Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
2.
Int J Mol Sci ; 22(24)2021 Dec 11.
Article in English | MEDLINE | ID: mdl-34948122

ABSTRACT

The pro-apoptotic tumor suppressor BIN1 inhibits the activities of the neoplastic transcription factor MYC, poly (ADP-ribose) polymerase-1 (PARP1), and ATM Ser/Thr kinase (ATM) by separate mechanisms. Although BIN1 deficits increase cancer-cell resistance to DNA-damaging chemotherapeutics, such as cisplatin, it is not fully understood when BIN1 deficiency occurs and how it provokes cisplatin resistance. Here, we report that the coordinated actions of MYC, PARP1, and ATM assist cancer cells in acquiring cisplatin resistance by BIN1 deficits. Forced BIN1 depletion compromised cisplatin sensitivity irrespective of Ser15-phosphorylated, pro-apoptotic TP53 tumor suppressor. The BIN1 deficit facilitated ATM to phosphorylate the DNA-damage-response (DDR) effectors, including MDC1. Consequently, another DDR protein, RNF8, bound to ATM-phosphorylated MDC1 and protected MDC1 from caspase-3-dependent proteolytic cleavage to hinder cisplatin sensitivity. Of note, long-term and repeated exposure to cisplatin naturally recapitulated the BIN1 loss and accompanying RNF8-dependent cisplatin resistance. Simultaneously, endogenous MYC was remarkably activated by PARP1, thereby repressing the BIN1 promoter, whereas PARP inhibition abolished the hyperactivated MYC-dependent BIN1 suppression and restored cisplatin sensitivity. Since the BIN1 gene rarely mutates in human cancers, our results suggest that simultaneous inhibition of PARP1 and ATM provokes a new BRCAness-independent synthetic lethal effect and ultimately re-establishes cisplatin sensitivity even in platinum-refractory cancer cells.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/enzymology , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Protein Kinase Inhibitors/chemistry
3.
J Biol Chem ; 294(14): 5700-5719, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30733337

ABSTRACT

The tumor suppressor bridging integrator 1 (BIN1) is a corepressor of the transcription factor E2F1 and inhibits cell-cycle progression. BIN1 also curbs cellular poly(ADP-ribosyl)ation (PARylation) and increases sensitivity of cancer cells to DNA-damaging therapeutic agents such as cisplatin. However, how BIN1 deficiency, a hallmark of advanced cancer cells, increases cisplatin resistance remains elusive. Here, we report that BIN1 inactivates ataxia telangiectasia-mutated (ATM) serine/threonine kinase, particularly when BIN1 binds E2F1. BIN1 + 12A (a cancer-associated BIN1 splicing variant) also inhibited cellular PARylation, but only BIN1 increased cisplatin sensitivity. BIN1 prevented E2F1 from transcriptionally activating the human ATM promoter, whereas BIN1 + 12A did not physically interact with E2F1. Conversely, BIN1 loss significantly increased E2F1-dependent formation of MRE11A/RAD50/NBS1 DNA end-binding protein complex and efficiently promoted ATM autophosphorylation. Even in the absence of dsDNA breaks (DSBs), BIN1 loss promoted ATM-dependent phosphorylation of histone H2A family member X (forming γH2AX, a DSB biomarker) and mediator of DNA damage checkpoint 1 (MDC1, a γH2AX-binding adaptor protein for DSB repair). Of note, even in the presence of transcriptionally active (i.e. proapoptotic) TP53 tumor suppressor, BIN1 loss generally increased cisplatin resistance, which was conversely alleviated by ATM inactivation or E2F1 reduction. However, E2F2 or E2F3 depletion did not recapitulate the cisplatin sensitivity elicited by E2F1 elimination. Our study unveils an E2F1-specific signaling circuit that constitutively activates ATM and provokes cisplatin resistance in BIN1-deficient cancer cells and further reveals that γH2AX emergence may not always reflect DSBs if BIN1 is absent.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Ataxia Telangiectasia Mutated Proteins/metabolism , Cisplatin/pharmacology , Drug Resistance, Neoplasm , E2F1 Transcription Factor/metabolism , Neoplasms/metabolism , Nuclear Proteins/deficiency , Transcription, Genetic , Tumor Suppressor Proteins/deficiency , Acid Anhydride Hydrolases , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Repair/drug effects , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , E2F1 Transcription Factor/genetics , Histones/genetics , Histones/metabolism , Humans , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , Neoplasms/genetics , Neoplasms/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Signal Transduction/drug effects
5.
Nat Commun ; 8(1): 2103, 2017 12 13.
Article in English | MEDLINE | ID: mdl-29235459

ABSTRACT

The c-myc oncogene stimulates ribosomal biogenesis and protein synthesis to promote cellular growth. However, the pathway by which cells sense and restore dysfunctional mRNA translation and how this is linked to cell proliferation and growth is not known. We here show that mRNA translation stress in cis triggered by the gly-ala repeat sequence of Epstein-Barr virus (EBV)-encoded EBNA1, results in PI3Kδ-dependent induction of E2F1 mRNA translation with the consequent activation of c-Myc and cell proliferation. Treatment with a specific PI3Kδ inhibitor Idelalisib (CAL-101) suppresses E2F1 and c-Myc levels and causes cell death in EBNA1-induced B cell lymphomas. Suppression of PI3Kδ prevents E2F1 activation also in non-EBV-infected cells. These data illustrate an mRNA translation stress-response pathway for E2F1 activation that is exploited by EBV to promote cell growth and proliferation, offering new strategies to treat EBV-carrying cancers.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , E2F1 Transcription Factor/genetics , Protein Biosynthesis , RNA, Messenger/genetics , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , E2F1 Transcription Factor/metabolism , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Gene Expression Regulation, Neoplastic , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Host-Pathogen Interactions/genetics , Humans , Mice , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/virology
6.
Mol Cell Oncol ; 2(3): e991225, 2015.
Article in English | MEDLINE | ID: mdl-27308472

ABSTRACT

Over the last decades, accumulating data have advanced our understanding of the mechanism of action of tumor suppressor proteins and therapeutic strategies to restore tumor suppressor pathways have emerged as a promising approach for cancer therapy. Based on our recent findings on bridging integrator-1 (BIN1), we outline potential advantages and disadvantages of chemical activation of tumor suppressors.

7.
J Pathol ; 235(2): 334-41, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25186125

ABSTRACT

The EBV-encoded EBNA1 was first discovered 40 years ago, approximately 10 years after the presence of EBV had been demonstrated in Burkitt's lymphoma cells. It took another 10 years before the functions of EBNA1 in maintaining the viral genome were revealed, and it has since been shown to be an essential viral factor expressed in all EBV-carrying cells. Apart from serving to maintain the viral episome and to control viral replication and gene expression, EBNA1 also harbours a cis-acting mechanism that allows virus-carrying host cells to evade the immune system. This relates to a particular glycine-alanine repeat (GAr) within EBNA1 that has the capacity to suppress antigen presentation to the major histocompatibility complex (MHC) class I pathway. We discuss the role of the GAr sequence at the level of mRNA translation initiation, rather than at the protein level, as at least part of the mechanism to avoid MHC presentation. Interfering with this mechanism has become the focus of the development of immune-based therapies against EBV-carrying cancers, and some lead compounds that affect translation of GAr-carrying mRNAs have been identified. In addition, we describe the EBV-encoded ZEBRA factor and the switch from the latent to the lytic cycle as an alternative virus-specific target for treating EBV-carrying cancers. Understanding the molecular mechanisms of how EBNA1 and ZEBRA interfere with cellular pathways not only opens new therapeutic approaches but continues to reveal new cell-biological insights on the interplay between host and virus. This review is a tale of discoveries relating to how EBNA1 and ZEBRA have emerged as targets for specific cancer therapies against EBV-carrying diseases, and serves as an illustration of how mRNA translation can play roles in future immune-based strategies to target viral disease.


Subject(s)
Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Nuclear Antigens/metabolism , Herpesvirus 4, Human/metabolism , Neoplasms/virology , Trans-Activators/metabolism , Animals , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/therapy , Epstein-Barr Virus Nuclear Antigens/genetics , Gene Expression Regulation, Viral , Genetic Therapy/methods , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions , Humans , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Trans-Activators/genetics , Virulence
9.
J Cell Biochem ; 112(10): 2992-3001, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21678469

ABSTRACT

Bridging integrator 1 (BIN1) is a nucleocytoplasmic adaptor protein with tumor suppressor properties. The protein interacts with and inhibits the c-MYC transcription factor through the BIN1 MYC-binding domain (MBD). However, in vitro colony formation assays have clearly demonstrated that the MBD is not essential for BIN1-mediated growth arrest. We hypothesized that BIN1 contains a MYC-independent effector domain (MID) for cancer suppression. Because a functionally unique domain frequently contains a distinct structure, the human full-length BIN1 protein was subjected to limited trypsin digestion and the digested peptides were analyzed with Edman sequencing and mass spectrometry. We identified a trypsin-resistant peptide that corresponds to amino acids 146-268 of BIN1. It encompassed part of the BAR region, a putative effector region of BIN1. Computational analysis predicted that the peptide is very likely to exhibit coiled-coil motifs, implying a potential role for this region in sustaining the BIN1 structure and function. Like MBD-deleted BIN1, the trypsin-resistant peptide of BIN1 was predominantly present in the cytoplasm and was sufficient to inhibit cancer growth, regardless of dysregulated c-MYC activity. Our results suggest that the coiled-coil BIN1 BAR peptide encodes a novel BIN1 MID domain, through which BIN1 acts as a MYC-independent cancer suppressor.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Neoplasms/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Line , Humans , Mass Spectrometry , Nuclear Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Trypsin/metabolism , Tumor Suppressor Proteins/genetics
10.
Sci Signal ; 4(166): ra19, 2011 Mar 29.
Article in English | MEDLINE | ID: mdl-21447800

ABSTRACT

Cancer cells acquire resistance to DNA-damaging therapeutic agents, such as cisplatin, but the genetic mechanisms through which this occurs remain unclear. We show that the c-MYC oncoprotein increases cisplatin resistance by decreasing production of the c-MYC inhibitor BIN1 (bridging integrator 1). The sensitivity of cancer cells to cisplatin depended on BIN1 abundance, regardless of the p53 gene status. BIN1 bound to the automodification domain of and suppressed the catalytic activity of poly(ADP-ribose) polymerase 1 (PARP1, EC 2.4.2.30), an enzyme essential for DNA repair, thereby reducing the stability of the genome. The inhibition of PARP1 activity was sufficient for BIN1 to suppress c-MYC-mediated transactivation, the G(2)-M transition, and cisplatin resistance. Conversely, overexpressed c-MYC repressed BIN1 expression by blocking its activation by the MYC-interacting zinc finger transcription factor 1 (MIZ1) and thereby released PARP1 activity. Thus, a c-MYC-mediated positive feedback loop may contribute to cancer cell resistance to cisplatin.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Neoplasms/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Division/drug effects , Cell Division/genetics , Cell Line, Tumor , DNA Damage/genetics , DNA Repair/drug effects , DNA Repair/genetics , Drug Resistance, Neoplasm/genetics , G2 Phase/drug effects , G2 Phase/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/metabolism , Proto-Oncogene Proteins c-myc/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases
11.
J Biol Chem ; 285(19): 14366-76, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20212045

ABSTRACT

Homeostatic scaling of glutamatergic and GABAergic transmission is triggered by prolonged alterations in synaptic neuronal activity. We have previously described a presynaptic mechanism for synaptic homeostasis and plasticity that involves scaling the level of vesicular glutamate (VGLUT1) and gamma-aminobutyric acid (GABA) (VGAT) transporter biosynthesis. These molecular determinants of vesicle filling and quantal size are regulated by neuronal activity in an opposite manner and bi-directionally. Here, we report that a striking induction of VGLUT2 mRNA and synaptic protein is triggered by a prolonged increase in glutamatergic synaptic activity in mature neocortical neuronal networks in vitro together with two determinants of inhibitory synaptic strength, the neuronal activity-regulated pentraxin (Narp), and glutamate decarboxylase (GAD65). Activity-dependent induction of VGLUT2 and Narp exhibits a similar intermediate-early gene response that is blocked by actinomycin D and tetrodotoxin, by inhibitors of ionotropic glutamate receptors and L-type voltage-gated calcium channels, and is dependent on downstream signaling via calmodulin, calcium/calmodulin-dependent protein kinase (CaMK) and extracellular signal-regulated kinase 1/2 (ERK1/2). The co-induction of VGLUT2 and Narp triggered by prolonged gamma-aminobutyric acid type A receptor blockade is independent of brain-derived nerve growth factor and TrkB receptor signaling. VGLUT2 protein induction occurs on a subset of cortically derived synaptic vesicles in excitatory synapses on somata and dendritic processes of multipolar GABAergic interneurons, recognized sites for the clustering of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate glutamate receptors by Narp. We propose that VGLUT2 and Narp induction by excitation-transcription coupling leads to increased glutamatergic transmission at synapses on GABAergic inhibitory feedback neurons as part of a coordinated program of Ca(2+)-signal transcription involved in mechanisms of homeostatic plasticity after prolonged hyperactivity.


Subject(s)
C-Reactive Protein/metabolism , Glutamic Acid/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Transcription, Genetic , Vesicular Glutamate Transport Protein 2/genetics , Animals , Animals, Newborn , Blotting, Western , Brain-Derived Neurotrophic Factor/metabolism , Calcium/metabolism , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/metabolism , Calmodulin/metabolism , Cells, Cultured , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists , Gene Expression Regulation, Developmental , Glutamate Decarboxylase/metabolism , Immunoenzyme Techniques , Neuronal Plasticity , Neurons/cytology , Neurons/drug effects , Pyridazines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptor, trkA/metabolism , Receptors, GABA-A/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Synaptic Vesicles/metabolism , Vesicular Glutamate Transport Protein 1/genetics , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Glutamate Transport Protein 2/metabolism
12.
Mol Cell Proteomics ; 6(2): 193-206, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17092930

ABSTRACT

The study of protein interactions constitutes an important domain to understand the physiology and pathogenesis of microorganisms. The two-dimensional blue native/SDS-PAGE was initially reported to analyze membrane protein complexes. In this study, both cytoplasmic and membrane complexes of a bacterium, the strain J99 of the gastric pathogen Helicobacter pylori, were analyzed by this method. It was possible to identify 34 different proteins grouped in 13 multiprotein complexes, 11 from the cytoplasm and two from the membrane, either previously reported partially or totally in the literature. Besides complexes involved in H. pylori physiology, this method allowed the description of interactions involving known pathogenic factors such as (i) urease with the heat shock protein GroEL or with the putative ketol-acid reductoisomerase IlvC and (ii) the cag pathogenicity island CagA protein with the DNA gyrase GyrA as well as insight on the partners of TsaA, a peroxide reductase/stress-dependent molecular chaperone. The two-dimensional blue native/SDS-PAGE combined with mass spectrometry is a potential tool to study the differences in complexes isolated in various situations and also to study the interactions between bacterial and eucaryotic cell proteins.


Subject(s)
Bacterial Proteins/analysis , Electrophoresis, Gel, Two-Dimensional/methods , Helicobacter pylori/chemistry , Multiprotein Complexes/analysis , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Cytoplasm/metabolism , Helicobacter pylori/enzymology , Helicobacter pylori/pathogenicity , Mass Spectrometry , Membrane Proteins/analysis , Molecular Sequence Data , Urease/metabolism , Virulence
13.
Electrophoresis ; 27(16): 3306-21, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16858726

ABSTRACT

Study of the complexome - all the protein complexes of the cell - is essential for a better understanding and more global vision of cell function. Using two-dimensional blue native/SDS-PAGE (2-D BN/SDS-PAGE) technology, the cytosolic and membrane protein complexes of Escherichia coli were separated. Then, the different partners of each protein complex were identified by LC-MS/MS. In this report, 306 protein complexes were separated and identified. Among these protein complexes, 50 heteromultimeric and 256 homomultimeric protein complexes were found. Among the 50 heteromultimeric protein complexes, 18 previously described protein complexes validate the technology. In this study, 109 new protein complexes were found, providing insight into the function of previously uncharacterized bacterial proteins.


Subject(s)
Electrophoresis, Gel, Two-Dimensional/methods , Electrophoresis, Polyacrylamide Gel/methods , Escherichia coli Proteins/analysis , Escherichia coli/chemistry , Proteome/analysis , Chromatography, Liquid , Escherichia coli/metabolism , Escherichia coli Proteins/classification , Escherichia coli Proteins/metabolism , Mass Spectrometry , Proteome/metabolism
14.
Helicobacter ; 10(3): 185-92, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15904476

ABSTRACT

BACKGROUND: Most strategies for direct mutagenesis of Helicobacter pylori primarily involve genomic DNA cloning which is a time-consuming and expensive technique. METHODS: To make a gene replacement, we propose a strategy using polymerase chain reaction (PCR) amplicons to allow a double homologous recombination in the genome of H. pylori. Different strains were used to validate this strategy and we describe how the amplicon insertion was made with accuracy. Moreover, we looked for the shortest homologous sequence needed to allow a specific gene replacement in H. pylori without any deletion, insertion or mutation at the recombination site. All of the experiments were performed at the flaA locus, whose gene encodes the major flagellin. RESULTS: Amplicons bearing 500 or 150 bp flanking regions of flaA on each side (depending on the strain) were sufficient to allow the specific insertion of a 1173 bp chloramphenicol cassette into the genome of H. pylori. The insertion was accurate with no substitutions at the insertion locus. CONCLUSIONS: This information opens the door to other strategies for mutagenesis used for the identification of virulence factors without deleting genes, which would not be based on a negative screening system. For example, they could be useful in performing protein fusion for a better understanding of the virulence factor's mechanism.


Subject(s)
Flagellin/genetics , Helicobacter pylori/genetics , Recombination, Genetic , Transformation, Bacterial , Alleles , DNA, Bacterial/analysis , DNA, Bacterial/genetics , Escherichia coli/genetics , Molecular Sequence Data , Plasmids , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...