Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
J Biol Chem ; 299(7): 104884, 2023 07.
Article in English | MEDLINE | ID: mdl-37269946

ABSTRACT

The unfolded protein response (UPR) is sensitive to proteotoxic and membrane bilayer stress, both of which are sensed by the ER protein Ire1. When activated, Ire1 splices HAC1 mRNA, producing a transcription factor that targets genes involved in proteostasis and lipid metabolism, among others. The major membrane lipid phosphatidylcholine (PC) is subject to phospholipase-mediated deacylation, producing glycerophosphocholine (GPC), followed by reacylation of GPC through the PC deacylation/reacylation pathway (PC-DRP). The reacylation events occur via a two-step process catalyzed first by the GPC acyltransferase Gpc1, followed by acylation of the lyso-PC molecule by Ale1. However, whether Gpc1 is critical for ER bilayer homeostasis is unclear. Using an improved method for C14-choline-GPC radiolabeling, we first show that loss of Gpc1 results in abrogation of PC synthesis through PC-DRP and that Gpc1 colocalizes with the ER. We then probe the role of Gpc1 as both a target and an effector of the UPR. Exposure to the UPR-inducing compounds tunicamycin, DTT, and canavanine results in a Hac1-dependent increase in GPC1 message. Further, cells lacking Gpc1 exhibit increased sensitivity to those proteotoxic stressors. Inositol limitation, known to induce the UPR via bilayer stress, also induces GPC1 expression. Finally, we show that loss of GPC1 induces the UPR. A gpc1Δ mutant displays upregulation of the UPR in strains expressing a mutant form of Ire1 that is unresponsive to unfolded proteins, indicating that bilayer stress is responsible for the observed upregulation. Collectively, our data indicate an important role for Gpc1 in yeast ER bilayer homeostasis.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Acyltransferases/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Phosphatidylcholines/metabolism , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Unfolded Protein Response
3.
J Am Chem Soc ; 137(5): 2056-66, 2015 Feb 11.
Article in English | MEDLINE | ID: mdl-25629952

ABSTRACT

Nanomaterials have great potential to offer effective treatment against devastating diseases by providing sustained release of high concentrations of therapeutic agents locally, especially when the route of administration allows for direct access to the diseased tissues. Biodegradable polyphosphoester-based polymeric micelles and shell cross-linked knedel-like nanoparticles (SCKs) have been designed from amphiphilic block-graft terpolymers, PEBP-b-PBYP-g-PEG, which effectively incorporate high concentrations of paclitaxel (PTX). Well-dispersed nanoparticles physically loaded with PTX were prepared, exhibiting desirable physiochemical characteristics. Encapsulation of 10 wt% PTX, into either micelles or SCKs, allowed for aqueous suspension of PTX at concentrations up to 4.8 mg/mL, as compared to <2.0 µg/mL for the aqueous solubility of the drug alone. Drug release studies indicated that PTX released from these nanostructures was defined through a structure-function relationship, whereby the half-life of sustained PTX release was doubled through cross-linking of the micellar structure to form SCKs. In vitro, physically loaded micellar and SCK nanotherapeutics demonstrated IC50 values against osteosarcoma cell lines, known to metastasize to the lungs (CCH-OS-O and SJSA), similar to the pharmaceutical Taxol formulation. Evaluation of these materials in vivo has provided an understanding of the effects of nanoparticle structure-function relationships on intratracheal delivery and related biodistribution and pharmacokinetics. Overall, we have demonstrated the potential of these novel nanotherapeutics toward future sustained release treatments via administration directly to the sites of lung metastases of osteosarcoma.


Subject(s)
Drug Carriers/chemistry , Nanoparticles/chemistry , Paclitaxel/chemistry , Polyethylene Glycols/chemistry , Polymers/chemistry , Alkynes/chemistry , Animals , Azides/chemistry , Bone Neoplasms/pathology , Catalysis , Cell Line, Tumor , Copper/chemistry , Drug Carriers/metabolism , Drug Carriers/pharmacokinetics , Drug Liberation , Esters , Half-Life , Humans , Hydrophobic and Hydrophilic Interactions , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Micelles , Models, Molecular , Molecular Conformation , Osteosarcoma/pathology , Polymers/metabolism , Polymers/pharmacokinetics , Tissue Distribution
4.
PLoS One ; 8(8): e70608, 2013.
Article in English | MEDLINE | ID: mdl-23936456

ABSTRACT

The presence of the Philadelphia chromosome in patients with acute lymphoblastic leukemia (Ph(+)ALL) is a negative prognostic indicator. Tyrosine kinase inhibitors (TKI) that target BCR/ABL, such as imatinib, have improved treatment of Ph(+)ALL and are generally incorporated into induction regimens. This approach has improved clinical responses, but molecular remissions are seen in less than 50% of patients leaving few treatment options in the event of relapse. Thus, identification of additional targets for therapeutic intervention has potential to improve outcomes for Ph+ALL. The human epidermal growth factor receptor 2 (ErbB2) is expressed in ~30% of B-ALLs, and numerous small molecule inhibitors are available to prevent its activation. We analyzed a cohort of 129 ALL patient samples using reverse phase protein array (RPPA) with ErbB2 and phospho-ErbB2 antibodies and found that activity of ErbB2 was elevated in 56% of Ph(+)ALL as compared to just 4.8% of Ph(-)ALL. In two human Ph+ALL cell lines, inhibition of ErbB kinase activity with canertinib resulted in a dose-dependent decrease in the phosphorylation of an ErbB kinase signaling target p70S6-kinase T389 (by 60% in Z119 and 39% in Z181 cells at 3 µM). Downstream, phosphorylation of S6-kinase was also diminished in both cell lines in a dose-dependent manner (by 91% in both cell lines at 3 µM). Canertinib treatment increased expression of the pro-apoptotic protein Bim by as much as 144% in Z119 cells and 49% in Z181 cells, and further produced caspase-3 activation and consequent apoptotic cell death. Both canertinib and the FDA-approved ErbB1/2-directed TKI lapatinib abrogated proliferation and increased sensitivity to BCR/ABL-directed TKIs at clinically relevant doses. Our results suggest that ErbB signaling is an additional molecular target in Ph(+)ALL and encourage the development of clinical strategies combining ErbB and BCR/ABL kinase inhibitors for this subset of ALL patients.


Subject(s)
Apoptosis/drug effects , ErbB Receptors/antagonists & inhibitors , Philadelphia Chromosome/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Adolescent , Adult , Aged , Aged, 80 and over , Caspase 3/metabolism , Cell Proliferation/drug effects , Enzyme Activation/drug effects , ErbB Receptors/metabolism , Female , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Middle Aged , Molecular Targeted Therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Young Adult
5.
Mol Cancer Res ; 11(9): 1017-28, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23723077

ABSTRACT

UNLABELLED: Malignant cell transformation, invasion, and metastasis are dependent on the coordinated rewiring of gene expression. A major component in the scaffold of these reprogramming events is one in which epithelial cells lose intercellular connections and polarity to adopt a more motile mesenchymal phenotype, which is largely supported by a robust transcriptional machinery consisting mostly of developmental transcription factors. This study demonstrates that the winged helix transcription factor, FOXQ1, contributes to this rewiring process, in part by directly modulating the transcription of TWIST1, itself a key mediator of metastasis that transcriptionally regulates the expression of important molecules involved in epithelial-to-mesenchymal transition. Forced expression and RNA-mediated silencing of FOXQ1 led to enhanced and suppressed mRNA and protein levels of TWIST1, respectively. Mechanistically, FOXQ1 enhanced the reporter activity of TWIST1 and directly interacted with its promoter. Furthermore, enhanced expression of FOXQ1 resulted in increased migration and invasion in colorectal cancer cell lines, whereas knockdown studies showed the opposite effect. Moreover, using the in vivo chicken chorioallantoic membrane metastasis assay model, FOXQ1 significantly enhanced distant metastasis with minimal effects on tumor growth. IMPLICATIONS: These findings reveal FOXQ1 as a modulator of TWIST1-mediated metastatic phenotypes and support its potential as a biomarker of metastasis.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Forkhead Transcription Factors/metabolism , Neoplasm Metastasis , Nuclear Proteins/metabolism , Twist-Related Protein 1/metabolism , Animals , Base Sequence , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Dogs , Epithelial Cells/metabolism , Epithelial Cells/pathology , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Madin Darby Canine Kidney Cells , Molecular Sequence Data , Neoplasm Invasiveness , Neoplasms, Experimental , Nuclear Proteins/genetics , Sequence Alignment , Twist-Related Protein 1/genetics
6.
Mol Cancer ; 11: 38, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22682314

ABSTRACT

BACKGROUND: Tri- and tetra-nucleotide repeats in mammalian genomes can induce formation of alternative non-B DNA structures such as triplexes and guanine (G)-quadruplexes. These structures can induce mutagenesis, chromosomal translocations and genomic instability. We wanted to determine if proteins that bind triplex DNA structures are quantitatively or qualitatively different between colorectal tumor and adjacent normal tissue and if this binding activity correlates with patient clinical characteristics. METHODS: Extracts from 63 human colorectal tumor and adjacent normal tissues were examined by gel shifts (EMSA) for triplex DNA-binding proteins, which were correlated with clinicopathological tumor characteristics using the Mann-Whitney U, Spearman's rho, Kaplan-Meier and Mantel-Cox log-rank tests. Biotinylated triplex DNA and streptavidin agarose affinity binding were used to purify triplex-binding proteins in RKO cells. Western blotting and reverse-phase protein array were used to measure protein expression in tissue extracts. RESULTS: Increased triplex DNA-binding activity in tumor extracts correlated significantly with lymphatic disease, metastasis, and reduced overall survival. We identified three multifunctional splicing factors with biotinylated triplex DNA affinity: U2AF65 in cytoplasmic extracts, and PSF and p54nrb in nuclear extracts. Super-shift EMSA with anti-U2AF65 antibodies produced a shifted band of the major EMSA H3 complex, identifying U2AF65 as the protein present in the major EMSA band. U2AF65 expression correlated significantly with EMSA H3 values in all extracts and was higher in extracts from Stage III/IV vs. Stage I/II colon tumors (p=0.024). EMSA H3 values and U2AF65 expression also correlated significantly with GSK3 beta, beta-catenin, and NF- B p65 expression, whereas p54nrb and PSF expression correlated with c-Myc, cyclin D1, and CDK4. EMSA values and expression of all three splicing factors correlated with ErbB1, mTOR, PTEN, and Stat5. Western blots confirmed that full-length and truncated beta-catenin expression correlated with U2AF65 expression in tumor extracts. CONCLUSIONS: Increased triplex DNA-binding activity in vitro correlates with lymph node disease, metastasis, and reduced overall survival in colorectal cancer, and increased U2AF65 expression is associated with total and truncated beta-catenin expression in high-stage colorectal tumors.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , DNA-Binding Proteins/metabolism , Proteomics , Colorectal Neoplasms/pathology , DNA/metabolism , DNA-Binding Proteins/genetics , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lymph Nodes/pathology , Male , Neoplasm Metastasis , Neoplasm Staging , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Octamer Transcription Factors/genetics , Octamer Transcription Factors/metabolism , PTB-Associated Splicing Factor , Protein Binding , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RecQ Helicases/genetics , RecQ Helicases/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Splicing Factor U2AF , Werner Syndrome Helicase , beta Catenin/genetics , beta Catenin/metabolism
7.
Biosci Rep ; 31(3): 185-97, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20815812

ABSTRACT

Curcumin has promising potential in cancer prevention and therapy by interacting with proteins and modifying their expression and activity, which includes transcription factors, inflammatory cytokines and factors of cell survival, proliferation and angiogenesis. miR-21 is overexpressed in many tumours, promoting progression and metastasis. In the present study, we examined the potential of curcumin to regulate miR-21, tumour growth, invasion and in vivo metastasis in colorectal cancer. In Rko and HCT116 cells, we identified two new transcriptional start sites of the miR-21 gene and delineated its promoter region. PMA stimulation induced miR-21 expression via motifs bound with AP-1 (activator protein 1) transcription factors. Curcumin treatment reduced miR-21 promoter activity and expression in a dose-dependent manner by inhibiting AP-1 binding to the promoter, and induced the expression of the tumour suppressor Pdcd4 (programmed cell death protein 4), which is a target of miR-21. Curcumin-treated Rko and HCT116 cells were arrested in the G2/M phase with increasing concentrations. Furthermore, curcumin inhibited tumour growth, invasion and in vivo metastasis in the chicken-embryo-metastasis assay [CAM (chorionallantoic membrane) assay]. Additionally, curcumin significantly inhibited miR-21 expression in primary tumours generated in vivo in the CAM assay by Rko and HCT116 cells (P<0.00006 and P<0.035 respectively). Taken together, this is the first paper to show that curcumin inhibits the transcriptional regulation of miR-21 via AP-1, suppresses cell proliferation, tumour growth, invasion and in vivo metastasis, and stabilizes the expression of the tumour suppressor Pdcd4 in colorectal cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/secondary , Curcumin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , MicroRNAs/genetics , Neoplasm Invasiveness/prevention & control , Animals , Antineoplastic Agents/therapeutic use , Apoptosis Regulatory Proteins/metabolism , Cell Cycle/drug effects , Chick Embryo , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Curcumin/therapeutic use , HCT116 Cells , Humans , MicroRNAs/antagonists & inhibitors , Neoplasm Metastasis/drug therapy , RNA-Binding Proteins/metabolism , Transcriptional Activation/drug effects
8.
Cancer Res ; 69(6): 2461-70, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19276367

ABSTRACT

Cetuximab, which blocks ligand binding to epidermal growth factor receptor (EGFR), is currently being studied as a novel treatment for non-small cell lung cancer (NSCLC). However, its mechanisms of action toward metastasis, and markers of drug sensitivity, have not been fully elucidated. This study was conducted to (a) determine the effect of Cetuximab on invasion and NSCLC-metastasis; (b) investigate urokinase-type plasminogen activator receptor (u-PAR), a major molecule promoting invasion and metastasis, as a target molecule; (c) delineate molecular mediators of Cetuximab-induced metastasis inhibition; and (d) identify biomarkers of drug sensitivity in NSCLC. Cetuximab treatment resulted in reduced growth and Matrigel invasion of H1395 and A549 NSCLC cell lines, in parallel with reduced u-PAR mRNA and protein. u-PAR down-regulation was brought about by suppressing the binding of JunD and c-Jun to u-PAR promoter motif -190/-171 in vivo, and an inhibition of MAP/ERK kinase signaling. Furthermore, Cetuximab inhibited NSCLC proliferation and metastasis to distant organs in vivo as indicated by the chicken embryo metastasis assay. Low E-cadherin and high u-PAR, but not EGFR, was associated with resistance to Cetuximab in seven NSCLC cell lines. Furthermore, siRNA knockdown of u-PAR led to a resensitization to Cetuximab. Moreover, low E-cadherin and high u-PAR was found in 63% of resected tumor tissues of NSCLC patients progressing under Cetuximab therapy. This is the first study to show u-PAR as a target and marker of sensitivity to Cetuximab, and to delineate novel mechanisms leading to metastasis suppression of NSCLC by Cetuximab.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/biosynthesis , Cadherins/biosynthesis , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Receptors, Urokinase Plasminogen Activator/biosynthesis , Animals , Antibodies, Monoclonal, Humanized , Biomarkers, Tumor/genetics , Cadherins/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cetuximab , Chick Embryo , Epidermal Growth Factor/antagonists & inhibitors , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Gene Expression , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MAP Kinase Signaling System , Neoplasm Metastasis , Promoter Regions, Genetic , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Urokinase Plasminogen Activator/genetics
9.
J Biol Chem ; 279(23): 24323-33, 2004 Jun 04.
Article in English | MEDLINE | ID: mdl-15044472

ABSTRACT

The Saccharomyces cerevisiae protein Stm1 was originally identified as a G4 quadruplex and purine motif triplex nucleic acid-binding protein. However, more recent studies have suggested a role for Stm1p in processes ranging from antiapoptosis to telomere maintenance. To better understand the biological role of Stm1p and its potential for G(*)G multiplex binding, we used epitope-tagged protein and immunological methods to identify the subcellular localization and protein and nucleic acid partners of Stm1p in vivo. Indirect immunofluorescence microscopy indicated that Stm1p is primarily a cytoplasmic protein, although a small percentage is also present in the nucleus. Conventional immunoprecipitation found that Stm1p is associated with ribosomal proteins and rRNA. This association was verified by rate zonal separation through sucrose gradients, which showed that Stm1p binds exclusively to mature 80 S ribosomes and polysomes. Chromatin immunoprecipitation experiments found that Stm1p preferentially binds telomere-proximal Y' element DNA sequences. Taken together, our data suggest that Stm1p is primarily a ribosome-associated protein, but one that can also interact with DNA, especially subtelomeric sequences. We discuss the implications of our findings in relation to prior genetic, genomic, and proteomic studies that have identified STM1 and/or Stm1p as well as the possible biological role of Stm1p.


Subject(s)
Membrane Proteins/chemistry , Membrane Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/metabolism , Amino Acid Motifs , Blotting, Western , Cell Nucleus/metabolism , Centrifugation, Density Gradient , Chromatin/chemistry , Cytoplasm/metabolism , Electrophoresis, Polyacrylamide Gel , Epitopes , Fluorescent Antibody Technique, Indirect , Microscopy, Fluorescence , Polyribosomes/chemistry , Precipitin Tests , Protein Binding , Purines/chemistry , RNA, Ribosomal/chemistry , Ribosomes/chemistry , Ribosomes/metabolism , Telomere/chemistry , Telomere/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL