Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Immunol ; 189(12): 5533-40, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23125418

RESUMEN

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.


Asunto(s)
Diferenciación Celular/inmunología , Oxidorreductasas Intramoleculares/fisiología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Factores Inhibidores de la Migración de Macrófagos/fisiología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Células Progenitoras Mieloides/inmunología , Animales , Línea Celular Tumoral , Femenino , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Oxidorreductasas Intramoleculares/deficiencia , Neoplasias Pulmonares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Células Progenitoras Mieloides/patología , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/secundario , Microambiente Tumoral/inmunología
2.
J Biol Chem ; 285(19): 14217-28, 2010 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-20220133

RESUMEN

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.


Asunto(s)
División Celular/fisiología , Daño del ADN , Fase G2/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Mitosis/fisiología , Fosfatasas cdc25/metabolismo , Animales , Western Blotting , Proliferación Celular , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Fosforilación
3.
Anal Chem ; 83(3): 856-65, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21192683

RESUMEN

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.


Asunto(s)
Isocianatos/análisis , Marcaje Isotópico/métodos , Espectrometría de Masas/métodos , Péptidos/análisis , Algoritmos , Isótopos de Carbono/análisis , Radioisótopos de Carbono/análisis , Cromatografía Liquida/métodos , Humanos , Isocianatos/química , Estructura Molecular
4.
Biochem J ; 423(3): 315-21, 2009 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-19723024

RESUMEN

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.


Asunto(s)
Oxidorreductasas Intramoleculares/metabolismo , Isotiocianatos/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Procesamiento Proteico-Postraduccional , Células HeLa , Humanos , Inflamación/genética , Inflamación/metabolismo , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Neoplasias/genética , Neoplasias/metabolismo
5.
Biochem Biophys Res Commun ; 369(4): 1215-20, 2008 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-18355442

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Metaloproteinasa 9 de la Matriz/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Proteínas de Fase Aguda/metabolismo , Neoplasias Encefálicas/enzimología , Activación Enzimática , Fibronectinas/química , Gelatina/química , Glioblastoma/enzimología , Humanos , Lipocalina 2 , Lipocalinas/metabolismo , Metaloproteinasa 9 de la Matriz/química , Invasividad Neoplásica , Proteínas Proto-Oncogénicas/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/química
6.
PLoS One ; 13(6): e0197702, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29864117

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/genética , Inmunidad Celular/genética , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Neoplasias Mamarias Animales/genética , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Proliferación Celular/genética , Células Dendríticas/inmunología , Células Dendríticas/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Oxidorreductasas Intramoleculares/inmunología , Factores Inhibidores de la Migración de Macrófagos/inmunología , Neoplasias Mamarias Animales/inmunología , Neoplasias Mamarias Animales/patología , Ratones
7.
BMC Cancer ; 7: 183, 2007 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-17894894

RESUMEN

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.


Asunto(s)
Alimentos , Isotiocianatos/farmacología , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Células HeLa , Humanos , Isotiocianatos/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo
8.
Mol Cell Biol ; 24(24): 10941-53, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15572695

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Cullin/metabolismo , Proteínas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Anticarcinógenos/farmacología , Antioxidantes/farmacología , Neoplasias de la Mama/patología , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Genes Reporteros , Humanos , Hidroquinonas/farmacología , Immunoblotting , Péptidos y Proteínas de Señalización Intracelular , Isotiocianatos , Proteína 1 Asociada A ECH Tipo Kelch , Luciferasas/metabolismo , Lisina/química , Factor 2 Relacionado con NF-E2 , Oxidación-Reducción , Estrés Oxidativo , Pruebas de Precipitina , Estructura Terciaria de Proteína , Proteínas/química , Proteínas/genética , Serina/metabolismo , Especificidad por Sustrato , Sulfóxidos , Tiocianatos/farmacología , Transactivadores/química , Transactivadores/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinas/metabolismo
9.
Antioxid Redox Signal ; 8(9-10): 1819-27, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16987034

RESUMEN

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.


Asunto(s)
Cisteína/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Animales , Humanos , Modelos Biológicos , Oxidación-Reducción , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/metabolismo
10.
Nucleic Acids Res ; 30(9): 1919-28, 2002 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11972328

RESUMEN

Synthesis of new ribosomes is an energy costly and thus highly regulated process. Ribosomal protein synthesis is controlled by regulating translation of the corresponding ribosomal protein (rp)mRNAs. In mammalian cells a 5'-terminal oligopyrimidine tract (TOP) is a conserved feature of these mRNAs that has been demonstrated to be essential for their translational regulation. Translation of TOP mRNAs has been proposed to be regulated by phosphorylation of ribosomal protein S6, which is a common effect of mitogenic stimulation of cells. However, as demonstrated here, S6 phosphorylation is not detectable in murine erythroleukemia (MEL) or other hematopoietic cells. The absence of S6 phosphorylation appears to be due to the action of a phosphatase that acts downstream of S6 kinase, presumably on S6 itself. Despite the absence of changes in S6 phosphorylation, translation of TOP mRNAs is repressed during differentiation of MEL cells. These data demonstrate the existence of a mechanism for regulating S6 phosphorylation that is distinct from kinase activation, as well as the existence of mechanisms for regulating translation of TOP mRNAs that are independent of S6 phosphorylation.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Biosíntesis de Proteínas , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Transducción de Señal , Región de Flanqueo 5' , Animales , Diferenciación Celular , Línea Celular , Linaje de la Célula , Regulación hacia Abajo , Células Madre Hematopoyéticas/fisiología , Humanos , Ratones , Fosforilación , Pirimidinas/análisis , ARN Mensajero/biosíntesis , ARN Mensajero/química , Proteína S6 Ribosómica , Proteínas Quinasas S6 Ribosómicas/metabolismo , Proteínas Ribosómicas/biosíntesis , Células Tumorales Cultivadas
11.
Cell Signal ; 15(7): 709-18, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12742231

RESUMEN

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.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclina B/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Estrés Fisiológico/enzimología , Fosfatasas cdc25/metabolismo , Secuencia de Aminoácidos/efectos de los fármacos , Secuencia de Aminoácidos/fisiología , Anticuerpos/farmacología , Sitios de Unión/efectos de los fármacos , Sitios de Unión/fisiología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Retroalimentación Fisiológica/efectos de los fármacos , Retroalimentación Fisiológica/fisiología , Fase G2/efectos de los fármacos , Fase G2/fisiología , Células HeLa , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Mitosis/efectos de los fármacos , Mitosis/fisiología , Fosforilación/efectos de los fármacos , Serina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Fosfatasas cdc25/antagonistas & inhibidores
12.
Mol Endocrinol ; 17(7): 1344-55, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12714703

RESUMEN

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.


Asunto(s)
Daño del ADN/efectos de los fármacos , Estradiol/análogos & derivados , Moduladores de los Receptores de Estrógeno/farmacología , Estrógenos/metabolismo , Guanina/análogos & derivados , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Tamoxifeno/análogos & derivados , 8-Hidroxi-2'-Desoxicoguanosina/análogos & derivados , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Daño del ADN/fisiología , Reparación del ADN/efectos de los fármacos , Reparación del ADN/fisiología , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Activación Enzimática/efectos de los fármacos , Células Epiteliales/metabolismo , Estradiol/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Estrógenos/farmacología , Femenino , Fulvestrant , Guanina/metabolismo , Humanos , NAD(P)H Deshidrogenasa (Quinona)/efectos de los fármacos , Estrés Oxidativo , Clorhidrato de Raloxifeno/farmacología , Receptores de Estrógenos/efectos de los fármacos , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Tamoxifeno/farmacología , Células Tumorales Cultivadas , Proteína de la Xerodermia Pigmentosa del Grupo A
13.
Biochem J ; 381(Pt 3): 675-83, 2004 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15139849

RESUMEN

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.


Asunto(s)
Adenosina Trifosfato/metabolismo , Glutatión/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Estrés Oxidativo/fisiología , Péptidos/metabolismo , Alquilación , Secuencia de Aminoácidos/genética , Secuencia de Aminoácidos/fisiología , Sustitución de Aminoácidos , Sitios de Unión/fisiología , Dominio Catalítico/efectos de los fármacos , Línea Celular Tumoral , Cisteína/metabolismo , Ditiotreitol/farmacología , Inhibidores Enzimáticos/farmacología , Etilmaleimida/farmacología , Humanos , Ganglios Linfáticos/enzimología , Ganglios Linfáticos/patología , Quinasa 1 de Quinasa de Quinasa MAP/química , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Quinasa 1 de Quinasa de Quinasa MAP/fisiología , Masculino , Datos de Secuencia Molecular , Mutación/fisiología , Oxidantes/antagonistas & inhibidores , Oxidantes/farmacología , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Péptidos/química , Péptidos/fisiología , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Estructura Terciaria de Proteína , Valina/metabolismo
14.
Antioxid Redox Signal ; 5(1): 103-13, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12626122

RESUMEN

The stress-activated protein kinases SAPK/JNK and p38/mHOG are activated by diverse classes of stress stimuli, many of which induce redox perturbations. We studied the effects of reactive quinones on stress signaling pathways. Menadione (2-methyl-1,4-naphthoquinone), which undergoes both one- and two-electron reduction, completely inhibited SAPK activity at high concentrations while activating SAPK at lower concentrations. Menadione activated p38/mHOG dose responsively. 2,3-Dimethyl-1,4-naphthoquinone (DMNQ), which preferentially undergoes two-electron reduction, had similar effects. In contrast, 1,4-naphthoquinone, which preferentially undergoes one-electron reduction, inhibited SAPK at high concentrations, but failed to activate SAPK at any concentration tested. In addition, this quinone activated p38 only at lower concentrations; high concentrations inhibited p38 activity. These activity profiles correlated with the activation state of the upstream kinase, indicating that the effects were mediated by an upstream step in the kinase pathway. The quinone reductase inhibitor dicoumarol blocked activation of SAPK by menadione and DMNQ, suggesting that two-electron reduction is important. Finally, addition of increasing amounts of hydrogen peroxide mimicked the effects of menadione and DMNQ, suggesting that hydrogen peroxide may be the relevant mediator. Differential activation of stress kinases by reactive quinones demonstrates that the cellular redox environment independently modulates these pathways.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/metabolismo , Quinonas/metabolismo , Animales , Antifibrinolíticos/farmacología , Dicumarol/farmacología , Relación Dosis-Respuesta a Droga , Electrones , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/farmacología , Ratones , Células 3T3 NIH , Naftoquinonas/farmacología , Ósmosis , Oxidación-Reducción , Isoformas de Proteínas , Desacopladores/farmacología , Vitamina K 3/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos
15.
PLoS One ; 7(2): e32527, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22389707

RESUMEN

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.


Asunto(s)
Proteínas/metabolismo , Proteómica/métodos , Células HeLa , Humanos , Espectrometría de Masas , Oxidación-Reducción , Proteínas/química
16.
PLoS One ; 7(11): e49744, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23166763

RESUMEN

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.


Asunto(s)
Dinoprostona/biosíntesis , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Tiocianatos/farmacología , Animales , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Isotiocianatos , Ratones , Regiones Promotoras Genéticas , Prostaglandina-E Sintasas , Unión Proteica/efectos de los fármacos , ARN Mensajero/genética , Sulfóxidos , Trasplante Heterólogo
17.
J Am Soc Mass Spectrom ; 21(1): 80-7, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19850495

RESUMEN

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.


Asunto(s)
Algoritmos , Péptidos/química , Proteómica/métodos , Espectrometría de Masas en Tándem/métodos , Secuencia de Aminoácidos , Bases de Datos de Proteínas , Humanos , Péptidos/análisis , Análisis de Secuencia de Proteína , Orina/química
18.
PLoS One ; 5(11): e15012, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21085594

RESUMEN

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.


Asunto(s)
Técnicas de Química Analítica/métodos , Cisteína/metabolismo , Proteínas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Cisteína/química , Cisteína/genética , Células HeLa , Humanos , Peróxido de Hidrógeno/farmacología , Immunoblotting , Proteína Quinasa 14 Activada por Mitógenos , Modelos Moleculares , Mutación , Oxidantes/farmacología , Oxidación-Reducción , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacología , Conformación Proteica , Proteínas/genética , Proteínas/aislamiento & purificación , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos , Compuestos de Sulfhidrilo/metabolismo , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/química , Proteínas Quinasas p38 Activadas por Mitógenos/genética
19.
Nat Methods ; 2(6): 435-41, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15908922

RESUMEN

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.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Genómica/métodos , Mutagénesis Sitio-Dirigida/genética , Mapeo de Interacción de Proteínas/métodos , Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Sustitución de Aminoácidos , Variación Genética/genética , Genómica/tendencias , Proteínas Quinasas/genética , Relación Estructura-Actividad
20.
J Cell Biochem ; 93(1): 104-11, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15352167

RESUMEN

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.


Asunto(s)
Apoptosis , Proteínas/metabolismo , Transducción de Señal , Compuestos de Sulfhidrilo , Animales , Humanos , Oxidación-Reducción , Fosforilación , Especies Reactivas de Oxígeno
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA