Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Mol Cancer Ther ; 16(9): 1866-1876, 2017 09.
Article in English | MEDLINE | ID: mdl-28522588

ABSTRACT

Neutropenia is a common adverse event in cancer patients treated with antibody-drug conjugates (ADC) and we aimed to elucidate the potential mechanism of this toxicity. To investigate whether ADCs affect neutrophil production from bone marrow, an in vitro assay was developed in which hematopoietic stem cells (HSC) were differentiated to neutrophils. Several antibodies against targets absent in HSCs and neutrophils were conjugated to MMAE via a cleavable valine-citrulline linker (vcMMAE-ADC) or MMAF via a noncleavable maleimidocaproyl linker (mcMMAF-ADC), and their cytotoxicity was tested in the neutrophil differentiation assay. Results showed that HSCs had similar sensitivity to vcMMAE-ADCs and mcMMAF-ADCs; however, vcMMAE-ADCs were more cytotoxic to differentiating neutrophils than the same antibody conjugated to mcMMAF. This inhibitory effect was not mediated by internalization of ADC either by macropinocytosis or FcγRs. Our results suggested that extracellular proteolysis of the cleavable valine-citrulline linker is responsible for the cytotoxicity to differentiating neutrophils. Mass spectrometry analyses indicated that free MMAE was released from vcMMAE-ADCs in the extracellular compartment when they were incubated with differentiating neutrophils or neutrophil conditioned medium, but not with HSC-conditioned medium. Using different protease inhibitors, our data suggested that serine, but not cysteine proteases, were responsible for the cleavage. In vitro experiments demonstrated that the purified serine protease, elastase, was capable of releasing free MMAE from a vcMMAE-ADC. Here we propose that ADCs containing protease cleavable linkers can contribute to neutropenia via extracellular cleavage mediated by serine proteases secreted by differentiating neutrophils in bone marrow. Mol Cancer Ther; 16(9); 1866-76. ©2017 AACRSee related article by Zhao et al., p. 1877.


Subject(s)
Antineoplastic Agents/adverse effects , Immunoconjugates/adverse effects , Myelopoiesis/drug effects , Neutropenia/blood , Neutropenia/etiology , Neutrophils/drug effects , Animals , Biomarkers , Cell Differentiation/drug effects , Cell Proliferation , Cell Survival/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Mice , Neutrophils/metabolism , Pinocytosis , Receptors, IgG/metabolism , Serine Proteases/metabolism
2.
Mol Cancer Ther ; 15(11): 2679-2687, 2016 11.
Article in English | MEDLINE | ID: mdl-27550944

ABSTRACT

Here, we report the development of an antibody-drug conjugate, ASG-5ME, which targets the solute carrier receptor SLC44A4. SLC44A4 is a member of a family of putative choline transporters that we show to be markedly upregulated in a variety of epithelial tumors, most notably prostate and pancreatic cancer. SLC44A4 is normally expressed on the apical surface of secretory epithelial cells, but in cancer we show expression is not restricted to the luminal surface in advanced and undifferentiated tumors. ASG-5ME consists of a human IgG2 anti-SLC44A4 antibody conjugated through a cleavable linker to the microtubule-disrupting agent monomethylauristatin E. It has potent antitumor activity in both cell line - and patient-derived xenograft models of pancreatic and prostate cancers. Combination studies with ASG-5ME and nab-paclitaxel demonstrated combination effect in both pancreatic and prostate tumor models. Altogether, the data presented here suggest that ASG-5ME may have the potential to offer a new therapeutic option for the treatment of pancreatic and prostate cancers. Mol Cancer Ther; 15(11); 2679-87. ©2016 AACR.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma/metabolism , Immunoconjugates/pharmacology , Membrane Transport Proteins/metabolism , Oligopeptides/pharmacology , Pancreatic Neoplasms/metabolism , Prostatic Neoplasms/metabolism , Animals , Carcinoma/drug therapy , Carcinoma/genetics , Carcinoma/pathology , Cell Line, Tumor , Disease Models, Animal , Drug Evaluation, Preclinical , Gene Expression Profiling , Humans , Male , Mice , Molecular Targeted Therapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
3.
PLoS One ; 9(11): e111515, 2014.
Article in English | MEDLINE | ID: mdl-25369402

ABSTRACT

Discoidin domain receptor 1 (DDR1) is a member of the receptor tyrosine kinase family. The receptor is activated upon binding to its ligand, collagen, and plays a crucial role in many fundamental processes such as cell differentiation, adhesion, migration and invasion. Although DDR1 is expressed in many normal tissues, upregulated expression of DDR1 in a variety of human cancers such as lung, colon and brain cancers is known to be associated with poor prognosis. Using shRNA silencing, we assessed the oncogenic potential of DDR1. DDR1 knockdown impaired tumor cell proliferation and migration in vitro and tumor growth in vivo. Microarray analysis of tumor cells demonstrated upregulation of TGFBI expression upon DDR1 knockdown, which was subsequently confirmed at the protein level. TGFBI is a TGFß-induced extracellular matrix protein secreted by the tumor cells and is known to act either as a tumor promoter or tumor suppressor, depending on the tumor environment. Here, we show that exogenous addition of recombinant TGFBI to BXPC3 tumor cells inhibited clonogenic growth and migration, thus recapitulating the phenotypic effect observed from DDR1 silencing. BXPC3 tumor xenografts demonstrated reduced growth with DDR1 knockdown, and the same xenograft tumors exhibited an increase in TGFBI expression level. Together, these data suggest that DDR1 expression level influences tumor growth in part via modulation of TGFBI expression. The reciprocal expression of DDR1 and TGFBI may help to elucidate the contribution of DDR1 in tumorigenesis and TGFBI may also be used as a biomarker for the therapeutic development of DDR1 specific inhibitors.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/metabolism , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Neoplastic , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Mitogen/metabolism , Transforming Growth Factor beta/genetics , Animals , Carcinogenesis/pathology , Cell Line, Tumor , Discoidin Domain Receptors , Humans , Mice, SCID , RNA Interference , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/genetics , Signal Transduction
4.
Exp Cell Res ; 322(1): 39-50, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24462598

ABSTRACT

Recent evidences indicated Nrf2 is more potent than Nrf1 in the activation of antioxidant genes. However, the roles of Nrf proteins in the regulation of copper-responsive transcription have not been well addressed. We took the toxicogenomic approach and the present network and Gene Ontology analyses results showed that Nrf1 and Nrf2 are distinctively involved in copper-responsive transcriptional regulation in HepG2 transcriptome. Cells deficient in either Nrf1 or Nrf2 were more susceptible to copper exposure than wild type cells. Nrf1 and Nrf2 null cells were transfected with the luciferase reporters containing either ARE(s) or a combination of ARE(s) and MREs, and then treated with copper. In Nrf2-null (Nrf2(-/-)) cells, copper did not activate transcription of reporter genes, whereas Nrf1 deficiency did not affect copper-inducible activation. Ectopic expression of Nrf2 restored copper-inducible transcription in Nrf2(-/-) cells. However, the changes in the intrinsic mRNA levels of MT-1 in Nrf null cells following copper treatment showed that Nrf1 and Nrf2 equally contributed to MT-1 activation after 4h, while Nrf1involved more than Nrf2 following 24h exposure. These results suggest that while Nrf2 is crucial for MRE/ARE-mediated transcription in response to copper, Nrf1 may activate MT-1 expression by a mechanism different from that Nrf2 employs.


Subject(s)
Copper/pharmacology , Gene Expression Regulation/drug effects , NF-E2-Related Factor 2/physiology , Nuclear Respiratory Factor 1/physiology , Animals , Cells, Cultured , Gene Expression Profiling , Gene Regulatory Networks , Hep G2 Cells , Humans , Metallothionein/genetics , Metallothionein/metabolism , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , Nuclear Respiratory Factor 1/genetics , Transcription, Genetic/drug effects
5.
PLoS One ; 6(9): e24004, 2011.
Article in English | MEDLINE | ID: mdl-21912659

ABSTRACT

BACKGROUND: Studies suggest that micronutrients may modify the risk or delay progression of prostate cancer; however, the molecular mechanisms involved are poorly understood. We examined the effects of lycopene and fish oil on prostate gene expression in a double-blind placebo-controlled randomized clinical trial. METHODS: Eighty-four men with low risk prostate cancer were stratified based on self-reported dietary consumption of fish and tomatoes and then randomly assigned to a 3-month intervention of lycopene (n = 29) or fish oil (n = 27) supplementation or placebo (n = 28). Gene expression in morphologically normal prostate tissue was studied at baseline and at 3 months via cDNA microarray analysis. Differential gene expression and pathway analyses were performed to identify genes and pathways modulated by these micronutrients. RESULTS: Global gene expression analysis revealed no significant individual genes that were associated with high intake of fish or tomato at baseline or after 3 months of supplementation with lycopene or fish oil. However, exploratory pathway analyses of rank-ordered genes (based on p-values not corrected for multiple comparisons) revealed the modulation of androgen and estrogen metabolism in men who routinely consumed more fish (p = 0.029) and tomato (p = 0.008) compared to men who ate less. In addition, modulation of arachidonic acid metabolism (p = 0.01) was observed after 3 months of fish oil supplementation compared with the placebo group; and modulation of nuclear factor (erythroid derived-2) factor 2 or Nrf2-mediated oxidative stress response for either supplement versus placebo (fish oil: p = 0.01, lycopene: p = 0.001). CONCLUSIONS: We did not detect significant individual genes associated with dietary intake and supplementation of lycopene and fish oil. However, exploratory analyses revealed candidate in vivo pathways that may be modulated by these micronutrients. TRIAL REGISTRATION: ClinicalTrials.gov NCT00402285.


Subject(s)
Carotenoids/pharmacology , Dietary Supplements , Fish Oils/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Carotenoids/therapeutic use , Diet , Double-Blind Method , Fish Oils/therapeutic use , Humans , Lycopene , Male , Oligonucleotide Array Sequence Analysis , Placebos , Prostatic Neoplasms/drug therapy
6.
J Biochem Mol Toxicol ; 24(5): 330-4, 2010.
Article in English | MEDLINE | ID: mdl-20979157

ABSTRACT

Metallothioneins (MTs) protect cells from oxidative damage by scavenging reactive oxygen species (ROS). Concurrent with protecting cells from ROS-mediated damage, MT transcription is induced by ROS. ROS activate transcription by affecting several signal transduction pathways, many of which have been implicated in regulating MT transcription. ROS-activated intracellular signaling is mediated by the stable lipid peroxide 4-hydroxynonenal (HNE). After determining the level of sensitivity of Hepa 1-6 cells to HNE, MT-1 mRNA expression was shown to be induced in a concentration and time-dependent manner after HNE exposure. Finally, using MT-based reporters, HNE was found to induce MT transcription via both antioxidant response and metal response elements. Thus, ROS may activate MT transcription by generating HNE that in turn affects signaling pathways that regulate MT transcription via the transcription factors AP-1 and MTF-1.


Subject(s)
Aldehydes/pharmacology , Metallothionein/genetics , Transcriptional Activation/genetics , Aldehydes/metabolism , Animals , Antioxidants/pharmacology , COS Cells , Cell Line , Chlorocebus aethiops , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Hepatocytes/cytology , Hepatocytes/metabolism , Lipid Peroxides/metabolism , Liver/metabolism , Metallothionein/biosynthesis , Metallothionein/metabolism , Metals/metabolism , Mice , Models, Biological , Oxidation-Reduction , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Response Elements/genetics , Signal Transduction/physiology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factor MTF-1
7.
J Mol Biol ; 383(5): 1008-18, 2008 Nov 28.
Article in English | MEDLINE | ID: mdl-18793645

ABSTRACT

Copper is an essential metal that is able to produce reactive oxygen species and to induce intracellular oxidative stress. Several studies have examined the effects of excessive copper and oxidative stress on various organisms and tissues, but few have addressed the molecular mechanisms by which copper affects transcription. Our results demonstrated that, in COS-7 cells, copper treatment caused an increase in the binding of nuclear proteins to activating protein-1 and antioxidant response elements. The level of copper-inducible nuclear protein binding was modulated by increasing or decreasing the level of intracellular oxidative stress. Copper exposure also led to an increase in the steady-state levels of c-fos, c-jun, and c-myc mRNAs. Exposure to copper resulted in an increase in the levels of phosphorylation and activation of the c-Jun N-terminal kinase/stress-activated protein kinase and p38 pathways. The activation of these pathways resulted in a concomitant increase in c-Jun phosphorylation. We investigated the hypothesis that copper-induced oxidative stress leads to the formation of stable lipid peroxidation by-products that activate mitogen-activated protein kinase (MAPK) pathways, ultimately affecting transcription. While exposure did result in the production of 4-hydroxynonenal, the timing of the increased levels of proto-oncogene mRNA, phosphorylation of c-jun, and phosphorylation and activation of MAPKs, as well as the inability of the lipophilic antioxidant vitamin E to abrogate MAPK phosphorylation, suggest that the formation of stable lipid peroxidation by-products may not be the primary mechanism by which copper activates MAPKs. These results further elucidate the effects of copper on signal transduction pathways to alter gene expression.


Subject(s)
Copper/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Transcription Factor AP-1/metabolism , Transcription, Genetic/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Aldehydes/metabolism , Animals , COS Cells , Chlorocebus aethiops , Extracellular Signal-Regulated MAP Kinases/metabolism , Lipid Peroxidation/drug effects , Metallothionein/genetics , Metallothionein/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements/genetics , Vitamin E/pharmacology
8.
Proc Natl Acad Sci U S A ; 105(24): 8369-74, 2008 Jun 17.
Article in English | MEDLINE | ID: mdl-18559852

ABSTRACT

Epidemiological and prospective studies indicate that comprehensive lifestyle changes may modify the progression of prostate cancer. However, the molecular mechanisms by which improvements in diet and lifestyle might affect the prostate microenvironment are poorly understood. We conducted a pilot study to examine changes in prostate gene expression in a unique population of men with low-risk prostate cancer who declined immediate surgery, hormonal therapy, or radiation and participated in an intensive nutrition and lifestyle intervention while undergoing careful surveillance for tumor progression. Consistent with previous studies, significant improvements in weight, abdominal obesity, blood pressure, and lipid profile were observed (all P < 0.05), and surveillance of low-risk patients was safe. Gene expression profiles were obtained from 30 participants, pairing RNA samples from control prostate needle biopsy taken before intervention to RNA from the same patient's 3-month postintervention biopsy. Quantitative real-time PCR was used to validate array observations for selected transcripts. Two-class paired analysis of global gene expression using significance analysis of microarrays detected 48 up-regulated and 453 down-regulated transcripts after the intervention. Pathway analysis identified significant modulation of biological processes that have critical roles in tumorigenesis, including protein metabolism and modification, intracellular protein traffic, and protein phosphorylation (all P < 0.05). Intensive nutrition and lifestyle changes may modulate gene expression in the prostate. Understanding the prostate molecular response to comprehensive lifestyle changes may strengthen efforts to develop effective prevention and treatment. Larger clinical trials are warranted to confirm the results of this pilot study.


Subject(s)
Diet , Gene Expression Profiling , Life Style , Prostate/metabolism , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/genetics , Abdominal Fat , Aged , Aged, 80 and over , Blood Pressure , Body Weight , Cardiovascular Diseases/epidemiology , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Obesity/epidemiology , Pilot Projects , Prospective Studies , Risk Factors , Up-Regulation
9.
Diabetes ; 56(7): 1751-60, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17456854

ABSTRACT

The objective of this study was to further establish and confirm the relationship of adipose mitochondrial biogenesis in diabetes/obesity and the effects of rosiglitazone (RSG), a peroxisome proliferator-activated receptor (PPAR) gamma agonist, by systematically analyzing mitochondrial gene expression and function in two mouse models of obesity and type 2 diabetes. Using microarray technology, adipose mitochondrial gene transcription was studied in db/db, high-fat diet-fed C57BL/6 (HFD) and respective control mice with or without RSG treatment. The findings were extended using mitochondrial staining, DNA quantification, and measurements of citrate synthase activity. In db/db and HFD mice, gene transcripts associated with mitochondrial ATP production, energy uncoupling, mitochondrial ribosomal proteins, outer and inner membrane translocases, and mitochondrial heat-shock proteins were decreased in abundance, compared with db/+ and standard-fat diet-fed control mice, respectively. RSG dose-dependently increased these transcripts in both db/db and HFD mice and induced transcription of mitochondrial structural proteins and cellular antioxidant enzymes responsible for removal of reactive oxygen species generated by increased mitochondrial activity. Transcription factors, including PPAR coactivator (PGC)-1beta, PGC-1alpha, estrogen-related receptor alpha, and PPARalpha, were suppressed in both models and induced by RSG. The effects of RSG on adipose mitochondrial genes were confirmed by quantitative RT-PCR and further supported by mitochondrial staining, mitochondrial DNA quantification, and citrate synthase activity. Adipose mitochondrial biogenesis was overwhelmingly suppressed in both mouse models of diabetes/obesity and globally induced by RSG. These findings suggest an important role of adipose mitochondria in diabetes/obesity and the potential for new treatment approaches targeting adipose mitochondria.


Subject(s)
Adipose Tissue/drug effects , Hypoglycemic Agents/pharmacology , Mitochondria/drug effects , PPAR gamma/agonists , Thiazolidinediones/pharmacology , Adipose Tissue/metabolism , Animals , Diabetes Mellitus, Type 2/drug therapy , Dietary Fats , Disease Models, Animal , Gene Expression , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Obesity/drug therapy , Rosiglitazone , Transcription, Genetic
10.
J Alzheimers Dis ; 11(1): 45-51, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17361034

ABSTRACT

Rosiglitazone was found to simulate mitochondrial biogenesis in mouse brain in an apolipoprotein (Apo) E isozyme-independent manner. Rosiglitazone induced both mitochondrial DNA (mtDNA) and estrogen-stimulated related receptor alpha (ESRRA) mRNA, a key regulator of mitochondrial biogenesis. Transcriptomics and proteomics analysis suggested the mitochondria produced in the presence of human ApoE3 and E4 were not as metabolically efficient as those in the wild type or ApoE knockout mice. Thus, we propose that PPARgamma agonism induces neuronal mitochondrial biogenesis and improves glucose utilization leading to improved cellular function and provides mechanistic support for the improvement in cognition observed in treatment of Alzheimer's patients with rosiglitazone.


Subject(s)
Brain/drug effects , DNA, Mitochondrial/genetics , Hypoglycemic Agents/pharmacology , Mitochondria/drug effects , Organelle Biogenesis , RNA, Messenger/genetics , Receptors, Estrogen/genetics , Thiazolidinediones/pharmacology , Alzheimer Disease/drug therapy , Animals , Apolipoprotein E3/metabolism , Apolipoprotein E4/metabolism , Apolipoproteins E/genetics , Cognition/drug effects , Frontal Lobe/drug effects , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/agonists , Reverse Transcriptase Polymerase Chain Reaction , Rosiglitazone , Thiazolidinediones/therapeutic use , ERRalpha Estrogen-Related Receptor
11.
Mol Cancer ; 5: 24, 2006 Jun 19.
Article in English | MEDLINE | ID: mdl-16784538

ABSTRACT

BACKGROUND: Recent studies indicate that microRNAs (miRNAs) are mechanistically involved in the development of various human malignancies, suggesting that they represent a promising new class of cancer biomarkers. However, previously reported methods for measuring miRNA expression consume large amounts of tissue, prohibiting high-throughput miRNA profiling from typically small clinical samples such as excision or core needle biopsies of breast or prostate cancer. Here we describe a novel combination of linear amplification and labeling of miRNA for highly sensitive expression microarray profiling requiring only picogram quantities of purified microRNA. RESULTS: Comparison of microarray and qRT-PCR measured miRNA levels from two different prostate cancer cell lines showed concordance between the two platforms (Pearson correlation R2 = 0.81); and extension of the amplification, labeling and microarray platform was successfully demonstrated using clinical core and excision biopsy samples from breast and prostate cancer patients. Unsupervised clustering analysis of the prostate biopsy microarrays separated advanced and metastatic prostate cancers from pooled normal prostatic samples and from a non-malignant precursor lesion. Unsupervised clustering of the breast cancer microarrays significantly distinguished ErbB2-positive/ER-negative, ErbB2-positive/ER-positive, and ErbB2-negative/ER-positive breast cancer phenotypes (Fisher exact test, p = 0.03); as well, supervised analysis of these microarray profiles identified distinct miRNA subsets distinguishing ErbB2-positive from ErbB2-negative and ER-positive from ER-negative breast cancers, independent of other clinically important parameters (patient age; tumor size, node status and proliferation index). CONCLUSION: In sum, these findings demonstrate that optimized high-throughput microRNA expression profiling offers novel biomarker identification from typically small clinical samples such as breast and prostate cancer biopsies.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/diagnosis , Gene Expression Profiling/methods , MicroRNAs/metabolism , Prostatic Neoplasms/diagnosis , Biopsy , Cluster Analysis , Female , Genes, erbB-2 , Humans , Male , Phenotype , Receptors, Estrogen/analysis , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction/methods , Sensitivity and Specificity , Tumor Cells, Cultured
12.
Cancer Res ; 66(3): 1277-81, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16452179

ABSTRACT

Improved understanding of the molecular mechanisms by which small-molecule inhibitors of histone deacetylases (HDAC) induce programs, such as cellular differentiation and apoptosis, would undoubtedly assist their clinical development as anticancer agents. As modulators of gene transcript levels, HDAC inhibitors (HDACi) typically affect only 5% to 10% of actively transcribed genes with approximately as many mRNA transcripts being up-regulated as down-regulated. Using microRNA (miRNA) array analysis, we report rapid alteration of miRNA levels in response to the potent hydroxamic acid HDACi LAQ824 in the breast cancer cell line SKBr3. Within 5 hours of exposure to a proapoptotic dose of LAQ824, significant changes were measured in 40% of the >60 different miRNA species expressed in SKBr3 cells with 22 miRNA species down-regulated and 5 miRNAs up-regulated. To explore a potential functional link between HDACi induced mRNA up-regulation and miRNA down-regulation, antisense experiments were done against miR-27a and miR-27b, both abundantly expressed and down-regulated in SKBr3 cells by LAQ824. Correlating a set of genes previously determined by cDNA array analysis to be rapidly up-regulated by LAQ824 in SKBr3 with a database of potential 3' untranslated region miRNA binding elements, two genes containing putative miR-27 anchor elements were identified as transcriptionally up-regulated following miR-27 antisense transfection, ZBTB10/RINZF, a Sp1 repressor, and RYBP/DEDAF, an apoptotic facilitator. These findings emphasize the importance of post-transcriptional mRNA regulation by HDACi in addition to their established effects on promoter-driven gene expression.


Subject(s)
Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , MicroRNAs/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Humans , MicroRNAs/genetics , RNA, Antisense/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection
13.
Am J Physiol Cell Physiol ; 286(2): C293-301, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14576086

ABSTRACT

Although copper is an essential metal, it is capable of catalyzing the formation of reactive oxygen species that can cause intracellular oxidative damage. We investigated the hypothesis that metal- and oxidative stress-responsive signal transduction pathways mediate the cellular and molecular responses associated with copper exposure. Transient transfection assays using COS-7 cells and mouse metallothionein-I (MT-I) or rat NAD(P)H:oxidoreductase 1-based reporter genes demonstrate that copper activates transcription via metal and antioxidant response elements. Concomitant with copper exposures is a decrease in the level of total glutathione and an increase in oxidized glutathione. Depletion of glutathione, before copper exposure, increases metal- and oxidative stress-inducible transcription and cytotoxicity. Pretreatment with the reactive oxygen scavengers aspirin or vitamin E provides partial protection against copper toxicity and reduces inducible transcription. Experiments using signal transduction inhibitors and a metal transcription factor (MTF)-1 null cell line demonstrate that copper-inducible MT-I transcription is regulated by protein kinase C and mitogen-activated protein kinase signaling pathways and requires MTF-1. The results of these studies indicate that copper activates transcription through both metal- and oxidative stress-responsive signal transduction pathways.


Subject(s)
Copper/pharmacology , Metals/pharmacology , Oxidative Stress/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription, Genetic/drug effects , Animals , Antioxidants/pharmacology , COS Cells , Chlorocebus aethiops , DNA-Binding Proteins , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Genes, Reporter/genetics , Glutathione/deficiency , Glutathione/metabolism , Metallothionein/genetics , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , NADH, NADPH Oxidoreductases/genetics , Protein Kinase C/antagonists & inhibitors , Rats , Response Elements/physiology , Transcription Factors/physiology , Transcription, Genetic/physiology , Transfection , Transcription Factor MTF-1
SELECTION OF CITATIONS
SEARCH DETAIL
...