Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cancer Res Commun ; 3(9): 1731-1742, 2023 09.
Article in English | MEDLINE | ID: mdl-37663435

ABSTRACT

DNA-dependent protein kinase (DNA-PK), a driver of the non-homologous end-joining (NHEJ) DNA damage response pathway, plays an instrumental role in repairing double-strand breaks (DSB) induced by DNA-damaging poisons. We evaluate ZL-2201, an orally bioavailable, highly potent, and selective pharmacologic inhibitor of DNA-PK activity, for the treatment of human cancerous malignancies. ZL-2201 demonstrated greater selectivity for DNA-PK and effectively inhibited DNA-PK autophosphorylation in a concentration- and time-dependent manner. Initial data suggested a potential correlation between ataxia-telangiectasia mutated (ATM) deficiency and ZL-2201 sensitivity. More so, ZL-2201 showed strong synergy with topoisomerase II inhibitors independent of ATM status in vitro. In vivo oral administration of ZL-2201 demonstrated dose-dependent antitumor activity in the NCI-H1703 xenograft model and significantly enhanced the activity of approved DNA-damaging agents in A549 and FaDu models. From a phosphoproteomic mass spectrometry screen, we identified and validated that ZL-2201 and PRKDC siRNA decreased Ser108 phosphorylation of MCM2, a key DNA replication factor. Collectively, we have characterized a potent and selective DNA-PK inhibitor with promising monotherapy and combinatory therapeutic potential with approved DNA-damaging agents. More importantly, we identified phospho-MCM2 (Ser108) as a potential proximal biomarker of DNA-PK inhibition that warrants further preclinical and clinical evaluation. Significance: ZL-2201, a potent and selective DNA-PK inhibitor, can target tumor models in combination with DNA DSB-inducing agents such as radiation or doxorubicin, with potential to improve recurrent therapies in the clinic.


Subject(s)
DNA-Activated Protein Kinase , Humans , Administration, Oral , Phosphorylation , Animals , DNA-Activated Protein Kinase/antagonists & inhibitors
2.
Prog Mol Biol Transl Sci ; 192(1): 179-204, 2022.
Article in English | MEDLINE | ID: mdl-36280319

ABSTRACT

Inflammatory bowel disease (IBD) is a complex multi-factorial chronic relapsing disease of the digestive tract where dysbiosis of autochthonous intestinal microbiota, environmental factors and host genetics are implicated in the disease development, severity, course and treatment outcomes. The two clinically well-defined forms of IBD are Crohn's disease (CD) and ulcerative colitis (UC). The CD affects the local immune response of the entire gastrointestinal tract whereas the inflammation in UC is mainly restricted to the colonic mucosa. Prolong progressive inflammation due to CD and UC often lead to colonic cancer. In healthy individuals, the enormous taxonomic diversity and functional potency of gut microbiota including members from the bacterial and fungal microbiota tune the host immunity and keep the gastric environment beneficial and protective. However, expansion of pathobionts, autochthonous microbes with the potency of pathogenicity in dysbiotic condition, in the gastrointestinal tract and subsequently enriched inflammatory microbial products in the gastrointestinal milieu attract different immune cells and activate aberrant host immune response which leads to excessive production and secretion of different cytokines that damage the colonic epithelial cells and manifest chronic inflammatory digestive disease. In the current chapter, we provided our updated understanding about the different bacterial and fungal pathobionts, their genomic and metabolic signatures, and geo-specific diversity of gut microbes linked with IBD across the globe at the molecular resolution. An improved understanding of IBD and the factors associated with the disease will be a boost for therapeutic development and disease management.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Humans , Dysbiosis , Inflammatory Bowel Diseases/microbiology , Inflammation , Bacteria , Cytokines
3.
Mol Cancer Res ; 17(2): 508-520, 2019 02.
Article in English | MEDLINE | ID: mdl-30266754

ABSTRACT

Isocitrate dehydrogenase 1 (IDH1) is the most commonly mutated metabolic enzyme in human malignancy. A heterozygous genetic alteration, arginine 132, promotes the conversion of α-ketoglutarate to D-2-hydroxyglutarate (2-HG). Although pharmacologic inhibitors of mutant IDH1 are promising, resistance mechanisms to targeted therapy are not understood. Additionally, the role of wild-type IDH1 (WT.IDH1) in cancer requires further study. Recently, it was observed that the regulatory RNA-binding protein, HuR (ELAVL1), protects nutrient-deprived cancer cells without IDH1 mutations, by stabilizing WT.IDH1 transcripts. In the present study, a similar regulatory effect on both mutant (Mut.IDH1) and WT.IDH1 transcripts in heterozygous IDH1-mutant tumors is observed. In ribonucleoprotein immunoprecipitation assays of IDH1-mutant cell lines, wild-type and mutant IDH1 mRNAs each bound to HuR. Both isoforms were profoundly downregulated at the mRNA and protein levels after genetic suppression of HuR (siRNAs or CRISPR deletion) in HT1080 (R132C IDH1 mutation) and BT054 cells (R132H). Proliferation and invasion were adversely affected after HuR suppression and metabolomic studies revealed a reduction in Pentose Phosphate Pathway metabolites, nucleotide precursors, and 2-HG levels. HuR-deficient cells were especially sensitive to stress, including low glucose conditions or a mutant IDH1 inhibitor (AGI-5198). IDH1-mutant cancer cells were rescued by WT.IDH1 overexpression to a greater extent than Mut.IDH1 overexpression under these conditions. This study reveals the importance of HuR's regulation of both mutant and wild-type IDH1 in tumors harboring a heterozygous IDH1 mutation with implications for therapy. IMPLICATIONS: This study highlights the HuR-IDH1 (mutant and wild-type IDH1) regulatory axis as a critical, actionable therapeutic target in IDH1-mutated cancer, and incomplete blockade of the entire HuR-IDH1 survival axis would likely diminish the efficacy of drugs that selectively target only the mutant isoenzyme.


Subject(s)
ELAV-Like Protein 1/metabolism , Isocitrate Dehydrogenase/metabolism , Animals , Cell Line, Tumor , Down-Regulation , ELAV-Like Protein 1/antagonists & inhibitors , ELAV-Like Protein 1/genetics , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Heterografts , Humans , Isocitrate Dehydrogenase/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Mice, Nude , Mutation
4.
Cancer Res ; 77(18): 5011-5025, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28687616

ABSTRACT

The majority of pancreatic ductal adenocarcinomas (PDAC) rely on the mRNA stability factor HuR (ELAV-L1) to drive cancer growth and progression. Here, we show that CRISPR-Cas9-mediated silencing of the HuR locus increases the relative sensitivity of PDAC cells to PARP inhibitors (PARPi). PDAC cells treated with PARPi stimulated translocation of HuR from the nucleus to the cytoplasm, specifically promoting stabilization of a new target, poly (ADP-ribose) glycohydrolase (PARG) mRNA, by binding a unique sequence embedded in its 3' untranslated region. HuR-dependent upregulation of PARG expression facilitated DNA repair via hydrolysis of polyADP-ribose on related repair proteins. Accordingly, strategies to inhibit HuR directly promoted DNA damage accumulation, inefficient PAR removal, and persistent PARP-1 residency on chromatin (PARP-1 trapping). Immunoprecipitation assays demonstrated that the PARP-1 protein binds and posttranslationally modifies HuR in PARPi-treated PDAC cells. In a mouse xenograft model of human PDAC, PARPi monotherapy combined with targeted silencing of HuR significantly reduced tumor growth compared with PARPi therapy alone. Our results highlight the HuR-PARG axis as an opportunity to enhance PARPi-based therapies. Cancer Res; 77(18); 5011-25. ©2017 AACR.


Subject(s)
DNA Repair/genetics , Drug Resistance, Neoplasm/genetics , ELAV-Like Protein 1/metabolism , Glycoside Hydrolases/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/chemistry , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Proliferation , DNA Damage/drug effects , DNA Damage/genetics , DNA Repair/drug effects , ELAV-Like Protein 1/antagonists & inhibitors , ELAV-Like Protein 1/genetics , Female , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Tumor Cells, Cultured , Up-Regulation , Xenograft Model Antitumor Assays , Pancreatic Neoplasms
5.
Cancer Res ; 77(16): 4460-4471, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28652247

ABSTRACT

Cancer aggressiveness may result from the selective pressure of a harsh nutrient-deprived microenvironment. Here we illustrate how such conditions promote chemotherapy resistance in pancreatic ductal adenocarcinoma (PDAC). Glucose or glutamine withdrawal resulted in a 5- to 10-fold protective effect with chemotherapy treatment. PDAC xenografts were less sensitive to gemcitabine in hypoglycemic mice compared with hyperglycemic mice. Consistent with this observation, patients receiving adjuvant gemcitabine (n = 107) with elevated serum glucose levels (HgbA1C > 6.5%) exhibited improved survival. We identified enhanced antioxidant defense as a driver of chemoresistance in this setting. ROS levels were doubled in vitro by either nutrient withdrawal or gemcitabine treatment, but depriving PDAC cells of nutrients before gemcitabine treatment attenuated this effect. Mechanistic investigations based on RNAi or CRISPR approaches implicated the RNA binding protein HuR in preserving survival under nutrient withdrawal, with or without gemcitabine. Notably, RNA deep sequencing and functional analyses in HuR-deficient PDAC cell lines identified isocitrate dehydrogenase 1 (IDH1) as the sole antioxidant enzyme under HuR regulation. HuR-deficient PDAC cells lacked the ability to engraft successfully in immunocompromised mice, but IDH1 overexpression in these cells was sufficient to fully restore chemoresistance under low nutrient conditions. Overall, our findings highlight the HuR-IDH1 regulatory axis as a critical, actionable therapeutic target in pancreatic cancer. Cancer Res; 77(16); 4460-71. ©2017 AACR.


Subject(s)
ELAV-Like Protein 1/metabolism , Isocitrate Dehydrogenase/metabolism , Pancreatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Cohort Studies , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm , ELAV-Like Protein 1/genetics , Humans , Isocitrate Dehydrogenase/genetics , Mice , Mice, Nude , Organoplatinum Compounds/pharmacology , Oxaliplatin , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phenotype , Protein Processing, Post-Translational , Survival Analysis , Transcriptional Activation , Transfection , Up-Regulation , Gemcitabine
6.
Mol Cancer Res ; 15(6): 696-707, 2017 06.
Article in English | MEDLINE | ID: mdl-28242812

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is the third leading cause of cancer-related deaths in the United States, whereas colorectal cancer is the third most common cancer. The RNA-binding protein HuR (ELAVL1) supports a pro-oncogenic network in gastrointestinal (GI) cancer cells through enhanced HuR expression. Using a publically available database, HuR expression levels were determined to be increased in primary PDA and colorectal cancer tumor cohorts as compared with normal pancreas and colon tissues, respectively. CRISPR/Cas9 technology was successfully used to delete the HuR gene in both PDA (MIA PaCa-2 and Hs 766T) and colorectal cancer (HCT116) cell lines. HuR deficiency has a mild phenotype, in vitro, as HuR-deficient MIA PaCa-2 (MIA.HuR-KO(-/-)) cells had increased apoptosis when compared with isogenic wild-type (MIA.HuR-WT(+/+)) cells. Using this isogenic system, mRNAs were identified that specifically bound to HuR and were required for transforming a two-dimensional culture into three dimensional (i.e., organoids). Importantly, HuR-deficient MIA PaCa-2 and Hs 766T cells were unable to engraft tumors in vivo compared with control HuR-proficient cells, demonstrating a unique xenograft lethal phenotype. Although not as a dramatic phenotype, CRISPR knockout HuR HCT116 colon cancer cells (HCT.HuR-KO(-/-)) showed significantly reduced in vivo tumor growth compared with controls (HCT.HuR-WT(+/+)). Finally, HuR deletion affects KRAS activity and controls a subset of pro-oncogenic genes.Implications: The work reported here supports the notion that targeting HuR is a promising therapeutic strategy to treat GI malignancies. Mol Cancer Res; 15(6); 696-707. ©2017 AACR.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Pancreatic Ductal/genetics , Colonic Neoplasms/genetics , ELAV-Like Protein 1/genetics , Pancreatic Neoplasms/genetics , Adenocarcinoma/pathology , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats , Colonic Neoplasms/pathology , ELAV-Like Protein 1/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Mice, Nude , Neoplasms, Experimental/genetics , Pancreatic Neoplasms/pathology , Phenotype , Xenograft Model Antitumor Assays
7.
Sci Rep ; 6: 33323, 2016 09 12.
Article in English | MEDLINE | ID: mdl-27616351

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is a lethal disease, in part, because of the lack of effective targeted therapeutic options. MK-1775 (also known as AZD1775), a mitotic inhibitor, has been demonstrated to enhance the anti-tumor effects of DNA damaging agents such as gemcitabine. We evaluated the efficacy of MK-1775 alone or in combination with DNA damaging agents (MMC or oxaliplatin) in PDA cell lines that are either DNA repair proficient (DDR-P) or deficient (DDR-D). PDA cell lines PL11, Hs 766T and Capan-1 harboring naturally selected mutations in DNA repair genes FANCC, FANCG and BRCA2 respectively, were less sensitive to MK-1775 as compared to two out of four representative DDR-P (MIA PaCa2 and PANC-1) cell lines. Accordingly, DDR-P cells exhibit reduced sensitivity to MK-1775 upon siRNA silencing of DNA repair genes, BRCA2 or FANCD2, compared to control cells. Only DDR-P cells showed increased apoptosis as a result of early mitotic entry and catastrophe compared to DDR-D cells. Taken together with other recently published reports, our results add another level of evidence that the efficacy of WEE1 inhibition is influenced by the DNA repair status of a cell and may also be dependent on the tumor type and model evaluated.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Cell Cycle Proteins/antagonists & inhibitors , DNA Repair/drug effects , Nuclear Proteins/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis , Carcinoma, Pancreatic Ductal/genetics , Cell Line, Tumor , DNA Damage , DNA Repair Enzymes/genetics , Drug Resistance, Neoplasm , Drug Synergism , Humans , Inhibitory Concentration 50 , Mitomycin/pharmacology , Mitosis , Mutagens/pharmacology , Mutation , Organoplatinum Compounds/pharmacology , Oxaliplatin , Pancreatic Neoplasms/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrimidinones
8.
Methods Mol Biol ; 1262: 239-46, 2015.
Article in English | MEDLINE | ID: mdl-25555585

ABSTRACT

Post-transcriptional regulation of mRNA can potently dictate protein expression patterns in eukaryotic cells. This mode of regulation occurs through cis-acting regulatory regions in the mRNA transcript that mediate direct interactions with trans-acting RNA-binding proteins (RBPs). This mRNA/protein interaction can be studied in numerous ways that range from in vitro to in vivo through messenger ribonucleoprotein immunoprecipitation (mRNP-IP or RIP) assays. This modified immunoprecipitation approach is an important and sensitive method to determine the regulation of gene expression by specific RBPs under different cellular stressors.


Subject(s)
Immunoradiometric Assay/methods , RNA, Messenger/isolation & purification , RNA, Messenger/metabolism , Animals , Humans , RNA Processing, Post-Transcriptional , Ribonucleoproteins/metabolism
9.
Mol Cancer Res ; 13(3): 439-48, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25336517

ABSTRACT

UNLABELLED: Mucin1 (MUC1) is overexpressed in pancreatic ductal adenocarcinoma (PDA) and is associated with tumor aggressiveness, suggesting that MUC1 is a promising therapeutic target for promoter-driven diphtheria toxin A (DTA). Endogenous MUC1 transcript levels were analyzed by quantitative PCR (qPCR) in multiple PDA cells (Capan1, HPAFII, Su.86.86, Capan2, Hs766T, MiaPaCa2, and Panc1). Expression levels were correlated with luciferase activity and cell death after transfection with MUC1 promoter-driven luciferase and DTA constructs. MUC1-positive (+) cells had significantly elevated MUC1 mRNA expression compared with MUC1-negative (-) cells. Luciferase activity was significantly higher in MUC1(+) cells when transfected with MUC1 promoter-driven luciferase and MUC1(+) cells underwent enhanced cell death after transfection with a single dose of MUC1 promoter-driven DTA. IFNγ pretreatment enhanced MUC1 expression in MUC1(-) cells and induced sensitivity to MUC1-DTA therapy. Matched primary and metastatic tumor lesions from clinical specimens revealed similar MUC1 IHC labeling patterns, and a tissue microarray of human PDA biopsies revealed increased immunolabeling with a combination of MUC1 and mesothelin (MSLN) antibodies, compared with either antibody alone. Combining MUC1 with MSLN-targeted DTA enhanced drug efficacy in an in vitro model of heterogeneous PDA. These data demonstrate that MUC1 promoter-driven DTA preferentially kills MUC1-expressing PDA cells and drugs that enhance MUC1 expression sensitize PDA cells with low MUC1 expression. IMPLICATIONS: MUC1 expression in primary and metastatic lesions provides a rationale for the development of a systemic MUC1 promoter-driven DTA therapy that may be further enhanced by combination with other promoter-driven DTA constructs.


Subject(s)
Carcinoma, Pancreatic Ductal/therapy , Diphtheria Toxin/pharmacology , Molecular Targeted Therapy/methods , Mucin-1/genetics , Pancreatic Neoplasms/therapy , Peptide Fragments/pharmacology , Promoter Regions, Genetic , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Death , Cell Line, Tumor , Diphtheria Toxin/genetics , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Neoplastic , Genetic Vectors/pharmacology , Humans , Interferon-gamma/pharmacology , Mesothelin , Mucin-1/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Peptide Fragments/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
10.
Cancer Biol Ther ; 15(6): 688-98, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24618665

ABSTRACT

Deoxycytidine kinase (dCK) and human antigen R (HuR) have been associated with response to gemcitabine in small studies. The present study investigates the prognostic and predictive value of dCK and HuR expression levels for sensitivity to gemcitabine and 5-fluorouracil (5-FU) in a large phase III adjuvant trial with chemoradiation backbone in pancreatic ductal adenocarcinoma (PDA). The dCK and HuR expression levels were determined by immunohistochemistry on a tissue microarray of 165 resected PDAs from the Radiation Therapy Oncology Group (RTOG) 9704 trial. Association with overall survival (OS) and disease-free survival (DFS) status were analyzed using the log-rank test and the Cox proportional hazards model. Experiments with cultured PDA cells were performed to explore mechanisms linking dCK and HuR expression to drug sensitivity. dCK expression levels were associated with improved OS for all patients analyzed from RTOG 9704 (HR: 0.66, 95% CI [0.47-0.93], P = 0.015). In a subset analysis based on treatment arm, the effect was restricted to patients receiving 5-FU (HR: 0.53, 95% CI [0.33-0.85], P = 0.0078). Studies in cultured cells confirmed that dCK expression rendered cells more sensitive to 5-FU. HuR cytoplasmic expression was neither prognostic nor predictive of treatment response. Previous studies along with drug sensitivity and biochemical studies demonstrate that radiation interferes with HuR's regulatory effects on dCK, and could account for the negative findings herein based on the clinical study design (i.e., inclusion of radiation). Finally, we demonstrate that 5-FU can increase HuR function by enhancing HuR translocation from the nucleus to the cytoplasm, similar to the effect of gemcitabine in PDA cells. For the first time, in the pre-treatment tumor samples, dCK and HuR cytoplasmic expression were strongly correlated (chi-square P = 0.015). This dual-institutional follow up study, in a multi-institutional PDA randomized clinical trial, observed that dCK expression levels were prognostic and had predictive value for sensitivity to 5-FU.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Pancreatic Ductal/metabolism , Deoxycytidine Kinase/metabolism , ELAV Proteins/metabolism , Fluorouracil/pharmacology , Pancreatic Neoplasms/metabolism , Antimetabolites, Antineoplastic/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/mortality , Cell Line, Tumor , Cell Nucleus/metabolism , Chemoradiotherapy , Cytoplasm/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine Kinase/genetics , Disease-Free Survival , ELAV-Like Protein 1 , Fluorouracil/therapeutic use , Follow-Up Studies , Gene Expression , Humans , Kaplan-Meier Estimate , Multivariate Analysis , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/mortality , Prognosis , Proportional Hazards Models , Protein Transport , Randomized Controlled Trials as Topic , Treatment Outcome , Gemcitabine
11.
Cancer Res ; 74(4): 1128-40, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24536047

ABSTRACT

HuR (ELAV1), an RNA-binding protein abundant in cancer cells, primarily resides in the nucleus, but under specific stress (e.g., gemcitabine), HuR translocates to the cytoplasm in which it tightly modulates the expression of mRNA survival cargo. Here, we demonstrate for the first time that stressing pancreatic ductal adenocarcinoma (PDA) cells by treatment with DNA-damaging anticancer agents (mitomycin C, oxaliplatin, cisplatin, carboplatin, and a PARP inhibitor) results in HuR's translocation from the nucleus to the cytoplasm. Importantly, silencing HuR in PDA cells sensitized the cells to these agents, whereas overexpressing HuR caused resistance. HuR's role in the efficacy of DNA-damaging agents in PDA cells was, in part, attributed to the acute upregulation of WEE1 by HuR. WEE1, a mitotic inhibitor kinase, regulates the DNA damage repair pathway, and therapeutic inhibition of WEE1 in combination with chemotherapy is currently in early phase trials for the treatment of cancer. We validate WEE1 as a HuR target in vitro and in vivo by demonstrating (i) direct binding of HuR to WEE1's mRNA (a discrete 56-bp region residing in the 3' untranslated region) and (ii) HuR siRNA silencing and overexpression directly affects the protein levels of WEE1, especially after DNA damage. HuR's positive regulation of WEE1 increases γ-H2AX levels, induces Cdk1 phosphorylation, and promotes cell-cycle arrest at the G2-M transition. We describe a novel mechanism that PDA cells use to protect against DNA damage in which HuR posttranscriptionally regulates the expression and downstream function of WEE1 upon exposure to DNA-damaging agents.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Cell Cycle Proteins/genetics , DNA Damage/physiology , ELAV Proteins/physiology , Nuclear Proteins/genetics , Pancreatic Neoplasms/genetics , Protein-Tyrosine Kinases/genetics , RNA Interference , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/metabolism , Cell Cycle Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice , Nuclear Proteins/metabolism , Pancreatic Neoplasms/metabolism , Protein Transport/drug effects , Protein-Tyrosine Kinases/metabolism , Tumor Cells, Cultured
12.
Cancer Res ; 74(1): 31-7, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24189460

ABSTRACT

PARP-1 is a nuclear protein that has important roles in maintenance of genomic integrity. During genotoxic stress, PARP-1 recruits to sites of DNA damage where PARP-1 domain architecture initiates catalytic activation and subsequent poly(ADP-ribose)-dependent DNA repair. PARP-1 inhibition is a promising new way to selectively target cancers harboring DNA repair deficiencies. However, current inhibitors target other PARPs, raising important questions about long-term off-target effects. Here, we propose a new strategy that targets PARP-1 allosteric regulation as a selective way of inhibiting PARP-1. We found that disruption of PARP-1 domain-domain contacts through mutagenesis held no cellular consequences on recruitment to DNA damage or a model system of transcriptional regulation, but prevented DNA-damage-dependent catalytic activation. Furthermore, PARP-1 mutant overexpression in a pancreatic cancer cell line (MIA PaCa-2) increased sensitivity to platinum-based anticancer agents. These results not only highlight the potential of a synergistic drug combination of allosteric PARP inhibitors with DNA-damaging agents in genomically unstable cancer cells (regardless of homologous recombination status), but also signify important applications of selective PARP-1 inhibition. Finally, the development of a high-throughput PARP-1 assay is described as a tool to promote discovery of novel PARP-1 selective inhibitors.


Subject(s)
Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/enzymology , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Allosteric Regulation , Animals , Cloning, Molecular , DNA Damage , HeLa Cells , High-Throughput Screening Assays , Humans , Mice , Models, Molecular , Molecular Targeted Therapy , Mutagenesis , Organoplatinum Compounds/pharmacology , Pancreatic Neoplasms/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/genetics , Protein Structure, Tertiary , Transfection
13.
Mol Cancer Res ; 11(8): 901-11, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23696131

ABSTRACT

UNLABELLED: Pancreatic ductal adenocarcinoma (PDA) is the fourth leading cause of cancer-related death in the United States, with a 95% five-year mortality rate. For over a decade, gemcitabine (GEM) has been the established first-line treatment for this disease despite suboptimal response rates. The development of PARP inhibitors that target the DNA damage repair (DDR) system in PDA cells has generated encouraging results. Ubiquitin-specific peptidase 11 (USP11), an enzyme that interacts with the DDR protein BRCA2, was recently discovered to play a key role in DNA double-strand break repair and may be a novel therapeutic target. A systematic high-throughput approach was used to biochemically screen 2,000 U.S. Food and Drug Administration (FDA)-approved compounds for inhibition of USP11 enzymatic activity. Six pharmacologically active small molecules that inhibit USP11 enzymatic activity were identified. An in vitro drug sensitivity assay demonstrated that one of these USP11 inhibitors, mitoxantrone, impacted PDA cell survival with an IC50 of less than 10 nM. Importantly, across six different PDA cell lines, two with defects in the Fanconi anemia/BRCA2 pathway (Hs766T and Capan-1), mitoxantrone is 40- to 20,000-fold more potent than GEM, with increased endogenous USP11 mRNA levels associated with increased sensitivity to mitoxantrone. Interestingly, USP11 silencing in PDA cells also enhanced sensitivity to GEM. These findings establish a preclinical model for the rapid discovery of FDA-approved compounds and identify USP11 as a target of mitoxantrone in PDA. IMPLICATIONS: This high-throughput approach provides a strong rationale to study mitoxantrone in an early-phase clinical setting for the treatment of PDA.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Mitoxantrone/therapeutic use , Pancreatic Neoplasms/drug therapy , Thiolester Hydrolases/antagonists & inhibitors , BRCA2 Protein/genetics , Benzimidazoles/therapeutic use , Carcinoma, Pancreatic Ductal/enzymology , Cell Line, Tumor , DNA Damage/genetics , Deoxycytidine/therapeutic use , Drug Screening Assays, Antitumor , Gene Silencing , High-Throughput Screening Assays , Humans , Pancreatic Neoplasms/enzymology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Thiolester Hydrolases/metabolism , Gemcitabine
14.
Mol Plant ; 4(6): 1123-32, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21653282

ABSTRACT

The morphology of inflorescences is regulated in part by the temporal and spatial events that regulate flower specification. In Arabidopsis, an endogenous flowering time pathway mediated by a subset of SQUAMOSA PROMOTER-BINDING PROTEIN-LIKE (SPL) transcription factors, including SPL3, SPL4, and SPL5, function to specify flowers by activating floral meristem identity genes. During shoot development, SPL3, SPL4, and SPL5 are post-transcriptionally regulated by microRNA156 (miR156). The photoperiod regulated florigenic signal, FLOWERING LOCUS T (FT), promotes floral induction, in part by activating SPL3, SPL4, and SPL5. In turn, these SPLs function in parallel with FT to specify flower meristems. Two related BELL1-like homeobox genes PENNYWISE (PNY) and POUND-FOOLISH (PNF) expressed in the shoot apical meristem are absolutely required for the specification of floral meristems. Genetic studies show that the floral specification function of FT depends upon PNY and PNF; however, the interplay between these homeodomain proteins and SPLs is not known. In this manuscript, we show that the photoperiodic floral induction of SPL3, SPL4, and SPL5 is dependent upon PNY and PNF. Further, PNY and PNF also control SPL3, SPL4, and SPL5 expression by negatively regulating miR156. Lastly, ectopic expression of SPL4 partially rescues the pny pnf non-flower-producing phenotype, while overexpression of SPL3 or SPL5 in pny pnf plants was unable to restore flower specification. These results suggest that: (1) SPL3, SPL4, and SPL5 function is dependent upon PNY and PNF, or (2) expression of multiple SPLs is required for floral specification in pny pnf plants.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Gene Expression Regulation, Plant , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , MicroRNAs/genetics , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Transcription Factors/genetics , Arabidopsis/genetics , Arabidopsis/growth & development , Arabidopsis/physiology , Flowers/genetics , Flowers/growth & development , Flowers/metabolism , Flowers/physiology , MicroRNAs/metabolism , Plant Shoots/genetics , Plant Shoots/growth & development , Plant Shoots/metabolism , Plant Shoots/physiology , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproduction/genetics , Up-Regulation/genetics
15.
Plant Physiol ; 156(2): 605-14, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21505100

ABSTRACT

Growth of the aerial part of the plant is dependent upon the maintenance of the shoot apical meristem (SAM). A balance between the self-renewing stem cells in the central zone (CZ) and organogenesis in the peripheral zone (PZ) is essential for the integrity, function, and maintenance of the SAM. Understanding how the SAM maintains a balance between stem cell perpetuation and organogenesis is a central question in plant biology. Two related BELL1-like homeodomain proteins, PENNYWISE (PNY) and POUND-FOOLISH (PNF), act to specify floral meristems during reproductive development. However, genetic studies also show that PNY and PNF regulate the maintenance of the SAM. To understand the role of PNY and PNF in meristem maintenance, the expression patterns for genes that specifically localize to the peripheral and central regions of the SAM were examined in Arabidopsis (Arabidopsis thaliana). Results from these experiments indicate that the integrity of the CZ is impaired in pny pnf plants, which alters the balance of stem cell renewal and organogenesis. As a result, pools of CZ cells may be allocated into initiating leaf primordia. Consistent with these results, the integrity of the central region of pny pnf SAMs can be partially restored by increasing the size of the CZ. Interestingly, flower specification is also reestablished by augmenting the size of the SAM in pny pnf plants. Taken together, we propose that PNY and PNF act to restrict organogenesis to the PZ by maintaining a boundary between the CZ and PZ.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/physiology , Homeodomain Proteins/metabolism , Meristem/physiology , Repressor Proteins/metabolism , Arabidopsis/cytology , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Body Patterning/genetics , Flowers/cytology , Flowers/genetics , Gene Expression Regulation, Plant , Genes, Plant/genetics , Homeodomain Proteins/genetics , Meristem/cytology , Meristem/genetics , Mutation/genetics , Reproduction/genetics , Transcription, Genetic , Up-Regulation/genetics
16.
Mar Genomics ; 4(1): 51-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21429465

ABSTRACT

Sea slugs (Gastropoda: Opisthobranchia) are characterized by extensive morphological homoplasy. In particular, reduced or absent shells are predominant throughout the group. This trend towards shell loss has resulted in a poor fossil record. DNA-based phylogenies have been helpful in improving our understanding of the evolution of this group and major clades are emerging. We report 13 new complete opisthobranch mitochondrial genomes that provide robust support for some of these emerging nodes. We name three new clades within the Opisthobranchia, the Actopleura (Acteonoidea plus Nudipleura), Placoesophaga (Cephalaspidea plus Anaspidea), and Siphoglossa (Sacoglossa plus the Siphonaria). Finally we use molecular clock dating that suggests an earlier opisthobranch divergence than previously reported. The implications of this evolutionary scenario are discussed.


Subject(s)
Biological Evolution , Gastropoda/genetics , Genome, Mitochondrial/genetics , Genomics/methods , Animals , Gastropoda/anatomy & histology , Gene Order
17.
J Exp Bot ; 62(2): 583-93, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20937733

ABSTRACT

In Arabidopsis floral meristems are specified on the periphery of the inflorescence meristem by the combined activities of the FLOWERING LOCUS T (FT)-FD complex and the flower meristem identity gene LEAFY. The floral specification activity of FT is dependent upon two related BELL1-like homeobox (BLH) genes PENNYWISE (PNY) and POUND-FOOLISH (PNF) which are required for floral evocation. PNY and PNF interact with a subset of KNOTTED1-LIKE homeobox proteins including SHOOT MERISTEMLESS (STM). Genetic analyses show that these BLH proteins function with STM to specify flowers and internodes during inflorescence development. In this study, experimental evidence demonstrates that the specification of flower and coflorescence meristems requires the combined activities of FT-FD and STM. FT and FD also regulate meristem maintenance during inflorescence development. In plants with reduced STM function, ectopic FT and FD promote the formation of axillary meristems during inflorescence development. Lastly, gene expression studies indicate that STM functions with FT-FD and AGAMOUS-LIKE 24 (AGL24)-SUPPRESSOR OF OVEREXPRESSION OF CONTANS1 (SOC1) complexes to up-regulate flower meristem identity genes during inflorescence development.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Inflorescence/growth & development , Meristem/metabolism , Transcription Factors/metabolism , Arabidopsis/genetics , Arabidopsis/growth & development , Arabidopsis Proteins/genetics , Flowers/genetics , Flowers/growth & development , Flowers/metabolism , Gene Expression Regulation, Plant , Homeodomain Proteins/genetics , Inflorescence/genetics , Inflorescence/metabolism , Meristem/genetics , Meristem/growth & development , Transcription Factors/genetics
18.
Cutan Ocul Toxicol ; 28(3): 93-103, 2009.
Article in English | MEDLINE | ID: mdl-19505226

ABSTRACT

Presence of a preservative in an ocular medication has often been considered a culprit in damaging the epithelium. However, the inclusion of a preservative is equally necessary, especially in multiple-dose containers, in order to protect against dangerous organisms accidentally gaining access during instillation. Benzalkonium chloride (BAK), chlorobutanol, chlorhexidine acetate (CHA), and phenylmercuric nitrate or acetate are some commonly used preservatives in eye preparations. New preservatives with a wide range of activity and good safety profiles have been introduced in the market, such as stabilized oxychloro complex (SOC), sofZia, and sodium perborate. In the present review, we discuss various conventional and newly proposed and patented preservative molecules for ocular use. Reasons for discontinuing traditional preservatives and the need for less-toxic molecules are discussed at length, along with newer options coming up in this area.


Subject(s)
Eye/drug effects , Ophthalmic Solutions/toxicity , Preservatives, Pharmaceutical/toxicity , Cells, Cultured , Epithelial Cells/drug effects , Humans , Ophthalmic Solutions/chemistry , Preservatives, Pharmaceutical/chemistry , Risk , Toxicity Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...