Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 117(32): 19415-19424, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32719125

ABSTRACT

Synthetic lethality strategies for cancer therapy exploit cancer-specific genetic defects to identify targets that are uniquely essential to the survival of tumor cells. Here we show RAD27/FEN1, which encodes flap endonuclease 1 (FEN1), a structure-specific nuclease with roles in DNA replication and repair, and has the greatest number of synthetic lethal interactions with Saccharomyces cerevisiae genome instability genes, is a druggable target for an inhibitor-based approach to kill cancers with defects in homologous recombination (HR). The vulnerability of cancers with HR defects to FEN1 loss was validated by studies showing that small-molecule FEN1 inhibitors and FEN1 small interfering RNAs (siRNAs) selectively killed BRCA1- and BRCA2-defective human cell lines. Furthermore, the differential sensitivity to FEN1 inhibition was recapitulated in mice, where a small-molecule FEN1 inhibitor reduced the growth of tumors established from drug-sensitive but not drug-resistant cancer cell lines. FEN1 inhibition induced a DNA damage response in both sensitive and resistant cell lines; however, sensitive cell lines were unable to recover and replicate DNA even when the inhibitor was removed. Although FEN1 inhibition activated caspase to higher levels in sensitive cells, this apoptotic response occurred in p53-defective cells and cell killing was not blocked by a pan-caspase inhibitor. These results suggest that FEN1 inhibitors have the potential for therapeutically targeting HR-defective cancers such as those resulting from BRCA1 and BRCA2 mutations, and other genetic defects.


Subject(s)
Antineoplastic Agents/pharmacology , Flap Endonucleases/antagonists & inhibitors , Homologous Recombination/drug effects , Neoplasms/genetics , Animals , BRCA1 Protein/deficiency , BRCA1 Protein/genetics , BRCA2 Protein/deficiency , BRCA2 Protein/genetics , Cell Line, Tumor , DNA Damage/drug effects , DNA Repair/drug effects , DNA Replication/drug effects , Flap Endonucleases/genetics , Genomic Instability/genetics , Humans , Mice , Neoplasms/drug therapy , RNA, Small Interfering/pharmacology , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/genetics , Small Molecule Libraries/pharmacology , Synthetic Lethal Mutations , Xenograft Model Antitumor Assays
2.
Yale J Biol Med ; 92(4): 771-779, 2019 12.
Article in English | MEDLINE | ID: mdl-31866794

ABSTRACT

The cell death response to DNA damage is discussed in this Perspectives piece with cancer as the backdrop because DNA damaging agents (DDA) are widely used to treat cancer. From decades of clinical results, we learn that DDA have cured some cancers but their toxicity is temporary in most cancers due to emergence of DDA-resistant cancer cells. Investigation of DDA-activated genes, proteins, and pathways, known collectively as the DNA damage response (DDR), has uncovered the inner workings of DDR that protect the genome to sustain life. Paradoxically, however, DDR can also activate death. Current knowledge on DDA-activated death and hypotheses for how DDR may determine when and where to execute death are discussed. Given that cancer cells suffer from DDR defects, which account for their initial sensitivity to DDA, future therapeutic development may exploit those cancer-specific DDR defects to selectively create death-inducing DNA lesions, without using DDA, to kill DDA-resistant cancers.


Subject(s)
Apoptosis , DNA Damage , Animals , Cell Cycle Checkpoints , DNA Repair , Humans , Necroptosis , Neoplasms/genetics , Neoplasms/pathology
3.
Nat Struct Mol Biol ; 25(11): 1047-1058, 2018 11.
Article in English | MEDLINE | ID: mdl-30374083

ABSTRACT

Transcriptome analysis reveals a strong positive correlation between human Schlafen family member 11 (SLFN11) expression and the sensitivity of tumor cells to DNA-damaging agents (DDAs). Here, we show that SLFN11 preferentially inhibits translation of the serine/threonine kinases ATR and ATM upon DDA treatment based on distinct codon usage without disrupting early DNA damage response signaling. Type II transfer RNAs (tRNAs), which include all serine and leucine tRNAs, are cleaved in a SLFN11-dependent manner in response to DDAs. Messenger RNAs encoded by genes with high TTA (Leu) codon usage, such as ATR, display utmost susceptibility to translational suppression by SLFN11. Specific attenuation of tRNA-Leu-TAA sufficed to ablate ATR protein expression and restore the DDA sensitivity of SLFN11-deficient cells. Our study uncovered a novel mechanism of codon-specific translational inhibition via SLFN11-dependent tRNA cleavage in the DNA damage response and supports the notion that SLFN11-deficient tumor cells can be resensitized to DDAs by targeting ATR or tRNA-Leu-TAA.


Subject(s)
Cell Death/physiology , DNA Damage , Nuclear Proteins/metabolism , RNA, Transfer/metabolism , Ataxia Telangiectasia Mutated Proteins/biosynthesis , Ataxia Telangiectasia Mutated Proteins/genetics , Camptothecin/pharmacology , Cell Death/drug effects , Cell Death/genetics , Cell Line, Tumor , Codon/genetics , HEK293 Cells , Humans , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Protein Biosynthesis/drug effects , RNA, Small Interfering/genetics , RNA, Transfer/classification , RNA, Transfer/genetics , RNA, Transfer, Leu/genetics , RNA, Transfer, Leu/metabolism , Topoisomerase I Inhibitors/pharmacology
4.
Oncogene ; 37(47): 6152-6165, 2018 11.
Article in English | MEDLINE | ID: mdl-29995872

ABSTRACT

Epithelial cells aggregate into spheroids when deprived of matrix, and the proclivity for spheroid formation and survival is a hallmark of normal and tumorigenic mammary stem cells. We show here that Nuclear Respiratory Factor 1 (NRF1) is a spheroid promoter by in silico identification of this transcription factor as highly connected to top shRNA-hits deduced from re-iterative selections for shRNAs enriched in MCF10A spheroids. NRF1-promoted spheroid survival is linked to its stimulation of mitochondrial OXPHOS, cell migration, invasion, and mesenchymal transition. Conversely, NRF1 knockdown in breast cancer MDA-MB-231 cells reduced spheroids, migration, invasion, and mesenchymal marker expression. NRF1 knockdown also reduced tumor burden in mammary fat pads and lungs of orthotopic- or tail vein-transplanted mice. With the Luminal A subtype of breast cancer, higher NRF1 expression is associated with lower survival. These results show that NRF1, an activator of mitochondrial metabolism, supports mammary spheroid survival and tumor development.


Subject(s)
Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/physiology , Mammary Glands, Human/metabolism , Nuclear Respiratory Factor 1/metabolism , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/physiology , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/metabolism , RNA, Small Interfering/metabolism , Transcription Factors/metabolism
5.
Mol Biol Cell ; 29(18): 2228-2242, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29975106

ABSTRACT

Ionizing radiation (IR) not only activates DNA damage response (DDR) in irradiated cells but also induces bystander effects (BE) in cells not directly targeted by radiation. How DDR pathways activated in irradiated cells stimulate BE is not well understood. We show here that extracellular vesicles secreted by irradiated cells (EV-IR), but not those from unirradiated controls (EV-C), inhibit colony formation in unirradiated cells by inducing reactive oxygen species (ROS). We found that µEV-IR from Abl nuclear localization signal-mutated ( Abl-µNLS) cells could not induce ROS, but expression of wild-type Abl restored that activity. Because nuclear Abl stimulates miR-34c biogenesis, we measured miR-34c in EV and found that its levels correlated with the ROS-inducing activity of EV. We then showed that EV from miR-34c minigene-transfected, but unirradiated cells induced ROS; and transfection with miR-34c-mimic, without radiation or EV addition, also induced ROS. Furthermore, EV-IR from miR34-family triple-knockout cells could not induce ROS, whereas EV-IR from wild-type cells could cause miR-34c increase and ROS induction in the miR-34 triple-knockout cells. These results establish a novel role for extracellular vesicles in transferring nuclear Abl-dependent and radiation-induced miR-34c into unirradiated cells to cause bystander oxidative stress.


Subject(s)
Bystander Effect/radiation effects , Extracellular Vesicles/radiation effects , Fibroblasts/radiation effects , MicroRNAs/biosynthesis , Proto-Oncogene Proteins c-abl/metabolism , Animals , Cell Culture Techniques , DNA Damage , DNA Repair , Extracellular Vesicles/metabolism , Fibroblasts/metabolism , HEK293 Cells , Humans , Mice , Oxidative Stress/radiation effects , Radiation, Ionizing , Reactive Oxygen Species/metabolism , Signal Transduction/radiation effects
6.
Mol Cell Oncol ; 3(2)2016.
Article in English | MEDLINE | ID: mdl-27182551

ABSTRACT

The Microprocessor complex consisting of DROSHA (a type III ribonuclease) and DGCR8 (DiGeorge syndrome critical region gene 8-encoded RNA binding protein) recognizes and cleaves the precursor microRNA hairpin (pre-miRNA) from the primary microRNA transcript (pri-miRNA). The Abelson tyrosine kinase 1 (ABL) phosphorylates DGCR8 to stimulate the cleavage of a subset of pro-apoptotic pri-miRNAs, thus expanding the nuclear functions of ABL to include regulation of RNA processing.

7.
J Clin Pathol ; 69(7): 627-31, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26658220

ABSTRACT

AIMS: The macroH2A histone variants are epigenetic marks for inactivated chromatin. In this study, we examined the expression of macroH2A2 in anal neoplasm from anal intraepithelial neoplasia (AIN) to anal squamous cell carcinoma (SCC). METHODS: AIN and anal SCC samples were analysed for macroH2A2 expression, HIV and human papilloma virus (HPV). The association of macroH2A2 expression with clinical grade, disease recurrence, overall survival and viral involvement was determined. RESULTS: macroH2A2 was expressed in normal squamous tissue and lower grade AIN (I and II). Expression was lost in 38% of high-grade AIN (III) and 71% of anal SCC (p=0.002). Patients with AIN with macroH2A2-negative lesions showed earlier recurrence than those with macroH2A2-positive neoplasm (p=0.017). With anal SCC, macroH2A2 loss was more prevalent in the HPV-negative tumours. CONCLUSIONS: Loss of histone variant macroH2A2 expression is associated with the progression of anal neoplasm and can be used as a prognostic biomarker for high-grade AIN and SCC.


Subject(s)
Anus Neoplasms/metabolism , Carcinoma in Situ/metabolism , Carcinoma, Squamous Cell/metabolism , Histones/metabolism , Papillomavirus Infections/metabolism , Adult , Anus Neoplasms/genetics , Anus Neoplasms/pathology , Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Disease Progression , Female , Histones/genetics , Humans , Male , Middle Aged , Papillomaviridae , Papillomavirus Infections/genetics , Papillomavirus Infections/pathology
8.
Stem Cell Res ; 15(3): 678-693, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26561938

ABSTRACT

A definitive cure for chronic myeloid leukemia (CML) requires identifying novel therapeutic targets to eradicate leukemia stem cells (LSCs). However, the rarity of LSCs within the primitive hematopoietic cell compartment remains a major limiting factor for their study in humans. Here we show that primitive hematopoietic cells with typical LSC features, including adhesion defect, increased long-term survival and proliferation, and innate resistance to tyrosine kinase inhibitor (TKI) imatinib, can be generated de novo from reprogrammed primary CML cells. Using CML iPSC-derived primitive leukemia cells, we discovered olfactomedin 4 (OLFM4) as a novel factor that contributes to survival and growth of somatic lin(-)CD34(+) cells from bone marrow of patients with CML in chronic phase, but not primitive hematopoietic cells from normal bone marrow. Overall, this study shows the feasibility and advantages of using reprogramming technology to develop strategies for targeting primitive leukemia cells.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Humans , Induced Pluripotent Stem Cells/cytology , Tumor Cells, Cultured
9.
PLoS One ; 10(10): e0140585, 2015.
Article in English | MEDLINE | ID: mdl-26473951

ABSTRACT

Knockout serum replacement (KOSR) is a nutrient supplement commonly used to replace serum for culturing stem cells. We show here that KOSR has pro-survival activity in chronic myelogenous leukemia (CML) cells transformed by the BCR-ABL oncogene. Inhibitors of BCR-ABL tyrosine kinase kill CML cells by stimulating pro-apoptotic BIM and inhibiting anti-apoptotic BCL2, BCLxL and MCL1. We found that KOSR protects CML cells from killing by BCR-ABL inhibitors--imatinib, dasatinib and nilotinib. The protective effect of KOSR is reversible and not due to the selective outgrowth of drug-resistant clones. In KOSR-protected CML cells, imatinib still inhibited the BCR-ABL tyrosine kinase, reduced the phosphorylation of STAT, ERK and AKT, down-regulated BCL2, BCLxL, MCL1 and up-regulated BIM. However, these pro-apoptotic alterations failed to cause cytochrome c release from the mitochondria. With mitochondria isolated from KOSR-cultured CML cells, we showed that addition of recombinant BIM protein also failed to cause cytochrome c release. Besides the kinase inhibitors, KOSR could protect cells from menadione, an inducer of oxidative stress, but it did not protect cells from DNA damaging agents. Switching from serum to KOSR caused a transient increase in reactive oxygen species and AKT phosphorylation in CML cells that were protected by KOSR but not in those that were not protected by this nutrient supplement. Treatment of KOSR-cultured cells with the PH-domain inhibitor MK2206 blocked AKT phosphorylation, abrogated the formation of BIM-resistant mitochondria and stimulated cell death. These results show that KOSR has cell-context dependent pro-survival activity that is linked to AKT activation and the inhibition of BIM-induced cytochrome c release from the mitochondria.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Culture Media/pharmacology , Cytochromes c/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Membrane Proteins/metabolism , Mitochondria/enzymology , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Cell Line, Tumor , Cell Survival/drug effects , Dasatinib/pharmacology , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imatinib Mesylate/pharmacology , Membrane Proteins/genetics , Mice , Proto-Oncogene Proteins/genetics , Pyrimidines/pharmacology
10.
BMC Res Notes ; 8: 616, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26515650

ABSTRACT

BACKGROUND: Eukaryotic RNA polymerase II contains a C-terminal repeated domain (CTD) consisting of 52 consensus heptad repeats of Y1S2P3T4S5P6S7 that mediate interactions with many cellular proteins to regulate transcription elongation, RNA processing and chromatin structure. A number of CTD-binding proteins have been identified and the crystal structures of several protein-CTD complexes have demonstrated considerable conformational flexibility of the heptad repeats in those interactions. Furthermore, phosphorylation of the CTD at tyrosine, serine and threonine residues can regulate the CTD-protein interactions. Although the interactions of CTD with specific proteins have been elucidated at the atomic level, the capacity and specificity of the CTD-interactome in mammalian cells is not yet determined. RESULTS: A proteomic study was conducted to examine the mammalian CTD-interactome. We utilized six synthetic peptides each consisting of four consensus CTD-repeats with different combinations of serine and tyrosine phosphorylation as affinity-probes to pull-down nuclear proteins from HeLa cells. The pull-down fractions were then analyzed by MUDPIT mass spectrometry, which identified 100 proteins with the majority from the phospho-CTD pull-downs. Proteins pulled-down by serine-phosphorylated CTD-peptides included those containing the previously defined CTD-interacting domain (CID). Using SILAC mass spectrometry, we showed that the in vivo interaction of RNA polymerase II with the mammalian CID-containing RPRD1B is disrupted by CID mutation. We also showed that the CID from four mammalian proteins interacted with pS2-phosphorylated but not pY1pS2-doubly phosphorylated CTD-peptides. However, we also found proteins that were preferentially pulled-down by pY1pS2- or pY1pS5-doubly phosphorylated CTD-peptides. We prepared an antibody against tyrosine phosphorylated CTD and showed that ionizing radiation (IR) induced a transient increase in CTD tyrosine phosphorylation by immunoblotting. Combining SILAC and IMAC purification of phospho-peptides, we found that IR regulated the phosphorylation at four CTD tyrosine sites in different ways. CONCLUSION: Upon phosphorylation, the 52 repeats of the CTD have the capacity to generate a large number of binding sites for cellular proteins. This study confirms previous findings that serine phosphorylation stimulates whereas tyrosine phosphorylation inhibits the protein-binding activity of the CTD. However, tyrosine phosphorylation of the CTD can also stimulate other CTD-protein interactions. The CTD-peptide affinity pull-down method described here can be adopted to survey the mammalian CTD-interactome in various cell types and under different biological conditions.


Subject(s)
Cell Cycle Proteins/chemistry , Neoplasm Proteins/chemistry , Peptides/metabolism , RNA Polymerase II/chemistry , Serine/chemistry , Tyrosine/chemistry , Amino Acid Sequence , Binding Sites , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Gamma Rays , Gene Expression Regulation , HeLa Cells , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Peptides/chemical synthesis , Phosphorylation/radiation effects , Protein Binding/radiation effects , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Proteomics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Sequence Alignment , Serine/metabolism , Signal Transduction , Tyrosine/metabolism
11.
Sci Signal ; 8(383): ra64, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26126715

ABSTRACT

The DNA damage response network stimulates microRNA (miRNA) biogenesis to coordinate repair, cell cycle checkpoints, and apoptosis. The multistep process of miRNA biogenesis involves the cleavage of primary miRNAs by the microprocessor complex composed of the ribonuclease Drosha and the RNA binding protein DGCR8. We found that the tyrosine kinase ABL phosphorylated DGCR8, a modification that was required for the induction of a subset of miRNAs after DNA damage. Focusing on the miR-34 family, ABL stimulated the production of miR-34c, but not miR-34a, through Drosha/DGCR8-dependent processing of primary miR-34c (pri-miR-34c). This miRNA-selective effect of ABL required the sequences flanking the precursor miR-34c (pre-miR-34c) stem-loop. In pri-miRNA processing, DGCR8 binds the pre-miR stem-loop and recruits Drosha to the miRNA. RNA cross-linking assays showed that DGCR8 and Drosha interacted with pri-miR-34c, but we found an inverse correlation between ABL-stimulated processing and DGCR8 association with pri-miR-34c. When coexpressed in HEK293T cells, ABL phosphorylated DGCR8 at Tyr(267). Ectopic expression of a Y267F-DGCR8 mutant reduced the recruitment of Drosha to pri-miR-34c and prevented ABL or Drosha from stimulating the processing of pri-miR-34c. In mice engineered to express a nuclear import-defective mutant of ABL, miR-34c, but not miR-34a, expression was reduced in the kidney, and apoptosis of the renal epithelial cells was impaired in response to cisplatin. These results reveal a new pathway in the DNA damage response wherein ABL-dependent tyrosine phosphorylation of DGCR8 stimulates the processing of selective primary miRNAs.


Subject(s)
DNA Damage , MicroRNAs/metabolism , Oncogene Proteins v-abl/metabolism , RNA Processing, Post-Transcriptional/physiology , RNA-Binding Proteins/metabolism , Animals , Humans , Mice , MicroRNAs/genetics , Oncogene Proteins v-abl/genetics , Phosphorylation/physiology , RNA-Binding Proteins/genetics
12.
Reprod Sci ; 21(9): 1161-70, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24784716

ABSTRACT

Uterine fibroids are the most common solid tumors found in women of reproductive age. It has been reported that deregulation of the mammalian target of rapamycin (mTOR) pathway plays an important role in the etiology of leiomyoma. Here, we investigated the effect of rapamycin, an inhibitor of mTORC1, on the growth of primary fibroid smooth muscle cells (fSMCs) and human telomerase reverse transcriptase (hTERT)-transduced and immortalized fSMCs. With the primary fSMCs, a 24-hour treatment with rapamycin was sufficient to trigger a growth arrest that was not reversible upon drug removal. By contrast, the growth inhibitory effect of rapamycin on the hTERT-transduced fSMCs was readily reversible, as these cells resumed proliferation upon the withdrawal of the drug. These results suggest that rapamycin-induced irreversible growth arrest of fSMCs is dependent on the senescence barrier that is abrogated by the ectopic expression of telomerase.


Subject(s)
Growth Inhibitors/pharmacology , Leiomyoma/enzymology , Myocytes, Smooth Muscle/enzymology , Sirolimus/pharmacology , Telomerase/biosynthesis , Uterine Neoplasms/enzymology , Animals , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/physiology , Cell Line, Transformed , Cells, Cultured , Female , Growth Inhibitors/therapeutic use , Humans , Leiomyoma/drug therapy , Leiomyoma/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Sirolimus/antagonists & inhibitors , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Uterine Neoplasms/drug therapy , Uterine Neoplasms/pathology
13.
Am J Physiol Cell Physiol ; 307(2): C180-9, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24848114

ABSTRACT

Enteropathogenic Escherichia coli (EPEC) and Citrobacter rodentium are attaching-and-effacing (A/E) pathogens that cause intestinal inflammation and diarrhea. The bacteria adhere to the intestinal epithelium, destroy microvilli, and induce actin-filled membranous pedestals but do not invade the mucosa. Adherence leads to activation of several host cell kinases, including FYN, n-SRC, YES, ABL, and ARG, phosphorylation of the bacterial translocated intimin receptor, and actin polymerization and pedestal formation in cultured cells. However, marked functional redundancy appears to exist between kinases, and their physiological importance in A/E pathogen infections has remained unclear. To address this question, we employed a novel dynamic in vitro infection model that mimics transient and short-term interactions in the intestinal tract. Screening of a kinase inhibitor library and RNA interference experiments in vitro revealed that ABL and platelet-derived growth factor (PDGF) receptor (PDGFR) kinases, as well as p38 MAP kinase, have unique, indispensable roles in early attachment of EPEC to epithelial cells under dynamic infection conditions. Studies with mutant EPEC showed that the attachment functions of ABL and PDGFR were independent of the intimin receptor but required bacterial bundle-forming pili. Furthermore, inhibition of ABL and PDGFR with imatinib protected against infection of mice with modest loads of C. rodentium, whereas the kinases were dispensable for high inocula or late after infection. These results indicate that ABL and PDGFR have indispensable roles in early A/E pathogen attachment to intestinal epithelial cells and for in vivo infection with limiting inocula but are not required for late intimate bacterial attachment or high inoculum infections.


Subject(s)
Bacterial Adhesion/physiology , Enteropathogenic Escherichia coli/metabolism , Epithelial Cells/physiology , Oncogene Proteins v-abl/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Animals , Cell Line , Enteropathogenic Escherichia coli/cytology , Enteropathogenic Escherichia coli/physiology , Escherichia coli Infections/microbiology , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Oncogene Proteins v-abl/genetics , Protein Kinase Inhibitors/pharmacology , Receptors, Platelet-Derived Growth Factor/genetics
14.
PLoS One ; 9(4): e94147, 2014.
Article in English | MEDLINE | ID: mdl-24710071

ABSTRACT

A new method, which allows for the identification and prioritization of predicted cancer genes for future analysis, is presented. This method generates a gene-specific score called the "S-Score" by incorporating data from different types of analysis including mutation screening, methylation status, copy-number variation and expression profiling. The method was applied to the data from The Cancer Genome Atlas and allowed the identification of known and potentially new oncogenes and tumor suppressors associated with different clinical features including shortest term of survival in ovarian cancer patients and hormonal subtypes in breast cancer patients. Furthermore, for the first time a genome-wide search for genes that behave as oncogenes and tumor suppressors in different tumor types was performed. We envisage that the S-score can be used as a standard method for the identification and prioritization of cancer genes for follow-up studies.


Subject(s)
Computational Biology/methods , Genes, Neoplasm/genetics , Neoplasms/genetics , DNA Copy Number Variations/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Genomics , Humans , Methylation , Mutation/genetics , Oncogenes/genetics
15.
Mol Cell Biol ; 34(7): 1188-97, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24421390

ABSTRACT

The mammalian ABL1 gene encodes the ubiquitously expressed nonreceptor tyrosine kinase ABL. In response to growth factors, cytokines, cell adhesion, DNA damage, oxidative stress, and other signals, ABL is activated to stimulate cell proliferation or differentiation, survival or death, retraction, or migration. ABL also regulates specialized functions such as antigen receptor signaling in lymphocytes, synapse formation in neurons, and bacterial adhesion to intestinal epithelial cells. Although discovered as the proto-oncogene from which the Abelson leukemia virus derived its Gag-v-Abl oncogene, recent results have linked ABL kinase activation to neuronal degeneration. This body of knowledge on ABL seems confusing because it does not fit the one-gene-one-function paradigm. Without question, ABL capabilities are encoded by its gene sequence and that molecular blueprint designs this kinase to be regulated by subcellular location-dependent interactions with inhibitors and substrate activators. Furthermore, ABL shuttles between the nucleus and the cytoplasm where it binds DNA and actin--two biopolymers with fundamental roles in almost all biological processes. Taken together, the cumulated results from analyses of ABL structure-function, ABL mutant mouse phenotypes, and ABL substrates suggest that this tyrosine kinase does not have its own agenda but that, instead, it has evolved to serve a variety of tissue-specific and context-dependent biological functions.


Subject(s)
Genes, abl , Oncogene Proteins v-abl/physiology , Proto-Oncogene Proteins c-abl/physiology , Animals , Humans , Mice , Models, Biological , Mutation , Nuclear Localization Signals/genetics , Oncogene Proteins v-abl/antagonists & inhibitors , Oncogene Proteins v-abl/genetics , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Mas , Proto-Oncogene Proteins c-abl/deficiency , Proto-Oncogene Proteins c-abl/genetics , Signal Transduction
16.
PLoS One ; 8(10): e77495, 2013.
Article in English | MEDLINE | ID: mdl-24147007

ABSTRACT

TNF-Related Apoptosis Inducing Ligand (TRAIL) binds to and activates death receptors to stimulate caspase-8 and apoptosis with higher efficiency in cancer than normal cells but the development of apoptosis resistance has limited its clinical efficacy. We found that stable, but not transient knockdown of the ABL tyrosine kinase enhanced the apoptotic response to TRAIL. Re-expression of Abl, but not its nuclear import- or kinase-defective mutant, in the ABL-knockdown cells re-established apoptosis suppression. TRAIL is known to stimulate caspase-8 ubiquitination (Ub-C8), which can facilitate caspase-8 activation or degradation by the lysosomes. In the ABL-knockdown cells, we found a higher basal level of Ub-C8 that was not further increased by lysosomal inhibition. Re-expression of Abl in the ABL-knockdown cells reduced the basal Ub-C8, correlating with apoptosis suppression. We found that lysosomal inhibition by chloroquine (CQ) could also enhance TRAIL-induced apoptosis. However, this pro-apoptotic effect of CQ was lost in the ABL-knockdown cells but restored by Abl re-expression. Interestingly, kinase inhibition at the time of TRAIL stimulation was not sufficient to enhance apoptosis. Instead, persistent treatment for several days with imatinib, an ABL kinase inhibitor, was required to cause the enhanced and the CQ-insensitive apoptotic response to TRAIL. Together, these results show that persistent loss of nuclear ABL tyrosine kinase function can sensitize cells to TRAIL and suggest that long-term exposure to the FDA-approved ABL kinase inhibitors may potentiate apoptotic response to TRAIL-based cancer therapy.


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Chloroquine/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Caspase 8/metabolism , Cell Line , Cell Nucleus/metabolism , Enzyme Activation , Gene Expression , Gene Knockdown Techniques , Humans , Lysosomes/metabolism , Protein Transport , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Ubiquitination
17.
J Immunol ; 191(2): 837-47, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23772033

ABSTRACT

The p53 protein has not only important tumor suppressor activity but also additional immunological and other functions, whose nature and extent are just beginning to be recognized. In this article, we show that p53 has a novel inflammation-promoting action in the intestinal tract, because loss of p53 or the upstream activating kinase, ATM, protects against acute intestinal inflammation in murine models. Mechanistically, deficiency in p53 leads to increased survival of epithelial cells and lamina propria macrophages, higher IL-6 expression owing to enhanced glucose-dependent NF-κB activation, and increased mucosal STAT3 activation. Blockade or loss of IL-6 signaling reverses the protective effects of p53 deficiency. Conversely, IL-6 treatment protects against acute colitis in a manner dependent on STAT3 signaling and induction of cytoprotective factors in epithelial cells. Together, these results indicate that p53 promotes inflammation in the intestinal tract through suppression of epithelium-protective factors, thus significantly expanding the spectrum of physiological and immunological p53 activities unrelated to cancer formation.


Subject(s)
Colitis/immunology , Colitis/prevention & control , Inflammation/immunology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Ataxia Telangiectasia Mutated Proteins , Bone Marrow Cells/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Colitis/metabolism , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dendritic Cells/metabolism , Enzyme Activation , Epithelial Cells/metabolism , Inflammation/prevention & control , Interleukin-6/biosynthesis , Interleukin-6/pharmacology , Interleukins/biosynthesis , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Oxidative Stress , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Interleukin-22
18.
Mol Cancer Res ; 11(3): 207-18, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23339186

ABSTRACT

Tumor necrosis factor (TNF)-α promotes tumor development under chronic inflammation. Because TNF also activates caspase-8, selective inhibition of TNF-induced extrinsic apoptosis would be required for inflammation-associated tumor growth. In a mouse model of inflammation-associated colon carcinogenesis, we found nuclear expression of ß-catenin in tumors of wild-type, but not mutant, mice that were made resistant to TNF-induced apoptosis by a germline mutation blocking caspase cleavage of the retinoblastoma (RB) protein, despite similar frequencies of ß-catenin exon-3 mutations in these two genetic backgrounds. TNF-induced apoptosis was also attenuated in human colon cancer cell lines with genetically activated ß-catenin. However, we found that HCT116 cells, which contain an activated allele of ß-catenin but do not express nuclear ß-catenin, were sensitive to TNF-induced apoptosis. In HCT116 cells, TNF stimulated efficient RB cleavage that preceded chromatin condensation. In contrast, TNF did not induce RB cleavage in colon cancer cells expressing nuclear ß-catenin and these cells could be sensitized to basal and/or TNF-induced apoptosis by the knockdown of ß-catenin or RB. In the apoptosis-resistant colon cancer cells, knockdown of ß-catenin led to a reduction in the RB protein without affecting RB mRNA. Furthermore, ectopic expression of the caspase-resistant, but not the wild-type, RB re-established resistance to TNF-induced caspase activation in colon cancer cells without ß-catenin. Together, these results suggest that nuclear ß-catenin-dependent RB stabilization suppresses TNF-induced apoptosis in caspase-8-positive colon cancer cells.


Subject(s)
Apoptosis/genetics , Colonic Neoplasms/genetics , Retinoblastoma Protein/genetics , beta Catenin/genetics , Animals , Caspase 8/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HCT116 Cells , Humans , Mice , Retinoblastoma Protein/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , beta Catenin/metabolism
19.
Mol Biol Cell ; 24(4): 465-73, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23264463

ABSTRACT

The Wnt/ß-catenin pathway is constitutively activated in more than 90% of human colorectal cancer. Activated ß-catenin stimulates cell proliferation and survival, however, its antiapoptotic mechanisms are not fully understood. We show here that activated ß-catenin is required to suppress caspase-8 activation, but only in colon cancer cells that are resistant to tumor necrosis factor-α (TNF)-induced apoptosis. We found that lysosomal delivery of internalized TNF occurred at a faster pace in apoptosis-resistant than in apoptosis-sensitive colon cancer cells. Retardation of endosomal trafficking through vacuolar ATPase (V-ATPase) inhibition enhanced caspase-8 activation in apoptosis-resistant but not apoptosis-sensitive cells. Interestingly, knockdown of ß-catenin also prolonged TNF association with the early endosome and enhanced caspase-8 activation in apoptosis-resistant but not apoptosis-sensitive colon cancer cells. In a mouse model of inflammation-associated colon tumors, we found nuclear expression of ß-catenin, resistance to TNF-induced apoptosis, and reactivation of apoptosis in vivo after cotreatment of TNF with a V-ATPase inhibitor. Together these results suggest that activated ß-catenin can facilitate endosomal trafficking of internalized TNF to suppress caspase-8 activation in colon cancer cells.


Subject(s)
Caspase 8/genetics , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Lysosomes/drug effects , Tumor Necrosis Factor-alpha/pharmacology , beta Catenin/genetics , Animals , Apoptosis/drug effects , Caspase 8/metabolism , Cell Proliferation/drug effects , Colonic Neoplasms/chemically induced , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Endocytosis/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Lysosomes/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Transport/drug effects , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL