tumorigenesis was the partial result of decreased expression of interleukin-8 (IL-8). Further, we identified that expression of IL-8 was possibly mediated through one of the ERK downstream molecules, Ets-1. CONCLUSIONS: Our results show that PEA15 phosphorylation status serves as an important regulator for PEA15's dual role as an oncogene or tumor suppressor and support the potential of PEA15-AA as a therapeutic strategy for treatment of TNBC.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Epithelial-Mesenchymal Transition , Triple Negative Breast Neoplasms , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Interleukin-8 , Mice , Triple Negative Breast Neoplasms/genetics
3.
Cancer ; 126(15): 3579-3592, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32484926

ABSTRACT

BACKGROUND: Poor outcomes for patients with ovarian cancer relate to dormant, drug-resistant cancer cells that survive after primary surgery and chemotherapy. Ovarian cancer (OvCa) cells persist in poorly vascularized scars on the peritoneal surface and depend on autophagy to survive nutrient deprivation. The authors have sought drugs that target autophagic cancer cells selectively to eliminate residual disease. METHODS: By using unbiased small-interfering RNA (siRNA) screens, the authors observed that knockdown of anaplastic lymphoma kinase (ALK) reduced the survival of autophagic OvCa cells. Small-molecule ALK inhibitors were evaluated for their selective toxicity against autophagic OvCa cell lines and xenografts. Autophagy was induced by reexpression of GTP-binding protein Di-Ras3 (DIRAS3) or serum starvation and was evaluated with Western blot analysis, fluorescence imaging, and transmission electron microscopy. Signaling pathways required for crizotinib-induced apoptosis of autophagic cells were explored with flow cytometric analysis, Western blot analysis, short-hairpin RNA knockdown of autophagic proteins, and small-molecule inhibitors of STAT3 and BCL-2. RESULTS: Induction of autophagy by reexpression of DIRAS3 or serum starvation in multiple OvCa cell lines significantly reduced the 50% inhibitory concentration of crizotinib and other ALK inhibitors. In 2 human OvCa xenograft models, the DIRAS3-expressing tumors treated with crizotinib had significantly decreased tumor burden and long-term survival in 67% to 79% of mice. Crizotinib treatment of autophagic cancer cells further enhanced autophagy and induced autophagy-mediated apoptosis by decreasing phosphorylated STAT3 and BCL-2 signaling. CONCLUSIONS: Crizotinib may eliminate dormant, autophagic, drug-resistant OvCa cells that remain after conventional cytoreductive surgery and combination chemotherapy. A clinical trial of ALK inhibitors as maintenance therapy after second-look operations should be seriously considered.


Subject(s)
Anaplastic Lymphoma Kinase/genetics , Ovarian Neoplasms/drug therapy , STAT3 Transcription Factor/genetics , rho GTP-Binding Proteins/genetics , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Animals , Autophagy/drug effects , Cell Line, Tumor , Cell Lineage/genetics , Cell Survival/genetics , Crizotinib/pharmacology , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Mice , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects
5.
Bioorg Med Chem Lett ; 27(24): 5436-5440, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29138030

ABSTRACT

This report details a search for alternative strains that produce the diterpenoid sphaeropsidin A (SphA) among A. candidus strains from the USDA Northern Regional Research Laboratories Culture Collection. We identified two strains that produced SphA using a limited set of test media. An initial scaled-up fermentation of NRRL 313 and isolation effort led to the procurement of sufficient quantities of SphA to prepare five semi-synthetic analogues (1-5) and evaluate their anticancer effects against glioblastoma cells D423 and Gli56 grown in 2D and 3D cultures. Although, the effectiveness of the synthetic analogues varied depending on the cell line and the type of cell culture, compound 5, bearing an aromatic ring at C16, displayed a stronger toxicity towards both D423 and Gli56 cell lines in 2D cultures and D423 spheroids in 3D culture than either SphA or compounds 1-4.


Subject(s)
Antineoplastic Agents, Phytogenic/chemical synthesis , Aspergillus/chemistry , Diterpenes/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , Aspergillus/metabolism , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Diterpenes/isolation & purification , Diterpenes/pharmacology , Humans
6.
J Nat Prod ; 80(3): 707-712, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28128947

ABSTRACT

Studies of the genome-sequenced, flutimide-producing coprophilous fungus Delitschia confertaspora (ATCC 74209), originally obtained from a sample of rock hyrax (Procavia capensis) dung collected in Namibia, led to the discovery of three new highly aromatic natural products named delicoferones A-B (1-2) and fimetarone B (3). The new benzophenone derivatives 1 and 2 have a somewhat unusual skeleton that incorporates three aromatic rings linked via two ketone carbonyl groups, while 3 contains a spiro[chroman-3,7'-isochromene]-4,6'(8'H) skeleton reported only once previously. The structures of these compounds were assigned mainly by analysis of 2D NMR and HRESITOFMS data.


Subject(s)
Benzophenones/isolation & purification , Fungi/chemistry , Spiro Compounds/isolation & purification , Animals , Benzophenones/chemistry , Hyraxes , Molecular Structure , Namibia , Nuclear Magnetic Resonance, Biomolecular , Spiro Compounds/chemistry
7.
J Nat Prod ; 80(7): 2101-2109, 2017 07 28.
Article in English | MEDLINE | ID: mdl-28657331

ABSTRACT

Auxarthrones A-E (1-5), five new phenalenones, and two new naturally occurring cyclic tetrapeptides, auxarthrides A (7) and B (8), were obtained from three different solvent extracts of cultures of the coprophilous fungus Auxarthron pseudauxarthron. Auxarthrones C (3) and E (5) possess an unusual 7a,8-dihydrocyclopenta[a]phenalene-7,9-dione ring system that has not been previously observed in natural products. Formation of 1-5 was found to be dependent on the solvent used for culture extraction. The structures of these new compounds were elucidated primarily by analysis of NMR and MS data. Auxarthrone A (1) was obtained as a mixture of chromatographically inseparable racemic diastereomers (1a and 1b) that cocrystallized, enabling confirmation of their structures by X-ray crystallography. The absolute configurations of 7 and 8 were assigned by analysis of their acid hydrolysates using Marfey's method. Compound 1 displayed moderate antifungal activity against Cryptococcus neoformans and Candida albicans, but did not affect human cancer cell lines.


Subject(s)
Antifungal Agents/isolation & purification , Antifungal Agents/pharmacology , Ascomycota/chemistry , Cryptococcus neoformans/drug effects , Peptides, Cyclic/isolation & purification , Peptides, Cyclic/pharmacology , Phenalenes/isolation & purification , Phenalenes/pharmacology , Antifungal Agents/chemistry , Breast Neoplasms/drug therapy , Candida albicans/drug effects , Crystallography, X-Ray , Female , Humans , Microbial Sensitivity Tests , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Peptides, Cyclic/chemistry , Phenalenes/chemistry
8.
Blood ; 117(11): 3151-62, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21248063

ABSTRACT

Although chronic myelogenous leukemia (CML) is effectively controlled by Bcr-Abl kinase inhibitors, resistance to inhibitors, progressive disease, and incomplete eradication of Bcr-Abl-expressing cells are concerns for the long-term control and suppression of this disease. We describe a novel approach to targeting key proteins in CML cells with a ubiquitin-cycle inhibitor, WP1130. Bcr-Abl is rapidly modified with K63-linked ubiquitin polymers in WP1130-treated CML cells, resulting in its accumulation in aggresomes, where is it unable to conduct signal transduction. Induction of apoptosis because of aggresomal compartmentalization of Bcr-Abl was observed in both imatinib-sensitive and -resistant cells. WP1130, but not Bcr-Abl kinase inhibitors, directly inhibits Usp9x deubiquitinase activity, resulting in the down-regulation of the prosurvival protein Mcl-1 and facilitating apoptosis. These results demonstrate that ubiquitin-cycle inhibition represents a novel and effective approach to blocking Bcr-Abl kinase signaling and reducing Mcl-1 levels to engage CML cell apoptosis. This approach may be a therapeutic option for kinase inhibitor-resistant CML patients.


Subject(s)
Apoptosis , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Signal Transduction , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitination , Apoptosis/drug effects , Benzamides , Cell Line, Tumor , Cyanoacrylates , Drug Resistance, Neoplasm/drug effects , Endopeptidases/metabolism , Fusion Proteins, bcr-abl/antagonists & inhibitors , Humans , Imatinib Mesylate , Models, Biological , Nitriles/pharmacology , Phosphorylation/drug effects , Piperazines/pharmacology , Protein Transport/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Substrate Specificity/drug effects , Ubiquitin Thiolesterase/metabolism , Ubiquitination/drug effects
9.
Cancer Cell ; 7(6): 575-89, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15950906

ABSTRACT

Epidermal growth factor receptor (EGFR) exists in the nucleus of highly proliferative cells where it functions as a transcription factor. Although EGFR has transactivational activity, it lacks a DNA binding domain and, therefore, may require a DNA binding transcription cofactor for its transcriptional function. Here, we report that EGFR physically interacts with signal transducers and activators of transcription 3 (STAT3) in the nucleus, leading to transcriptional activation of inducible nitric oxide synthase (iNOS). In breast carcinomas, nuclear EGFR positively correlates with iNOS. This study describes a mode of transcriptional control involving cooperated efforts of STAT3 and nuclear EGFR. Our work suggests that the deregulated iNOS/NO pathway may partly contribute to the malignant biology of tumor cells with high levels of nuclear EGFR and STAT3.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , ErbB Receptors/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/physiology , Trans-Activators/metabolism , Animals , Base Sequence , Binding Sites/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CHO Cells , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Cell Survival/drug effects , Chromatin Immunoprecipitation , Cricetinae , Cricetulus , Drug Synergism , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , Female , Gene Expression/drug effects , Gene Expression Regulation, Neoplastic , Genes, bcl-1/genetics , Genes, fos/genetics , HeLa Cells , Humans , Janus Kinase 2 , Microscopy, Fluorescence , Microscopy, Immunoelectron , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Phosphorylation/drug effects , Prognosis , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , S-Nitroso-N-Acetylpenicillamine/pharmacology , STAT3 Transcription Factor , Signal Transduction/drug effects , Survival Analysis
10.
Cancer Res Commun ; 3(6): 1078-1092, 2023 06.
Article in English | MEDLINE | ID: mdl-37377604

ABSTRACT

Triple-negative breast cancer (TNBC) has high relapse and metastasis rates and a high proportion of cancer stem-like cells (CSC), which possess self-renewal and tumor initiation capacity. MELK (maternal embryonic leucine zipper kinase), a protein kinase of the Snf1/AMPK kinase family, is known to promote CSC maintenance and malignant transformation. However, the role of MELK in TNBC metastasis is unknown; we sought to address this in the current study. We found that MELK mRNA levels were higher in TNBC tumors [8.11 (3.79-10.95)] than in HR+HER2- tumors [6.54 (2.90-9.26)]; P < 0.001]. In univariate analysis, patients with breast cancer with high-MELK-expressing tumors had worse overall survival (P < 0.001) and distant metastasis-free survival (P < 0.01) than patients with low-MELK-expressing tumors. In a multicovariate Cox regression model, high MELK expression was associated with shorter overall survival after adjusting for other baseline risk factors. MELK knockdown using siRNA or MELK inhibition using the MELK inhibitor MELK-In-17 significantly reduced invasiveness, reversed epithelial-to-mesenchymal transition, and reduced CSC self-renewal and maintenance in TNBC cells. Nude mice injected with CRISPR MELK-knockout MDA-MB-231 cells exhibited suppression of lung metastasis and improved overall survival compared with mice injected with control cells (P < 0.05). Furthermore, MELK-In-17 suppressed 4T1 tumor growth in syngeneic BALB/c mice (P < 0.001). Our findings indicate that MELK supports metastasis by promoting epithelial-to-mesenchymal transition and the CSC phenotype in TNBC. Significance: These findings indicate that MELK is a driver of aggressiveness and metastasis in TNBC.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Animals , Mice , Triple Negative Breast Neoplasms/genetics , Mice, Nude , Leucine Zippers , Cell Proliferation/physiology , Neoplasm Recurrence, Local , Protein Serine-Threonine Kinases/genetics
11.
Cancer Cell ; 6(3): 251-61, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15380516

ABSTRACT

Pathological expression of human ErbB-2 protein, also known as HER-2, is common in many types of cancer. ErbB-2 is a member of the EGF receptor tyrosine kinase family and has been rigorously studied as a signaling molecule on the cell membrane. Here, we report that ErbB-2 is also expressed in the nucleus in cultured cells as well as primary tumor tissues. Nuclear ErbB-2 was found to associate with multiple genomic targets in vivo, including the cyclooxygenase enzyme COX-2 gene promoter. ErbB-2 forms a complex at a specific nucleotide sequence of the COX-2 promoter and is able to stimulate its transcription. This study demonstrates the presence of ErbB-2 in the nucleus and identifies the function of ErbB-2 as a transcriptional regulator.


Subject(s)
Isoenzymes/genetics , Prostaglandin-Endoperoxide Synthases/genetics , Receptor, ErbB-2/physiology , Transcriptional Activation , Base Sequence , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cell Nucleus/metabolism , Cyclooxygenase 2 , DNA-Binding Proteins/metabolism , Humans , Membrane Proteins , Promoter Regions, Genetic , Tumor Cells, Cultured
12.
Cancer Drug Resist ; 4(4): 888-902, 2021.
Article in English | MEDLINE | ID: mdl-34888496

ABSTRACT

AIM: Multiple myeloma (MM) is a hematological malignancy of antibody-producing mature B cells or plasma cells. The proteasome inhibitor, bortezomib, was the first-in-class compound to be FDA approved for MM and is frequently utilized in induction therapy. However, bortezomib refractory disease is a major clinical concern, and the efficacy of the pan-histone deacetylase inhibitor (HDACi), panobinostat, in bortezomib refractory disease indicates that HDAC targeting is a viable strategy. Here, we utilized isogenic bortezomib resistant models to profile HDAC expression and define baseline and HDACi-induced expression patterns of individual HDAC family members in sensitive vs. resistant cells to better understanding the potential for targeting these enzymes. METHODS: Gene expression of HDAC family members in two sets of isogenic bortezomib sensitive or resistant myeloma cell lines was examined. These cell lines were subsequently treated with HDAC inhibitors: panobinostat or vorinostat, and HDAC expression was evaluated. CRISPR/Cas9 knockdown and pharmacological inhibition of specific HDAC family members were conducted. RESULTS: Interestingly, HDAC6 and HDAC7 were significantly upregulated and downregulated, respectively, in bortezomib-resistant cells. Panobinostat was effective at inducing cell death in these lines and modulated HDAC expression in cell lines and patient samples. Knockdown of HDAC7 inhibited cell growth while pharmacologically inhibiting HDAC6 augmented cell death by panobinostat. CONCLUSION: Our data revealed heterogeneous expression of individual HDACs in bortezomib sensitive vs. resistant isogenic cell lines and patient samples treated with panobinostat. Cumulatively our findings highlight distinct roles for HDAC6 and HDAC7 in regulating cell death in the context of bortezomib resistance.

13.
JCI Insight ; 5(1)2020 01 16.
Article in English | MEDLINE | ID: mdl-31941838

ABSTRACT

Myelofibrosis (MF) is a myeloproliferative neoplasm characterized by cytopenia and extramedullary hematopoiesis, resulting in splenomegaly. Multiple pathological mechanisms (e.g., circulating cytokines and genetic alterations, such as JAKV617F mutation) have been implicated in the etiology of MF, but the molecular mechanism causing resistance to JAK2V617F inhibitor therapy remains unknown. Among MF patients who were treated with the JAK inhibitor ruxolitinib, we compared noncoding RNA profiles of ruxolitinib therapy responders versus nonresponders and found miR-543 was significantly upregulated in nonresponders. We validated these findings by reverse transcription-quantitative PCR. in this same cohort, in 2 additional independent MF patient cohorts from the United States and Romania, and in a JAK2V617F mouse model of MF. Both in vitro and in vivo models were used to determine the underlying molecular mechanism of miR-543 in MF. Here, we demonstrate that miR-543 targets the dioxygenases ten-eleven translocation 1 (TET1) and 2 (TET2) in patients and in vitro, causing increased levels of global 5-methylcytosine, while decreasing the acetylation of histone 3, STAT3, and tumor protein p53. Mechanistically, we found that activation of STAT3 by JAKs epigenetically controls miR-543 expression via binding the promoter region of miR-543. Furthermore, miR-543 upregulation promotes the expression of genes related to drug metabolism, including CYP3A4, which is involved in ruxolitinib metabolism. Our findings suggest miR-543 as a potentially novel biomarker for the prognosis of MF patients with a high risk of treatment resistance and as a potentially new target for the development of new treatment options.


Subject(s)
DNA-Binding Proteins/drug effects , Epigenesis, Genetic/drug effects , MicroRNAs/metabolism , MicroRNAs/pharmacology , Primary Myelofibrosis/drug therapy , Proto-Oncogene Proteins/drug effects , Animals , Cytokines/metabolism , DNA-Binding Proteins/genetics , Dioxygenases , Disease Models, Animal , Histones , Humans , Janus Kinase Inhibitors/therapeutic use , Janus Kinases/metabolism , Mice , MicroRNAs/genetics , Mixed Function Oxygenases , Mutation , Myeloproliferative Disorders , Nitriles , Primary Myelofibrosis/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/genetics , Pyrazoles/therapeutic use , Pyrimidines , STAT3 Transcription Factor , Transcriptome , United States
14.
Cancer Res ; 67(8): 3912-8, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17440106

ABSTRACT

c-Myc is a highly unstable transcription factor whose deregulation and increased expression are associated with cancer. Degrasyn, a small synthetic molecule, induces rapid degradation of c-Myc protein in MM-1 multiple myeloma and other tumor cell lines. Destruction of c-Myc by degrasyn requires the presence of a region of c-Myc between amino acid residues 316 and 378 that has not previously been associated with c-Myc stability. Degrasyn-induced degradation of c-Myc depends on proteasomes but is independent of the degron regions previously shown to be important for ubiquitin-mediated targeting and proteasomal destruction of the protein. Degrasyn-dependent c-Myc proteolysis is not mediated by any previously identified c-Myc regulatory mechanism, does not require new protein synthesis, and does not depend on the nuclear localization of c-Myc. Degrasyn reduced c-Myc levels in A375 melanoma cells and in A375 tumors in nude mice, and this activity correlated with tumor growth inhibition. Together, these results suggest that degrasyn reduces the stability of c-Myc in vitro and in vivo through a unique signaling process that uses c-Myc domains not previously associated with c-Myc regulation.


Subject(s)
Melanoma/drug therapy , Nitriles/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Pyridines/pharmacology , Animals , Cell Line, Tumor , Cyanoacrylates , Down-Regulation/drug effects , Enzyme Activation/drug effects , Female , HeLa Cells , Humans , Melanoma/enzymology , Melanoma/metabolism , Mice , Mice, Nude , Multiple Myeloma/drug therapy , Multiple Myeloma/enzymology , Multiple Myeloma/metabolism , Peptide Mapping , Protein Structure, Tertiary , Xenograft Model Antitumor Assays
15.
Clin Cancer Res ; 25(18): 5702-5716, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31391192

ABSTRACT

PURPOSE: Paclitaxel is an integral component of primary therapy for breast and epithelial ovarian cancers, but less than half of these cancers respond to the drug. Enhancing the response to primary therapy with paclitaxel could improve outcomes for women with both diseases.Experimental Design: Twelve kinases that regulate metabolism were depleted in multiple ovarian and breast cancer cell lines to determine whether they regulate sensitivity to paclitaxel in Sulforhodamine B assays. The effects of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 2 (PFKFB2) depletion on cell metabolomics, extracellular acidification rate, nicotinamide adenine dinucleotide phosphate, reactive oxygen species (ROS), and apoptosis were studied in multiple ovarian and breast cancer cell lines. Four breast and ovarian human xenografts and a breast cancer patient-derived xenograft (PDX) were used to examine the knockdown effect of PFKFB2 on tumor cell growth in vivo. RESULTS: Knockdown of PFKFB2 inhibited clonogenic growth and enhanced paclitaxel sensitivity in ovarian and breast cancer cell lines with wild-type TP53 (wtTP53). Silencing PFKFB2 significantly inhibited tumor growth and enhanced paclitaxel sensitivity in four xenografts derived from two ovarian and two breast cancer cell lines, and prolonged survival in a triple-negative breast cancer PDX. Transfection of siPFKFB2 increased the glycolysis rate, but decreased the flow of intermediates through the pentose-phosphate pathway in cancer cells with wtTP53, decreasing NADPH. ROS accumulated after PFKFB2 knockdown, which stimulated Jun N-terminal kinase and p53 phosphorylation, and induced apoptosis that depended upon upregulation of p21 and Puma. CONCLUSIONS: PFKFB2 is a novel target whose inhibition can enhance the effect of paclitaxel-based primary chemotherapy upon ovarian and breast cancers retaining wtTP53.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Ovarian Neoplasms/metabolism , Paclitaxel/pharmacology , Phosphofructokinase-2/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Female , Gene Expression , Gene Silencing , Humans , Immunohistochemistry , Metabolic Networks and Pathways , Mice , Mutation , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Oxidative Stress , Phosphofructokinase-2/genetics , RNA Interference , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
16.
Mol Cancer Ther ; 18(1): 162-172, 2019 01.
Article in English | MEDLINE | ID: mdl-30305341

ABSTRACT

For mucinous ovarian cancer (MOC), standard platinum-based therapy is largely ineffective. We sought to identify possible mechanisms of oxaliplatin resistance of MOC and develop strategies to overcome this resistance. A kinome-based siRNA library screen was carried out using human MOC cells to identify novel targets to enhance the efficacy of chemotherapy. In vitro and in vivo validations of antitumor effects were performed using mouse MOC models. Specifically, the role of PRKRA/PACT in oxaliplatin resistance was interrogated. We focused on PRKRA, a known activator of PKR kinase, and its encoded protein PACT because it was one of the five most significantly downregulated genes in the siRNA screen. In orthotopic mouse models of MOC, we observed a significant antitumor effect of PRKRA siRNA plus oxaliplatin. In addition, expression of miR-515-3p was regulated by PACT-Dicer interaction, and miR-515-3p increased the sensitivity of MOC to oxaliplatin. Mechanistically, miR-515-3p regulated chemosensitivity, in part, by targeting AXL. The PRKRA/PACT axis represents an important therapeutic target in MOC to enhance sensitivity to oxaliplatin.


Subject(s)
Adenocarcinoma, Mucinous/pathology , Drug Resistance, Neoplasm , Ovarian Neoplasms/pathology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Up-Regulation , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/metabolism , Animals , Cell Line, Tumor , Cell Survival , DEAD-box RNA Helicases/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , MicroRNAs/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Oxaliplatin , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/pharmacology , Receptor Protein-Tyrosine Kinases/genetics , Ribonuclease III/metabolism , Axl Receptor Tyrosine Kinase
19.
Mol Cell Biol ; 25(24): 11005-18, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16314522

ABSTRACT

The cell membrane receptor ErbB-2 migrates to the nucleus. However, the mechanism of its nuclear translocation is unclear. Here, we report a novel mechanism of its nuclear localization that involves interaction with the transport receptor importin beta1, nuclear pore protein Nup358, and a host of players in endocytic internalization. Knocking down importin beta1 using small interfering RNA oligonucleotides or inactivation of small GTPase Ran by RanQ69L, a dominant-negative mutant of Ran, causes a nuclear transport defect of ErbB-2. Mutation of a putative nuclear localization signal in ErbB-2 destroys its interaction with importin beta1 and arrests nuclear translocation, while inactivation of nuclear export receptor piles up ErbB-2 within the nucleus. Additionally, blocking of internalization by a dominant-negative mutant of dynamin halts its nuclear localization. Thus, the cell membrane-embedded ErbB-2, through endocytosis using the endocytic vesicle as a vehicle, importin beta1 as a driver and Nup358 as a traffic light, migrates from the cell surface to the nucleus. This novel mechanism explains how a receptor tyrosine kinase on the cell surface can be translocated into the nucleus. This pathway may serve as a general mechanism to allow direct communication between cell surface receptors and the nucleus, and our findings thus open a new era in understanding direct trafficking between the cell membrane and nucleus.


Subject(s)
Cell Nucleus/metabolism , Endocytosis , Molecular Chaperones/metabolism , Nuclear Pore Complex Proteins/metabolism , Receptor, ErbB-2/metabolism , Receptors, Cell Surface/metabolism , beta Karyopherins/metabolism , Active Transport, Cell Nucleus , Cell Nucleus/chemistry , Cells, Cultured , Clathrin/metabolism , Endosomes/metabolism , Humans , Karyopherins/genetics , Karyopherins/metabolism , Mutation , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Receptor, ErbB-2/analysis , Receptors, Cell Surface/analysis , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , beta Karyopherins/analysis , ran GTP-Binding Protein/genetics , ran GTP-Binding Protein/metabolism , Exportin 1 Protein
20.
PLoS One ; 13(5): e0195932, 2018.
Article in English | MEDLINE | ID: mdl-29768500

ABSTRACT

Inflammatory breast cancer (IBC) is a rare and aggressive presentation of invasive breast cancer with a 62% to 68% 5-year survival rate. It is the most lethal form of breast cancer, and early recognition and treatment is important for patient survival. Like non-inflammatory breast cancer, IBC comprises multiple subtypes, with the triple-negative subtype being overrepresented. Although the current multimodality treatment regime of anthracycline- and taxane-based neoadjuvant therapy, surgery, and radiotherapy has improved the outcome of patients with triple-negative IBC, overall survival continues to be worse than in patients with non-inflammatory locally advanced breast cancer. Translation of new therapies into the clinics to successfully treat IBC has been poor, in part because of the lack of in vitro preclinical models that can accurately predict the response of the original tumor to therapy. We report the generation of a preclinical IBC patient-derived xenograft (PDX)-derived ex vivo (PDXEx) model and show that it closely replicates the tissue architecture of the original PDX tumor harvested from mice. The gene expression profile of our IBC PDXEx model had a high degree of correlation to that of the original tumor. This suggests that the process of generating the PDXEx model did not significantly alter the molecular signature of the original tumor. We demonstrate a high degree of similarity in drug response profile between a PDX mouse model and our PDXEx model generated from the same original PDX tumor tissue and treated with the same panel of drugs, indicating that our PDXEx model had high predictive value in identifying effective tumor-specific therapies. Finally, we used our PDXEx model as a platform for a robotic-based high-throughput drug screen of a 386-drug anti-cancer compound library. The top candidates identified from this drug screen all demonstrated greater therapeutic efficacy than the standard-of-care drugs used in the clinic to treat triple-negative IBC, doxorubicin and paclitaxel. Our PDXEx model is simple, and we are confident that it can be incorporated into a PDX mouse system for use as a first-pass screening platform. This will permit the identification of effective tumor-specific therapies with high predictive value in a resource-, time-, and cost-efficient manner.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Inflammatory Breast Neoplasms/pathology , Xenograft Model Antitumor Assays , Animals , Female , Gene Expression Profiling , Humans , Inflammatory Breast Neoplasms/drug therapy , Inflammatory Breast Neoplasms/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL