Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Sci Adv ; 10(7): eadk1835, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38354236

ABSTRACT

The TP53 tumor suppressor gene is mutated early in most of the patients with triple-negative breast cancer (TNBC). The most frequent TP53 alterations are missense mutations that contribute to tumor aggressiveness. Here, we used an autochthonous somatic TNBC mouse model, in which mutant p53 can be toggled on and off genetically while leaving the tumor microenvironment intact and wild-type for p53 to identify physiological dependencies on mutant p53. In TNBCs that develop in this model, deletion of two different hotspot p53R172H and p53R245W mutants triggers ferroptosis in vivo, a cell death mechanism involving iron-dependent lipid peroxidation. Mutant p53 protects cells from ferroptosis inducers, and ferroptosis inhibitors reverse the effects of mutant p53 loss in vivo. Single-cell transcriptomic data revealed that mutant p53 protects cells from undergoing ferroptosis through NRF2-dependent regulation of Mgst3 and Prdx6, which encode two glutathione-dependent peroxidases that detoxify lipid peroxides. Thus, mutant p53 protects TNBCs from ferroptotic death.


Subject(s)
Adenocarcinoma , Ferroptosis , Triple Negative Breast Neoplasms , Animals , Humans , Mice , Cell Line, Tumor , Ferroptosis/genetics , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
2.
Cancer Res Commun ; 3(12): 2640-2652, 2023 12 29.
Article in English | MEDLINE | ID: mdl-38047594

ABSTRACT

Obesity is a significant global health concern. Non-alcoholic fatty liver disease and non-alcoholic steatohepatitis (NASH) are common risk factors for hepatocellular carcinoma (HCC) and are closely associated with metabolic comorbidities, including obesity and diabetes. The TP53 tumor suppressor is the most frequently mutated gene in liver cancers, with half of these alterations being missense mutations. These mutations produce highly abundant proteins in cancer cells which have both inhibitory effects on wildtype (WT) p53, and gain-of-function (GOF) activities that contribute to tumor progression. A Western diet increases p53 activity in the liver. To elucidate the functional consequences of Trp53 mutations in a NASH-driven liver tumorigenesis model, we generated somatic mouse models with Trp53 deletion or the missense hotspot mutant p53R245W only in hepatocytes and placed mice on a high-fat, choline-deficient diet. p53R245W in the presence of diet increased fatty liver, compensatory proliferation in the liver parenchyma, and enriched genes of tumor-promoting pathways such as KRAS signaling, MYC, and epithelial-mesenchymal transition when compared with controls in the premalignant liver. Moreover, p53R245W suppressed transcriptional activity of WT p53 in the liver in vivo under metabolic challenges, and shortened survival and doubling of HCC incidence as compared with control heterozygous mice. Complete loss of Trp53 also significantly accelerated liver tumor incidence and lowered time-to-tumor development compared with WT controls. p53R245W GOF properties increased carcinoma initiation, fueled mixed hepatocholangial carcinoma incidence, and tripled metastatic disease. Collectively, our in vivo studies indicate that p53R245W has stronger tumor promoting activities than Trp53 loss in the context of NASH. SIGNIFICANCE: Using somatic NASH-driven mouse models with p53 deletion or mutant p53R245W only in hepatocytes, we discovered that p53R245W increased carcinoma initiation, fueled hepatocholangial carcinoma incidence, and tripled metastases.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Mice , Animals , Non-alcoholic Fatty Liver Disease/genetics , Carcinoma, Hepatocellular/genetics , Tumor Suppressor Protein p53/genetics , Liver Neoplasms/genetics , Carcinogenesis/genetics , Mutation , Disease Models, Animal , Obesity/complications
3.
Proc Natl Acad Sci U S A ; 120(34): e2308807120, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37579145

ABSTRACT

The TP53 tumor suppressor gene is mutated early in the majority of patients with triple-negative breast cancer (TNBC). The most frequent TP53 alterations are missense mutations that contribute to tumor aggressiveness. We developed an autochthonous somatic K14-Cre driven TNBC mouse model with p53R172H and p53R245W mutations in which mutant p53 can be toggled on and off genetically while leaving the tumor microenvironment intact and wild-type for p53. These mice develop TNBCs with a median latency of 1 y. Deletion of mutant p53R172H or p53R245W in vivo in these tumors blunts their tumor growth and significantly extends survival of mice. Downstream analyses revealed that deletion of mutant Trp53 activated the cyclic GMP-AMP Synthase-Stimulator of Interferon Genes pathway but did not cause apoptosis implicating other mechanisms of tumor regression. Furthermore, we determined that only tumors with stable mutant p53 are dependent on mutant p53 for growth.


Subject(s)
Triple Negative Breast Neoplasms , Tumor Suppressor Protein p53 , Animals , Humans , Mice , Genes, p53 , Mutation , Mutation, Missense , Triple Negative Breast Neoplasms/metabolism , Tumor Microenvironment , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
PLoS One ; 13(3): e0193485, 2018.
Article in English | MEDLINE | ID: mdl-29494633

ABSTRACT

Spondyloarthropathies, the second most frequently occurring form of chronic inflammatory arthritis, affects young adults in particular. However, a proper model with which to study the biology of this disease and to develop therapeutics is lacking. One of the most accepted animal models for this disease uses HLA-B27/Hu-ß2m transgenic rats; however, only 30%-50% of male HLA-B27/Hu-ß2m rats develop spontaneous, clinically apparent spondylitis and have a variable time until disease onset. Here, we report a high-incidence, low-variation spontaneous mouse model that delineates how the combination of inflammatory cytokine interleukin-27 (IL-27) signaling deficiency and mitogenic signaling (mutant p53R172H) in vivo, leads to bone loss in the vertebral bodies and ossification of the cartilage in the intervertebral discs. In this human disease-like mouse model, bone loss and pathogenic bone development are seen as early as 4 months of age in the absence of inflammatory aggregates in the enthesis or intervertebral disc.


Subject(s)
Disease Models, Animal , Receptors, Cytokine/genetics , Spondylarthropathies/pathology , Tumor Suppressor Protein p53/genetics , Animals , Humans , Male , Mice , Mutation , Rats , Receptors, Interleukin , Signal Transduction , Spondylarthropathies/diagnostic imaging , Spondylarthropathies/genetics , Spondylarthropathies/veterinary , X-Ray Microtomography/veterinary
5.
Oncotarget ; 7(47): 77138-77151, 2016 Nov 22.
Article in English | MEDLINE | ID: mdl-27738312

ABSTRACT

Establishment of a permissive pre-malignant niche in concert with mutant stem are key triggers to initiate skin carcinogenesis. An understudied area of research is finding upstream regulators of both these triggers. IL27, a pleiotropic cytokine with both pro- and anti-inflammatory properties, was found to be a key regulator of both. Two step skin carcinogenesis model and K15-KRASG12D mouse model were used to understand the role of IL27 in skin tumors. CD11b-/- mice and small-molecule of ETAR signaling (ZD4054) inhibitor were used in vivo to understand mechanistically how IL27 promotes skin carcinogenesis. Interestingly, using in vivo studies, IL27 promoted papilloma incidence primarily through IL27 signaling in bone-marrow derived cells. Mechanistically, IL27 initiated the establishment of the pre-malignant niche and expansion of mutated stem cells in K15-KRASG12D mouse model by driving the accumulation of Endothelin A receptor (ETAR)-positive CD11b cells in the skin-a novel category of pro-tumor inflammatory identified in this study. These findings are clinically relevant, as the number of IL27RA-positive cells in the stroma is highly related to tumor de-differentiation in patients with squamous cell carcinomas.


Subject(s)
CD11b Antigen/genetics , Interleukins/metabolism , Pyrrolidines/pharmacology , Skin Neoplasms/pathology , Animals , CD11b Antigen/metabolism , Mice , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Receptor, Endothelin A/metabolism , Receptors, Cytokine/genetics , Receptors, Interleukin , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Stem Cell Niche
6.
Clin Cancer Res ; 22(15): 3876-83, 2016 08 01.
Article in English | MEDLINE | ID: mdl-26979394

ABSTRACT

PURPOSE: p53 is mutated in about 50% of human cancers, mostly through missense mutations. Expression of mutant p53 is associated with poor clinical outcomes or metastasis. Although mutant p53 is inherently instable, various stressors such as DNA damage or expression of the oncogenic Kras or c-myc affect the oncogenic properties of mutant p53. However, the effects of inflammation on mutant p53 are largely unknown. IL27 is an important immunomodulatory cytokine, but its impact on mutant p53-driven tumorigenesis has not been reported. EXPERIMENTAL DESIGN: IL27RA(-/-) mice were bred with mutant p53 heterozygous (p53(R172H/+)) mice to obtain IL27RA(-/-)p53(H/+) and IL27RA(-/-)p53(H/H) mice. Mouse survival and tumor spectra for the cohort were analyzed. Stability of p53 protein was analyzed via IHC and Western blot analysis. RESULTS: This study unraveled that lack of IL27 signaling significantly shortened the survival duration of mice with tumors expressing both copies of the mutant p53 gene (Li-Fraumeni mouse model). Interestingly, in mice that were heterozygous for mutant p53, lack of IL27 signaling not only significantly shortened survival time but also doubled the incidence of osteosarcomas. Furthermore, lack of IL27 signaling is closely associated with increased mutant p53 stability in vivo from early age. CONCLUSIONS: These results suggest that IL27 signaling modulates the oncogenic properties of mutant p53 in vivo Clin Cancer Res; 22(15); 3876-83. ©2016 AACR.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Immunomodulation , Interleukin-27/metabolism , Mutation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Genotype , Heterografts , Humans , Immunohistochemistry , Mice , Mice, Knockout , Receptors, Interleukin/metabolism , Signal Transduction , X-Ray Microtomography
7.
Hepatology ; 63(3): 1000-12, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26637970

ABSTRACT

UNLABELLED: The cellular and molecular etiology of unresolved chronic liver inflammation remains obscure. Whereas mutant p53 has gain-of-function properties in tumors, the role of this protein in liver inflammation is unknown. Herein, mutant p53(R172H) is mechanistically linked to spontaneous and sustained liver inflammation and steatosis when combined with the absence of interleukin-27 (IL27) signaling (IL27RA), resembling the phenotype observed in nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) patients. Indeed, these mice develop, with age, hepatocyte necrosis, immune cell infiltration, fibrosis, and micro- and macrosteatosis; however, these phenotypes are absent in mutant p53(R172H) or IL27RA(-/-) mice. Mechanistically, endothelin A receptor (ETAR)-positive macrophages are highly accumulated in the inflamed liver, and chemical inhibition of ETAR signaling reverses the observed phenotype and negatively regulates mutant p53 levels in macrophages. CONCLUSION: The combination of mutant p53 and IL27RA(-/-) causes spontaneous liver inflammation, steatosis, and fibrosis in vivo, whereas either gene alone in vivo has no effects on the liver.


Subject(s)
Genes, p53 , Macrophages/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Receptor, Endothelin A/metabolism , Receptors, Cytokine/deficiency , Animals , Mice, Inbred C57BL , Mice, Transgenic , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, Interleukin
8.
J Natl Cancer Inst ; 107(8)2015 Aug.
Article in English | MEDLINE | ID: mdl-25971300

ABSTRACT

BACKGROUND: Fibrinogen-like protein 2 (FGL2) may promote glioblastoma multiforme (GBM) cancer development by inducing multiple immune-suppression mechanisms. METHODS: The biological significance of FGL2 expression was assessed using the The Cancer Genome Atlast (TCGA) glioma database and tumor lysates analysis. The therapeutic effects of an anti-Fgl2 antibody and the role of immune suppression regulation by Fgl2 were determined in immune-competent, NOD-scid IL2Rgammanull (NSG), and FcÉ£RIIB-/- mice (n = 3-18 per group). Data were analyzed with two-way analysis of variance, log-rank survival analysis, and Pearson correlation. All statistical tests were two-sided. RESULTS: In low-grade gliomas, 72.5% of patients maintained two copies of the FGL2 gene, whereas 83.8% of GBM patients had gene amplification or copy gain. Patients with high levels of FGL2 mRNA in glioma tissues had a lower overall survival (P = .009). Protein levels of FGL2 in GBM lysates were higher relative to low-grade glioma lysates (11.48±5.75ng/mg vs 3.96±1.01ng/mg, P = .003). In GL261 mice treated with an anti-FGL2 antibody, median survival was 27 days compared with only 17 days for mice treated with an isotype control antibody (P = .01). The anti-FGL2 antibody treatment reduced CD39(+) Tregs, M2 macrophages, programmed cell death protein 1 (PD-1), and myeloid-derived suppressor cells (MDSCs). FGL2-induced increases in M2, CD39, and PD-1 were ablated in FcÉ£RIIB-/- mice. CONCLUSIONS: FGL2 augments glioma immunosuppression by increasing the expression levels of PD-1 and CD39, expanding the frequency of tumor-supportive M2 macrophages via the FcγRIIB pathway, and enhancing the number of MDSCs and CD39(+) regulatory T cells. Collectively, these results show that FGL2 functions as a key immune-suppressive modulator and has potential as an immunotherapeutic target for treating GBM.


Subject(s)
Biomarkers, Tumor/immunology , Brain Neoplasms/immunology , Fibrinogen/immunology , Glioblastoma/immunology , Immunosuppression Therapy , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Analysis of Variance , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/blood , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Female , Fibrinogen/genetics , Fibrinogen/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/blood , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Immunologic Factors/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Middle Aged , RNA, Messenger/metabolism , Survival Analysis
9.
Biochim Biophys Acta ; 1846(1): 152-60, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24821201

ABSTRACT

Inflammation plays an integral part in tumor initiation. Specifically, patients with colitis, pancreatitis, or hepatitis have an increased susceptibility to cancer. The activation, mutation, and overexpression of oncogenes have been well documented in cell proliferation and transformation. Recently, oncogenes were found to also regulate the inflammatory milieu in tumors. Similarly, the inflammatory milieu can promote oncogene activation. In this review, we summarize advances of the symbiotic relationship oncogene activation and inflammation in gastrointestinal tumors such as colorectal, hepatic, and pancreatic tumors. NF-κB and STAT3 are the two most common pathways that are deregulated via these oncogenes. Understanding these interactions may yield effective therapeutic strategies for tumor prevention and treatment.


Subject(s)
Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , Inflammation/genetics , Oncogenes/physiology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Gastrointestinal Neoplasms/immunology , Gene Expression Regulation, Neoplastic , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Permeability , Signal Transduction/genetics
10.
Cancer Res ; 74(6): 1645-50, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24448245

ABSTRACT

To date, no specific marker exists for the detection of circulating tumor cells (CTC) from different types of sarcomas, though tools are available for detection of CTCs in peripheral blood of patients with cancer for epithelial cancers. Here, we report cell-surface vimentin (CSV) as an exclusive marker on sarcoma CTC regardless of the tissue origin of the sarcoma as detected by a novel monoclonal antibody. Utilizing CSV as a probe, we isolated and enumerated sarcoma CTCs with high sensitivity and specificity from the blood of patients bearing different types of sarcoma, validating their phenotype by single cell genomic amplification, mutation detection, and FISH. Our results establish the first universal and specific CTC marker described for enumerating CTCs from different types of sarcoma, thereby providing a key prognosis tool to monitor cancer metastasis and relapse.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplastic Cells, Circulating/metabolism , Sarcoma/pathology , Vimentin/metabolism , Antibodies, Monoclonal/chemistry , Base Sequence , Cell Line, Tumor , DNA Mutational Analysis , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Gene Amplification , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Microscopy, Fluorescence , Prognosis , Sarcoma/diagnosis , Sarcoma/metabolism , Sensitivity and Specificity , Single-Cell Analysis , Tumor Suppressor Protein p53/genetics , Vimentin/immunology
11.
Mediators Inflamm ; 2013: 378971, 2013.
Article in English | MEDLINE | ID: mdl-24369443

ABSTRACT

The safest and most effective cytokine therapies require the favorable accumulation of the cytokine in the tumor environment. While direct treatment into the neoplasm is ideal, systemic tumor-targeted therapies will be more feasible. Electroporation-mediated transfection of cytokine plasmid DNA including a tumor-targeting peptide-encoding sequence is one method for obtaining a tumor-targeted cytokine produced by the tumor-bearing patient's tissues. Here, the impact on efficacy of the location of targeting peptide, choice of targeting peptide, tumor histotype, and cytokine utilization are studied in multiple syngeneic murine tumor models. Within the same tumor model, the location of the targeting peptide could either improve or reduce the antitumor effect of interleukin (IL)12 gene treatments, yet in other tumor models the tumor-targeted IL12 plasmid DNAs were equally effective regardless of the peptide location. Similarly, the same targeting peptide that enhances IL12 therapies in one model fails to improve the effect of either IL15 or PF4 for inhibiting tumor growth in the same model. These interesting and sometimes contrasting results highlight both the efficacy and personalization of tumor-targeted cytokine gene therapies while exposing important aspects of these same therapies which must be considered before progressing into approved treatment options.


Subject(s)
Cytokines/metabolism , Gene Expression Regulation, Neoplastic , Genetic Therapy/methods , Neoplasms/therapy , Protein Processing, Post-Translational , Animals , Cell Line, Tumor , DNA/metabolism , Disease Models, Animal , Electroporation , Female , Interleukin-12/genetics , Interleukin-15/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Neoplasms/genetics , Peptides/chemistry , Plasmids/metabolism , Transfection
12.
Cell Commun Signal ; 11: 95, 2013 Dec 13.
Article in English | MEDLINE | ID: mdl-24330710

ABSTRACT

BACKGROUND: Studies into the regulation of interleukin-10 (IL-10), have focused only on the molecular or single-cell level. The cues that induce IL-10 in the context of cell-to-cell communication are scarce. To fill this gap, this study elucidates the cell-to-cell interaction dependent regulation of IL-10. RESULTS: The simultaneous activation of CD4+ T cells via CD3/CD28 and stimulation of macrophages via CpG and their intercellular communication with each other in the same microenvironment is necessary to induce a synergistic expression of IL-10. NF-κB1, ERK, and STAT3 are positive regulators of this cell-to-cell communication mediated molecular change of IL-10 induction. Strikingly, the activation of CD40/CD154 signaling is a negative regulator of IL-10 levels by CD3/CD28/CpG. CONCLUSIONS: These findings are of prominence as CD3/CD28/CpG treatment can induce the anti-inflammatory cytokines IL-10 and IL-30, and the activation or inhibition of the CD40/CD154 acts as molecular rheostat of the expression of IL-10 or IL-30. More importantly, this not only serves as an example of IL-10 regulation at the cellular via coordination of two signals from two cell types, but these findings also lay the molecular and cellular groundwork for future studies to investigate how to manipulate IL-10 or IL-30 production during inflammation, cancer, or autoimmune diseases.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Interleukin-10/metabolism , Macrophages, Peritoneal/metabolism , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cell Communication , CpG Islands , Mice
13.
J Immunol ; 188(8): 3709-15, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22407920

ABSTRACT

IL-30, the p28 subunit of IL-27, interacts with EBV-induced gene 3 to form IL-27, which modulates both proinflammatory and anti-inflammatory responses during autoimmune or infectious disease. It also acts as a natural antagonist of gp130, thereby attenuating the signals of other gp130-associated cytokines. IL-30 regulation via LPS has been reported by others, but the intercellular communication that induces IL-30 expression is unknown. In this study, we show that treatment with anti-CD3/CD28 Abs plus CpG oligodeoxynucleotides induces robust expression of IL-30, whereas either treatment alone induces only low expression of IL-30. This observation in vitro mirrors the murine model in which administration of CpG under inflammatory conditions in vivo induces IL-30 expression. This robust induction of IL-30 occurs through the coordination of helper CD4(+) T cells and innate immune cells (e.g., macrophages) and, to a lesser degree, B cells via the CD40/CD154 signaling pathway. These findings reveal a previously unrecognized mechanism that integrates signaling pathways from T cells and macrophages at the cellular level to induce IL-30 expression.


Subject(s)
CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , Interleukins/immunology , Macrophages/immunology , Animals , Antibodies, Neutralizing , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Communication , Gene Expression Regulation , Interleukins/genetics , Interleukins/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/immunology , Oligodeoxyribonucleotides/pharmacology , Signal Transduction , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology
14.
Hepatology ; 55(4): 1204-14, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22105582

ABSTRACT

UNLABELLED: The liver is the major metabolic organ and is subjected to constant attacks from chronic viral infection, uptake of therapeutic drugs, life behavior (alcoholic), and environmental contaminants, all of which result in chronic inflammation, fibrosis, and, ultimately, cancer. Therefore, there is an urgent need to discover effective therapeutic agents for the prevention and treatment of liver injury, the ideal drug being a naturally occurring biological inhibitor. Here we establish the role of IL30 as a potent antiinflammatory cytokine that can inhibit inflammation-induced liver injury. In contrast, interleukin (IL)27, which contains IL30 as a subunit, is not hepatoprotective. Interestingly, IL30 is induced by the proinflammatory signal such as IL12 through interferon-gamma (IFN-γ)/signal transducer and activator of transcription 1 signaling. In animal models, administration of IL30 by way of a gene therapy approach prevents and treats both IL12-, IFN-γ-, and concanavalin A-induced liver toxicity. Likewise, immunohistochemistry analysis of human tissue samples revealed that IL30 is highly expressed in hepatocytes, yet barely expressed in inflammation-induced tissue such as fibrous/connective tissue. CONCLUSION: These novel observations reveal a novel role of IL30 as a therapeutic cytokine that suppresses proinflammatory cytokine-associated liver toxicity.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/therapy , Cytokines/adverse effects , Genetic Therapy , Interleukins/genetics , Animals , Concanavalin A/adverse effects , Disease Models, Animal , Humans , In Vitro Techniques , Interferon-gamma/adverse effects , Interferon-gamma/metabolism , Interleukin-12/adverse effects , Interleukin-12/genetics , Interleukin-12/metabolism , Interleukins/metabolism , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , Receptors, Cytokine/deficiency , Receptors, Cytokine/genetics , Receptors, Interleukin , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics
15.
PLoS One ; 6(4): e19072, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21559505

ABSTRACT

Crosstalk between tumor cells and the cognate microenvironment plays a crucial role in tumor initiation and progression. However, only a few genes are known to affect such a crosstalk. This study reveals that WSX1 plays such a role when highly expressed in tumor cells. The expression of WSX1 in Lewis Lung Carcinoma (LLC) and the melanoma cell line AGS induces the death of T cells and inhibits the production of the effector cytokine IFNγ from NK and T cells, resulting in the promotion of tumor growth. These pro-tumorigenic properties of WSX1 are independent of IL27. This key observation reveals a new pathway of tumor-host interaction, which will ultimately lead to better strategies in immune therapy to reverse tumor tolerance.


Subject(s)
Carcinoma, Lewis Lung/metabolism , Melanoma/metabolism , Receptors, Cytokine/biosynthesis , Animals , Carcinoma, Lewis Lung/immunology , Cell Line, Tumor , Coculture Techniques , Cytokines/metabolism , Immune System , Immune Tolerance , Interferon-gamma/metabolism , Interleukin-17/metabolism , Killer Cells, Natural/cytology , Melanoma/immunology , Mice , Mice, Inbred C57BL , Mice, SCID , Microscopy, Confocal/methods , Receptors, Cytokine/physiology , Receptors, Interleukin , T-Lymphocytes/cytology , T-Lymphocytes/immunology
16.
Mol Ther ; 19(8): 1468-77, 2011 08.
Article in English | MEDLINE | ID: mdl-21386825

ABSTRACT

Like many effective therapeutics, interleukin-12 (IL-12) therapy often causes side effects. Tumor targeted delivery may improve the efficacy and decrease the toxicity of systemic IL-12 treatments. In this study, a novel targeting approach was investigated. A secreted alkaline phosphatase (SEAP) reporter gene-based screening process was used to identify a mini-peptide which can be produced in vivo to target gene products to tumors. The coding region for the best peptide was inserted into an IL-12 gene to determine the antitumor efficacy. Affinity chromatography, mass spectrometry analysis, and binding studies were used to identify a receptor for this peptide. We discovered that the linear peptide VNTANST increased the tumor accumulation of the reporter gene products in five independent tumor models including one human xenogeneic model. The product from VNTANST-IL-12 fusion gene therapy increased accumulation of IL-12 in the tumor environment, and in three tumor models, VNTANST-IL-12 gene therapy inhibited distal tumor growth. In a spontaneous lung metastasis model, inhibition of metastatic tumor growth was improved compared to wild-type IL-12 gene therapy, and in a squamous cell carcinoma model, toxic liver lesions were reduced. The receptor for VNTANST was identified as vimentin. These results show the promise of using VNTANST to improve IL-12 treatments.


Subject(s)
Carcinoma, Squamous Cell/genetics , Genetic Therapy/methods , Interleukin-12/genetics , Lung Neoplasms/genetics , Recombinant Fusion Proteins/metabolism , Animals , Antineoplastic Agents , Carcinoma, Squamous Cell/drug therapy , Carrier Proteins , Chromatography, Affinity , Genes, Reporter , Interleukin-12/pharmacology , Interleukin-12/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Mass Spectrometry , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Nude , Vimentin/metabolism
17.
Cancer Res ; 69(13): 5505-13, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19549909

ABSTRACT

It is well known that the interleukin (IL)-27 receptor WSX1 is expressed in immune cells and induces an IL-27-dependent immune response. Opposing this conventional dogma, this study reveals a much higher level of WSX1 expression in multiple types of epithelial tumor cells when compared with normal epithelial cells. Expression of exogenous WSX1 in epithelial tumor cells suppresses tumorigenicity in vitro and inhibits tumor growth in vivo. Different from the role of WSX1 in immune cells, the antitumor activity of WSX1 in epithelial tumor cells is independent of IL-27 signaling but is mainly dependent on natural killer (NK) cell surveillance. Deficiency of either the IL-27 subunit EBV-induced gene 3 or the IL-27 receptor WSX1 in the host animals had no effect on tumor growth inhibition induced by WSX1 expression in tumor cells. Expression of WSX1 in epithelial tumor cells enhances NK cell cytolytic activity against tumor cells, whereas the absence of functional NK cells impairs the WSX1-mediated inhibition of epithelial tumor growth. The underlying mechanism by which WSX1 expression in tumor cells enhances NK cytolytic activity is dependent on up-regulation of NKG2D ligand expression. Our results reveal an IL-27-independent function of WSX1: sensitizing NK cell-mediated antitumor surveillance via a NKG2D-dependent mechanism.


Subject(s)
Interleukin-17/immunology , Killer Cells, Natural/immunology , Receptors, Cytokine/genetics , Receptors, Interleukin/genetics , Animals , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor/immunology , Cell Line, Tumor/pathology , Colon , Flow Cytometry , Gene Expression Regulation , HeLa Cells , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Humans , Interleukin-17/physiology , Mice , Mice, Knockout , Papillomaviridae/immunology , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...