Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Anticancer Res ; 41(3): 1485-1496, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33788741

ABSTRACT

BACKGROUND: As the prognosis of biliary tract cancer (BTC) is extremely poor and treatment options are limited, new treatment modalities are urgently needed. We designed a phase II clinical trial to investigate the immune responses and clinical benefits of OCV-C01, an HLA-A*24:02-restricted three-peptide cancer vaccine targeting VEGFR1, VEGFR2, and KIF20A. PATIENTS AND METHODS: Participants were patients with advanced BTC who had unresectable tumours and were refractory to standard chemotherapy. OCV-C01 was injected weekly until the discontinuance criteria were met. RESULTS: Six participants, including four patients positive for HLA-A*24:02, were enrolled in this study for assessment of efficacy. Four out of six patients exhibited vaccine-specific T-cell responses to one or more of three antigens. Log-rank tests revealed that vaccine-specific T cell responses contributed significantly to overall survival. CONCLUSION: The cancer vaccine had positive effects on survival, indicating that this approach warrants further clinical studies.


Subject(s)
Biliary Tract Neoplasms/drug therapy , Cancer Vaccines/administration & dosage , Kinesins/antagonists & inhibitors , Vaccines, Subunit/administration & dosage , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Aged , Aged, 80 and over , Biliary Tract Neoplasms/immunology , Biliary Tract Neoplasms/metabolism , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Disease-Free Survival , Female , Fever/chemically induced , Headache/chemically induced , Humans , Kinesins/immunology , Male , Middle Aged , Molecular Targeted Therapy/methods , Prognosis , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Vaccines, Subunit/adverse effects , Vaccines, Subunit/immunology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology
2.
Anticancer Res ; 38(11): 6121-6126, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30396927

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is a major cause of morbidity and mortality throughout the world. It is the third most common cancer worldwide and the fourth most common cause of cancer-related death. FOLFOX, a combination of leucovorin calcium, fluorouracil, and oxaliplatin, is the first-line chemotherapy for stage III and stage IV CRC. However, patients with FOLFOX-resistant CRC have a poor prognosis. In recent years, virochemotherapy has been proposed as a potential treatment for chemotherapy-resistant cancer. MATERIALS AND METHODS: Through our first screening assay, we found that coxsackievirus A11 (CVA11) displayed potent oncolytic activities. We tested whether coxsackievirus A11 (CVA11) has oncolytic activity in human CRC cells in vitro and in vivo. We also examined whether pretreatment of oxaliplatin-resistant CRC cells with oxaliplatin enhances the oncolytic activity of CVA11. RESULTS: We found that CVA11 was potently oncolytic against the oxaliplatin-sensitive Caco-2 cell line, but had little effect on the oxaliplatin-resistant line WiDr. However, pretreatment of WiDr cells with oxaliplatin enhanced the oncolytic activity of CVA11, and the combination therapy was more cytotoxic than either oxaliplatin treatment or CVA11 infection alone. Furthermore, growth of subcutaneous WiDr tumors in a xenograft model was significantly lower in mice treated with oxaliplatin followed by intratumoral CVA11 injection compared with mice receiving either treatment alone. CONCLUSION: Oxaliplatin pretreatment sensitized oxaliplatin-resistant CRC cells to lysis by CVA11 infection in vitro and in vivo. Taken together, these findings identify a novel potential chemovirotherapeutic modality for the treatment of oxaliplatin-resistant human CRC.


Subject(s)
Colorectal Neoplasms/therapy , Enterovirus/physiology , Oncolytic Virotherapy/methods , Oxaliplatin/pharmacology , Animals , CD55 Antigens/biosynthesis , Caco-2 Cells , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/virology , Combined Modality Therapy , Drug Resistance, Neoplasm , Enterovirus/drug effects , Female , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
3.
Anticancer Res ; 38(8): 4543-4547, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30061220

ABSTRACT

BACKGROUND/AIM: Small cell lung cancer (SCLC) is still a deadly type of cancer for which there are few effective therapeutic strategies. Development of a new molecule targeting agent is urgently desired. Previously we showed that recombination signal binding protein for immunoglobulin-kappa-J region (RBPJ) and mastermind-like 3 (MAML3) are new therapeutic targets for pancreatic cancer. In the present study, we analyzed whether RBPJ/MAML3 inhibition could also be a new therapeutic strategy for SCLC. MATERIALS AND METHODS: Using silencing of RBPJ/MAML3, proliferation, invasion, migration and chemosensitivity of SBC-5 cells were investigated. RESULTS: RBPJ/MAML3 inhibition reduced Smoothened and HES1 expression, suggesting that RBPJ/MAML3 signaling was through Hedgehog and NOTCH pathways. In the analysis of cell functions, RBPJ/MAML3 inhibition significantly reduced proliferation and invasiveness via reduction of expression of matrix metalloproteinases. On the other hand, RBPJ/MAML3 inhibition also reduced chemosensitivity to cis-diamminedichlo-roplatinum and gemcitabine. CONCLUSION: These results suggest that RBPJ and MAML3 could be new therapeutic targets for SCLC, however, chemosensitivity may be reduced in combinational use with other chemo-therapeutic agents.


Subject(s)
DNA-Binding Proteins/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Lung Neoplasms/metabolism , Nuclear Proteins/metabolism , Small Cell Lung Carcinoma/metabolism , Transcription Factors/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Hedgehog Proteins/metabolism , Humans , Lung Neoplasms/drug therapy , Matrix Metalloproteinases/metabolism , Neoplasm Invasiveness/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Small Cell Lung Carcinoma/drug therapy , Trans-Activators , Gemcitabine
4.
Cancer Cell ; 26(3): 331-343, 2014 Sep 08.
Article in English | MEDLINE | ID: mdl-25132496

ABSTRACT

Endoplasmic reticulum (ER) stress has been implicated in the pathogenesis of viral hepatitis, insulin resistance, hepatosteatosis, and nonalcoholic steatohepatitis (NASH), disorders that increase risk of hepatocellular carcinoma (HCC). To determine whether and how ER stress contributes to obesity-driven hepatic tumorigenesis we fed wild-type (WT) and MUP-uPA mice, in which hepatocyte ER stress is induced by plasminogen activator expression, with high-fat diet. Although both strains were equally insulin resistant, the MUP-uPA mice exhibited more liver damage, more immune infiltration, and increased lipogenesis and, as a result, displayed classical NASH signs and developed typical steatohepatitic HCC. Both NASH and HCC development were dependent on TNF produced by inflammatory macrophages that accumulate in the MUP-uPA liver in response to hepatocyte ER stress.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Endoplasmic Reticulum Stress , Liver Neoplasms, Experimental/metabolism , Overnutrition/complications , Tumor Necrosis Factor-alpha/physiology , Animals , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/pathology , Cells, Cultured , Diet, High-Fat/adverse effects , Endoplasmic Reticulum Chaperone BiP , Fatty Liver/etiology , Fatty Liver/metabolism , Heat-Shock Proteins/metabolism , Lipogenesis , Liver Neoplasms, Experimental/etiology , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Transgenic , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Tumor Burden , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
5.
Cell ; 155(2): 384-96, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24120137

ABSTRACT

Hepatocellular carcinoma (HCC) is a slowly developing malignancy postulated to evolve from premalignant lesions in chronically damaged livers. However, it was never established that premalignant lesions actually contain tumor progenitors that give rise to cancer. Here, we describe isolation and characterization of HCC progenitor cells (HcPCs) from different mouse HCC models. Unlike fully malignant HCC, HcPCs give rise to cancer only when introduced into a liver undergoing chronic damage and compensatory proliferation. Although HcPCs exhibit a similar transcriptomic profile to bipotential hepatobiliary progenitors, the latter do not give rise to tumors. Cells resembling HcPCs reside within dysplastic lesions that appear several months before HCC nodules. Unlike early hepatocarcinogenesis, which depends on paracrine IL-6 production by inflammatory cells, due to upregulation of LIN28 expression, HcPCs had acquired autocrine IL-6 signaling that stimulates their in vivo growth and malignant progression. This may be a general mechanism that drives other IL-6-producing malignancies.


Subject(s)
Autocrine Communication , Gene Expression Regulation, Neoplastic , Interleukin-6/metabolism , Liver Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Disease Progression , Hepacivirus , Hepatitis C/genetics , Hepatitis C/metabolism , Hepatitis C/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mice, Inbred C57BL
6.
J Clin Invest ; 123(5): 2231-43, 2013 May.
Article in English | MEDLINE | ID: mdl-23563314

ABSTRACT

Chronic pancreatitis is an inflammatory disease that causes progressive destruction of pancreatic acinar cells and, ultimately, loss of pancreatic function. We investigated the role of IκB kinase α (IKKα) in pancreatic homeostasis. Pancreas-specific ablation of IKKα (Ikkα(Δpan)) caused spontaneous and progressive acinar cell vacuolization and death, interstitial fibrosis, inflammation, and circulatory release of pancreatic enzymes, clinical signs resembling those of human chronic pancreatitis. Loss of pancreatic IKKα causes defective autophagic protein degradation, leading to accumulation of p62-mediated protein aggregates and enhanced oxidative and ER stress in acinar cells, but none of these effects is related to NF-κB. Pancreas-specific p62 ablation prevented ER and oxidative stresses and attenuated pancreatitis in Ikkα(Δpan) mice, suggesting that cellular stress induced by p62 aggregates promotes development of pancreatitis. Importantly, downregulation of IKKα and accumulation of p62 aggregates were also observed in chronic human pancreatitis. Our studies demonstrate that IKKα, which may control autophagic protein degradation through its interaction with ATG16L2, plays a critical role in maintaining pancreatic acinar cell homeostasis, whose dysregulation promotes pancreatitis through p62 aggregate accumulation.


Subject(s)
Acinar Cells/cytology , Gene Expression Regulation, Enzymologic , I-kappa B Kinase/metabolism , Pancreatitis/metabolism , Animals , Autophagy , Carrier Proteins/metabolism , Cell Proliferation , Down-Regulation , Endoplasmic Reticulum/metabolism , Fibrosis , Immunohistochemistry , Inflammation , Mice , Mice, Transgenic , NF-kappa B/metabolism , Oxidative Stress , Transcription Factor TFIIH , Transcription Factors/metabolism
7.
Cell ; 147(6): 1233-47, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22153071

ABSTRACT

Hepatocyte nuclear factor 4α (HNF4α) is essential for liver development and hepatocyte function. Here, we show that transient inhibition of HNF4α initiates hepatocellular transformation through a microRNA-inflammatory feedback loop circuit consisting of miR-124, IL6R, STAT3, miR-24, and miR-629. Moreover, we show that, once this circuit is activated, it maintains suppression of HNF4α and sustains oncogenesis. Systemic administration of miR-124, which modulates inflammatory signaling, prevents and suppresses hepatocellular carcinogenesis by inducing tumor-specific apoptosis without toxic side effects. As we also show that this HNF4α circuit is perturbed in human hepatocellular carcinomas, our data raise the possibility that manipulation of this microRNA feedback-inflammatory loop has therapeutic potential for treating liver cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic , Hepatocyte Nuclear Factor 4/metabolism , Inflammation/metabolism , Liver Neoplasms/metabolism , MicroRNAs/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Receptors, Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism
8.
Liver Int ; 30(8): 1229-39, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20536716

ABSTRACT

BACKGROUND: Methionine-choline-deficient (MCD) diets cause steatohepatitis in rodents and are used to model fatty liver disease in human beings. Recent studies have identified sucrose as a major contributor to MCD-related liver disease through its ability to promote hepatic de novo lipogenesis. AIMS: To determine whether glucose and fructose, the two constitutents of sucrose, differ in their capacity to provoke steatohepatitis when incorporated individually into MCD formulas. MATERIALS & METHODS: MCD and control formulas prepared with either glucose or fructose as the sole source of carbohydrate were fed to mice for 21 days. Liver injury was assessed biochemically and histologically together with hepatic gene expression and fatty acid analysis. RESULTS: Mice fed MCD formulas developed similar degrees of hepatic steatosis whether they contained glucose or fructose. By contrast, mice fed MCD-fructose developed significantly more hepatocellular injury than mice fed MCD-glucose, judged by histology, apoptosis staining and serum alanine aminotransferase. Liver injury in MCD-fructose mice coincided with an exaggerated rise in the ratio of long-chain saturated to unsaturated fatty acids in the liver. Notably, hepatic inflammation was not enhanced in mice fed MCD-fructose, correlating instead with hepatic lipid peroxidation, which was equivalent in the two MCD groups. DISCUSSION: Fructose is more cytotoxic than glucose when used as the source of carbohydrate in MCD formulas. CONCLUSION: The data suggest the enhanced cytotoxicity of fructose in the MCD model is related to its ability to stimulate de novo lipogenesis, which yields harmful long-chain saturated fatty acids.


Subject(s)
Diet/adverse effects , Dietary Sucrose/adverse effects , Fatty Liver/etiology , Fructose/adverse effects , Gene Expression Regulation/drug effects , Liver/pathology , Alanine Transaminase/blood , Analysis of Variance , Animals , Blood Glucose , Cholesterol/blood , Choline Deficiency , Fatty Liver/pathology , Gene Expression Profiling , Lipid Peroxidation/physiology , Lipogenesis/drug effects , Liver/metabolism , Male , Methionine/deficiency , Mice , Mice, Mutant Strains , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/blood
9.
Cancer Cell ; 17(3): 286-97, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20227042

ABSTRACT

The NF-kappaB activating kinase IKKbeta suppresses early chemically induced liver tumorigenesis by inhibiting hepatocyte death and compensatory proliferation. To study IKKbeta's role in late tumor promotion and progression, we developed a transplant system that allows initiated mouse hepatocytes to form hepatocellular carcinomas (HCC) in host liver after a long latency. Deletion of IKKbeta long after initiation accelerated HCC development and enhanced proliferation of tumor initiating cells. These effects of IKKbeta/NF-kappaB were cell autonomous and correlated with increased accumulation of reactive oxygen species that led to JNK and STAT3 activation. Hepatocyte-specific STAT3 ablation prevented HCC development. The negative crosstalk between NF-kappaB and STAT3, which is also evident in human HCC, is a critical regulator of liver cancer development and progression.


Subject(s)
Hepatocytes/metabolism , I-kappa B Kinase/physiology , Liver Neoplasms/metabolism , NF-kappa B/physiology , Oxidative Stress , STAT3 Transcription Factor/metabolism , Animals , Gene Deletion , Hepatocytes/transplantation , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Transcriptional Activation
10.
Cancer Cell ; 17(1): 89-97, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20129250

ABSTRACT

Chronic exposure to tobacco smoke, which contains over 60 tumor-initiating carcinogens, is the major risk factor for development of lung cancer, accounting for a large portion of cancer-related deaths worldwide. It is well established that tobacco smoke is a tumor initiator, but we asked whether it also acts as a tumor promoter once malignant initiation, such as caused by K-ras activation, has taken place. Here we demonstrate that repetitive exposure to tobacco smoke promotes tumor development both in carcinogen-treated mice and in transgenic mice undergoing sporadic K-ras activation in lung epithelial cells. Tumor promotion is due to induction of inflammation that results in enhanced pneumocyte proliferation and is abrogated by IKKbeta ablation in myeloid cells or inactivation of JNK1.


Subject(s)
Cell Transformation, Neoplastic/chemically induced , I-kappa B Kinase/metabolism , Inflammation/chemically induced , Lung Neoplasms/chemically induced , Mitogen-Activated Protein Kinase 8/metabolism , Tobacco Smoke Pollution/adverse effects , Animals , Carcinogens/toxicity , Cell Transformation, Neoplastic/metabolism , Enzyme-Linked Immunosorbent Assay , Genes, ras/genetics , Immunohistochemistry , Inflammation/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Transgenic , Nitrosamines/toxicity , Reverse Transcriptase Polymerase Chain Reaction
11.
J Lipid Res ; 50(10): 2072-82, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19295183

ABSTRACT

Methionine-choline-deficient (MCD) diets cause steatohepatitis in rodents and are used to study the pathophysiology of fatty liver disease in human beings. The most widely used commercial MCD formulas not only lack methionine and choline but also contain excess sucrose and fat. The objective of this study was to determine whether dietary sucrose in the MCD formula plays a role in the pathogenesis of MCD-related liver disease. We prepared two custom MCD formulas, one containing sucrose as the principal carbohydrate and the other substituting sucrose with starch. Mice fed the sucrose-enriched formula developed typical features of MCD-related liver disease, including hepatic steatosis, hepatocellular apoptosis, alanine aminotransferase elevation, lipid peroxidation, and hepatic inflammation. In contrast, mice fed MCD-starch were significantly protected against liver injury. MCD-sucrose and MCD-starch mice displayed identical diet-related abnormalities in hepatic fatty acid uptake and triglyceride secretion. Hepatic de novo lipogenesis and triglyceride synthesis, however, were 2 times higher in MCD-sucrose mice than MCD-starch mice (P < 0.01). Hepatic lipid analysis revealed accumulation of excess saturated fatty acids in MCD-sucrose mice that correlated with hepatocellular injury. Overall, the results indicate that dietary sucrose is critical to the pathogenesis of MCD-mediated steatohepatitis. They suggest that saturated fatty acids, which are products of de novo lipogenesis, are mediators of hepatic toxicity in this model of liver disease.


Subject(s)
Choline Deficiency/physiopathology , Fatty Liver/chemically induced , Fatty Liver/metabolism , Liver/drug effects , Liver/metabolism , Methionine/deficiency , Sucrose/pharmacology , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Choline Deficiency/genetics , Fatty Acids/blood , Fatty Acids/metabolism , Fatty Liver/genetics , In Situ Nick-End Labeling , Lipid Peroxidation/drug effects , Lipids/blood , Liver/pathology , Male , Mice , Polymerase Chain Reaction
12.
Gastroenterology ; 131(1): 179-93, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16831601

ABSTRACT

BACKGROUND & AIMS: A recent study has suggested that the methylation silencing of the suppressor of cytokine signaling-3 (SOCS3), a negative regulator of interleukin-6-related cytokines, could be involved in hepatocellular carcinoma (HCC). However, the roles of SOCS3 in hepatocellular carcinogenesis and hepatitis have not been established. We investigated the effect of deleting the SOCS3 gene on the development of hepatitis and HCC in hepatitis C virus-infected patients and mouse models. METHODS: The expression of SOCS genes in HCC and non-HCC regions of patient samples was determined by real-time reverse-transcription polymerase chain reaction and immunoblotting. The conditional knockout approach in mice was used to determine the hepatocyte-specific roles of SOCS3. To generate a liver-specific deletion, floxed SOCS3 (SOCS3(fl/fl)) mice were crossed with albumin-Cre transgenic mice. Hepatitis and HCC were induced by administering concanavalin A and diethylnitrosamine, respectively. RESULTS: SOCS3 expression was reduced in the HCC regions compared with the non-HCC regions. Carcinogen-induced hepatic tumor development was enhanced by deletion of the SOCS3 gene, which was associated with higher levels of the targets of signal transducers and activators of transcription (ie, B-cell lymphoma-XL, B-cell lymphoma-2, C-myelocytomatosis, cyclin D1, and vascular endothelial growth factor). In the concanavalin A-mediated hepatitis model, deletion of the SOCS3 gene in the hepatocytes protected against liver injury through suppression of interferon-gamma signaling and induction of the antiapoptotic protein Bcl-XL. CONCLUSIONS: Deletion of the SOCS3 gene in hepatocytes promotes the activation of STAT3, resistance to apoptosis, and an acceleration of proliferation, resulting in enhanced hepatitis-induced hepatocarcinogenesis.


Subject(s)
Carcinoma, Hepatocellular/genetics , Chemical and Drug Induced Liver Injury/complications , Gene Deletion , Hepatocytes/pathology , Liver Neoplasms/genetics , RNA, Neoplasm/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Adult , Aged , Animals , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/pathology , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Concanavalin A/toxicity , Female , Hepatocytes/drug effects , Humans , Immunohistochemistry , In Vitro Techniques , Liver Neoplasms/etiology , Liver Neoplasms/pathology , Male , Mice , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Suppressor of Cytokine Signaling 3 Protein
13.
Gastroenterology ; 130(2): 373-88, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16472593

ABSTRACT

BACKGROUND & AIMS: The suppressor of cytokine signaling-1 (SOCS1) is a potent negative regulator of various cytokines and it has been implicated in the regulation of immune responses. However, the role of SOCS1 in inflammatory bowel diseases (IBDs) has not been clarified. To determine the role of SOCS1 in colitis, we generated SOCS1/T-cell receptor alpha (TCRalpha) double knockout (DKO) mice. METHODS: The depletion of interferon gamma (IFNgamma) and IL-4 was achieved by crossing the DKO mice with IFNgamma knockout (KO) mice and by the administration of anti-IL-4 antibody, respectively. The activation of cytokine-induced transcription factors was determined by Western blotting with phosphorylation-specific antibodies, and the induction of inflammatory factors was measured by reverse-transcription polymerase chain reaction. RESULTS: Much more severe colitis developed in 100% of the DKO mice within 9 weeks of age than in TCRalpha-KO mice. Although the proportion and the activation status of CD4(+) TCRalpha(-)beta(+) T cells in DKO mice were similar to those in TCRalpha-KO mice, signal transducer and activator of transcription 1, nuclear factor kappaB, and their target genes were hyperactivated in infiltrated mononuclear cells and colonic epithelial cells in DKO mice. Cytokine-depletion experiments showed that exacerbated colitis in the DKO mice was dependent on both IFNgamma and IL-4. SOCS1-deficient cells were hypersensitive to IFNgamma, IL-4, and lipopolysaccharides, depending on the target genes. CONCLUSIONS: SOCS1 plays an important role in preventing murine colitis by restricting the cytokine signals. SOCS1/TCRalpha DKO mice could be a useful model for investigating human IBD.


Subject(s)
Carrier Proteins/physiology , Inflammatory Bowel Diseases/physiopathology , Interferon-gamma/physiology , Interleukin-4/physiology , Repressor Proteins/physiology , Suppressor of Cytokine Signaling Proteins/physiology , Animals , Carrier Proteins/genetics , Inflammatory Bowel Diseases/genetics , Mice , Mice, Knockout , NF-kappa B/genetics , Receptors, Antigen, T-Cell/deficiency , Receptors, Antigen, T-Cell/genetics , Repressor Proteins/genetics , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/deficiency , Suppressor of Cytokine Signaling Proteins/genetics , T-Lymphocytes/immunology
14.
Int Immunol ; 17(7): 889-97, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15908443

ABSTRACT

WSX-1 is a component of the IL-27R. Analyses of WSX-1 knockout (WSX-1(-/-)) mice have shown that IL-27/WSX-1 signaling is essential for the proper development of T(h)1 responses and that WSX-1 can suppress cellular activation and pro-inflammatory cytokine production. We have generated transgenic mouse lines over-expressing the WSX-1 gene under the control of the T cell-specific CD2 promoter (WSX-1 Tg mice). Unexpectedly, like activated CD4(+) T cells from WSX-1(-/-) mice, activated CD4(+) T cells from WSX-1 Tg mice showed increased proliferation, augmented IL-2 production and up-regulated surface expression of activation markers. IL-27-mediated tyrosine phosphorylation of STAT1 was also enhanced in WSX-1 Tg CD4(+) T cells, but STAT3 activation was normal. Exogenous IL-27 supported the proliferation of wild-type CD4(+) T cells but suppressed that of WSX-1 Tg cells. WSX-1 over-expression increased IFN-gamma production in T(h)1-polarized CD4(+) T cells, but also promoted T(h)2 cytokine production under T(h)1-polarizing conditions. Importantly, WSX-1 over-expression failed to suppress T(h)2 cytokine production under T(h)2-polarizing conditions. Cytokine hyperproduction was also observed in vivo in WSX-1 Tg mice injected with Con A. Our data suggest that WSX-1 plays a pivotal role in regulating T cell responsiveness to TCR stimulation and that the correct balance of STAT1/STAT3 activation downstream of IL-27R engagement is crucial for the physiological function of IL-27.


Subject(s)
Interleukin-2/biosynthesis , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Cytokine/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , CD2 Antigens/genetics , CD2 Antigens/immunology , Cell Proliferation/drug effects , Cells, Cultured , Concanavalin A/administration & dosage , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Interleukins/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Knockout , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Receptors, Cytokine/genetics , Receptors, Interleukin , STAT3 Transcription Factor/immunology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology
15.
Am J Pathol ; 165(5): 1499-508, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15509521

ABSTRACT

The pathogenesis of hepatitis C virus (HCV)-associated insulin resistance remains unclear. Therefore, we investigated mechanisms for HCV-associated insulin resistance. Homeostasis model assessment for insulin resistance was increased in patients with HCV infection. An increase in fasting insulin levels was associated with the presence of serum HCV core, the severity of hepatic fibrosis and a decrease in expression of insulin receptor substrate (IRS) 1 and IRS2, central molecules of the insulin-signaling cascade, in patients with HCV infection. Down-regulation of IRS1 and IRS2 was also seen in HCV core-transgenic mice livers and HCV core-transfected human hepatoma cells. Carbobenzoxy-l-leucyl-l-leucyl-l-leucinal, a potent proteosomal proteolysis inhibitor, blocked down-regulation of IRS1 and IRS2 in HCV core-transfected hepatoma cells. In human hepatoma cells, HCV core up-regulated suppressor of cytokine signaling (SOCS) 3 and caused ubiquitination of IRS1 and IRS2. HCV core-induced down-regulation of IRS1 and IRS2 was not seen in SOCS3(-/-) mouse embryonic fibroblast cells. Furthermore, HCV core suppressed insulin-induced phosphorylation of p85 subunit of phosphatidylinositol 3-kinase and Akt, activation of 6-phosphofructo-2-kinase, and glucose uptake. In conclusion, HCV infection changes a subset of hepatic molecules regulating glucose metabolism. A possible mechanism is that HCV core-induced SOCS3 promotes proteosomal degradation of IRS1 and IRS2 through ubiquitination.


Subject(s)
Down-Regulation , Hepacivirus/metabolism , Phosphoproteins/biosynthesis , Repressor Proteins/biosynthesis , Transcription Factors/biosynthesis , Up-Regulation , Adult , Animals , Cell Line, Tumor , Cells, Cultured , DNA, Complementary/metabolism , Female , Genes, Viral , Genotype , Glucose/metabolism , Humans , Hyperinsulinism/virology , Immunoblotting , Immunohistochemistry , Insulin Receptor Substrate Proteins , Insulin Resistance , Intracellular Signaling Peptides and Proteins , Liver/virology , Liver Diseases/metabolism , Liver Diseases/virology , Male , Mice , Mice, Transgenic , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Phosphofructokinase-2/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Time Factors , Transfection , Ubiquitin/metabolism
16.
J Exp Med ; 199(12): 1701-7, 2004 Jun 21.
Article in English | MEDLINE | ID: mdl-15197228

ABSTRACT

Hepatocellular carcinomas (HCCs) mainly develop from liver cirrhosis and severe liver fibrosis that are established with long-lasting inflammation of the liver. Silencing of the suppressor of the cytokine signaling-1 (SOCS1) gene, a negative regulator of cytokine signaling, by DNA methylation has been implicated in development or progress of HCC. However, how SOCS1 contributes to HCC is unknown. We examined SOCS1 gene methylation in >200 patients with chronic liver disease and found that the severity of liver fibrosis is strongly correlated with SOCS1 gene methylation. In murine liver fibrosis models using dimethylnitrosamine, mice with haploinsufficiency of the SOCS1 gene (SOCS1(-/+) mice) developed more severe liver fibrosis than did wild-type littermates (SOCS1(+/+) mice). Moreover, carcinogen-induced HCC development was also enhanced by heterozygous deletion of the SOCS1 gene. These findings suggest that SOCS1 contributes to protection against hepatic injury and fibrosis, and may also protect against hepatocarcinogenesis.


Subject(s)
Carcinoma, Hepatocellular/immunology , Carrier Proteins/physiology , Intracellular Signaling Peptides and Proteins , Liver Cirrhosis/genetics , Liver Neoplasms/immunology , Repressor Proteins/physiology , Animals , Biopsy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/prevention & control , Carrier Proteins/genetics , Choline Deficiency/genetics , Choline Deficiency/immunology , Choline Deficiency/pathology , DNA Methylation , DNA, Neoplasm/genetics , Disease Models, Animal , Gene Silencing , Humans , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/prevention & control , Mice , Mice, Knockout , Polymerase Chain Reaction/methods , Repressor Proteins/genetics , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins
17.
Oncogene ; 23(17): 3107-15, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15021916

ABSTRACT

Human papilloma viruses (HPVs) are small double-stranded DNA viruses that infect mucosal and cutaneous epithelium and induce cervical cancer. It has been shown that interferon (IFN)gamma suppresses proliferation of HPV-infected cells by suppressing expression of HPV E7. Here, we found that IFNgamma induces not only suppression of E7 transcription but also proteasome-dependent degradation. Suppressor of cytokine signaling-1 (SOCS1)/JAB, a suppressor of cytokine signaling, is known to be induced by IFNgamma, and functions as an antioncogene against various hematopoietic oncogenic proteins. SOCS1 contains the SOCS-box, which is shown to recruit ubiquitin transferase to the molecules that interact with SOCS1. We found that SOCS1 interacted with HPV E7 protein and induced ubiquitination and degradation of E7 in a SOCS-box-dependent manner. SOCS1 overexpression also increased Rb protein levels and suppressed proliferation of cervical cancer cell lines infected with HPV. Moreover, E7 protein levels were higher and Rb protein levels were lower in SOCS1-deficient fibroblasts infected with retrovirus vector carrying E7 gene than in wild-type fibroblasts. E7 induced anchorage-independent growth in SOCS1-deficient fibroblasts, but not in wild-type cells. These data suggested that SOCS1 plays an important role in regulating the levels of E7 protein and their transforming potential, and could be a new therapeutic tool for HPV-mediated tumors.


Subject(s)
Cell Transformation, Viral/physiology , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomaviridae/genetics , Uterine Cervical Neoplasms/pathology , Animals , Base Sequence , Cell Division , Cell Line, Tumor , Cysteine Endopeptidases/metabolism , DNA Primers , DNA, Intergenic/genetics , Female , Genetic Vectors , HeLa Cells/transplantation , Humans , Mice , Mice, SCID , Multienzyme Complexes/metabolism , Papillomavirus E7 Proteins , Proteasome Endopeptidase Complex , Restriction Mapping , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Transplantation, Heterologous , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...