Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Proc Natl Acad Sci U S A ; 119(49): e2213146119, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36449545

ABSTRACT

Activation of endogenous retrotransposons frequently occurs in cancer cells and contributes to tumor genomic instability. To test whether inhibition of retrotranspositions has an anticancer effect, we used treatment with the nucleoside reverse transcriptase inhibitor (NRTI) stavudine (STV) in mouse cancer models, MMTV-HER2/Neu and Th-MYCN, that spontaneously develop breast cancer and neuroblastoma, respectively. In both cases, STV in drinking water did not affect tumor incidence nor demonstrate direct antitumor effects. However, STV dramatically extended progression-free survival in both models following an initial complete response to chemotherapy. To approach the mechanism underlying this phenomenon, we analyzed the effect of NRTI on the selection of treatment-resistant variants in tumor cells in culture. Cultivation of mouse breast carcinoma 4T1 in the presence of STV dramatically reduced the frequency of cells capable of surviving treatment with anticancer drugs. Global transcriptome analysis demonstrated that the acquisition of drug resistance by 4T1 cells was accompanied by an increase in the constitutive activity of interferon type I and NF-κB pathways and an elevated expression of LINE-1 elements, which are known to induce inflammatory responses via their products of reverse transcription. Treatment with NRTI reduced NF-κB activity and reverted drug resistance. Furthermore, the inducible expression of LINE-1 stimulated inflammatory response and increased the frequency of drug-resistant variants in a tumor cell population. These results indicate a mechanism by which retrotransposon desilencing can stimulate tumor cell survival during treatment and suggest reverse transcriptase inhibition as a potential therapeutic approach for targeting the development of drug-resistant cancers.


Subject(s)
Retroelements , Reverse Transcriptase Inhibitors , Animals , Mice , Reverse Transcriptase Inhibitors/pharmacology , Retroelements/genetics , NF-kappa B , Drug Resistance, Neoplasm/genetics , Long Interspersed Nucleotide Elements
2.
Int J Cancer ; 146(7): 1902-1916, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31325323

ABSTRACT

Around 10% of acute leukemias harbor a rearrangement of the MLL/KMT2A gene, and the presence of this translocation results in a highly aggressive, therapy-resistant leukemia subtype with survival rates below 50%. There is a high unmet need to identify safer and more potent therapies for MLL-rearranged (MLL-r) leukemia that can be combined with established chemotherapeutics to decrease treatment-related toxicities. The curaxin, CBL0137, has demonstrated nongenotoxic anticancer and chemopotentiating effects in a number of preclinical cancer models and is currently in adult Phase I clinical trials for solid tumors and hematological malignancies. The aim of our study was to investigate whether CBL0137 has potential as a therapeutic and chemopotentiating compound in MLL-r leukemia through a comprehensive analysis of its efficacy in preclinical models of the disease. CBL0137 decreased the viability of a panel of MLL-r leukemia cell lines (n = 12) and xenograft cells derived from patients with MLL-r acute lymphoblastic leukemia (ALL, n = 3) in vitro with submicromolar IC50s. The small molecule drug was well-tolerated in vivo and significantly reduced leukemia burden in a subcutaneous MV4;11 MLL-r acute myeloid leukemia model and in patient-derived xenograft models of MLL-r ALL (n = 5). The in vivo efficacy of standard of care drugs used in remission induction for pediatric ALL was also potentiated by CBL0137. CBL0137 exerted its anticancer effect by trapping Facilitator of Chromatin Transcription (FACT) into chromatin, activating the p53 pathway and inducing an Interferon response. Our findings support further preclinical evaluation of CBL0137 as a new approach for the treatment of MLL-r leukemia.


Subject(s)
Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Gene Rearrangement , Histone-Lysine N-Methyltransferase/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/genetics , Carbazoles/therapeutic use , Cell Line, Tumor , DNA-Binding Proteins/genetics , Disease Models, Animal , Gene Expression Profiling , High Mobility Group Proteins/genetics , Humans , Kaplan-Meier Estimate , Leukemia, Biphenotypic, Acute/diagnosis , Leukemia, Biphenotypic, Acute/drug therapy , Leukemia, Biphenotypic, Acute/genetics , Leukemia, Biphenotypic, Acute/mortality , Mice , Signal Transduction/drug effects , Transcriptional Elongation Factors/genetics , Transcriptome , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
3.
J Virol ; 84(18): 9390-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20631142

ABSTRACT

The 9-aminoacridine (9AA) derivative quinacrine (QC) has a long history of safe human use as an antiprotozoal and antirheumatic agent. QC intercalates into DNA and RNA and can inhibit DNA replication, RNA transcription, and protein synthesis. The extent of QC intercalation into RNA depends on the complexity of its secondary and tertiary structure. Internal ribosome entry sites (IRESs) that are required for initiation of translation of some viral and cellular mRNAs typically have complex structures. Recent work has shown that some intercalating drugs, including QC, are capable of inhibiting hepatitis C virus IRES-mediated translation in a cell-free system. Here, we show that QC suppresses translation directed by the encephalomyocarditis virus (EMCV) and poliovirus IRESs in a cell-free system and in virus-infected HeLa cells. In contrast, IRESs present in the mammalian p53 transcript that are predicted to have less-complex structures were not sensitive to QC. Inhibition of IRES-mediated translation by QC correlated with the affinity of binding between QC and the particular IRES. Expression of viral capsid proteins, replication of viral RNAs, and production of virus were all strongly inhibited by QC (and 9AA). These results suggest that QC and similar intercalating drugs could potentially be used for treatment of viral infections.


Subject(s)
Antiviral Agents/pharmacology , Encephalomyocarditis virus/drug effects , Poliovirus/drug effects , Quinacrine/pharmacology , Virus Replication/drug effects , Binding Sites , Encephalomyocarditis virus/physiology , HeLa Cells , Humans , Nucleic Acid Conformation , Poliovirus/physiology , Protein Biosynthesis/drug effects , RNA, Viral/metabolism , Viral Proteins/biosynthesis
4.
FASEB J ; 19(8): 1030-2, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15811878

ABSTRACT

Chronic inflammation is known to promote cancer, suggesting that negative regulation of inflammation is likely to be tumor suppressive. We found that p53 is a general inhibitor of inflammation that acts as an antagonist of nuclear factor kappaB (NFkappaB). We first observed striking similarities in global gene expression profiles in human prostate cancer cells LNCaP transduced with p53 inhibitory genetic element or treated with TNF, suggesting that p53 inhibits transcription of TNF-inducible genes that are largely regulated by NFkappaB. Consistently, ectopically expressed p53 acts as an inhibitor of transcription of NFkappaB-dependent promoters. Furthermore, suppression of inflammatory response by p53 was observed in vivo in mice by comparing wild-type and p53 null animals at molecular (inhibition of transcription of genes encoding cytokines and chemokines, reducing accumulation of reactive oxygen species and protein oxidation products), cellular (activation of macrophages and neutrophil clearance) and organismal (high levels of metabolic markers of inflammation in tissues of p53-deficient mice and their hypersensitivity to LPS) levels. These observations indicate that p53, acting through suppression of NFkappaB, plays the role of a general "buffer" of innate immune response in vivo that is well consistent with its tumor suppressor function and frequent constitutive activation of NFkappaB in tumors.


Subject(s)
Inflammation/prevention & control , NF-kappa B/antagonists & inhibitors , Tumor Suppressor Protein p53/physiology , Animals , Cecum/surgery , Chemokines/genetics , Cytokines/genetics , DNA/metabolism , Humans , Inflammation/chemically induced , Ligation , Lipopolysaccharides/pharmacology , Macrophage Activation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/analysis , NF-kappa B/physiology , Neutrophils/physiology , Oligonucleotide Array Sequence Analysis , Oxidation-Reduction , Peritonitis/etiology , Peroxidase/blood , Phagocytosis , Promoter Regions, Genetic/genetics , Punctures , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Shock, Septic/mortality , Thioglycolates , Transcription, Genetic/drug effects , Transcriptional Activation/physiology , Transfection , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
5.
Cancer Res ; 76(22): 6620-6630, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27680682

ABSTRACT

Isolated limb perfusion (ILP) with the chemotherapeutic agent melphalan is an effective treatment option for extremity in-transit melanoma but is toxic and technically challenging to deliver locoregionally. CBL0137 is an experimental clinical drug with broad anticancer activity in animal models, owing to its ability to bind DNA in a nongenotoxic manner and inactivate the FACT chromatin modulator essential for tumor cell viability. Here, we report that CBL0137 delivered by ILP in a murine melanoma model is as efficacious as melphalan, displaying antitumor activity at doses corresponding to only a fraction of the systemic MTD of CBL0137. The ability to bind DNA quickly combined with a favorable safety profile made it possible to substitute CBL0137 in the ILP protocol, using an intra-arterial infusion method, to safely achieve effective tumor suppression. Our findings of a preclinical proof of concept for CBL0137 and its administration via intra-arterial infusion as a superior treatment compared with melphalan ILP allows for locoregional treatment anywhere a catheter can be placed. Cancer Res; 76(22); 6620-30. ©2016 AACR.


Subject(s)
Extremities/pathology , Infusion Pumps , Melanoma/drug therapy , Animals , Female , Humans , Melanoma/pathology , Mice , Mice, Inbred C57BL , Treatment Outcome , Validation Studies as Topic
6.
DNA Cell Biol ; 24(12): 777-85, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16332174

ABSTRACT

Tumor necrosis factor (TNF) induces apoptosis in sensitive cells in culture when used in combination with inhibitors of transcription or translation. We applied the genetic suppressor element (GSE) methodology to search for the genetic elements protecting NIH3T3 cells from TNF-stimulated death. Ten putative GSEs were isolated from TNF-resistant cells, one of which (GSE0-1) corresponded to the cDNA sequence known as the mouse homolog of human serologically defined colon cancer antigen 3 (SDCCAG3). SDCCAG3 protein contains the region similar to the coiled-coil domain of the myosin tail. The same domain is present in the proteins related to the organelles/proteins trafficking, such as kinesin, Golgin-160, and dynein. We proposed that the SDCCAG3 function might be related to protein trafficking and secretion. The expression of the coiledcoil domain as the dominant negative mutant form of SDCCAG3 made the NIH3T3 and HeLa cells resistant to TNF-specific apoptosis. The presentation of TNFR1 at the surface of these cells was reduced, which affected the sensitivity of the cells to the TNF treatment. We recently showed that the inhibition of protein trafficking and secretion depleted the unstable TNFR1 from plasma membrane. The inhibition of SDCCAG3 activity by its dominant negative mutant suppressed the protein trafficking and secretion, and decreased TNFR1 presentation on the cell surface. Based on these results, we presume that SDCCAG3 is important for protein trafficking and presentation of TNFR1 on the cell surface. Therefore, SDCCAG3 can be viewed as a potential target for modulation of TNF response.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Apoptosis/immunology , Receptors, Tumor Necrosis Factor, Type I/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Blotting, Western , DNA Primers , Flow Cytometry , Gene Library , HeLa Cells , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Luciferases , Mice , Mutation/genetics , NIH 3T3 Cells , Protein Transport/immunology , Suppression, Genetic/genetics , Suppression, Genetic/immunology , Vesicular Transport Proteins
7.
DNA Cell Biol ; 23(3): 175-82, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15068587

ABSTRACT

In contrast with hematopoietic cells and fibroblasts, which express mainly one form of protein tyrosine phosphatase (PTP) SHP-1 or SHP-2, epithelial cells like A431, HeLa, and 293 express both forms of PTP. These two PTP regulate NFkappaB activity differently; SHP-1 inhibits and SHP-2 stimulates NFkappaB activation. In epithelial cells the process of NFkappaB activation depends on the combination of two PTP activities. The activity of PTP SHP-1 dominates in this tandem according to our data. The signal regulatory protein (SIRPalpha) is the adapter and the substrate of PTP SHP-1 and SHP-2. We investigated the role of SIRPalpha and its dominant negative mutant in PTP activities in 293 cells. The overexpression of wild-type SIRPalpha suppresses the activities of both PTP, but has a stronger effect on PTP SHP-2, especially when this protein is overexpressed in 293 cells. In contrast with wild-type SIRPalpha, its dominant negative mutant acts predominantly against PTP SHP-1, and can be detected in the complex with PTP SHP-1. The expression of dominant negative mutant of SIRPalpha has an effect similar to the expression of dominant negative PTP SHP-1 in the process of NFkappaB activation.


Subject(s)
Antigens, Differentiation , Membrane Glycoproteins/genetics , NF-kappa B/metabolism , Neural Cell Adhesion Molecule L1/genetics , Protein Tyrosine Phosphatases/metabolism , Receptors, Immunologic/genetics , Fibroblasts , Genes, Dominant , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Receptors, Immunologic/metabolism
8.
Oncotarget ; 2(3): 209-21, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21444945

ABSTRACT

Proteotoxic stress (PS) is generated in cells under a variety of conditions involving accumulation of misfolded proteins. To avoid the toxicity of unmitigated PS, cells activate the heat shock response (HSR). HSR involves upregulation of factors such as ubiquitin and the non-housekeeping chaperone Hsp70 which assist with metabolism of aberrant proteins. The PS-HSR axis is a potential anticancer treatment target since many tumor cells display constitutive PS and dependence on HSR due to their rapid rates of proliferation and translation. In fact, induction of PS via stimulation of protein misfolding (hyperthermia), inhibition of proteasomes (bortezomib) or inhibition of Hsp90 (geldanamycin) have all been considered or used for cancer treatment. We found that combination of bortezomib with an inducer of protein misfolding (hyperthermia or puromycin) resulted in enhanced PS. HSR was also induced, but could not mitigate the elevated PS and the cells died via largely p53-independent apoptosis. Thus, combination treatments were more cytotoxic in vitro than the component single treatments. Consistent with this, combination of non-toxic doses of puromycin with bortezomib significantly increased the antitumor activity of bortezomib in a mouse model of multiple myeloma. These results provide support for using combination treatments that disrupt the balance of PS and HSR to increase the therapeutic index of anticancer therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Multiple Myeloma/metabolism , Multiple Myeloma/therapy , Proteasome Inhibitors , Proteostasis Deficiencies/metabolism , Pyrazines/pharmacology , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Boronic Acids/administration & dosage , Bortezomib , Cell Line, Tumor , Combined Modality Therapy , Drug Synergism , HCT116 Cells , HSP70 Heat-Shock Proteins/biosynthesis , HSP90 Heat-Shock Proteins/biosynthesis , HeLa Cells , Heat-Shock Response/drug effects , Humans , Hyperthermia, Induced , Mice , Mice, Inbred BALB C , Multiple Myeloma/drug therapy , Proteasome Endopeptidase Complex/metabolism , Proteostasis Deficiencies/chemically induced , Puromycin/administration & dosage , Puromycin/pharmacology , Pyrazines/administration & dosage
10.
Cell Cycle ; 8(23): 3960-70, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19901558

ABSTRACT

The number of physical conditions and chemical agents induce accumulation of misfolded proteins creating proteotoxic stress. This leads to activation of adaptive pro-survival pathway, known as heat shock response (HSR), resulting in expression of additional chaperones. Several cancer treatment approaches, such as proteasome inhibitor Bortezomib and hsp90 inhibitor geldanamycin, involve activation of proteotoxic stress. Low efficacy of these therapies is likely due to the protective effects of HSR induced in treated cells, making this pathway an attractive target for pharmacological suppression. We found that the anti-malaria drugs quinacrine (QC) and emetine prevented HSR in cancer cells, as judged by induction of hsp70 expression. As opposed to emetine, which inhibited general translation, QC did not affect protein synthesis, but rather suppressed inducible HSF1-dependent transcription of the hsp70 gene in a relatively selective manner. The treatment of tumor cells in vitro with a combination of non-toxic concentrations of QC and proteotoxic stress inducers resulted in rapid induction of apoptosis. The effect was similar if QC was substituted by siRNA against hsp70, suggesting that the HSR inhibitory activity of QC was responsible for cell sensitization to proteotoxic stress inducers. QC was also found to enhance the antitumor efficacy of proteotoxic stress inducers in vivo: combinatorial treatment with 17-DMAG + QC resulted in suppression of tumor growth in two mouse syngeneic models. These results reveal that QC is an inhibitor of HSF1-mediated HSR. As such, this compound has significant clinical potential as an adjuvant in therapeutic strategies aimed at exploiting the cytotoxic potential of proteotoxic stress.


Subject(s)
Antimalarials/pharmacology , Antineoplastic Agents/pharmacology , Heat-Shock Response/drug effects , Quinacrine/pharmacology , Apoptosis , Benzoquinones/pharmacology , Boronic Acids/pharmacology , Bortezomib , DNA-Binding Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , HeLa Cells , Heat Shock Transcription Factors , Humans , Lactams, Macrocyclic/pharmacology , Neoplasms/drug therapy , Pyrazines/pharmacology , RNA, Small Interfering/metabolism , Transcription Factors/metabolism
11.
DNA Cell Biol ; 27(4): 191-8, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18067462

ABSTRACT

Flavonoid quercetin and its derivative, methylquercetin, inhibit the replication of poliovirus in several cell lines. Here, we show that replication of poliovirus is inhibited by quercetin and that the extent of this inhibition depends on the intracellular content of pirin, a quercetinase. HeLa cells contain higher content of pirin protein than normal kidney human epithelial (NKE) or 293 cells do. Poliovirus replication in HeLa cells is significantly more resistant to quercetin than its replication in NKE and 293 cells. Overexpression of pirin reduced antiviral inhibitory effect of quercetin, while siRNA-induced suppression of pirin level made poliovirus replication more sensitive to the flavonoid. The results suggest that quercetinase activity of pirin determines the resistance of poliovirus infection to quercetin.


Subject(s)
Antioxidants/pharmacology , Carrier Proteins/metabolism , Drug Resistance, Viral , Nuclear Proteins/metabolism , Poliovirus/drug effects , Poliovirus/physiology , Quercetin/pharmacology , Virus Replication/drug effects , Androstadienes/pharmacology , Cell Line , Dioxygenases/metabolism , Drug Resistance, Viral/drug effects , HeLa Cells , Humans , Protein Kinase Inhibitors/pharmacology , Wortmannin
12.
PLoS One ; 3(4): e1887, 2008 Apr 02.
Article in English | MEDLINE | ID: mdl-18382670

ABSTRACT

Proteasome activity is an important part of viral replication. In this study, we examined the effect of proteasome inhibitors on the replication of vesicular stomatitis virus (VSV) and poliovirus. We found that the proteasome inhibitors significantly suppressed VSV protein synthesis, virus accumulation, and protected infected cells from toxic effect of VSV replication. In contrast, poliovirus replication was delayed, but not diminished in the presence of the proteasome inhibitors MG132 and Bortezomib. We also found that inhibition of proteasomes stimulated stress-related processes, such as accumulation of chaperone hsp70, phosphorylation of eIF2alpha, and overall inhibition of translation. VSV replication was sensitive to this stress with significant decline in replication process. Poliovirus growth was less sensitive with only delay in replication. Inhibition of proteasome activity suppressed cellular and VSV protein synthesis, but did not reduce poliovirus protein synthesis. Protein kinase GCN2 supported the ability of proteasome inhibitors to attenuate general translation and to suppress VSV replication. We propose that different mechanisms of translational initiation by VSV and poliovirus determine their sensitivity to stress induced by the inhibition of proteasomes. To our knowledge, this is the first study that connects the effect of stress induced by proteasome inhibition with the efficiency of viral infection.


Subject(s)
Poliovirus/metabolism , Proteasome Inhibitors , Vesiculovirus/metabolism , Virus Replication , Boronic Acids/pharmacology , Bortezomib , DNA/metabolism , DNA Replication , Eukaryotic Initiation Factor-2/metabolism , HeLa Cells , Humans , Leupeptins/pharmacology , Models, Biological , Proteasome Endopeptidase Complex/metabolism , Pyrazines/pharmacology , Transfection , Viral Proteins/metabolism
13.
Cell Cycle ; 4(10): 1403-10, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16138011

ABSTRACT

Many viruses encode anti-apoptotic proteins that have been used as valuable tools for identification and analysis of key cellular regulators of programmed cell death. Here we demonstrate that the poliovirus protein 3A, previously shown to exhibit anti-apoptotic activity, binds and inactivates LIS1, a component of the dynein/dynactin motor complex, encoded by the gene mutated in patients with type I lissencephaly ("smooth brain"), thereby causing deregulation of endoplasmatic reticilum-to-Golgi vesicular transport, resulting in rapid disappearance of short-living receptors from the plasma membrane and loss of cell sensitivity to TNF and interferon. Truncated derivatives of LIS1, acting in a dominant negative manner, cause similar effects. However, 3A, being an endoplasmic reticulum-bound protein, locks Golgi-targeted YFP in the endoplasmatic reticilum, while expression of LIS1 mutants results in a dispersed cytoplasmic localization of the reporter protein. LIS1 dysfunction caused by ectopic expressing 3A or LIS1 mutants, as well as by overexpression of wild type LIS1, leads to cell blocking at the postmitotic stage associated with inability to undergo cytokinesis. Thus, the use of poliovirus protein as a research tool allowed us to reveal the role of cellular protein LIS1 in membrane protein trafficking, maintenance of Golgi integrity, surface presentation of unstable receptors, cell sensitivity to TNF-induced apoptosis and cell cycle progression.


Subject(s)
Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Viral Core Proteins/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase , Animals , Cell Division , Cell Line , Cell Membrane/metabolism , Gene Expression Regulation , Golgi Apparatus/metabolism , Humans , Mice , Microtubule-Associated Proteins/genetics , Mutation/genetics , Poliovirus , Protein Binding , Protein Transport , Viral Core Proteins/genetics
14.
J Biol Chem ; 280(25): 24153-8, 2005 Jun 24.
Article in English | MEDLINE | ID: mdl-15845545

ABSTRACT

Activation of NF-kappaB during viral infection is one of the critical elements in innate immune response. Several virus-specific factors, such as double-stranded RNA, can trigger host defense mechanisms by inducing NF-kappaB-mediated expression of cytokines and interferons. Early stages of poliovirus infection are also associated with degradation of IkappaB alpha and translocation of NF-kappaB into the nucleus. However, at later stages of poliovirus replication the p65-RelA component of the NF-kappaB complex undergoes a specific cleavage that coincides with the onset of intensive poliovirus protein synthesis and the appearance of the activity of poliovirus protease 3C. Indeed, the p65-RelA amino acid sequence contains the recognition site for 3C, and recombinant protein 3C was shown to be capable of proteolytic cleavage of p65-RelA, generating truncated product similar to that observed during poliovirus infection. Cleavage of p65-RelA occurs during replication of ECHO-1 and rhinovirus 14, suggesting that inactivation of NF-kappaB function by proteolytic cleavage of p65-RelA is the common mechanism by which picornaviruses suppress the innate immune response.


Subject(s)
NF-kappa B/metabolism , Poliomyelitis/metabolism , Amino Acid Sequence , Base Sequence , DNA Primers , Electrophoretic Mobility Shift Assay , HeLa Cells , Humans , Hydrolysis , Molecular Sequence Data , NF-kappa B/chemistry , Transcription Factor RelA
15.
Proc Natl Acad Sci U S A ; 101(25): 9327-32, 2004 Jun 22.
Article in English | MEDLINE | ID: mdl-15187233

ABSTRACT

Screening of expression libraries for bioactive clones that modulate the growth of mammalian cells has been limited largely to positive selections incapable of revealing growth suppressive or lethal genetic elements. We have developed a technique, selection-subtraction approach (SSA), that allows growth-modulating clones to be isolated based on alterations in their relative abundance in growing cell populations that have been transduced with an expression library. SSA utilizes tagged retroviral libraries in bacteriophage lambda vectors (retrophages). Nylon prints from retrophage libraries are used to determine the relative abundance of tags in library-transduced cells to identify biological activity of individual clones. Applications of SSA for gene discovery, target discovery, and generation of mutant proteins have been demonstrated, by using p53 and ataxia telangiectasia mutated (ATM) as models to isolate growth inhibitory proteins, peptides and antisense RNAs, and temperature-sensitive mutant proteins.


Subject(s)
Genetic Testing/methods , Polydeoxyribonucleotides/chemistry , Base Sequence , Databases, Nucleic Acid , Gene Library , Genetic Vectors , Humans , Male , Molecular Sequence Data , Mutagenesis, Site-Directed , Prostate , Proteins/chemistry , Proteins/genetics , RNA, Antisense/genetics , Restriction Mapping , Selection, Genetic
16.
Med Sci Monit ; 8(10): BR391-6, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12388911

ABSTRACT

BACKGROUND: Cellular receptors play a significant role in pathogenesis of viral infections. Previously, we demonstrated that TNFa receptor (TNFR1) rapidly disappeared from the cell surface upon poliovirus infection, whereas FAS was much more stable [1]. We suggested that the rate of decrease in receptor presentation on the surface of infected cells might reflect its turnover rate on uninfected cells. MATERIAL/METHODS: To test this hypothesis, we estimated by FACS analysis the turnover rates of receptors for TRAIL (TRAILR1 and TRAILR2), signal regulatory protein SIRPa, receptor for alpha/beta interferon (INFR1), and poliovirus receptor (CD155) on the surface of HeLa cells after the treatment with brefeldin A (to stop receptor replenishment through the Golgi-mediated trafficking) or poliovirus infection. RESULTS: A good correlation between turnover rates caused by the two interventions was observed, with the stability of receptor presentation changing in the following order: TRAILR1, TRAILR2, SIRPa (half-life on infected cells between 2-4 h) < INFR1 (4-6 h) < CD155 (>8 h, besides some early masking of the receptor by its binding of the virus). CONCLUSIONS: Our results suggest that disruption of the protein trafficking pathway during poliovirus infection leads to the diminished sensitivity of infected cells to pro-apoptotic factors, and thus represents one of the mechanisms by which virus modulates the host defense reactions.


Subject(s)
Membrane Proteins , Poliomyelitis/metabolism , Poliovirus/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Virus/metabolism , Animals , Apoptosis/physiology , Brefeldin A/metabolism , Cell Line , Cell Separation , Flow Cytometry , HeLa Cells , Humans , Protein Synthesis Inhibitors/metabolism , Protein Transport/physiology
17.
J Biol Chem ; 278(18): 15465-8, 2003 May 02.
Article in English | MEDLINE | ID: mdl-12637507

ABSTRACT

Pifithrin alpha (PFTalpha) is a chemical compound isolated for its ability to suppress p53-mediated transactivation. It can protect cells from p53-mediated apoptosis induced by various stimuli and reduce sensitivity of mice to gamma radiation. Identification of molecular targets of PFTalpha is likely to provide new insights into mechanisms of regulation of p53 pathway and is important for predicting potential risks associated with administration of PFTalpha-like p53 inhibitors in vivo. We found that PFTalpha, in addition to p53, can suppress heat shock and glucocorticoid receptor signaling but has no effect on nuclear factor-kappaB signaling. PFTalpha reduces activation of heat shock transcription factor (HSF1) and increases cell sensitivity to heat. Moreover, it reduces activation of glucocorticoid receptor and rescues mouse thymocytes in vitro and in vivo from apoptotic death after dexamethasone treatment. PFTalpha affected both signaling pathways in a p53-independent manner. These observations suggest that PFTalpha targets some unknown factor that is common for three major signal transduction pathways.


Subject(s)
Glucocorticoids/pharmacology , Heat-Shock Response/drug effects , Signal Transduction/drug effects , Thiazoles/pharmacology , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Benzothiazoles , DNA-Binding Proteins/biosynthesis , Heat Shock Transcription Factors , Mice , NF-kappa B/metabolism , Receptors, Glucocorticoid/physiology , Transcription Factors
18.
J Biol Chem ; 278(6): 3809-15, 2003 Feb 07.
Article in English | MEDLINE | ID: mdl-12446684

ABSTRACT

Genetic suppressor element (GSE) methodology was applied to identify new genes controlling cell response to tumor necrosis factor (TNF). A retroviral library of randomly fragmented normalized cDNA from mouse fibroblasts was screened for GSEs capable of protecting NIH3T3 cells from TNF-induced apoptosis. The most abundant among isolated GSEs represented a fragment of cDNA encoding the C-terminal cytoplasmic region of the immunoglobulin family inhibitory receptor, SHPS-1 (mouse homologue of human SIRPalpha). Ectopic expression of this fragment (both from human and mouse versions) increased the NF-kappaB-dependent transcription in three cell lines tested; this effect could be reduced by the expression of full-length SIRPalpha, suggesting that the isolated GSE acts through a dominant negative mechanism. GSE-mediated activation of NF-kappaB depended on the presence of serum, was abrogated by wortmannin, and was associated with phosphorylation of PKB/Akt, suggesting that Akt mediates it. These data indicate that SIRPalpha/SHPS-1 is involved in negative regulation of NF-kappaB signaling.


Subject(s)
Antigens, Differentiation , Apoptosis/physiology , Membrane Glycoproteins/physiology , NF-kappa B/metabolism , Neural Cell Adhesion Molecule L1/physiology , Receptors, Immunologic , Tumor Necrosis Factor-alpha/antagonists & inhibitors , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Cytoplasm/metabolism , DNA Primers , HeLa Cells , Humans , Membrane Glycoproteins/chemistry , Mice , Molecular Sequence Data , Neural Cell Adhesion Molecule L1/chemistry , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL