Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Virologie (Montrouge) ; 12(1): 39-52, 2008 Feb 01.
Article in French | MEDLINE | ID: mdl-36131433

ABSTRACT

Discovered in 1957 for their antiviral properties, interferons (IFNs) are a growing cytokine family with diverse biological activities including antitumor and immunoregulatory activities. IFN are classified in three types I, II and III. They bind to different specific cell receptors and induce via the Jak/Stat pathway the expression of more than 300 genes, the products of which are believed to mediate their biological effects. Several proteins have been implicated in resistance to viral infection in IFN-treated cells, i.e. the dsRNAdependent protein kinase PKR, the 2'5' oligoadenylate synthetase/RNaseL and Mx proteins. However, it was demonstrated that cells from triple knockout mice lacking PKR, RNase L and Mx are still sensitive to the IFN-induced antiviral state, indicating that other pathways exist. One of these pathways implicates promyelocytic leukemia (PML) protein. This article reviews the potential antiviral activities of the different IFN-induced mediators focusing onPMLpathway and how viruses from different families overcome this defence.

2.
Cell Mol Biol (Noisy-le-grand) ; 52(1): 9-15, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16914093

ABSTRACT

Interferons (IFNs) and arsenic trioxide (As2O3) are known inhibitors of cell proliferation and have been used in the treatment of certain forms of malignancy. IFNgamma treatment of cells leads to tyrosine phosphorylation of STAT1 followed by dimerization that accumulates in the nucleus. This is followed by DNA binding, activation of target gene transcription, dephosphorylation, and return to the cytoplasm. We have shown earlier that IFNgamma and As2O3 act synergistically in acute promyelocytic leukemia cells to upregulate IRF-1 expression and to induce apoptosis. Here, we show that in the human fibrosarcoma cell line 2fTGH, As2O3 prolongs IFNgamma-induced STAT1 phosphorylation resulting in persistent binding of STAT1 to GAS motif leading to an increase in IRF-1 expression which correlated with both higher anti-proliferative effect and increased apoptosis. These biological responses induced by IFNgamma alone or in combination with As2O3 were abolished when IRF-1 expression was down-regulated by RNA interference, thus demonstrating the key role of IRF-1.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Arsenic/pharmacology , Interferon Regulatory Factor-1/physiology , Interferon-gamma/pharmacology , Amino Acid Motifs , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , GPI-Linked Proteins , Gene Expression/drug effects , Growth Inhibitors/pharmacology , Humans , Interferon Regulatory Factor-1/metabolism , Membrane Proteins/metabolism , Models, Biological , Phosphorylation/drug effects , Protein Binding/drug effects , STAT1 Transcription Factor/metabolism , Tumor Cells, Cultured
3.
Oncogene ; 20(49): 7274-86, 2001 Oct 29.
Article in English | MEDLINE | ID: mdl-11704856

ABSTRACT

Interferons (IFNs) are a family of secreted proteins with antiviral, antiproliferative and immunomodulatory activities. The different biological actions of IFN are believed to be mediated by the products of specifically induced cellular genes in the target cells. The promyelocytic leukaemia (PML) protein localizes both in the nucleoplasm and in matrix-associated multi-protein complexes known as nuclear bodies (NBs). PML is essential for the proper formation and the integrity of the NBs. Modification of PML by the Small Ubiquitin MOdifier (SUMO) was shown to be required for its localization in NBs. The number and the intensity of PML NBs increase in response to interferon (IFN). Inactivation of the IFN-induced PML gene by its fusion to retinoic acid receptor alpha alters the normal localization of PML from the punctuate nuclear patterns of NBs to micro-dispersed tiny dots and results in uncontrolled growth in Acute Promyelocytic Leukaemia. The NBs-associated proteins, PML, Sp100, Sp140, Sp110, ISG20 and PA28 are induced by IFN suggesting that nuclear bodies could play a role in IFN response. Although the function of PML NBs is still unclear, some results indicate that they may represent preferential targets for viral infections and that PML could play a role in the mechanism of the antiviral action of IFNs. Viruses, which require the cellular machinery for their replication, have evolved different ways to counteract the action of IFN by inhibiting IFN signalling, by blocking the activities of specific antiviral mediators or by altering PML expression and/or localization on nuclear bodies.


Subject(s)
Cell Nucleus Structures/metabolism , DNA Virus Infections/metabolism , GTP-Binding Proteins , Interferons/metabolism , Neoplasm Proteins/metabolism , Organelles/metabolism , RNA Virus Infections/metabolism , Transcription Factors/metabolism , Animals , Cell Nucleus Structures/drug effects , Enzymes/metabolism , Gene Expression/drug effects , Humans , Interferons/pharmacology , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , Myxovirus Resistance Proteins , Nuclear Proteins/metabolism , Organelles/drug effects , Promyelocytic Leukemia Protein , Proteins/metabolism , Signal Transduction/drug effects , Tumor Suppressor Proteins
4.
Oncogene ; 18(4): 935-41, 1999 Jan 28.
Article in English | MEDLINE | ID: mdl-10023669

ABSTRACT

The PML protein is associated to nuclear bodies (NBs) whose functions are as yet unknown. PML and two other NBs-associated proteins, Sp100 And ISG20 are directly induced by interferons (IFN). PML and Sp100 proteins are covalently linked to SUMO-1, and ubiquitin-like peptide. PML NBs are disorganized in acute promyelocytic leukemia and during several DNA virus infections. In particular, the HSV-1 ICP0 protein is known to delocalize PML from NBs. Thus, NBs could play an important role in oncogenesis, IFN response and viral infections. Here, we show that HSV-1 induced PML protein degradation without altering its mRNA level. This degradation was time- and multiplicity of infection-dependent. Sp100 protein was also degraded, while another SUMO-1 conjugated protein, RanGAP1 and the IFN-induced protein kinase PKR were not. The proteasome inhibitor MG132 abrogated the HSV-1-induced PML and Sp100 degradation and partially restored their NB-localization. HSV-1 induced PML and Sp100 degradation constitutes a new example of viral inactivation of IFN target gene products.


Subject(s)
Antigens, Nuclear , Autoantigens/metabolism , GTPase-Activating Proteins , Herpes Simplex/metabolism , Herpesvirus 1, Human , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Autoantigens/chemistry , Carrier Proteins/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Humans , Interferons/pharmacology , Leupeptins/pharmacology , Molecular Weight , Neoplasm Proteins/chemistry , Nuclear Proteins/chemistry , Promyelocytic Leukemia Protein , Protein Kinases/metabolism , RNA, Messenger/metabolism , SUMO-1 Protein , Time Factors , Transcription Factors/chemistry , Tumor Cells, Cultured , Tumor Suppressor Proteins , Ubiquitins/metabolism
5.
Oncogene ; 15(19): 2349-59, 1997 Nov 06.
Article in English | MEDLINE | ID: mdl-9393879

ABSTRACT

Retinoic acid (RA) and interferons (IFNs) are negative regulators of cell proliferation. In vitro and in vivo, their combination leads to a more potent growth inhibition. However, the molecular mechanisms by which RA and IFNs potentiate each other are not fully understood. As some IFN-induced gene products regulate cell growth and/or antiviral activity, we analysed the effects of RA on their expressions. RA increases the level of 2'5'oligoadenylate synthetase, p68 kinase, the promyelocytic leukemia protein (PML) and Sp100 in both HL-60 and WISH cells. Moreover, RA and IFN act cooperatively to increase the expression of these proteins. RA also inhibits vesicular stomatitis virus replication and induces a higher antiviral state and growth inhibition when combined with IFN. RA stimulates the IFN regulatory factor 1 (IRF-1) gene expression directly through the GAS motif and causes the induction and secretion of IFNalpha. Additional mechanisms could be involved as RA increases the level of signal transducing activators of transcription (STAT) proteins, and enhances the IFN-induced STAT activation, suggesting that cooperative effects by RA and IFN are mediated through multiple pathways.


Subject(s)
Antigens, Nuclear , Interferon-alpha/pharmacology , Tretinoin/pharmacology , 2',5'-Oligoadenylate Synthetase/metabolism , Autoantigens/metabolism , Cell Division/drug effects , Cell Line , Drug Synergism , HL-60 Cells , HeLa Cells , Humans , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Signal Transduction , Vesicular stomatitis Indiana virus/drug effects , Virus Replication/drug effects
6.
Oncogene ; 18(27): 3944-53, 1999 Jul 08.
Article in English | MEDLINE | ID: mdl-10435617

ABSTRACT

In the t(15;17) acute promyelocytic leukaemia (APL), all trans-retinoic (RA) treatment induces maturation leading to clinically complete but not durable remission, as RA resistance develops in the treated patients as well as in vitro. RA and interferons (IFNs) are known inhibitors of proliferation in various cells including those from APL. In this report, we show that they can act cooperatively to inhibit growth and to induce differentiation of NB4 cells but not of two RA-resistant NB4 derived cell lines, NB4-R1 and NB4-R2. However, the resistant cell lines respond to IFN. In NB4 cells, RA increases the expression of Stat1, p48 and IRF-1, three transcription factors playing a central role in the IFN response and induces the synthesis and the secretion of IFN alpha. RA-induced IFN alpha seems to play a role in inhibition of NB4 cell growth but not in their differentiation. In the resistant cells, NB4-R1 and NB4-R2, both the induction of IFN and the increase of Statl and p48 expression by RA are completely blocked. In contrast, IRF-1 mRNA and protein expressions are induced in the three cell lines. This suggests that increase of IRF-1 expression is not sufficient for IFN induction. Our results identify some defects linked to RA-resistance in APL and support the hypothesis that RA-induced Stat1 expression and IFN secretion may be one of the mechanisms mediating growth inhibition by RA.


Subject(s)
DNA-Binding Proteins/biosynthesis , Interferon-alpha/biosynthesis , Leukemia, Promyelocytic, Acute/metabolism , Trans-Activators/biosynthesis , Transcription Factors/biosynthesis , Tretinoin/pharmacology , Cell Differentiation/drug effects , Cell Division/drug effects , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm , Drug Synergism , Growth Inhibitors/physiology , Humans , Interferon Regulatory Factor-1 , Interferon Regulatory Factor-3 , Interferon-Stimulated Gene Factor 3 , Interferon-Stimulated Gene Factor 3, gamma Subunit , Interferon-alpha/pharmacology , Interferon-alpha/physiology , Leukemia, Promyelocytic, Acute/pathology , Phosphoproteins/biosynthesis , Phosphoproteins/metabolism , Phosphorylation , STAT1 Transcription Factor , Signal Transduction/drug effects , Trans-Activators/metabolism , Transcription Factors/metabolism , Tumor Cells, Cultured
7.
Oncogene ; 19(44): 5041-53, 2000 Oct 19.
Article in English | MEDLINE | ID: mdl-11042692

ABSTRACT

Type I Interferon (IFN) and all-trans retinoic acid (RA) inhibit cell proliferation of squamous carcinoma cell lines (SCC). Examinations of growth-affected cell populations show that SCC lines ME-180 and SiHa treated with IFN-beta undergo a specific slower progression through the S phase that seems to trigger cellular death. In combination treatment RA potentiates IFN-beta effect in SCC ME-180 but not in SiHa cell line, partially resistant to RA antiproliferative action. RA added as single agent affects cell proliferation differently by inducing a slight G1 accumulation. The IFN-beta-induced S phase lengthening parallels the increased expression of PML, a nuclear phosphoprotein specifically up-regulated at transcriptional level by IFN, whose overexpression induces cell growth inhibition and tumor suppression. We report that PML up-regulation may account for the alteration of cell cycle progression induced by IFN-beta in SCC by infecting cells with PML-PINCO recombinant retrovirus carrying the PML-3 cDNA under the control of the 5' LTR. In fact PML overexpression reproduces the IFN-beta-induced S phase lengthening. These findings provide important insight into the mechanism of tumor suppressing function of PML and could allow PML to be included in the pathways responsible for IFN-induced cell growth suppression.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/pathology , Interferon Type I/pharmacology , Neoplasm Proteins/biosynthesis , Nuclear Proteins , S Phase/drug effects , Transcription Factors/biosynthesis , Uterine Cervical Neoplasms/pathology , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Cell Division/drug effects , DNA, Neoplasm/biosynthesis , Female , Gene Expression Regulation, Neoplastic/drug effects , Growth Inhibitors/administration & dosage , Growth Inhibitors/pharmacology , Humans , Interferon Type I/administration & dosage , Neoplasm Proteins/genetics , Promyelocytic Leukemia Protein , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Recombinant Proteins , Transcription Factors/genetics , Tretinoin/administration & dosage , Tretinoin/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Proteins , Up-Regulation/drug effects , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism
8.
Oncogene ; 10(7): 1315-24, 1995 Apr 06.
Article in English | MEDLINE | ID: mdl-7731682

ABSTRACT

Altered sub-nuclear localisation of the nuclear body-associated PML protein in acute promyelocytic leukaemia, has been proposed to contribute to leukaemogenesis. We have recently shown that PML is a primary target gene of interferons. Here, it is shown that PML has growth suppressive properties and displays an altered expression pattern during human oncogenesis. PML is widely expressed in cell-lines and is cell-cycle regulated. Overexpression of the protein induces a sharp reduction in growth rates in vitro and in vivo. In contrast with cell-lines, in normal tissues (including those that rapidly proliferate) only a few cells have detectable PML levels. However, these can be upregulated by soluble factors (e.g. IFN, estrogens). Human epithelial tumors show a gradual increase of PML levels as the lesion progresses from benign dysplasia to carcinoma. A similar induction is found in the surrounding stroma and vessels, which likely results from paracrine interactions. Strikingly, when malignant cells turn invasive, they loose PML expression, while expression is conserved in the stromal compartment. These observations point to the existence of a consistent deregulation in the expression of the PML growth-suppressor during human oncogenesis.


Subject(s)
Cell Cycle , Growth Inhibitors , Neoplasm Proteins , Neoplasms/metabolism , Nuclear Proteins , Transcription Factors/metabolism , Animals , Estrogens/physiology , Female , Humans , Immunohistochemistry , In Vitro Techniques , Mice , Mice, Nude , Neoplasm Transplantation , Promyelocytic Leukemia Protein , Skin/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured , Tumor Suppressor Proteins
9.
Oncogene ; 11(12): 2565-73, 1995 Dec 21.
Article in English | MEDLINE | ID: mdl-8545113

ABSTRACT

PML is a nuclear matrix protein with growth suppressing properties, whose expression is deregulated during oncogenesis. Moreover, in the t(15;17) translocation of acute promyelocytic leukaemia (APL), PML fusion to the retinoic acid receptor alpha (RAR alpha) is the likely molecular basis of leukaemogenesis. Here we show that interferons (IFNs) alpha, beta, and gamma upregulate PML mRNA expression. Analysis of 5' genomic sequences of the PML gene revealed an IFN-alpha/-beta stimulated response element (ISRE) and an IFN-gamma activation site (GAS) in the untranslated first exon. Binding of IFN signal transducers and activators of transcription (STATs) was demonstrated to be weak for the PML GAS, but strong for the PML ISRE which also seemed to contribute substantially to the IFN-gamma response. Thus, PML is a primary target gene of IFNs and would appear as a suitable candidate for mediating some of their antiproliferative effects. Abnormalities of PML structure, localisation or expression in human malignancy, constitute examples of how an IFN target gene may be altered in oncogenesis.


Subject(s)
Interferons/pharmacology , Neoplasm Proteins , Nuclear Proteins , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription, Genetic/drug effects , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Cricetinae , HeLa Cells , Humans , Molecular Sequence Data , Promyelocytic Leukemia Protein , Tumor Suppressor Proteins
10.
Leukemia ; 13(8): 1167-74, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10450744

ABSTRACT

Retinoic acid (RA) and interferon (IFN) potentiate each other to induce biological responses. Their combination has shown synergistic differentiating, antiproliferative and antiviral activities in various cell lines including those derived from the acute promyelocytic leukemia (APL). IFNs have demonstrated broad applications in cancer, as well as in virologic diseases. RA has variable effectiveness in therapy. Its real success is in APL where it provides the first example of a differentiation therapy. However, complete clinical remission with RA alone is always transient as RA resistance develops in the treated patients as well as in vitro. In various cell lines, including those derived from APL, RA induces directly the expression of two transcription factors, Stat1 and IRF-1 which play central roles in the IFN signal transduction. In addition, RA induces IFN-alpha synthesis and enhances the IFN-induced Stat activation. Here, we review the molecular mechanisms by which RA and IFNs can cooperate in inducing differentiation, inhibition of cell growth or viral replication focusing on recent results derived from normal and RA-resistant APL cells.


Subject(s)
Antineoplastic Agents/pharmacology , Interferons/pharmacology , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , Receptors, Interferon/metabolism , Receptors, Retinoic Acid/metabolism , Signal Transduction , Tretinoin/pharmacology , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Humans , Interferons/therapeutic use , Receptor Cross-Talk , Tretinoin/therapeutic use
11.
Leukemia ; 9(12): 2027-33, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8609713

ABSTRACT

PML has been identified through its fusion to the RAR alpha gene in acute promyelocytic leukemia (APL). The PML protein is specifically associated to nuclear bodies (NBs) whose alterations in APL were proposed to contribute to leukemogenesis. The role of this nuclear domain (which also harbors the Sp100 autoantigen and the NDP52 protein) is unknown. Here, we show that the PML protein, like Sp100 and NDP52, is induced by interferons (IFNs alpha, beta and gamma) in a large variety of human cells. Interestingly, the NBs that contain the three IFN-induced proteins appear to be associated to speckles labelled by the IFN-mediator Mx1. These observations link NBs to IFN response pathways, which may contribute to the elucidation of the biological role of these structures. In APL cells, IFNs induced both PML and PML/RAR alpha expression, resulting in an increased sequestration of PML and RXRs in the microspeckles induced by the fusion protein. As PML has growth suppressing properties, it may mediate some of the antiproliferative effects of IFN. In APL, inactivation of PML may result in disruption of growth control.


Subject(s)
Antineoplastic Agents/pharmacology , GTP-Binding Proteins , Interferons/pharmacology , Leukemia, Promyelocytic, Acute/metabolism , Neoplasm Proteins , Proteins/metabolism , Transcription Factors/biosynthesis , Blotting, Western , Humans , Microscopy, Confocal , Myxovirus Resistance Proteins , Nuclear Proteins/biosynthesis , Promyelocytic Leukemia Protein , Tumor Cells, Cultured , Tumor Suppressor Proteins
12.
Leukemia ; 14(4): 716-21, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10764160

ABSTRACT

The combination of the anti-viral agents, zidovudine (AZT) and interferon-alpha (IFN), is a potent treatment of HTLV-I-associated adult T cell leukemia/lymphoma (ATL). In this study we investigate the possible mechanism of action of this combination by examining several cellular parameters including cell proliferation, cell cycle distribution and apoptosis. The ATL-derived T cell lines HuT-102 and MT-2 served as models. HTLV-I negative T cell lines (CEM and Jurkat) were used as controls. No significant modification of cell growth was observed except at suprapharmacological doses of AZT and IFN. Moreover, these effects were less pronounced in HTLV-I-infected cell lines compared to control cell lines. AZT and IFN treatment did not induce any significant modification of the expression of bcl-2 and p53. Interestingly no in vitro cytotoxic effect of AZT/IFN combination was observed on fresh leukemic cells derived from an acute ATL patient at diagnosis despite achievement of in vivo complete remission using the same therapy. These results suggest that the therapeutic effect of AZT and IFN is not through a direct cytotoxic effect of these drugs on the leukemic cells.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Immunologic Factors/pharmacology , Interferon-alpha/pharmacology , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Neoplastic Stem Cells/drug effects , T-Lymphocytes/drug effects , Zidovudine/pharmacology , Aged , Antimetabolites, Antineoplastic/therapeutic use , Apoptosis/drug effects , Biomarkers, Tumor/analysis , Cell Survival/drug effects , Combined Modality Therapy , DNA, Neoplasm/analysis , Drug Synergism , Female , Human T-lymphotropic virus 1/isolation & purification , Humans , Immunologic Factors/therapeutic use , Interferon-alpha/therapeutic use , Jurkat Cells/drug effects , Leukemia-Lymphoma, Adult T-Cell/pathology , Remission Induction , Tumor Cells, Cultured/drug effects , Zidovudine/therapeutic use
13.
Peptides ; 12(5): 1085-93, 1991.
Article in English | MEDLINE | ID: mdl-1724797

ABSTRACT

Vasoactive intestinal peptide (VIP), composed of 28 amino acids, is a multifunctional neurotransmitter. We have demonstrated here that its action on human transformed colonic epithelial (HT-29) cells is mediated through the induction of interferon (IFN) synthesis. We have found that these cells have a functional receptor for IFN alpha 2; binding was specific to either IFN alpha 2 or IFN beta but not to IFN gamma. VIP induced the 2'5'oligoadenylate synthetase (2'5'A synthetase) and the antiviral state with the same efficiency as poly (I).poly (C). The induction of 2'5'A synthetase activity required cellular RNA and protein synthesis, and the maximum induction occurred with 10(-7) M VIP at 24 h. VIP, like some IFN inducers, induced the synthesis of the 70 hsp which, however, preceded the expression of 2'5'A synthetase. VIP treatment caused the induction and secretion of IFN, having a titer value of 32 international units/ml. This IFN has been identified as type beta/alpha, because both 2'5'A synthetase and the antiviral activities were abolished by anti-human IFN beta/alpha antibodies, but not by anti-IFN gamma antibodies. Thus the pathway of VIP action on HT-29 cells may be outlined as 1) binding of VIP, 2) synthesis of 70 hsp, 3) induction of IFN synthesis and its secretion, 4) binding of the secreted IFN to cell surface receptors and 5) turning on the induction of 2'5'A synthetase and antiviral activities.


Subject(s)
2',5'-Oligoadenylate Synthetase/biosynthesis , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Poly I-C/pharmacology , Receptors, Immunologic/metabolism , Vasoactive Intestinal Peptide/pharmacology , Vesicular stomatitis Indiana virus/physiology , 2-Aminopurine/pharmacology , Binding, Competitive , Cell Line, Transformed , Colonic Neoplasms , Cyclic AMP/metabolism , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Enzyme Induction , Humans , Interferons/metabolism , Interferons/pharmacology , Kinetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/isolation & purification , Polyribonucleotides/pharmacology , Receptors, Interferon , Time Factors , Virus Replication
14.
Bull Cancer ; 85(4): 313-8, 1998 Apr.
Article in French | MEDLINE | ID: mdl-9752294

ABSTRACT

Retinoic acid (RA) and interferons (IFN) are negative regulators of cell proliferation. A number of clinical trials were thus carried out in cancer therapy with RA and/or IFN. In vitro and in vivo, their combination leads to a more potent cell growth inhibition. Moreover, RA and IFN act cooperatively to increase the expression of many IFN-stimulated genes, leading also to a higher cell differentiation and inhibition of viral replication. However, the molecular mechanisms by which RA and IFN potentiate each other are not fully understood. The cooperative effects by RA and IFN are mediated through multiple pathways. RA causes the induction and secretion of IFN alpha. RA also stimulates the IFN regulatory factor gene expression (IRF1 and p48). Additional mechanisms could be involved as RA increases the level of signal transducing activators of transcription (Stat) proteins, and thus enhances the IFN-induced Stat activation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Interferons/therapeutic use , Tretinoin/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/metabolism , Drug Interactions , Humans , Interferon-alpha/metabolism , Interferon-beta/metabolism , Interferons/administration & dosage , Interferons/classification , Interferons/metabolism , Phosphorylation , Signal Transduction , Trans-Activators/metabolism , Tretinoin/administration & dosage , Tretinoin/metabolism
17.
J Biol Chem ; 267(3): 1741-5, 1992 Jan 25.
Article in English | MEDLINE | ID: mdl-1309794

ABSTRACT

We demonstrate here that ethanol, in contrast to heat shock (Chousterman, S., Chelbi-Alix, M.K., and Thang, M.N. (1987) J. Biol. Chem. 262, 4806-4811), induces interferon (IFN) synthesis and its related activities in Madin-Darby bovine kidney (MDBK) cells. The induced IFN is secreted maximally at 6 h, whereas the induction of 2',5'-oligoadenylate synthetase mRNA peaks between 9 and 12 h and its activity at 15 h. The appearance of both 2',5'-oligoadenylate synthetase activity and the antiviral state upon ethanol treatment is prevented by anti-bovine recombinant IFN-beta antibodies. Bovine diarrhea virus infection-free MDBK cells cultured in medium supplemented with serum substitute also gave similar results, thus indicating that IFN synthesis induced by ethanol is not mediated by the activation of bovine diarrhea virus. Together, these results show that: 1) ethanol induces the 2',5'-oligoadenylate synthetase and antiviral activities through IFN-beta production; and 2) the IFN produced does not act directly from inside the cells, but has to be first secreted to bind to its receptor. In MDBK cells, ethanol induces the synthesis of the 70-kDa protein, which precedes the expression of 2',5'-oligoadenylate synthetase; moreover, the transient nature of the synthesis of the hsp 70 in these cells is similar after both heat shock and ethanol treatment.


Subject(s)
2',5'-Oligoadenylate Synthetase/biosynthesis , Diarrhea Viruses, Bovine Viral/physiology , Ethanol/pharmacology , Interferon-beta/biosynthesis , Animals , Antiviral Agents , Cattle , Cell Division/drug effects , Cell Line , Enzyme Induction , Heat-Shock Proteins/biosynthesis , Kinetics , Molecular Weight
18.
J Biol Chem ; 262(10): 4806-11, 1987 Apr 05.
Article in English | MEDLINE | ID: mdl-2435717

ABSTRACT

Hyperthermia (45 degrees C) induced the synthesis of a characteristic heat-shock protein of 70,000 daltons (70 hsp) in Madin-Darby bovine kidney (MDBK) cells. In addition, subsequent to heat shock, there was a substantial increase in the 2',5'-oligoadenylate synthetase (2',5'-A synthetase) activity in both MDBK and human WISH cells. However, in contrast to 70 hsp synthesis, which reached its maximum 3 h after cell transfer from 45 to 37 degrees C, increase in 2',5'-A synthetase expression, conspicuous after 6 h, attained its maximum only 18 h after transfer. Another interesting observation is that, during recovery at 37 degrees C, the cells released into the medium heat-shock-induced factor(s) (HSIF) capable of inducing an increase in 2',5'-A synthetase activity in fresh MDBK cells. HSIF behaves as a polypeptide with a molecular weight of more than 5,000; it is relatively heat stable and sensitive to acidic treatment. HISF seems different from interferon (IFN) since: 1) no detectable antiviral state developed after infection in cells treated with HSIF; 2) antibovine IFN antibodies did not abolish the inducing capacity of HSIF; 3) IFN had an additive effect on the inducing capacity of HSIF, and 4) HSIF released from bovine cells induced a net enhancement of 2',5'-A synthetase activity in human WISH cells. The first three of these observations applied also to heat-shocked MDBK cells.


Subject(s)
2',5'-Oligoadenylate Synthetase/biosynthesis , Hot Temperature , Animals , Cattle , Cell Line , Enzyme Induction , Heat-Shock Proteins/biosynthesis , Interferons/biosynthesis , Kinetics
19.
Biochem Biophys Res Commun ; 145(1): 426-35, 1987 May 29.
Article in English | MEDLINE | ID: mdl-3036128

ABSTRACT

The HuIFN-alpha A and HuIFN-alpha D interferons, produced by two independent recombinant bacterial clones, have different affinities for polyinosinic acid (poly I). The monomeric form HuIFN-alpha A (FMM), but not the HuIFN-alpha D, binds to poly (I)-agarose and is protected by poly (I) from thermal inactivation. Other subtypes of HuIFN-alpha A including the monomer SMM and oligomers have no affinity for this polynucleotide. In addition, these interferons show different target cell preferences in agreement with our previous suggestion (23) that the polynucleotide binding domain may be responsible for species specificity. Two significant observations are 1) the fractions of HuIFN-alpha D and HuIFN-alpha A unbound on poly (I)-agarose show higher antiviral inducing activity on heterologous (MDBK) than on homologous (WISH) cells, whereas they induce about the same activity of 2'5' oligoadenylate synthetase in these two cell lines. These fractions are also active on L929 cells. 2) The bound fraction of HuIFN-alpha A induces almost the same antiviral and 2'5' oligoadenylate synthetase activities in MDBK and in WISH cells but neither activity in L929 cells.


Subject(s)
Interferon Type I/metabolism , Poly I/metabolism , Polyribonucleotides/metabolism , Recombinant Proteins/metabolism , 2',5'-Oligoadenylate Synthetase/biosynthesis , Animals , Cell Line , Drug Stability , Enzyme Induction , Humans , Interferon Type I/pharmacology , Poly I/pharmacology , Protein Binding , Recombinant Proteins/pharmacology , Species Specificity , Structure-Activity Relationship , Vesicular stomatitis Indiana virus/drug effects
20.
Exp Cell Res ; 213(2): 383-90, 1994 Aug.
Article in English | MEDLINE | ID: mdl-7519564

ABSTRACT

We have examined the ability of insulin and dsRNA, a well-known interferon inducer, in relation to their effects on cell growth, to induce the expression of hsp 70 and the synthesis of interferon in epithelial HT-29 and fibroblast Madin-Darby bovine kidney (MDBK) cells. Insulin was mitogenic in both MDBK and HT-29 cells; MDBK cells nevertheless required much higher concentrations. DsRNA stimulated the growth of MDBK but inhibited that of HT-29 cells. Both substances induced a transient synthesis of hsp 70 in HT-29 and MDBK cells with similar kinetics. However, whereas both insulin and dsRNA efficiently induced 2'5' oligoadenylate synthetase and an antiviral state through interferon synthesis in HT-29 cells, only dsRNA caused these effects in MDBK cells. Thus, insulin cannot, unlike dsRNA, elicit an antiviral state in all cell systems, although, like dsRNA, it can induce hsp 70, thereby suggesting the cell specificity of insulin action. These results reveal that the mitogenic and IFN-inducing effects of insulin and dsRNA are dependent on the cell type and unrelated to hsp 70 expression.


Subject(s)
Heat-Shock Proteins/metabolism , Insulin/physiology , Interferons/metabolism , RNA, Double-Stranded/physiology , 2',5'-Oligoadenylate Synthetase/metabolism , Animals , Cell Division , Cell Line , Dogs , Epithelial Cells , Epithelium/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Poly I-C/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL