Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 21(6): 1030-1043, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35313341

ABSTRACT

This article investigates mechanisms of resistance to the VEGF receptor inhibitor cediranib in high-grade serous ovarian cancer (HGSOC), and defines rational combination therapies. We used three different syngeneic orthotopic mouse HGSOC models that replicated the human tumor microenvironment (TME). After 4 to 5 weeks treatment of established tumors, cediranib had antitumor activity with increased tumor T-cell infiltrates and alterations in myeloid cells. However, continued cediranib treatment did not change overall survival or the immune microenvironment in two of the three models. Moreover, treated mice developed additional peritoneal metastases not seen in controls. Cediranib-resistant tumors had intrinsically high levels of IL6 and JAK/STAT signaling and treatment increased endothelial STAT3 activation. Combination of cediranib with a murine anti-IL6 antibody was superior to monotherapy, increasing mouse survival, reducing blood vessel density, and pSTAT3, with increased T-cell infiltrates in both models. In a third HGSOC model, that had lower inherent IL6 JAK/STAT3 signaling in the TME but high programmed cell death protein 1 (PD-1) signaling, long-term cediranib treatment significantly increased overall survival. When the mice eventually relapsed, pSTAT3 was still reduced in the tumors but there were high levels of immune cell PD-1 and Programmed death-ligand 1. Combining cediranib with an anti-PD-1 antibody was superior to monotherapy in this model, increasing T cells and decreasing blood vessel densities. Bioinformatics analysis of two human HGSOC transcriptional datasets revealed distinct clusters of tumors with IL6 and PD-1 pathway expression patterns that replicated the mouse tumors. Combination of anti-IL6 or anti-PD-1 in these patients may increase activity of VEGFR inhibitors and prolong disease-free survival.


Subject(s)
Ovarian Neoplasms , Programmed Cell Death 1 Receptor , Angiogenesis Inhibitors/pharmacology , Animals , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Female , Humans , Indoles , Interleukin-6 , Mice , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Quinazolines , Tumor Microenvironment
2.
Cancer Res ; 81(22): 5706-5719, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34561272

ABSTRACT

The tumor microenvironment evolves during malignant progression, with major changes in nonmalignant cells, cytokine networks, and the extracellular matrix (ECM). In this study, we aimed to understand how the ECM changes during neoplastic transformation of serous tubal intraepithelial carcinoma lesions (STIC) into high-grade serous ovarian cancers (HGSOC). Analysis of the mechanical properties of human fallopian tubes (FT) and ovaries revealed that normal FT and fimbria had a lower tissue modulus, a measure of stiffness, than normal or diseased ovaries. Proteomic analysis of the matrisome fraction between FT, fimbria, and ovaries showed significant differences in the ECM protein TGF beta induced (TGFBI, also known as ßig-h3). STIC lesions in the fimbria expressed high levels of TGFBI, which was predominantly produced by CD163-positive macrophages proximal to STIC epithelial cells. In vitro stimulation of macrophages with TGFß and IL4 induced secretion of TGFBI, whereas IFNγ/LPS downregulated macrophage TGFBI expression. Immortalized FT secretory epithelial cells carrying clinically relevant TP53 mutations stimulated macrophages to secrete TGFBI and upregulated integrin αvß3, a putative TGFBI receptor. Transcriptomic HGSOC datasets showed a significant correlation between TGFBI expression and alternatively activated macrophage signatures. Fibroblasts in HGSOC metastases expressed TGFBI and stimulated macrophage TGFBI production in vitro. Treatment of orthotopic mouse HGSOC tumors with an anti-TGFBI antibody reduced peritoneal tumor size, increased tumor monocytes, and activated ß3-expressing unconventional T cells. In conclusion, TGFBI may favor an immunosuppressive microenvironment in STICs that persists in advanced HGSOC. Furthermore, TGFBI may be an effector of the tumor-promoting actions of TGFß and a potential therapeutic target. SIGNIFICANCE: Analysis of ECM changes during neoplastic transformation reveals a role for TGFBI secreted by macrophages in immunosuppression in early ovarian cancer.


Subject(s)
Cystadenocarcinoma, Serous/pathology , Extracellular Matrix/pathology , Macrophages/immunology , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/pathology , Transforming Growth Factor beta1/metabolism , Tumor Microenvironment , Animals , Apoptosis , Cell Proliferation , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/metabolism , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Female , Humans , Immunosuppressive Agents , Mice , Mice, Inbred C57BL , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/metabolism , Prognosis , Transforming Growth Factor beta1/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Cancer Immunol Res ; 9(6): 665-681, 2021 06.
Article in English | MEDLINE | ID: mdl-33839687

ABSTRACT

Neoadjuvant chemotherapy (NACT) may stimulate anticancer adaptive immune responses in high-grade serous ovarian cancer (HGSOC), but little is known about effects on innate immunity. Using omental biopsies from HGSOC, and omental tumors from orthotopic mouse HGSOC models that replicate the human tumor microenvironment, we studied the impact of platinum-based NACT on tumor-associated macrophages (TAM). We found that chemotherapy reduces markers associated with alternative macrophage activation while increasing expression of proinflammatory pathways, with evidence of inflammasome activation. Further evidence of a shift in TAM functions came from macrophage depletion via CSF1R inhibitors (CSF1Ri) in the mouse models. Although macrophage depletion in established disease had no impact on tumor weight or survival, CSF1Ri treatment after chemotherapy significantly decreased disease-free and overall survival. This decrease in survival was accompanied by significant inhibition of adaptive immune response pathways in the tumors. We conclude that chemotherapy skews the TAM population in HSGOC toward an antitumor phenotype that may aid adaptive immune responses, and therapies that enhance or sustain this during remission may delay relapse.


Subject(s)
Cystadenocarcinoma, Serous/immunology , Ovarian Neoplasms/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Tumor-Associated Macrophages/immunology , Adaptive Immunity , Animals , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/mortality , Cystadenocarcinoma, Serous/pathology , Disease Models, Animal , Disease-Free Survival , Female , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Neoadjuvant Therapy/methods , Neoplasm Grading , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Tumor Microenvironment/immunology
4.
Cell Rep ; 30(2): 525-540.e7, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31940494

ABSTRACT

Although there are many prospective targets in the tumor microenvironment (TME) of high-grade serous ovarian cancer (HGSOC), pre-clinical testing is challenging, especially as there is limited information on the murine TME. Here, we characterize the TME of six orthotopic, transplantable syngeneic murine HGSOC lines established from genetic models and compare these to patient biopsies. We identify significant correlations between the transcriptome, host cell infiltrates, matrisome, vasculature, and tissue modulus of mouse and human TMEs, with several stromal and malignant targets in common. However, each model shows distinct differences and potential vulnerabilities that enabled us to test predictions about response to chemotherapy and an anti-IL-6 antibody. Using machine learning, the transcriptional profiles of the mouse tumors that differed in chemotherapy response are able to classify chemotherapy-sensitive and -refractory patient tumors. These models provide useful pre-clinical tools and may help identify subgroups of HGSOC patients who are most likely to respond to specific therapies.


Subject(s)
Ovarian Neoplasms/genetics , Tumor Microenvironment/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Mice , Ovarian Neoplasms/pathology
5.
J Clin Invest ; 127(3): 801-813, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28134623

ABSTRACT

Elevated expression of the chemokine receptor CCR4 in tumors is associated with poor prognosis in several cancers. Here, we have determined that CCR4 was highly expressed in human renal cell carcinoma (RCC) biopsies and observed abnormal levels of CCR4 ligands in RCC patient plasma. An antagonistic anti-CCR4 antibody had antitumor activity in the RENCA mouse model of RCC. CCR4 inhibition did not reduce the proportion of infiltrating leukocytes in the tumor microenvironment but altered the phenotype of myeloid cells, increased NK cell and Th1 cytokine levels, and reduced immature myeloid cell infiltrate and blood chemokine levels. In spite of prominent changes in the myeloid compartment, the anti-CCR4 antibody did not affect RENCA tumors in T cell-deficient mice, and treatment with an anti-class II MHC antibody abrogated its antitumor activity. We concluded that the effects of the anti-CCR4 antibody required the adaptive immune system and CD4+ T cells. Moreover, CCL17-induced IFN-γ production was reduced when Th1-polarized normal CD4+ T cells were exposed to the CCR4 ligand, evidencing the involvement of CCR4 in Th1/Th2 regulation. The anti-CCR4 antibody, alone or in combination with other immune modulators, is a potential treatment approach to human solid cancers with high levels of CCR4-expressing tumor-infiltrating leukocytes and abnormal plasma CCR4 ligand levels.


Subject(s)
Antibodies, Neoplasm/pharmacology , Carcinoma, Renal Cell/immunology , Killer Cells, Natural/immunology , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Experimental/immunology , Receptors, CCR4/antagonists & inhibitors , Th1 Cells/immunology , Tumor Microenvironment/immunology , Animals , Antibodies, Neoplasm/immunology , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Chemokine CCL17/genetics , Chemokine CCL17/immunology , Chemokine CCL17/pharmacology , Female , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Kidney Neoplasms , Male , Mice , Mice, Inbred BALB C , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Receptors, CCR4/genetics , Receptors, CCR4/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
6.
Mol Cell Biol ; 32(9): 1633-44, 2012 May.
Article in English | MEDLINE | ID: mdl-22371483

ABSTRACT

The TFAP2C transcription factor has been shown to downregulate transcription of the universal cell cycle inhibitor p21(cip) (CDKN1A). In examining the mechanism of TFAP2C-mediated repression, we have identified a ternary complex at the proximal promoter containing TFAP2C, the oncoprotein Myc, and the trimethylated lysine 4 of histone H3 (H3K4me3) demethylase, KDM5B. We demonstrated that while TFAP2C and Myc can downregulate the CDKN1A promoter independently, KDM5B acts as a corepressor dependent on the other two proteins. All three factors collaborate for optimal CDKN1A repression, which requires the AP-2 binding site at -111/-103 and KDM5B demethylase activity. Silencing of TFAP2C-KDM5B-Myc led to increased H3K4me3 at the endogenous promoter and full induction of CDKN1A expression. Coimmunoprecipitation assays showed that TFAP2C and Myc associate with distinct domains of KDM5B and the TFAP2C C-terminal 270 amino acids (aa) are required for Myc and KDM5B interaction. Overexpression of all three proteins resulted in forced S-phase entry and attenuation of checkpoint activation, even in the presence of chemotherapy drugs. Since each protein has been linked to poor prognosis in breast cancer, our findings suggest that the TFAP2C-Myc-KDM5B complex promotes cell cycle progression via direct CDKN1A repression, thereby contributing to tumorigenesis and therapy failure.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Genes, myc , Jumonji Domain-Containing Histone Demethylases/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factor AP-2/metabolism , Adaptor Protein Complex 2/genetics , Adaptor Protein Complex 2/metabolism , Binding Sites , Cell Cycle , Cell Line, Tumor , Down-Regulation , Genetic Loci , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Binding , Repressor Proteins/genetics , Transcription Factor AP-2/genetics
7.
Breast Cancer Res ; 13(2): R23, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21375726

ABSTRACT

INTRODUCTION: AP-2α is a transcription factor implicated in the regulation of differentiation and proliferation in certain tissues, including the mammary gland. In breast tumours, continued expression of AP-2α has been correlated with a better prognosis, but this is hard to reconcile with a reported role in the upregulation of the ERBB2 oncogene. The existence of TFAP2A isoforms, deriving from alternative first exons and differing in their N-terminal sequence, has been described in some mammals, but their relative abundance and activity has not been investigated in the human breast. METHODS: Expression levels of four TFAP2A isoforms were assayed at the level of RNA and protein (via the generation of isoform-specific antibodies) in a panel of breast tumour cell lines and in tissue from normal breast and primary tumour samples. Expression constructs for each isoform were used in reporter assays with synthetic and natural promoters (cyclin D3 and ERBB2) to compare the activation and repression activity of the isoforms. RESULTS: We demonstrate that the two isoforms AP-2α 1b and AP-2α 1c, in addition to the originally cloned, AP-2α 1a, are conserved throughout evolution in vertebrates. Moreover, we show that isoform 1c in particular is expressed at levels at least on a par with the 1a isoform in breast epithelial lines and tissues and may be more highly expressed in tamoxifen resistant tumours. The isoforms share a similar transactivation mechanism involving the recruitment of the adaptors CITED2 or 4 and the transactivators p300 or CBP. However, isoform 1b and 1c are stronger transactivators of the ERBB2 promoter than isoform 1a. In contrast, AP-2α 1a is the only isoform able to act as a repressor, an activity that requires an intact sumoylation motif present within the N-terminus of the protein, and which the other two isoforms lack. CONCLUSIONS: Our findings suggest that TFAP2A isoforms may be differentially regulated during breast tumourigenesis and this, coupled with differences in their transcriptional activity, may impact on tumour responses to tamoxifen therapy. These data also have implications for the interpretation of tumour studies that seek to correlate outcomes with TFAP2A expression level.


Subject(s)
Breast Neoplasms/genetics , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism , Animals , Antibodies, Monoclonal/immunology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cyclin D3/genetics , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Humans , Promoter Regions, Genetic , Protein Isoforms/immunology , Protein Isoforms/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, ErbB-2/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Tamoxifen/therapeutic use , Transcription Factor AP-2/immunology , Transcription, Genetic , Transcriptional Activation , Xenopus , Xenopus Proteins/genetics , Zebrafish , Zebrafish Proteins/genetics
8.
EMBO J ; 28(22): 3591-601, 2009 Nov 18.
Article in English | MEDLINE | ID: mdl-19798054

ABSTRACT

Overexpression of the activator protein (AP)-2gamma transcription factor in breast tumours has been identified as an independent predictor of poor outcome and failure of hormone therapy. To understand further the function of AP-2gamma in breast carcinoma, we have used an RNA interference and gene expression profiling strategy with the MCF-7 cell line as a model. Gene expression changes between control and silenced cells implicate AP-2gamma in the control of cell cycle progression and developmental signalling. A function for AP-2gamma in cell cycle control was verified using flow cytometry: AP-2gamma silencing led to a partial G1/S arrest and induction of the cyclin-dependent kinase inhibitor, p21cip/CDKN1A. Reporter and chromatin immunoprecipitation assays demonstrated a direct, functional interaction by AP-2gamma at the CDKN1A proximal promoter. AP-2gamma silencing coincided with acquisition of an active chromatin conformation at the CDKN1A locus and increased gene expression. These data provide a mechanism whereby AP-2gamma overexpression can promote breast epithelial proliferation and, coupled with previously published data, suggest how loss of oestrogen regulation of AP-2gamma may contribute to the failure of hormone therapy in patients.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Transcription Factor AP-2/physiology , Cell Cycle/genetics , Down-Regulation , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Oligonucleotide Array Sequence Analysis , Tumor Cells, Cultured
9.
Endocrinology ; 150(6): 2924-33, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19246539

ABSTRACT

The terminal differentiation of the mouse mammary gland epithelium during lactation has been shown to require IGFs and/or superphysiological levels of insulin. It has been suggested that IGF receptor I (IGF-IR), in addition to its well-established role in the mammary gland during puberty and pregnancy, serves as the principal mediator of IGFs at this stage of development. However, our analysis of the expression levels of IGF-IR and the two insulin receptor (IR) splice variants, IR-A and IR-B, has revealed a 3- to 4-fold up-regulation of IR-B transcripts and a 6-fold down-regulation of IGF-IR transcripts and protein during terminal differentiation in the developing mammary gland. IR-B expression was also more than 10-fold up-regulated in murine mammary epithelial cell line HC11 during differentiation in vitro. As already described for the human form, murine IR-B cloned from HC11 exhibited selectivity for insulin as compared with IGFs. When differentiated HC11 cells were stimulated by 10 nm insulin, a concentration that is unable to activate IGF-IR, induction of milk protein and lipid synthetic enzyme gene expression, lactate production, and phosphorylation of Akt were observed. In contrast, on differentiated HC11 cells 10 nm IGF-I or 10 nm IGF-II were able to exert growth-promoting effects only. The lack of response of differentiated cells to low levels of IGFs could not be explained by inactivation of IGFs by IGF binding proteins. Our results suggest a previously unrecognized predominant role for IR-B in the differentiated mammary epithelium.


Subject(s)
Insulin-Like Growth Factor II/pharmacology , Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Mammary Glands, Animal/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Up-Regulation/drug effects , Animals , Base Sequence , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Epithelium/drug effects , Epithelium/metabolism , Female , Lactation/physiology , Mammary Glands, Animal/cytology , Mammary Glands, Animal/drug effects , Mice , Models, Animal , Molecular Sequence Data , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism
10.
J Biol Chem ; 283(32): 22089-96, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18524780

ABSTRACT

Cytokine and glucocorticoid (GC) hormone signaling act in an integrated fashion to control inflammation and immune response. Here we establish a new mode of interaction of these two pathways and propose Suppressor of Cytokine Signaling (SOCS)-1 as an essential player in mediating cross-talk. We observed that glucocorticoid receptor (GR) and SOCS1 form an intracellular complex through an interaction, which required the SH2 domain of SOCS1 and the ligand binding domain of GR. Furthermore, GC stimulation was found to increase the nuclear level of SOCS1. SOCS1 binding to the GR did not require ligand binding of the receptor; however, it was abolished after long term GC stimulation, suggesting a functional role of the interaction for the early phase of GC action. The interaction between GR and SOCS1 appeared to negatively influence the transcription of the two GR-regulated genes, FKBP5 and MKP1, because the GC-dependent expression of these genes was inhibited by the SOCS1 inducer IFNgamma and enhanced in SOCS1-deficient murine embryonic fibroblasts as compared with IFNgamma treated wild-type cells. Our results suggest a prominent role of SOCS1 in the early phase of cross-talk between GR and cytokine signaling.


Subject(s)
Receptors, Glucocorticoid/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , COS Cells , Cell Line, Tumor , Cell Nucleus/metabolism , Chlorocebus aethiops , Glucocorticoids/metabolism , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Protein Binding , Rats , Suppressor of Cytokine Signaling 1 Protein , src Homology Domains
11.
Traffic ; 8(12): 1815-1828, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17892529

ABSTRACT

In addition to its extracellular function as a secreted protein, IGF-binding protein (IGFBP)-5 has been postulated to act as a signaling molecule in the nucleus. This study aims to assess the significance of this postulated nuclear localization. By confocal immunofluorescence microscopy, we detected IGFBP-5 in the vesicular compartment of mammary epithelial cells in culture, while no nuclear staining was observed. Immunohistochemistry performed on paraffin sections of the involuting mammary gland revealed IGFBP-5 positive staining of epithelial cells only outside the nucleus. To evaluate the contribution of reuptake of extracellular IGFBP-5, T47D cells were incubated with Alexa Fluor 647-labeled IGFBP-5. The protein was taken up into intracellular vesicles and again was neither detectable in the cytoplasm outside of vesicular structures nor in the nucleus. Quantification of the time and concentration dependence of uptake by immunoblotting revealed that the process was saturable at IGFBP-5 concentrations between 1 and 2 mum and partially reversible with 30% remaining in the cell after a 1-h chase. The observation of nuclear uptake of IGFBP-5 was restricted to artificial conditions such as expression of non-secreted forms of IGFBP-5 or selective permeabilization of the plasma membrane by digitonin.


Subject(s)
Cell Membrane/metabolism , Cell Nucleus/metabolism , Digitonin/metabolism , Insulin-Like Growth Factor Binding Protein 5/metabolism , Active Transport, Cell Nucleus , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Cytoplasm/metabolism , Epithelial Cells/metabolism , Humans , Mammary Glands, Animal/metabolism , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Models, Biological , Time Factors
12.
J Mammary Gland Biol Neoplasia ; 11(1): 63-73, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16900390

ABSTRACT

Nuclear Factor-kappaB (NF-kappaB) has been implicated in the lobuloalveolar development of the mammary gland. In breast cancer its activation has been linked to tumor progression via stimulation of cell proliferation, pro-survival, and angiogenesis pathways and metastasis. Whether NF-kappaB activation in the immune system influences mammary cancer remains unclear. In addition to the constitutive activation frequently found in mammary carcinoma tissue, radio- and chemotherapeutic agents used in the treatment of mammary cancer can lead to activation of NF-kappaB. This effect has been postulated to contribute to the development of resistance to these agents and suggests the use of NF-kappaB inhibitors as sensitizers for therapy. The review describes principle targets and drugs used to inhibit NF-kappaB function and discusses future perspectives in the use of these inhibitors for the treatment of mammary cancer.


Subject(s)
Antineoplastic Agents , Breast Neoplasms/drug therapy , Breast Neoplasms/physiopathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Animals , Apoptosis , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis/physiopathology , Rodentia
13.
J Biol Chem ; 279(14): 13746-54, 2004 Apr 02.
Article in English | MEDLINE | ID: mdl-14742442

ABSTRACT

SOCS-3 (suppressor of cytokine signaling 3) is an intracellular protein that is selectively and rapidly induced by appropriate agonists and that modulates responses of immune cells to cytokines by interfering with the Janus kinase/signal transducer and activator of transcription (Jak/STAT) pathway. On the basis of the observations that interferon gamma (IFNgamma) up-regulates SOCS-3 gene and protein expression in primary mouse macrophages, J774 macrophage cell line and embryonal fibroblasts, we investigated which sequences of the 5' SOCS-3 gene are responsive to IFNgamma. By promoter deletion analysis we identified a functional IFNgamma-responsive element, located at nucleotides -72/-64 upstream from the transcription initiation, whose presence and integrity is necessary to ensure responsiveness to IFNgamma. This element contains a STAT consensus binding sequence (SOCS-3/STAT-binding element (SBE)) whose specific mutation totally abolished the responsiveness to IFNgamma. In contrast, discrete deletion of other 5' regions of the SOCS-3 promoter did not substantially modify the inducibility by IFNgamma. Electromobility shift assay analyses revealed that IFNgamma promotes specific DNA binding activities to an oligonucleotide probe containing the SOCS-3/SBE sequence. Even though IFNgamma triggered tyrosine phosphorylation of both STAT1 and STAT3 in macrophages and J774 cells, only STAT1 was appropriately activated and thus found to specifically bind to the SOCS-3/SBE oligonucleotide probe. Accordingly, IFNgamma-induced SOCS-3 protein expression was not impaired in STAT3-deficient embryonal fibroblasts. Taken together, these results demonstrate that the induction of SOCS-3 by IFNgamma depends upon the presence of a STAT-binding element in the SOCS-3 promoter that is specifically activated by STAT1.


Subject(s)
Antineoplastic Agents/pharmacology , Interferon-gamma/pharmacology , Macrophages, Peritoneal/physiology , Repressor Proteins/genetics , Transcription Factors/genetics , Animals , Cells, Cultured , DNA-Binding Proteins/metabolism , Gene Expression/drug effects , Gene Expression/immunology , Macrophages, Peritoneal/cytology , Mice , Mice, Inbred C57BL , Mutagenesis , Promoter Regions, Genetic/genetics , STAT1 Transcription Factor , STAT3 Transcription Factor , Signal Transduction/drug effects , Signal Transduction/immunology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Trans-Activators/metabolism
14.
J Immunol ; 168(12): 6404-11, 2002 Jun 15.
Article in English | MEDLINE | ID: mdl-12055259

ABSTRACT

Previous studies have shown that IL-10 can induce the expression of the suppressor of cytokine signaling 3 (SOCS-3) mRNA in human monocytes and neutrophils, suggesting that the capacity of IL-10 to inhibit the expression of LPS-inducible proinflammatory genes may depend on SOCS-3 induction. However, no direct experimental evidence has been provided to support such hypothesis. Herein, we show that stable transfection of SOCS-3 into the mouse macrophage cell line J774 resulted in an inhibition of NO, TNF-alpha, IL-6, and GM-CSF secretion in response to LPS at levels similar to those exerted by IL-10 in LPS-stimulated wild-type J774. Constitutive SOCS-3 expression also down-regulated the mRNA expression of inducible NO synthase and IL-6 and impaired the production of TNF-alpha, mainly at a post-transcriptional level. In addition, SOCS-3-transfected cells displayed a constitutive expression of the IL-1R antagonist gene, consistent with the observation that IL-10 enhances IL-1R antagonist mRNA in LPS-stimulated wild-type cells. Furthermore, in peritoneal macrophages harvested from mice carrying heterozygous disruption of the SOCS-3 gene, IL-10 was less effective in repressing LPS-stimulated TNF-alpha and NO production. Taken together, our data show that SOCS-3 inhibits LPS-induced macrophage activation, strongly supporting the idea that it plays a role in the molecular mechanism by which IL-10 down-modulates the effector functions of LPS-activated macrophages. Finally, we show that forced expression of SOCS-3 significantly suppresses the ability of IL-10 to trigger tyrosine phosphorylation of STAT3. Therefore, SOCS-3 functions both as an LPS signal inhibitor and as a negative feedback regulator of IL-10/STAT3 signaling.


Subject(s)
Interleukin-10/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Macrophage Activation/immunology , Proteins/physiology , Repressor Proteins , Signal Transduction/immunology , Transcription Factors , Animals , Cell Line , Cells, Cultured , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Interleukin 1 Receptor Antagonist Protein , Interleukin-10/physiology , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Interleukin-6/metabolism , MAP Kinase Signaling System/immunology , Macrophage Activation/genetics , Macrophages/immunology , Macrophages/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Protein Biosynthesis , Proteins/genetics , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , RNA, Messenger/metabolism , Receptors, Interleukin-1/antagonists & inhibitors , STAT3 Transcription Factor , Sialoglycoproteins/biosynthesis , Sialoglycoproteins/genetics , Signal Transduction/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , Transfection , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Up-Regulation/genetics , Up-Regulation/immunology
15.
Eur Cytokine Netw ; 13(1): 47-53, 2002.
Article in English | MEDLINE | ID: mdl-11956020

ABSTRACT

We have recently shown that IL-10 represents an efficient stimulus for suppressor of cytokine signalling (SOCS)-3 mRNA expression in human neutrophils and PBMC. Herein, we identify cAMP-elevating agents such as prostaglandin E2 (PGE2), PGE1, forskolin, dibutyryl cAMP (dbcAMP) and cholera toxin as a novel class of agonists able to induce SOCS-3 mRNA and protein expression in human leukocytes, cooperating with interleukin-10 (IL-10) in such activities. While PGE2 or dbcAMP prolonged the stability of SOCS-3 mRNA isolated from IL-10-treated leukocytes, inhibitors of cAMP-dependent protein kinase A (H89, KT5720, and St-Ht31 peptide) did not influence the action of PGE2/dbcAMP and/or IL-10 on SOCS-3 mRNA and protein expression, implying that their effect are mediated through a PKA-independent pathway. Taken together, our data identify cAMP-elevating substances as a novel class of agonists able to trigger SOCS-3 expression, and suggest that SOCS-3 might be involved in the regulatory effects of cAMP-elevating substances.


Subject(s)
Bucladesine/pharmacology , Dinoprostone/pharmacology , Interleukin-10/pharmacology , Leukocytes, Mononuclear/drug effects , Proteins/metabolism , Repressor Proteins , Transcription Factors , Cyclic AMP/analysis , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/physiology , Drug Combinations , Humans , Interleukin-10/metabolism , Leukocytes, Mononuclear/metabolism , Proteins/genetics , RNA Stability/drug effects , RNA, Messenger/biosynthesis , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...