Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
NPJ Breast Cancer ; 8(1): 126, 2022 Nov 29.
Article in English | MEDLINE | ID: mdl-36446794

ABSTRACT

Despite the biological and therapeutic relevance of CDK4/6 for the treatment of HR+, HER2- advanced breast cancer, the detailed mode of action of CDK4/6 inhibitors is not completely understood. Of particular interest, phosphorylation of CDK4 at T172 (pT172) is critical for generating the active conformation, yet no such crystal structure has been reported to date. We describe here the x-ray structure of active CDK4-cyclin D3 bound to the CDK4/6 inhibitor abemaciclib and discuss the key aspects of the catalytically-competent complex. Furthermore, the effect of CDK4/6 inhibitors on CDK4 T172 phosphorylation has not been explored, despite its role as a potential biomarker of CDK4/6 inhibitor response. We show mechanistically that CDK4/6i stabilize primed (pT172) CDK4-cyclin D complex and selectively displace p21 in responsive tumor cells. Stabilization of active CDK4-cyclin D1 complex can lead to pathway reactivation following alternate dosing regimen. Consequently, sustained binding of abemaciclib to CDK4 leads to potent cell cycle inhibition in breast cancer cell lines and prevents rebound activation of downstream signaling. Overall, our study provides key insights demonstrating that prolonged treatment with CDK4/6 inhibitors and composition of the CDK4/6-cyclin D complex are both critical determinants of abemaciclib efficacy, with implications for this class of anticancer therapy.

3.
Sci Rep ; 12(1): 2673, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35177665

ABSTRACT

Cellular models recapitulating features of tauopathies are useful tools to investigate the causes and consequences of tau aggregation and the identification of novel treatments. We seeded rat primary cortical neurons with tau isolated from Alzheimer's disease brains to induce a time-dependent increase in endogenous tau inclusions. Transcriptomics of seeded and control cells identified 1075 differentially expressed genes (including 26 altered at two time points). These were enriched for lipid/steroid metabolism and neuronal/glial cell development genes. 50 genes were correlated with tau inclusion formation at both transcriptomic and proteomic levels, including several microtubule and cytoskeleton-related proteins such as Tubb2a, Tubb4a, Nefl and Snca. Several genes (such as Fyn kinase and PTBP1, a tau exon 10 repressor) interact directly with or regulate tau. We conclude that this neuronal model may be a suitable platform for high-throughput screens for target or hit compound identification and validation.


Subject(s)
Alzheimer Disease/metabolism , Gene Expression Regulation , Neurons/metabolism , Transcriptome , tau Proteins/metabolism , Humans
4.
Sci Rep ; 9(1): 7187, 2019 05 10.
Article in English | MEDLINE | ID: mdl-31076588

ABSTRACT

We have used cryo Electron Tomography, proteomics and immunolabeling to study centrosomes isolated from the young lamb thymus, an efficient source of quiescent differentiated cells. We compared the proteome of thymocyte centrosomes to data published for KE37 cells, focusing on proteins associated with centriole disengagement and centrosome separation. The data obtained enhances our understanding of the protein system joining the centrioles, a system comprised of a branched network of fibers linked to an apparently amorphous density that was partially characterized here. A number of proteins were localized to the amorphous density by immunolabeling (C-NAP1, cohesin SMC1, condensin SMC4 and NCAPD2), yet not DNA. In conjuction, these data not only extend our understanding of centrosomes but they will help refine the model that focus on the protein system associated with the centriolar junction.


Subject(s)
Centrosome/metabolism , Proteomics/methods , Thymocytes/cytology , Animals , Cell Line , Cryoelectron Microscopy , Electron Microscope Tomography , Gene Regulatory Networks , Sheep , Thymocytes/metabolism
5.
Traffic ; 18(8): 491-504, 2017 08.
Article in English | MEDLINE | ID: mdl-28477369

ABSTRACT

T Lymphocyte recognition of antigens leads to the formation of a highly organized structure termed immune synapse (IS) by analogy with the neuronals synapse. Sorting nexin 27 (SNX27) controls the endosomal traffic of PSD95, Dlg1, ZO-1 (PDZ) domain-interacting proteins, and its alteration is associated with impaired synaptic function and neurological diseases. In T-lymphocytes, SNX27-positive vesicles polarize to the IS, the identity of SNX27 interactors in these conditions nonetheless remains unknown. Here we used proteomics to analyze the SNX27 interactome purified from IS-forming T cells, and confirmed the conserved nature of the SNX27/WASH/retromer association in hematopoietic cells. Furthermore, our comparative interactome analysis of SNX27 wild-type and a mutant-deficient for PDZ cargo recognition identified the epithelial cell-cell junction protein zona occludens-2 (ZO-2) as an IS component. Biochemistry and microscopy approaches in T cells confirmed SNX27/ZO-2 PDZ-dependent interaction, and demonstrated its role controlling the dynamic localization of ZO-2 at the IS. This study broadens our knowledge of SNX27 function in T lymphocytes, and suggests that pathways that delimit polarized structures in nervous and epithelial systems also participate in IS regulation.


Subject(s)
Immunological Synapses/metabolism , Protein Interaction Maps , Sorting Nexins/metabolism , T-Lymphocytes/metabolism , Zonula Occludens-2 Protein/metabolism , Cell Line, Tumor , Humans , Protein Transport
6.
Mol Pharmacol ; 89(1): 42-52, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26487510

ABSTRACT

Aldose reductase (AKR1B1) is a critical drug target because of its involvement in diabetic complications, inflammation, and tumorigenesis. However, to date, development of clinically useful inhibitors has been largely unsuccessful. Cyclopentenone prostaglandins (cyPGs) are reactive lipid mediators that bind covalently to proteins and exert anti-inflammatory and antiproliferative effects in numerous settings. By pursuing targets for modification by cyPGs we have found that the cyPG PGA1 binds to and inactivates AKR1B1. A PGA1-AKR1B1 adduct was observed, both by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry and by SDS-PAGE using biotinylated PGA1 (PGA1-B). Insight into the molecular interactions between AKR1B1 and PGA1 was advanced by molecular modeling. This anticipated the addition of PGA1 to active site Cys298 and the potential reversibility of the adduct, which was supported experimentally. Indeed, loss of biotin label from the AKR1B1-PGA1-B adduct was favored by glutathione, indicating a retro-Michael reaction, which unveils new implications of cyPG-protein interaction. PGA1 elicited only marginal inhibition of aldehyde reductase (AKR1A1), considered responsible for the severe adverse effects of many AKR1B1 inhibitors. Interestingly, other prostaglandins (PGs) inhibited the enzyme, including non-electrophilic PGE1 and PGE2, currently used in clinical practice. Moreover, both PGA1 and PGE1 reduced the formation of sorbitol in an ex-vivo model of diabetic cataract to an extent comparable to that attained by the known AKR inhibitor epalrestat. Taken together, these results highlight the role of PGs as AKR1B1 inhibitors and the interest in PG-related molecules as leads for the development of novel pharmacological tools.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Aldehyde Reductase/metabolism , Prostaglandins A/metabolism , Prostaglandins A/pharmacology , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Male , Prostaglandins/metabolism , Prostaglandins/pharmacology , Protein Binding/physiology , Rats , Rats, Wistar
7.
Sci Signal ; 8(374): re6, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25921290

ABSTRACT

The diacylglycerol kinases (DGKs) attenuate diacylglycerol (DAG)-mediated signals by catalyzing the conversion of DAG to phosphatidic acid. In T lymphocytes, the antigen-stimulated generation of DAG links signal strength to the intensity and duration of signaling by the Ras-extracellular signal-regulated kinase (ERK) and protein kinase C (PKC)-dependent pathways. The generation of DAG at the plasma membrane of T cells lies at the core of the mechanisms that delimit T cell functions. DGKα and DGKζ are the two main isoforms that are found in T cells, and several approaches define their precise contribution to T cell responses. Each of these isoforms has specialized and redundant functions that limit the intensity of DAG-regulated signals downstream of antigenic stimulation. This ability, which in normal T cells contributes to maintaining homeostasis and function, is exploited by tumors to evade immune surveillance. Modification of DGK activity offers new perspectives for the therapeutic manipulation of T cell functions for treatment of autoimmune pathologies, or for overcoming tumor-induced T cell tolerance. Precise knowledge of the mechanisms that sustain DGK isoform-specific regulation in T lymphocytes is indispensable for the development of new tools for pharmacological intervention.


Subject(s)
Autoimmune Diseases/immunology , Diacylglycerol Kinase/immunology , Diglycerides/immunology , Neoplasms/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Tumor Escape , Animals , Autoimmune Diseases/pathology , Extracellular Signal-Regulated MAP Kinases/immunology , Humans , Immune Tolerance , Neoplasms/pathology , Protein Kinase C/immunology , T-Lymphocytes/pathology
8.
EMBO J ; 33(6): 559-77, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24502978

ABSTRACT

T-cell receptors (TCR) recognize their antigen ligand at the interface between T cells and antigen-presenting cells, known as the immunological synapse (IS). The IS provides a means of sustaining the TCR signal which requires the continual supply of new TCRs. These are endocytosed and redirected from distal membrane locations to the IS. In our search for novel cytoplasmic effectors, we have identified ß-arrestin-1 as a ligand of non-phosphorylated resting TCRs. Using dominant-negative and knockdown approaches we demonstrate that ß-arrestin-1 is required for the internalization and downregulation of non-engaged bystander TCRs. Furthermore, TCR triggering provokes the ß-arrestin-1-mediated downregulation of the G-protein coupled chemokine receptor CXCR4, but not of other control receptors. We demonstrate that ß-arrestin-1 recruitment to the TCR, and bystander TCR and CXCR4 downregulation, are mechanistically mediated by the TCR-triggered PKC-mediated phosphorylation of ß-arrestin-1 at Ser163. This mechanism allows the first triggered TCRs to deliver a stop migration signal, and to promote the internalization of distal TCRs and CXCR4 and their translocation to the IS. This receptor crosstalk mechanism is critical to sustain the TCR signal.


Subject(s)
Arrestins/metabolism , Gene Expression Regulation/immunology , Immunological Synapses/metabolism , Models, Immunological , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , Animals , Blotting, Western , Electroporation , Fluorescent Antibody Technique , Gene Knockdown Techniques , HEK293 Cells , Humans , Immunoprecipitation , Jurkat Cells , Mice , Mice, Transgenic , Microscopy, Fluorescence , Pyrimidines , Receptors, CXCR4/metabolism , Time-Lapse Imaging , beta-Arrestin 1 , beta-Arrestins
9.
J Proteome Res ; 13(1): 158-72, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24138474

ABSTRACT

The Spanish team of the Human Proteome Project (SpHPP) marked the annotation of Chr16 and data analysis as one of its priorities. Precise annotation of Chromosome 16 proteins according to C-HPP criteria is presented. Moreover, Human Body Map 2.0 RNA-Seq and Encyclopedia of DNA Elements (ENCODE) data sets were used to obtain further information relative to cell/tissue specific chromosome 16 coding gene expression patterns and to infer the presence of missing proteins. Twenty-four shotgun 2D-LC-MS/MS and gel/LC-MS/MS MIAPE compliant experiments, representing 41% coverage of chromosome 16 proteins, were performed. Furthermore, mapping of large-scale multicenter mass spectrometry data sets from CCD18, MCF7, Jurkat, and Ramos cell lines into RNA-Seq data allowed further insights relative to correlation of chromosome 16 transcripts and proteins. Detection and quantification of chromosome 16 proteins in biological matrices by SRM procedures are also primary goals of the SpHPP. Two strategies were undertaken: one focused on known proteins, taking advantage of MS data already available, and the second, aimed at the detection of the missing proteins, is based on the expression of recombinant proteins to gather MS information and optimize SRM methods that will be used in real biological samples. SRM methods for 49 known proteins and for recombinant forms of 24 missing proteins are reported in this study.


Subject(s)
Chromosomes, Human, Pair 16 , Proteome , Transcriptome , Chromatography, Liquid , Humans , Mass Spectrometry , Sequence Analysis, RNA
10.
J Cell Sci ; 126(Pt 10): 2176-86, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23525016

ABSTRACT

Considerable evidence indicates that diacylglycerol (DAG) generation at the immunological synapse (IS) determines T cell functions by regulating the duration and amplitude of Ras/ERK signals. The exact mechanism by which DAG regulates Ras/ERK activation downstream of the T cell receptor (TCR) nonetheless remains poorly understood. Here we characterize PKCα as a previously unrecognized component of the machinery that translates cell receptor occupancy into Ras/ERK-propagated signals. We show transient translocation of PKCα to the IS, mediated by DAG generation at the contact area. Diacylglycerol kinase (DGK)ζ negatively regulated PKCα translocation kinetics, whereas PKCα activity limited its own persistence at the IS. Coordinated activation of DGKζ and PKCα in response to antigen recognition regulated the amplitude and duration of Ras/ERK activation; this in turn mediated early processes of T cell surface proteolysis such as L-selectin shedding. Analysis of DGKζ-deficient mice further showed that increased DAG signaling is translated to downstream elements of this pathway, as reflected by enhanced PKCα-dependent L-selectin shedding. We propose that early activation of a DAG-PKCα axis contributes to the mechanisms by which antigen affinity translates into TCR biological responses.


Subject(s)
Cell Membrane/metabolism , Diacylglycerol Kinase/metabolism , Immunological Synapses/metabolism , Protein Kinase C-alpha/metabolism , T-Lymphocytes/immunology , Animals , Antigens/immunology , Diacylglycerol Kinase/genetics , Diacylglycerol Kinase/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback, Physiological , Humans , Jurkat Cells , L-Selectin/metabolism , Lymphocyte Activation/genetics , Mice , Mice, Knockout , Oncogene Protein p21(ras)/metabolism , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/immunology , Protein Transport/genetics , Protein Transport/immunology , RNA, Small Interfering/genetics , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/genetics
11.
Prog Lipid Res ; 51(1): 1-10, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22067957

ABSTRACT

Diacylglycerol (DAG) and phosphatidic acid (PA) are lipids with unique functions as metabolic intermediates, basic membrane constituents, and second-signal components. Diacylglycerol kinases (DGK) regulate the levels of these two lipids, catalyzing the interconversion of one to the other. The DGK family of enzymes is composed of 10 isoforms, grouped into five subfamilies based on the presence of distinct regulatory domains. From its initial characterization as a type IV DGK to the generation of mouse models showing its importance in cardiac dysfunction and immune pathologies, diacylglycerol kinase ζ (DGKζ) has proved an excellent example of the critical role of lipid-metabolizing enzymes in the control of cell responses. Although the mechanism that regulates this enzyme is not well known, many studies demonstrate its subtle regulation and its strategic function in specific signaling and as part of adaptor protein complexes. These data suggest that DGKζ offers new opportunities for therapeutic manipulation of lipid metabolism.


Subject(s)
Diacylglycerol Kinase/metabolism , Lipid Metabolism , Animals , Diacylglycerol Kinase/chemistry , Diglycerides/metabolism , Humans , Intracellular Space/enzymology , Intracellular Space/metabolism , Phosphatidic Acids/metabolism , Protein Structure, Tertiary
12.
Mol Biol Cell ; 22(22): 4406-14, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21937721

ABSTRACT

Diacylglycerol (DAG) generation at the T cell immunological synapse (IS) determines the correct activation of antigen-specific immune responses. DAG kinases (DGKs) α and ζ act as negative regulators of DAG-mediated signals by catalyzing DAG conversion to phosphatidic acid (PA). Nonetheless, the specific input of each enzyme and their spatial regulation during IS formation remain uncharacterized. Here we report recruitment of endogenous DGKα and DGKζ to the T cell receptor (TCR) complex following TCR/CD28 engagement. Specific DGK gene silencing shows that PA production at the activated complex depends mainly on DGKζ, indicating functional differences between these proteins. DGKζ kinase activity at the TCR is enhanced by phorbol-12-myristate-13-acetate cotreatment, suggesting DAG-mediated regulation of DGKζ responsiveness. We used GFP-DGKζ and -DGKα chimeras to assess translocation dynamics during IS formation. Only GFP-DGKζ translocated rapidly to the plasma membrane at early stages of IS formation, independent of enzyme activity. Finally, use of a fluorescent DAG sensor confirmed rapid, sustained DAG accumulation at the IS and allowed us to directly correlate membrane translocation of active DGKζ with DAG consumption at the IS. This study highlights a DGKζ-specific function for local DAG metabolism at the IS and offers new clues to its mode of regulation.


Subject(s)
Diacylglycerol Kinase/metabolism , Diglycerides/metabolism , Immunological Synapses/metabolism , T-Lymphocytes/immunology , CD28 Antigens/metabolism , Cell Line , Cell Membrane , Diacylglycerol Kinase/genetics , Diacylglycerol Kinase/immunology , Humans , Jurkat Cells , Phorbol Esters/pharmacology , Phosphatidic Acids/biosynthesis , RNA Interference , Receptors, Antigen, T-Cell/metabolism , Signal Transduction
13.
Cancer Res ; 71(12): 4161-71, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21507934

ABSTRACT

Cyclopentenone prostaglandins (cyPG) are reactive eicosanoids that may display anti-inflammatory and antiproliferative actions, possibly offering therapeutic potential. Here we report the identification of members of the aldo-keto reductase (AKR) family as selective targets of the cyPG prostaglandin A(1) (PGA(1)). AKR enzymes metabolize aldehydes and drugs containing carbonyl groups and are involved in inflammation and tumorigenesis. Thus, these enzymes represent a class of targets to develop small molecule inhibitors with therapeutic activity. Molecular modeling studies pointed to the covalent binding of PGA(1) to Cys299, close to the active site of AKR, with His111 and Tyr49, which are highly conserved in the AKR family, playing a role in PGA(1) orientation. Among AKR enzymes, AKR1B10 is considered as a tumor marker and contributes to tumor development and chemoresistance. We validated the direct modification of AKR1B10 by biotinylated PGA(1) (PGA(1)-B) in cells, and confirmed that mutation of Cys299 abolishes PGA(1)-B incorporation, whereas substitution of His111 or Tyr49 reduced the interaction. Modification of AKR1B10 by PGA(1) correlated with loss of enzymatic activity and both effects were increased by depletion of cellular glutathione. Moreover, in lung cancer cells PGA(1) reduced tumorigenic potential and increased accumulation of the AKR substrate doxorubicin, potentiating cell-cycle arrest induced by this chemotherapeutic agent. Our findings define PGA(1) as a new AKR inhibitor and they offer a framework to develop compounds that could counteract cancer chemoresistance.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Prostaglandins A/pharmacology , Aldehyde Reductase/chemistry , Aldo-Keto Reductases , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Drug Resistance, Neoplasm , Humans , Mice , Models, Molecular , Molecular Sequence Data , NIH 3T3 Cells
14.
J Cell Sci ; 124(Pt 5): 776-88, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21303929

ABSTRACT

Sorting nexin 27 (SNX27) belongs to the sorting nexin family of proteins, which participate in vesicular and protein trafficking. Similarly to all sorting nexin proteins, SNX27 has a functional PX domain that is important for endosome binding, but it is the only sorting nexin with a PDZ domain. We identified SNX27 as a partner of diacylglycerol kinase ζ (DGKζ), a negative regulator of T cell function that metabolises diacylglycerol to yield phosphatidic acid. SNX27 interacts with the DGKζ PDZ-binding motif in early/recycling endosomes in resting T cells; however, the dynamics and mechanisms underlying SNX27 subcellular localisation during T cell activation are unknown. We demonstrate that in T cells that encounter pulsed antigen-presenting cells, SNX27 in transit on early/recycling endosomes polarise to the immunological synapse. A fraction of SNX27 accumulates at the mature immunological synapse in a process that is dependent on vesicular trafficking, binding of the PX domain to phosphatidylinositol 3-phosphate and the presence of the PDZ region. Downmodulation of expression of either SNX27 or DGKζ results in enhanced basal and antigen-triggered ERK phosphorylation. These results identify SNX27 as a PDZ-containing component of the T cell immunological synapse, and demonstrate a role for this protein in the regulation of the Ras-ERK pathway, suggesting a functional relationship between SNX27 and DGKζ.


Subject(s)
Lymphocyte Activation , Protein Transport/physiology , Sorting Nexins/metabolism , T-Lymphocytes/metabolism , Diacylglycerol Kinase/metabolism , Endosomes/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Immunological Synapses/metabolism , Jurkat Cells , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sorting Nexins/genetics , T-Lymphocytes/cytology , ras Proteins/metabolism
15.
Methods Mol Biol ; 616: 69-82, 2010.
Article in English | MEDLINE | ID: mdl-20379869

ABSTRACT

Leucocyte transendothelial migration (TEM) or diapedesis is pivotal in leucocyte trafficking during the inflammatory and immune responses. The endothelium plays an active role in this process, triggering an array of signalling pathways and reorganizing its cytoskeleton and membrane to facilitate leucocyte TEM. Diapedesis can occur between endothelial cells (paracellular) or through individual endothelial cells (transcellular). This latter route accounts for up to 30% of the total diapedesis in certain endothelial cell types in vitro. Mechanisms underlying both routes of diapedesis have been subjected to intense investigation during recent years. Here we describe a method to discriminate between the paracellular and the transcellular routes of diapedesis in vitro. The method is based on a transmigration assay of human T lymphoblasts through TNF-alpha-stimulated human primary endothelial monolayers, a triple fluorescence labelling of F-actin, the adhesion receptor ICAM-1 and the junctional protein beta-catenin and a subsequent acquisition of z-stacks of high-resolution confocal sections.


Subject(s)
Biological Assay/methods , Cell Movement , Endothelial Cells/cytology , Leukocytes/cytology , Cell Movement/drug effects , Cells, Cultured , Dermis/cytology , Endothelial Cells/drug effects , Fluorescent Antibody Technique , Humans , Inflammation/pathology , Leukocytes/drug effects , Microscopy, Confocal , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Umbilical Veins/cytology
16.
Chem Biol Interact ; 183(1): 212-21, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-19800325

ABSTRACT

The cyclopentenone prostaglandin (cyPG) PGA(1) displays potent anti-proliferative and anti-inflammatory effects. Therefore, PGA(1) derivatives are being studied as therapeutic agents. One major mechanism for cyPG action is the modification of protein cysteine residues, the nature of the modified proteins being highly dependent on the structure of the cyPG. Biotinylated cyPGs may aid in the proteomic identification of cyPG targets of therapeutic interest. However, for the identified targets to be relevant it is critical to assess whether biotinylated cyPGs retain the desired biological activity. Here we have explored the anti-inflammatory, anti-proliferative and cell stress-inducing effects of a biotinylated analog of PGA(1) (PGA(1)-biotinamide, PGA(1)-B), to establish its validity to identify cyPG-protein interactions of potential therapeutic interest. PGA(1) and PGA(1)-B displayed similar effects on cell viability, Hsp70 and heme oxygenase-1 induction and pro-inflammatory gene inhibition. Remarkably, PGA(1)-B did not activate PPAR. Therefore, this biotinylated analog can be useful to identify PPAR-independent effects of cyPGs. Protein modification and subcellular distribution of PGA(1)-B targets were cell-type-dependent. Through proteomic and biochemical approaches we have identified a novel set of PGA(1)-B targets including proteins involved in stress response, protein synthesis, cytoskeletal regulation and carbohydrate metabolism. Moreover, the modification of several of the targets identified could be reproduced in vitro. These results unveil novel interactions of PGA(1) that will contribute to delineate the mechanisms for the anti-proliferative and metabolic actions of this cyPG.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Biotin/analogs & derivatives , Peroxisome Proliferator-Activated Receptors/metabolism , Prostaglandins A/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Biotin/chemistry , Biotin/pharmacology , Biotinylation , Cell Line , HSP70 Heat-Shock Proteins/metabolism , Heme Oxygenase-1/metabolism , Mice , NIH 3T3 Cells , Prostaglandins A/chemistry , Protein Processing, Post-Translational , Rats
17.
J Mass Spectrom ; 42(11): 1474-84, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17960581

ABSTRACT

Prostaglandins with cyclopentenone structure (cyPG) display potent antiproliferative actions that have elicited their study as potential anticancer agents. Several natural and synthetic analogs of the cyPG prostaglandin A(1) (PGA(1)) have proven antitumoral efficacy in cancer cell lines and animal models. In addition, PGA(1) has been used as an inhibitor of transcription factor NF-kappaB-mediated processes, including inflammatory gene expression and viral replication. An important determinant for these effects is the ability of cyPG to form Michael adducts with free thiol groups. The chemical nature of this interaction implies that PGA(1) could covalently modify cysteine residues in a large number of cellular proteins potentially involved in its beneficial effects. However, only a few targets of PGA(1) have been identified. In previous work, we have observed that a biotinylated analog of PGA(1) that retains the cyclopentenone moiety (PGA(1)-B) binds to multiple targets in fibroblasts. Here, we have addressed the identification of these targets through a proteomic approach. Cell fractionation followed by avidin affinity chromatography yielded a fraction enriched in proteins modified by PGA(1)-B. Analysis of this fraction by SDS-PAGE and LC-MS/MS allowed the identification of the chaperone Hsp90, elongation and initiation factors for protein synthesis and cytoskeletal proteins including actin, tubulin and vimentin. Furthermore, we have characterized the modification of vimentin both in vitro and in intact cells. Our observations indicate that cysteine 328 is the main site for PGA(1) addition. These results may contribute to a better understanding of the mechanism of action of PGA(1) and the potential of cyPG-based therapeutic strategies.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Antineoplastic Agents/metabolism , Prostaglandins A/metabolism , Vimentin/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Avidin/chemistry , Biotinylation , COS Cells , Chlorocebus aethiops , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Eukaryotic Initiation Factor-4A/chemistry , Eukaryotic Initiation Factor-4A/metabolism , Glial Fibrillary Acidic Protein/chemistry , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice , Mutation , NIH 3T3 Cells , Peptide Elongation Factor 1/chemistry , Peptide Elongation Factor 1/metabolism , Prostaglandins A/chemistry , Prostaglandins A/pharmacology , Protein Processing, Post-Translational/drug effects , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , Transfection , Tubulin/chemistry , Tubulin/metabolism , Vimentin/chemistry , Vimentin/genetics
18.
Proc Natl Acad Sci U S A ; 104(19): 7809-14, 2007 May 08.
Article in English | MEDLINE | ID: mdl-17470792

ABSTRACT

Class IA phosphoinositide 3-kinases (PI3Ks) signal downstream of tyrosine kinases and Ras and control a wide variety of biological responses. In mammals, these heterodimeric PI3Ks consist of a p110 catalytic subunit (p110alpha, p110beta, or p110delta) bound to any of five distinct regulatory subunits (p85alpha, p85beta, p55gamma, p55alpha, and p50alpha, collectively referred to as "p85s"). The relative expression levels of p85 and p110 have been invoked to explain key features of PI3K signaling. For example, free (i.e., non-p110-bound) p85alpha has been proposed to negatively regulate PI3K signaling by competition with p85/p110 for recruitment to phosphotyrosine docking sites. Using affinity and ion exchange chromatography and quantitative mass spectrometry, we demonstrate that the p85 and p110 subunits are present in equimolar amounts in mammalian cell lines and tissues. No evidence for free p85 or p110 subunits could be obtained. Cell lines contain 10,000-15,000 p85/p110 complexes per cell, with p110beta and p110delta being the most prevalent catalytic subunits in nonleukocytes and leukocytes, respectively. These results argue against a role of free p85 in PI3K signaling and provide insights into the nonredundant functions of the different class IA PI3K isoforms.


Subject(s)
Phosphatidylinositol 3-Kinases/chemistry , Animals , Catalytic Domain , Dimerization , Mass Spectrometry , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Phosphatidylinositol 3-Kinases/analysis , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/physiology , Protein Subunits , RNA, Messenger/analysis , Signal Transduction
19.
Biochem J ; 404(1): 15-21, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17302559

ABSTRACT

The PI3Ks (phosphatidylinositol 3-kinases) regulate cellular signalling networks that are involved in processes linked to the survival, growth, proliferation, metabolism and specialized differentiated functions of cells. The subversion of this network is common in cancer and has also been linked to disorders of inflammation. The elucidation of the physiological function of PI3K has come from pharmacological studies, which use the enzyme inhibitors Wortmannin and LY294002, and from PI3K genetic knockout models of the effects of loss of PI3K function. Several reports have shown that LY294002 is not exclusively selective for the PI3Ks, and could in fact act on other lipid kinases and additional apparently unrelated proteins. Since this inhibitor still remains a drug of choice in numerous PI3K studies (over 500 in the last year), it is important to establish the precise specificity of this compound. We report here the use of a chemical proteomic strategy in which an analogue of LY294002, PI828, was immobilized onto epoxy-activated Sepharose beads. This affinity material was then used as a bait to fish-out potential protein targets from cellular extracts. Proteins with high affinity for immobilized PI828 were separated by one-dimensional gel electrophoresis and identified by liquid chromatography-tandem MS. The present study reveals that LY294002 not only binds to class I PI3Ks and other PI3K-related kinases, but also to novel targets seemingly unrelated to the PI3K family.


Subject(s)
Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Morpholines/pharmacology , Proteins/antagonists & inhibitors , Animals , Cell Line , Cell Survival/drug effects , Cells, Cultured , Culture Media , Humans , Kinetics , Models, Animal , Proteasome Endopeptidase Complex , Proteins/chemical synthesis , Proteins/genetics , Signal Transduction/drug effects
20.
Proc Natl Acad Sci U S A ; 103(24): 8959-64, 2006 Jun 13.
Article in English | MEDLINE | ID: mdl-16751276

ABSTRACT

The phosphoinositide 3-kinase (PI3K)/Akt pathway controls a vast array of normal physiological processes and is frequently aberrantly activated in cancer, thus identifying PI3K/Akt-signaling components as promising drug targets in oncology. However, implementation of rational cancer therapies for this pathway needs robust and simple tools to stratify patients according to PI3K pathway activation and to validate and measure the impact of targeted inhibition on primary cancer tissues. Herein we present a technique for the quantification of the PI3K/Akt-signaling pathway based on the mass spectrometric measurement of PI3K-dependent protein kinase activity in cell lysates. The concept of this application of MS is to exploit enzymatic activity to amplify the signal of the enzyme under study analogous to the PCR used to amplify nucleic acid sequences. We show that this approach allows quantitative analysis of a cell-signaling pathway with high sensitivity, precision of quantification, and specificity. Due to its special analytical capabilities and potential for multiplexing, this approach could contribute significantly to cell-signaling studies and to the development and implementation of personalized cancer therapies.


Subject(s)
Mass Spectrometry/methods , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Animals , Cell Line, Tumor , Enzyme Inhibitors/metabolism , Isotopes/chemistry , Mice , Neoplasms/enzymology , Phosphatidylinositol 3-Kinases/chemistry , Proto-Oncogene Proteins c-akt/chemistry , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...