Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
PLoS One ; 13(6): e0197702, 2018.
Article in English | MEDLINE | ID: mdl-29864117

ABSTRACT

The Macrophage Migration Inhibitory Factor (MIF) is an inflammatory cytokine that is overexpressed in a number of cancer types, with increased MIF expression often correlating with tumor aggressiveness and poor patient outcomes. In this study, we aimed to better understand the link between primary tumor expression of MIF and increased tumor growth. Using the MMTV-PyMT murine model of breast cancer, we observed that elevated MIF expression promoted tumor appearance and growth. Supporting this, we confirmed our previous observation that higher MIF expression supported tumor growth in the 4T1 murine model of breast cancer. We subsequently discovered that loss of MIF expression in 4T1 cells led to decreased cell numbers and increased apoptosis in vitro under reduced serum culture conditions. We hypothesized that this increase in cell death would promote detection by the host immune system in vivo, which could explain the observed impairment in tumor growth. Supporting this, we demonstrated that loss of MIF expression in the primary tumor led to an increased abundance of intra-tumoral IFNgamma-producing CD4+ and CD8+ T cells, and that depletion of T cells from mice bearing MIF-deficient tumors restored growth to the level of MIF-expressing tumors. Furthermore, we found that MIF depletion from the tumor cells resulted in greater numbers of activated intra-tumoral dendritic cells (DCs). Lastly, we demonstrated that loss of MIF expression led to a robust induction of a specialized form of cell death, immunogenic cell death (ICD), in vitro. Together, our data suggests a model in which MIF expression in the primary tumor dampens the anti-tumor immune response, promoting tumor growth.


Subject(s)
Breast Neoplasms/genetics , Immunity, Cellular/genetics , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Mammary Neoplasms, Animal/genetics , Animals , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation/genetics , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/immunology , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/pathology , Mice
2.
PLoS One ; 7(11): e49744, 2012.
Article in English | MEDLINE | ID: mdl-23166763

ABSTRACT

Sulforaphane (SFN) is a dietary cancer preventive with incompletely characterized mechanism(s) of cancer prevention. Since prostaglandin E2 (PGE2) promotes cancer progression, we hypothesized that SFN may block PGE2 synthesis in cancer cells. We found that SFN indeed blocked PGE2 production in human A549 cancer cells not by inhibiting COX-2, but rather by suppressing the expression of microsomal prostaglandin E synthase (mPGES-1), the enzyme that directly synthesizes PGE2. We identified the Hypoxia Inducible Factor 1 alpha (HIF-1α) as the target of SFN-mediated mPGES-1 suppression. SFN suppressed HIF-1α protein expression and the presence of HIF-1α at the mPGES-1 promoter, resulting in reduced transcription of mPGES-1. Finally, SFN also reduced expression of mPGES-1 and PGE2 production in A549 xenograft tumors in mice. Together, these results point to the HIF-1α, mPGES-1 and PGE2 axis as a potential mediator of the anti-cancer effects of SFN, and illustrate the potential of SFN for therapeutic control of cancer and inflammation. Harmful side effects in patients taking agents that target the more upstream COX-2 enzyme render the downstream target mPGES-1 a significant target for anti-inflammatory therapy. Thus, SFN could prove to be an important therapeutic approach to both cancer and inflammation.


Subject(s)
Dinoprostone/biosynthesis , Intramolecular Oxidoreductases/antagonists & inhibitors , Thiocyanates/pharmacology , Animals , Cell Line, Tumor , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Isothiocyanates , Mice , Promoter Regions, Genetic , Prostaglandin-E Synthases , Protein Binding/drug effects , RNA, Messenger/genetics , Sulfoxides , Transplantation, Heterologous
3.
J Immunol ; 189(12): 5533-40, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23125418

ABSTRACT

The macrophage migration inhibitory factor (MIF), an inflammatory cytokine, is overexpressed in many solid tumors and is associated with poor prognosis. We previously identified inhibitors of MIF within a class of natural products with demonstrated anti-cancer activities. We therefore sought to determine how MIF contributes to tumor growth and progression. We show in this study that in murine tumors including the 4T1 model of aggressive, spontaneously metastatic breast cancer in immunologically intact mice, tumor-derived MIF promotes tumor growth and pulmonary metastasis through control of inflammatory cells within the tumor. Specifically, MIF increases the prevalence of a highly immune suppressive subpopulation of myeloid-derived suppressor cells (MDSCs) within the tumor. In vitro, MIF promotes differentiation of myeloid cells into the same population of MDSCs. Pharmacologic inhibition of MIF reduces MDSC accumulation in the tumor similar to MIF depletion and blocks the MIF-dependent in vitro differentiation of MDSCs. Our results demonstrate that MIF is a therapeutically targetable mechanism for control of tumor growth and metastasis through regulation of the host immune response and support the potential utility of MIF inhibitors, either alone or in combination with standard tumor-targeting therapeutic or immunotherapy approaches.


Subject(s)
Cell Differentiation/immunology , Intramolecular Oxidoreductases/physiology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Macrophage Migration-Inhibitory Factors/physiology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Myeloid Progenitor Cells/immunology , Animals , Cell Line, Tumor , Female , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Intramolecular Oxidoreductases/deficiency , Lung Neoplasms/metabolism , Macrophage Migration-Inhibitory Factors/deficiency , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, SCID , Myeloid Progenitor Cells/pathology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Skin Neoplasms/secondary , Tumor Microenvironment/immunology
4.
PLoS One ; 7(2): e32527, 2012.
Article in English | MEDLINE | ID: mdl-22389707

ABSTRACT

Signal transduction pathways that are modulated by thiol oxidation events are beginning to be uncovered, but these discoveries are limited by the availability of relatively few analytical methods to examine protein oxidation compared to other signaling events such as protein phosphorylation. We report here the coupling of PROP, a method to purify reversibly oxidized proteins, with the proteomic identification of the purified mixture using mass spectrometry. A gene ontology (GO), KEGG enrichment and Wikipathways analysis of the identified proteins indicated a significant enrichment in proteins associated with both translation and mRNA splicing. This methodology also enabled the identification of some of the specific cysteine residue targets within identified proteins that are reversibly oxidized by hydrogen peroxide treatment of intact cells. From these identifications, we determined a potential consensus sequence motif associated with oxidized cysteine residues. Furthermore, because we identified proteins and specific sites of oxidation from both abundant proteins and from far less abundant signaling proteins (e.g. hepatoma derived growth factor, prostaglandin E synthase 3), the results suggest that the PROP procedure was efficient. Thus, this PROP-proteomics methodology offers a sensitive means to identify biologically relevant redox signaling events that occur within intact cells.


Subject(s)
Proteins/metabolism , Proteomics/methods , HeLa Cells , Humans , Mass Spectrometry , Oxidation-Reduction , Proteins/chemistry
5.
Anal Chem ; 83(3): 856-65, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21192683

ABSTRACT

We have developed a complete system for the isotopic labeling, fractionation, and automated quantification of differentially expressed peptides that significantly facilitates candidate biomarker discovery. We describe a new stable mass tagging reagent pair, (12)C(6)- and (13)C(6)-phenyl isocyanate (PIC), that offers significant advantages over currently available tags. Peptides are labeled predominantly at their amino termini and exhibit elution profiles that are independent of label isotope. Importantly, PIC-labeled peptides have unique neutral-mass losses upon CID fragmentation that enable charge state and label isotope identification and, thereby, decouple the sequence identification from the quantification of candidate biomarkers. To exploit these properties, we have coupled peptide fractionation protocols with a Thermo LTQ-XL LC-MS(2) data acquisition strategy and a suite of automated spectrum analysis software that identifies quantitative differences between labeled samples. This approach, dubbed the PICquant platform, is independent of protein sequence identification and excludes unlabeled peptides that otherwise confound biomarker discovery. Application of the PICquant platform to a set of complex clinical samples showed that the system allows rapid identification of peptides that are differentially expressed between control and patient groups.


Subject(s)
Isocyanates/analysis , Isotope Labeling/methods , Mass Spectrometry/methods , Peptides/analysis , Algorithms , Carbon Isotopes/analysis , Carbon Radioisotopes/analysis , Chromatography, Liquid/methods , Humans , Isocyanates/chemistry , Molecular Structure
6.
PLoS One ; 5(11): e15012, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-21085594

ABSTRACT

Oxidation of cysteine residues of proteins is emerging as an important means of regulation of signal transduction, particularly of protein kinase function. Tools to detect and quantify cysteine oxidation of proteins have been a limiting factor in understanding the role of cysteine oxidation in signal transduction. As an example, the p38 MAP kinase is activated by several stress-related stimuli that are often accompanied by in vitro generation of hydrogen peroxide. We noted that hydrogen peroxide inhibited p38 activity despite paradoxically increasing the activating phosphorylation of p38. To address the possibility that cysteine oxidation may provide a negative regulatory effect on p38 activity, we developed a biochemical assay to detect reversible cysteine oxidation in intact cells. This procedure, PROP, demonstrated in vivo oxidation of p38 in response to hydrogen peroxide and also to the natural inflammatory lipid prostaglandin J2. Mutagenesis of the potential target cysteines showed that oxidation occurred preferentially on residues near the surface of the p38 molecule. Cysteine oxidation thus controls a functional redox switch regulating the intensity or duration of p38 activity that would not be revealed by immunodetection of phosphoprotein commonly interpreted as reflective of p38 activity.


Subject(s)
Chemistry Techniques, Analytical/methods , Cysteine/metabolism , Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Cysteine/chemistry , Cysteine/genetics , HeLa Cells , Humans , Hydrogen Peroxide/pharmacology , Immunoblotting , Mitogen-Activated Protein Kinase 14 , Models, Molecular , Mutation , Oxidants/pharmacology , Oxidation-Reduction , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/pharmacology , Protein Conformation , Proteins/genetics , Proteins/isolation & purification , Reproducibility of Results , Signal Transduction/drug effects , Sulfhydryl Compounds/metabolism , Transfection , p38 Mitogen-Activated Protein Kinases/chemistry , p38 Mitogen-Activated Protein Kinases/genetics
7.
J Biol Chem ; 285(19): 14217-28, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20220133

ABSTRACT

c-Jun NH(2)-terminal Kinases (JNKs) play a central role in the cellular response to a wide variety of stress signals. After their activation, JNKs induce phosphorylation of substrates, which control proliferation, migration, survival, and differentiation. Recent studies suggest that JNKs may also play a role in cell cycle control, although the underlying mechanisms are largely unexplored. Here we show that JNK directly phosphorylates Cdc25C at serine 168 during G(2) phase of the cell cycle. Cdc25C phosphorylation by JNK negatively regulates its phosphatase activity and thereby Cdk1 activation, enabling a timely control of mitosis onset. Unrestrained phosphorylation by JNK, as obtained by a cell cycle-stabilized form of JNK or as seen in some human tumors, results in aberrant cell cycle progression. Additionally, UV irradiation-induced G(2)/M checkpoint requires inactivation of Cdc25C by JNK phosphorylation. JNK phosphorylation of Cdc25C as well as Cdc25A establishes a novel link between stress signaling and unperturbed cell cycle and checkpoint pathways.


Subject(s)
Cell Division/physiology , DNA Damage , G2 Phase/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Mitosis/physiology , cdc25 Phosphatases/metabolism , Animals , Blotting, Western , Cell Proliferation , Cells, Cultured , Fluorescent Antibody Technique , Humans , Mice , Phosphorylation
8.
J Am Soc Mass Spectrom ; 21(1): 80-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19850495

ABSTRACT

Peptide sequence identification using tandem mass spectroscopy remains a major challenge for complex proteomic studies. Peptide matching algorithms require the accurate determination of both the mass and charge of the precursor ion and accommodate uncertainties in these properties by using a wide precursor mass tolerance and by testing, for each spectrum, several possible candidate charges. Using a data acquisition strategy that includes obtaining narrow mass-range MS(1) "zoom" scans, we describe here a post-acquisition algorithm dubbed mass and charge (Z) inference engine (MAZIE), which accurately determines the charge and monoisotopic mass of precursor ions on a low-resolution Thermo LTQ-XL mass spectrometer. This is achieved by examining the isotopic distribution obtained in the preceding MS(1) zoom spectrum and comparing to theoretical distributions for candidate charge states from +1 to +4. MAZIE then writes modified data files with the corrected monoisotopic mass and charge. We have validated MAZIE results by comparing the sequence search results obtained with the MAZIE-generated data files to results using the unmodified data files. Using two different search algorithms and a false discovery rate filter, we found that MAZIE-interpreted data resulted in 80% (using SEQUEST) and 30% (using OMSSA) more high-confidence sequence identifications. Analyses of these results indicate that the accurate determination of the precursor ion mass greatly facilitates the ability to differentiate between true and false positive matches, while the determination of the precursor ion charge reduces the overall search time but does not significantly reduce the ambiguity of interpreting the search results. MAZIE is distributed as an open-source PERL script.


Subject(s)
Algorithms , Peptides/chemistry , Proteomics/methods , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Databases, Protein , Humans , Peptides/analysis , Sequence Analysis, Protein , Urine/chemistry
9.
Biochem J ; 423(3): 315-21, 2009 Oct 12.
Article in English | MEDLINE | ID: mdl-19723024

ABSTRACT

Dietary ITCs (isothiocyanates) prevent cancer and show other bioactivities in vivo. As electrophiles, ITCs may covalently modify cellular proteins. Using a novel proteomics screen, we identified MIF (macrophage migration inhibitory factor) as the principal target of nutrient ITCs in intact cells. ITCs covalently modify the N-terminal proline residue of MIF and extinguish its catalytic tautomerase activity. MIF deficiency does not prevent induction of Phase 2 gene expression, a hallmark of many cancer chemopreventives, including ITCs. Due to the emerging role of MIF in the control of malignant cell growth and its clear involvement in inflammation, inhibition of MIF by nutrient ITCs suggests therapeutic strategies for inflammatory diseases and cancer.


Subject(s)
Intramolecular Oxidoreductases/metabolism , Isothiocyanates/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Protein Processing, Post-Translational , HeLa Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Neoplasms/genetics , Neoplasms/metabolism
10.
Biochem Biophys Res Commun ; 369(4): 1215-20, 2008 May 16.
Article in English | MEDLINE | ID: mdl-18355442

ABSTRACT

Previous reports showed that urokinase plasminogen activator (uPA) converts plasminogen to plasmin which then activates matrix metalloproteinases (MMPs). Here, we report that uPA directly cleaved pro-MMP-9 in a time-dependent manner at both C- and N-terminus and generated two gelatinolytic bands. uPA-activated-MMP-9 efficiently degraded fibronectin and blocked by uPA inhibitor B428 and recombinant tissue inhibitor of metalloproteinase-1 (TIMP-1). B428 inhibited basal and PMA-induced active MMP-9 in glioblastomas (GBM) U1242 cell media as well as cell invasion in vitro. A combination of MMP-9 and uPA antibodies more significantly inhibited U1242 cell invasion than uPA or MMP-9 antibody alone. Both uPA and MMP-9 were highly expressed in U1242 cell and GBM patient specimens. Furthermore, two active MMP-9 fragments with identical molecular weights to the uPA-activated MMP-9 products were detected in GBM patient specimens. These results suggest that uPA-mediated direct activation of MMP-9 may promote GBM cell invasion.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Matrix Metalloproteinase 9/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Acute-Phase Proteins/metabolism , Brain Neoplasms/enzymology , Enzyme Activation , Fibronectins/chemistry , Gelatin/chemistry , Glioblastoma/enzymology , Humans , Lipocalin-2 , Lipocalins/metabolism , Matrix Metalloproteinase 9/chemistry , Neoplasm Invasiveness , Proto-Oncogene Proteins/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Urokinase-Type Plasminogen Activator/chemistry
11.
BMC Cancer ; 7: 183, 2007 Sep 25.
Article in English | MEDLINE | ID: mdl-17894894

ABSTRACT

BACKGROUND: Dietary isothiocyanates (ITCs) are electrophilic compounds that have diverse biological activities including induction of apoptosis and effects on cell cycle. They protect against experimental carcinogenesis in animals, an activity believed to result from the transcriptional induction of "Phase 2" enzymes. The molecular mechanism of action of ITCs is unknown. Since ITCs are electrophiles capable of reacting with sulfhydryl groups on amino acids, we hypothesized that ITCs induce their biological effects through covalent modification of proteins, leading to changes in cell regulatory events. We previously demonstrated that stress-signaling kinase pathways are inhibited by other electrophilic compounds such as menadione. We therefore tested the effects of nutritional ITCs on MEKK1, an upstream regulator of the SAPK/JNK signal transduction pathway. METHODS: The activity of MEKK1 expressed in cells was monitored using in vitro kinase assays to measure changes in catalytic activity. The activity of endogenous MEKK1, immunopurified from ITC treated and untreated LnCAP cells was also measured by in vitro kinase assay. A novel labeling and affinity reagent for detection of protein modification by ITCs was synthesized and used in competition assays to monitor direct modification of MEKK1 by ITC. Finally, immunoblots with phospho-specific antibodies were used to measure the activity of MAPK protein kinases. RESULTS: ITCs inhibited the MEKK1 protein kinase in a manner dependent on a specific cysteine residue in the ATP binding pocket. Inhibition of MEKK1 catalytic activity was due to direct, covalent and irreversible modification of the MEKK1 protein itself. In addition, ITCs inhibited the catalytic activity of endogenous MEKK1. This correlated with inhibition of the downstream target of MEKK1 activity, i.e. the SAPK/JNK kinase. This inhibition was specific to SAPK, as parallel MAPK pathways were unaffected. CONCLUSION: These results demonstrate that MEKK1 is directly modified and inhibited by ITCs, and that this correlates with inhibition of downstream activation of SAPK. These results support the conclusion that ITCs may carry out many of their actions by directly targeting important cell regulatory proteins.


Subject(s)
Food , Isothiocyanates/pharmacology , MAP Kinase Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase Kinase 1/metabolism , Protein Kinase Inhibitors/pharmacology , HeLa Cells , Humans , Isothiocyanates/metabolism , Protein Kinase Inhibitors/metabolism
12.
Antioxid Redox Signal ; 8(9-10): 1819-27, 2006.
Article in English | MEDLINE | ID: mdl-16987034

ABSTRACT

The production of reactive oxygen species (ROS) accompanies many signaling events. Antioxidants and ROS scavenging enzymes in general have effects that indicate a critical role for ROS in downstream signaling, but a mechanistic understanding of the contribution of ROS as second messengers is incomplete. Here, the role of reactive oxygen species in cell signaling is discussed, emphasizing the ability of ROS to directly modify signaling proteins through thiol oxidation. Examples are provided of protein thiol modifications that control signal transduction effectors that include protein kinases, phosphatases, and transcription factors. Whereas the effects of cysteine oxidation on these proteins in experimental systems is clear, it has proven more difficult to demonstrate these modifications in response to physiologic stimuli. Improved detection methods for analysis of thiol modification will be essential to define these regulatory mechanisms. Bridging these two areas of research could reveal new regulatory mechanisms in signaling pathways, and identify new therapeutic targets.


Subject(s)
Cysteine/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Animals , Humans , Models, Biological , Oxidation-Reduction , Phosphoprotein Phosphatases/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism
13.
Nat Methods ; 2(6): 435-41, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15908922

ABSTRACT

Chemical genetic analysis of protein kinases involves engineering kinases to be uniquely sensitive to inhibitors and ATP analogs that are not recognized by wild-type kinases. Despite the successful application of this approach to over two dozen kinases, several kinases do not tolerate the necessary modification to the ATP binding pocket, as they lose catalytic activity or cellular function upon mutation of the 'gatekeeper' residue that governs inhibitor and nucleotide substrate specificity. Here we describe the identification of second-site suppressor mutations to rescue the activity of 'intolerant' kinases. A bacterial genetic selection for second-site suppressors using an aminoglycoside kinase APH(3')-IIIa revealed several suppressor hotspots in the kinase domain. Informed by results from this selection, we focused on the beta sheet in the N-terminal subdomain and generated a structure-based sequence alignment of protein kinases in this region. From this alignment, we identified second-site suppressors for several divergent kinases including Cdc5, MEKK1, GRK2 and Pto. The ability to identify second-site suppressors to rescue the activity of intolerant kinases should facilitate chemical genetic analysis of the majority of protein kinases in the genome.


Subject(s)
Gene Expression Profiling/methods , Genomics/methods , Mutagenesis, Site-Directed/genetics , Protein Interaction Mapping/methods , Protein Kinases/chemistry , Protein Kinases/metabolism , Amino Acid Substitution , Genetic Variation/genetics , Genomics/trends , Protein Kinases/genetics , Structure-Activity Relationship
14.
Mol Cell Biol ; 24(24): 10941-53, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15572695

ABSTRACT

The bZIP transcription factor Nrf2 controls a genetic program that protects cells from oxidative damage and maintains cellular redox homeostasis. Keap1, a BTB-Kelch protein, is the major upstream regulator of Nrf2 and controls both the subcellular localization and steady-state levels of Nrf2. In this report, we demonstrate that Keap1 functions as a substrate adaptor protein for a Cul3-dependent E3 ubiquitin ligase complex. Keap1 assembles into a functional E3 ubiquitin ligase complex with Cul3 and Rbx1 that targets multiple lysine residues located in the N-terminal Neh2 domain of Nrf2 for ubiquitin conjugation both in vivo and in vitro. Keap1-dependent ubiquitination of Nrf2 is inhibited following exposure of cells to quinone-induced oxidative stress and sulforaphane, a cancer-preventive isothiocyanate. A mutant Keap1 protein containing a single cysteine-to-serine substitution at residue 151 within the BTB domain of Keap1 is markedly resistant to inhibition by either quinone-induced oxidative stress or sulforaphane. Inhibition of Keap1-dependent ubiquitination of Nrf2 correlates with decreased association of Keap1 with Cul3. Neither quinone-induced oxidative stress nor sulforaphane disrupts association between Keap1 and Nrf2. Our results suggest that the ability of Keap1 to assemble into a functional E3 ubiquitin ligase complex is the critical determinant that controls steady-state levels of Nrf2 in response to cancer-preventive compounds and oxidative stress.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Cullin Proteins/metabolism , Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Anticarcinogenic Agents/pharmacology , Antioxidants/pharmacology , Breast Neoplasms/pathology , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Genes, Reporter , Humans , Hydroquinones/pharmacology , Immunoblotting , Intracellular Signaling Peptides and Proteins , Isothiocyanates , Kelch-Like ECH-Associated Protein 1 , Luciferases/metabolism , Lysine/chemistry , NF-E2-Related Factor 2 , Oxidation-Reduction , Oxidative Stress , Precipitin Tests , Protein Structure, Tertiary , Proteins/chemistry , Proteins/genetics , Serine/metabolism , Substrate Specificity , Sulfoxides , Thiocyanates/pharmacology , Trans-Activators/chemistry , Trans-Activators/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitins/metabolism
15.
J Cell Biochem ; 93(1): 104-11, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15352167

ABSTRACT

Studies of cell signal transduction have predominantly focused on regulation of protein function by phosphorylation. However, recent efforts have begun to uncover another layer of regulation mediated by direct oxidation of cysteine residues in signaling proteins. Typically induced during signaling responses accompanied by generation of reactive oxygen species, these thiol modifications have a variety of functional consequences for target proteins. Using specific signaling protein targets as examples, we discuss how thiol oxidation generally activates pro-apoptotic signaling pathways while inhibiting pathways that promote cell survival. We propose a model in which thiol oxidation acts to control the equilibrium between survival and apoptosis, fine tuning cellular responses that play a central role in the apoptotic decision-making process. We identify areas of focus for future work, including a better understanding of specificity in thiol oxidation events, and a critical need for approaches to examine these modifications under physiologically relevant signaling conditions.


Subject(s)
Apoptosis , Proteins/metabolism , Signal Transduction , Sulfhydryl Compounds , Animals , Humans , Oxidation-Reduction , Phosphorylation , Reactive Oxygen Species
16.
Biochem J ; 381(Pt 3): 675-83, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15139849

ABSTRACT

Many intracellular signalling events are accompanied by generation of reactive oxygen species in cells. Oxidation of protein thiol groups is an emerging theme in signal-transduction research. We have found that MEKK1 [MAPK (mitogen-activated protein kinase)/ERK (extracellular-signal-regulated kinase) kinase kinase 1], an upstream activator of the SAPK/JNK (stress-activated protein kinase/c-Jun N-terminal kinase) pathway, is directly inhibited by cysteine alkylation using NEM (N-ethylmaleimide). The related kinase, ASK1 (apoptosis signal-regulating kinase 1), was not inhibited, but was instead activated by NEM. Inhibition of MEKK1 requires a single unique cysteine residue (Cys1238) in the ATP-binding domain of MEKK1. Oxidative stress induced by menadione (2-methyl-1,4-naphthoquinone) also inhibited MEKK1, but activated ASK1, in cells. MEKK1 inhibition by menadione also required Cys1238. Oxidant-inhibited MEKK1 was re-activated by dithiothreitol and glutathione, supporting reversible cysteine oxidation as a mechanism. Using various chemical probes, we excluded modification by S-nitrosylation or oxidation of cysteine to sulphenic acid. Oxidant-inhibited MEKK1 migrated normally on non-reducing gels, excluding the possibility of intra- or inter-molecular disulphide bond formation. MEKK1 was inhibited by glutathionylation in vitro, and MEKK1 isolated from menadione-treated cells was shown by MS to be modified by glutathione on Cys1238. Our results support a model whereby the redox environment within the cell selectively regulates stress signalling through MEKK1 versus ASK1, and may thereby participate in the induction of apoptosis by oxidative stress.


Subject(s)
Adenosine Triphosphate/metabolism , Glutathione/metabolism , MAP Kinase Kinase Kinase 1/antagonists & inhibitors , Oxidative Stress/physiology , Peptides/metabolism , Alkylation , Amino Acid Sequence/genetics , Amino Acid Sequence/physiology , Amino Acid Substitution , Binding Sites/physiology , Catalytic Domain/drug effects , Cell Line, Tumor , Cysteine/metabolism , Dithiothreitol/pharmacology , Enzyme Inhibitors/pharmacology , Ethylmaleimide/pharmacology , Humans , Lymph Nodes/enzymology , Lymph Nodes/pathology , MAP Kinase Kinase Kinase 1/chemistry , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 1/physiology , Male , Molecular Sequence Data , Mutation/physiology , Oxidants/antagonists & inhibitors , Oxidants/pharmacology , Oxidation-Reduction , Oxidative Stress/drug effects , Peptides/chemistry , Peptides/physiology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Protein Structure, Tertiary , Valine/metabolism
17.
J Biomed Sci ; 10(6 Pt 2): 792-804, 2003.
Article in English | MEDLINE | ID: mdl-14631119

ABSTRACT

The human cell lines VACO-235 and VACO-411 constitute a novel in vitro model of colon adenoma to carcinoma progression. By differential display RT-PCR we identified a transcript that is expressed in the parental nontumorigenic adenoma line (VACO-235E), but is not expressed in the tumorigenic daughter (VACO-235L) or granddaughter (VACO-411) lines. This cDNA represents a previously uncharacterized portion of the 3'UTR of human PIGR. Human PIGR mRNA was found to be highly expressed in normal colon epithelium, but was decreased in 6 of 8 colon tumors and was negligible in 8 of 10 colon tumor cell lines. We sequenced the entire 1.8 kb 3'UTR of human PIGR, and found it to contain multiple repetitive elements as well as elements that could affect the processing and stability of PIGR mRNA. We hypothesize that differential regulation of PIGR mRNA stability may contribute to its downregulation in colon cancer.


Subject(s)
Adenoma/genetics , Carcinoma/genetics , Colonic Neoplasms/genetics , Gene Expression/physiology , Receptors, Polymeric Immunoglobulin/genetics , Base Sequence , Blotting, Northern , Cell Line, Tumor , Chromosome Mapping , DNA Primers , Humans , In Vitro Techniques , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Analysis, DNA
18.
Oncogene ; 22(40): 6129-41, 2003 Sep 18.
Article in English | MEDLINE | ID: mdl-13679851

ABSTRACT

The Sterile-20 or Ste20 family of serine/threonine kinases is a group of signaling molecules whose physiological roles within mammalian cells are just starting to be elucidated. Here, in this report we present the characterization of three human Ste20-like kinases with greater than 90% similarity within their catalytic domains that define a novel subfamily of Ste20s. Members of this kinase family include rat thousand and one (TAO1) and chicken KFC (kinase from chicken). For the lack of a consensus nomenclature in the literature, in this report, we shall call this family hKFC (for their homology to chicken KFC) and the three members hKFC-A, hKFC-B, and hKFC-C, respectively. These kinases have many similarities including an aminoterminal kinase domain, a serine-rich region, and a coiled-coil configuration within the C-terminus. All three kinases are able to activate the p38 MAP kinase pathway through the specific activation of the upstream MKK3 kinase. We also offer evidence, both theoretical and biochemical, showing that these kinases can undergo self-association. Despite these similarities, these kinases differ in tissue distribution, apparent subcellular localization, and feature structural differences largely within the carboxyl-terminal sequence.


Subject(s)
Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Dimerization , Gene Expression Regulation, Enzymologic , Humans , Intracellular Signaling Peptides and Proteins , Isoenzymes/chemistry , Isoenzymes/metabolism , Jurkat Cells , MAP Kinase Kinase 3 , MAP Kinase Kinase Kinases , Male , Molecular Sequence Data , Phylogeny , Protein Conformation , Sequence Homology, Amino Acid , Subcellular Fractions/enzymology , Tissue Distribution , p38 Mitogen-Activated Protein Kinases
19.
Cell Signal ; 15(7): 709-18, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12742231

ABSTRACT

Cells undergo M phase arrest in response to stresses like UV irradiation or DNA damage. Stress-activated protein kinase (SAPK, also known as c-Jun N-terminal kinase, JNK) is activated by such stress stimuli. We addressed the potential effects of SAPK activation on cell cycle regulatory proteins. Activation of SAPK strongly correlated with inhibition of cdc2/cyclin B kinase, an important regulator of G2/M phase. SAPK directly phosphorylated the cdc2 regulator, cdc25c, in vitro on serine 168 (S168). This residue was highly phosphorylated in vivo in response to stress stimuli. cdc25c phosphorylated on S168 in cells lacks phosphatase activity, and expression of a S168A mutant of cdc25c reversed the inhibition of cdc2/cyclin B kinase activity by cell stress. Antibodies directed against phosphorylated S168 detect increased phosphorylation of S168 after cell stress. We conclude that SAPK regulates cdc2/cyclin B kinase following stress events by a novel mechanism involving inhibitory phosphorylation of the cdc2-activating phosphatase cdc25c on S168.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Cyclin B/metabolism , Mitogen-Activated Protein Kinases/metabolism , Stress, Physiological/enzymology , cdc25 Phosphatases/metabolism , Amino Acid Sequence/drug effects , Amino Acid Sequence/physiology , Antibodies/pharmacology , Binding Sites/drug effects , Binding Sites/physiology , Cell Cycle Proteins/antagonists & inhibitors , Feedback, Physiological/drug effects , Feedback, Physiological/physiology , G2 Phase/drug effects , G2 Phase/physiology , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases , Mitosis/drug effects , Mitosis/physiology , Phosphorylation/drug effects , Serine/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , cdc25 Phosphatases/antagonists & inhibitors
20.
Mol Endocrinol ; 17(7): 1344-55, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12714703

ABSTRACT

Recent studies have shown that the antiestrogens tamoxifen and raloxifene may protect against breast cancer, presumably because of a blockade of estrogen receptor (ER)-mediated transcription. Another possible explanation is that antiestrogen-liganded ER transcriptionally induces genes that are protective against cancer. We previously reported that antiestrogen-liganded ERbeta transcriptionally activates the major detoxifying enzyme quinone reductase (QR) [NAD(P)H:quinone oxidoreductase]. It has been established that metabolites of estrogen, termed catecholestrogens, can form DNA adducts and cause oxidative DNA damage. We hypothesize that QR inhibits estrogen-induced DNA damage by detoxification of reactive catecholestrogens. We report here that physiological concentrations of 17beta-estradiol cause oxidative DNA damage, as measured by levels of 8- hydroxydeoxyguanine, in ER-positive MCF7 breast cancer cells, MDA-MB-231 breast cancer cells (ERalpha negative/ERbeta positive) and nontumorigenic MCF10A breast epithelial cells (very low ER), which is dependent on estrogen metabolism. Estrogen-induced 8-hydroxydeoxyguanine was inversely correlated to QR and ERbeta levels and was followed by downstream induction of the DNA repair enzyme XPA. Trans-hydroxytamoxifen, raloxifene, and the pure antiestrogen ICI-182,780 protected against estradiol-mediated damage in breast cancer cells containing ERbeta. This is most likely due to the ability of these antiestrogens to activate expression of QR via ERbeta. We conclude that up-regulation of QR, either by overexpression or induction by tamoxifen, can protect breast cells against oxidative DNA damage caused by estrogen metabolites, representing a possible novel mechanism of tamoxifen prevention against breast cancer.


Subject(s)
DNA Damage/drug effects , Estradiol/analogs & derivatives , Estrogen Receptor Modulators/pharmacology , Estrogens/metabolism , Guanine/analogs & derivatives , NAD(P)H Dehydrogenase (Quinone)/metabolism , Tamoxifen/analogs & derivatives , 8-Hydroxy-2'-Deoxyguanosine/analogs & derivatives , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , DNA Damage/physiology , DNA Repair/drug effects , DNA Repair/physiology , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Enzyme Activation/drug effects , Epithelial Cells/metabolism , Estradiol/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha , Estrogen Receptor beta , Estrogens/pharmacology , Female , Fulvestrant , Guanine/metabolism , Humans , NAD(P)H Dehydrogenase (Quinone)/drug effects , Oxidative Stress , Raloxifene Hydrochloride/pharmacology , Receptors, Estrogen/drug effects , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology , Tumor Cells, Cultured , Xeroderma Pigmentosum Group A Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...