Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Cell Rep ; 38(2): 110090, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35021075

ABSTRACT

Alpha-synuclein (αS) is causally involved in the development of Parkinson disease (PD); however, its role in normal vertebrate physiology has remained unknown. Recent studies demonstrate that αS is induced by noroviral infection in the enteric nervous system of children and protects mice against lethal neurotropic viral infection. Additionally, αS is a potent chemotactic activator of phagocytes. In this report, using both wild-type and αS knockout mice, we show that αS is a critical mediator of inflammatory and immune responses. αS is required for the development of a normal inflammatory response to bacterial peptidoglycan introduced into the peritoneal cavity as well as antigen-specific and T cell responses following intraperitoneal immunization. Furthermore, we show that neural cells are the sources of αS required for immune competence. Our report supports the hypothesis that αS accumulates within the nervous system of PD individuals because of an inflammatory/immune response.


Subject(s)
Immunity/physiology , alpha-Synuclein/metabolism , alpha-Synuclein/physiology , Animals , Brain/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System/metabolism , Neurons/metabolism , Toll-Like Receptor 4/immunology , alpha-Synuclein/genetics
2.
Cancer Res ; 81(12): 3374-3386, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33837043

ABSTRACT

Screening for sensitizers of cancer cells to TRAIL-mediated apoptosis identified a natural product of the 17ß-hydroxywithanolide (17-BHW) class, physachenolide C (PCC), as a promising hit. In this study, we show that PCC was also able to sensitize melanoma and renal carcinoma cells to apoptosis in response not only to TRAIL, but also to the synthetic polynucleotide poly I:C, a viral mimetic and immune activator, by reducing levels of antiapoptotic proteins cFLIP and Livin. Both death receptor and TLR3 signaling elicited subsequent increased assembly of a proapoptotic ripoptosome signaling complex. Administration of a combination of PCC and poly I:C in human M14 melanoma xenograft and a syngeneic B16 melanoma model provided significant therapeutic benefit as compared with individual agents. In addition, PCC enhanced melanoma cell death in response to activated human T cells in vitro and in vivo in a death ligand-dependent manner. Biochemical mechanism-of-action studies established bromo and extraterminal domain (BET) proteins as major cellular targets of PCC. Thus, by targeting of BET proteins to reduce antiapoptotic proteins and enhance caspase-8-dependent apoptosis of cancer cells, PCC represents a unique agent that can potentially be used in combination with various immunotherapeutic approaches to promote tumor regression and improve outcome. SIGNIFICANCE: These findings demonstrate that PCC selectively sensitizes cancer cells to immune-mediated cell death, potentially improving the efficacy of cancer immunotherapies. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/12/3374/F1.large.jpg.


Subject(s)
Biological Products/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Immunotherapy/methods , Melanoma, Experimental/drug therapy , Poly I-C/pharmacology , Transcription Factors/antagonists & inhibitors , Withanolides/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Cell Proliferation , Drug Therapy, Combination , Female , Humans , Interferon Inducers/pharmacology , Kidney Neoplasms/drug therapy , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
J Vis Exp ; (157)2020 03 20.
Article in English | MEDLINE | ID: mdl-32250353

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with limited therapeutic options. When compared to patients with less aggressive breast tumors, the 5-year survival rate of TNBC patients is 77% due to their characteristic drug-resistant phenotype and metastatic burden. Toward this end, murine models have been established aimed at identifying novel therapeutic strategies limiting TNBC tumor growth and metastatic spread. This work describes a practical guide for the TNBC orthotopic model where MDA-MB-231 breast cancer cells suspended in a basement membrane matrix are implanted in the fourth mammary fat pad, which closely mimics the cancer cell behavior in humans. Measurement of tumors by caliper, lung metastasis assessment via in vivo and ex vivo imaging, and molecular detection are discussed. This model provides an excellent platform to study therapeutic efficacy and is especially suitable for the study of the interaction between the primary tumor and distal metastatic sites.


Subject(s)
Triple Negative Breast Neoplasms/pathology , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Lung Neoplasms/secondary , Mice , Phenotype , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Xenograft Model Antitumor Assays
4.
Oncotarget ; 9(32): 22605-22617, 2018 Apr 27.
Article in English | MEDLINE | ID: mdl-29854301

ABSTRACT

Acute lymphoblastic leukemia (ALL) is the most common cancer in children. Current chemotherapy is quite toxic in growing children and more directed therapeutics are being sought. The IL-7R pathway is a major driver of ALL and here we evaluate two drugs directed to that pathway using a model of T cell ALL. Mutant gain-of-function IL-7Rα was transduced into an IL-7-dependent murine thymocyte line conferring ligand-independent survival and growth. JAK1 is associated with IL-7Rα and mediates signaling from the mutant receptor. In vitro, treating the transformed cell line with the JAK1/2 inhibitor ruxolitinib inhibited ligand-independent signaling and induced cell death. Transfer of the transformed cell line into mice resulted in aggressive leukemia and untreated mice succumbed in about three weeks. Treatment with ruxolitinib incorporated into chow showed a potent therapeutic benefit with reduction in leukemic burden and extension of survival. BCL-2 is an anti-apoptotic downstream mediator of the IL-7R survival mechanism. Venetoclax, an inhibitor of BCL-2, showed activity against the transformed cell line in vitro and could be combined with ruxolitinib in vivo. These findings support the therapeutic potential of treating T-ALL by targeting the IL-7R pathway.

5.
Gut ; 65(10): 1765-75, 2016 10.
Article in English | MEDLINE | ID: mdl-26206664

ABSTRACT

OBJECTIVES: The relative contributions of inflammatory signalling and sequential oncogenic dysregulation driving liver cancer pathogenesis remain incompletely understood. Lymphotoxin-ß receptor (LTßR) signalling is critically involved in hepatitis and liver tumorigenesis. Therefore, we explored the interdependence of inflammatory lymphotoxin signalling and specific oncogenic pathways in the progression of hepatic cancer. DESIGN: Pathologically distinct liver tumours were initiated by hydrodynamic transfection of oncogenic V-Akt Murine Thymoma Viral Oncogene Homolog 1 (AKT)/ß-catenin or AKT/Notch expressing plasmids. To investigate the relationship of LTßR signalling and specific oncogenic pathways, LTßR antagonist (LTßR-Fc) or agonist (anti-LTßR) were administered post oncogene transfection. Initiated livers/tumours were investigated for changes in oncogene expression, tumour proliferation, progression, latency and pathology. Moreover, specific LTßR-mediated molecular events were investigated in human liver cancer cell lines and through transcriptional analyses of samples from patients with intrahepatic cholangiocarcinoma (ICC). RESULTS: AKT/ß-catenin-transfected livers displayed increased expression of LTß and LTßR, with antagonism of LTßR signalling reducing tumour progression and enhancing survival. Conversely, enforced LTßR-activation of AKT/ß-catenin-initiated tumours induced robust increases in proliferation and progression of hepatic tumour phenotypes in an AKT-dependent manner. LTßR-activation also rapidly accelerated ICC progression initiated by AKT/Notch, but not Notch alone. Moreover, LTßR-accelerated development coincides with increases of Notch, Hes1, c-MYC, pAKT and ß-catenin. We further demonstrate LTßR signalling in human liver cancer cell lines to be a regulator of Notch, pAKTser473 and ß-catenin. Transcriptome analysis of samples from patients with ICC links increased LTßR network expression with poor patient survival, increased Notch1 expression and Notch and AKT/PI3K signalling. CONCLUSIONS: Our findings link LTßR and oncogenic AKT signalling in the development of ICC.


Subject(s)
Carcinogenesis/metabolism , Cholangiocarcinoma , Liver Neoplasms , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-beta/metabolism , Signal Transduction/physiology , Animals , Cell Proliferation/physiology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Disease Progression , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Statistics as Topic
6.
J Hepatol ; 63(5): 1181-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26143441

ABSTRACT

BACKGROUND & AIMS: Liver inflammatory diseases associated with cancer promoting somatic oncogene mutations are increasing in frequency. Preclinical cancer models that allow for the study of early tumor progression are often protracted, which limits the experimental study parameters due to time and expense. Here we report a robust inexpensive approach using Sleeping Beauty transposition (SBT) delivery of oncogenes along with Gaussia Luciferase expression vector GLuc, to assess de novo liver tumor progression, as well as the detection of innate immune responses or responses induced by therapeutic intervention. METHODS: Tracking de novo liver tumor progression with GLuc was demonstrated in models of hepatocellular carcinoma (HCC) or adenoma (HCA) initiated by hydrodynamic delivery of SBT oncogenes. RESULTS: Rising serum luciferase levels correlated directly with increasing liver tumor burden and eventual morbidity. Early detection of hepatocyte apoptosis from mice with MET+CAT transfected hepatocytes was associated with a transient delay in HCC growth mediated by a CD8(+) T-cell response against transformed hepatocytes. Furthermore, mice that lack B cells or macrophages had an increase in TUNEL(+) hepatocytes following liver MET transfection demonstrating that these cells provide protection from MET-induced hepatocyte apoptosis. Treatment with IL-18+IL-12 of mice displaying established HCC decreased tumor burden which was associated with decreased levels of serum luciferase. CONCLUSIONS: Hydrodynamic delivery of the SBT vector GLuc to hepatocytes serves as a simple blood-based approach for real-time tracking of pathologically distinct types of liver cancer. This revealed tumor-induced immunologic responses and was beneficial in monitoring the efficacy of therapeutic interventions.


Subject(s)
Adenoma, Liver Cell/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Hepatocellular/immunology , Immunity, Cellular , Liver Neoplasms, Experimental/immunology , Luciferases/blood , Recombinant Proteins/therapeutic use , Adenoma, Liver Cell/drug therapy , Adenoma, Liver Cell/pathology , Animals , Apoptosis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Disease Progression , Hepatocytes/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Interleukin-12/therapeutic use , Interleukin-18/therapeutic use , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/pathology , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Signal Transduction
7.
Science ; 342(6161): 967-70, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24264989

ABSTRACT

The gut microbiota influences both local and systemic inflammation. Inflammation contributes to development, progression, and treatment of cancer, but it remains unclear whether commensal bacteria affect inflammation in the sterile tumor microenvironment. Here, we show that disruption of the microbiota impairs the response of subcutaneous tumors to CpG-oligonucleotide immunotherapy and platinum chemotherapy. In antibiotics-treated or germ-free mice, tumor-infiltrating myeloid-derived cells responded poorly to therapy, resulting in lower cytokine production and tumor necrosis after CpG-oligonucleotide treatment and deficient production of reactive oxygen species and cytotoxicity after chemotherapy. Thus, optimal responses to cancer therapy require an intact commensal microbiota that mediates its effects by modulating myeloid-derived cell functions in the tumor microenvironment. These findings underscore the importance of the microbiota in the outcome of disease treatment.


Subject(s)
Intestines/microbiology , Microbiota/physiology , Neoplasms/immunology , Neoplasms/therapy , Tumor Microenvironment/immunology , Animals , Anti-Bacterial Agents/administration & dosage , Antigen Presentation/genetics , Antineoplastic Agents/therapeutic use , Bacteria/drug effects , Bacterial Physiological Phenomena/drug effects , Down-Regulation , Gene Expression Regulation , Germ-Free Life , Immunotherapy , Inflammation/genetics , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Microbiota/drug effects , Neoplasm Transplantation , Neoplasms/microbiology , Oligodeoxyribonucleotides/therapeutic use , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Phagocytosis/genetics , Reactive Oxygen Species/metabolism , Symbiosis , Tumor Necrosis Factor-alpha/metabolism
8.
Cancer Cell ; 23(4): 527-40, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23597566

ABSTRACT

Here, we report that kinase-dead IKKα knockin mice develop spontaneous lung squamous cell carcinomas (SCCs) associated with IKKα downregulation and marked pulmonary inflammation. IKKα reduction upregulated the expression of p63, Trim29, and keratin 5 (K5), which serve as diagnostic markers for human lung SCCs. IKKα(low)K5(+)p63(hi) cell expansion and SCC formation were accompanied by inflammation-associated deregulation of oncogenes, tumor suppressors, and stem cell regulators. Reintroducing transgenic K5.IKKα, depleting macrophages, and reconstituting irradiated mutant animals with wild-type bone marrow (BM) prevented SCC development, suggesting that BM-derived IKKα mutant macrophages promote the transition of IKKα(low)K5(+)p63(hi) cells to tumor cells. This mouse model resembles human lung SCCs, sheds light on the mechanisms underlying lung malignancy development, and identifies targets for therapy of lung SCCs.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Cell Transformation, Neoplastic/metabolism , I-kappa B Kinase/physiology , Lung Neoplasms/enzymology , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Phosphoproteins/biosynthesis , Trans-Activators/biosynthesis , Transcription Factors/biosynthesis
9.
Int J Cancer ; 132(4): 785-94, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-22821831

ABSTRACT

MIF is a proinflammatory cytokine and is implicated in cancer. A higher MIF level is found in many human cancer and cancer-prone inflammatory diseases, including chronic pancreatitis and pancreatic cancer. We tested the hypothesis that MIF contributes to pancreatic cancer aggressiveness and predicts disease outcome in resected cases. Consistent with our hypothesis we found that an elevated MIF mRNA expression in tumors was significantly associated with poor outcome in resected cases. Multivariate Cox-regression analysis further showed that MIF is independently associated with patients' survival (HR = 2.26, 95% CI = 1.17-4.37, p = 0.015). Mechanistic analyses revealed that MIF overexpression decreased E-cadherin and increased vimentin mRNA and protein levels in pancreatic cancer cell lines, consistent with the features of epithelial-to-mesenchymal transition (EMT). Furthermore, MIF-overexpression significantly increased ZEB1/2 and decreased miR-200b expression, while shRNA-mediated inhibition of MIF increased E-cadherin and miR-200b expression, and reduced the expression of ZEB1/2 in Panc1 cells. Re-expression of miR-200b in MIF overexpressing cells restored the epithelial characteristics, as indicated by an increase in E-cadherin and decrease in ZEB1/2 and vimentin expression. A reduced sensitivity to the chemotherapeutic drug, gemcitabine, occurred in MIF-overexpressing cells. Indicative of an increased malignant potential, MIF over-expressing cells showed significant increase in their invasion ability in vitro, and tumor growth and metastasis in an orthotopic xenograft mouse model. These results support a role of MIF in disease aggressiveness, indicating its potential usefulness as a candidate target for designing improved treatment in pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Epithelial-Mesenchymal Transition/genetics , Macrophage Migration-Inhibitory Factors/genetics , Pancreatic Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Macrophage Migration-Inhibitory Factors/metabolism , Male , Mice, Nude , MicroRNAs/genetics , Middle Aged , Neoplasm Invasiveness , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prognosis , RNA Interference , Transplantation, Heterologous , Gemcitabine
10.
Cancer Invest ; 30(5): 343-63, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22571338

ABSTRACT

Using two MYCN transgenic mouse strains, we established 10 transplantable neuroblastoma cell lines via serial orthotopic passage in the adrenal gland. Tissue arrays demonstrate that by histochemistry, vascularity, immunohistochemical staining for neuroblastoma markers, catecholamine analysis, and concurrent cDNA microarray analysis, there is a close correspondence between the transplantable lines and the spontaneous tumors. Several genes closely associated with the pathobiology and immune evasion of neuroblastoma, novel targets that warrant evaluation, and decreased expression of tumor suppressor genes are demonstrated. These studies describe a unique and generalizable approach to expand the utility of transgenic models of spontaneous tumor, providing new tools for preclinical investigation.


Subject(s)
Drug Discovery , Gene Expression Profiling , Neuroblastoma/pathology , Animals , Apoptosis , Cell Line, Tumor , Cyclin-Dependent Kinase 4/analysis , Genes, Tumor Suppressor , High-Throughput Screening Assays , Humans , Mice , Mice, Inbred C57BL , N-Myc Proto-Oncogene Protein , Neoplasm Transplantation , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/ultrastructure , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Principal Component Analysis , Tissue Array Analysis
11.
PLoS One ; 7(3): e33303, 2012.
Article in English | MEDLINE | ID: mdl-22428016

ABSTRACT

The liver is an immunologically unique organ containing tolerogenic dendritic cells (DC) that maintain an immunosuppressive microenvironment. Although systemic IL-12 administration can improve responses to tumors, the effects of IL-12-based treatments on DC, in particular hepatic DC, remain incompletely understood. In this study, we demonstrate systemic IL-12 administration induces a 2-3 fold increase in conventional, but not plasmacytoid, DC subsets in the liver. Following IL-12 administration, hepatic DC became more phenotypically and functionally mature, resembling the function of splenic DC, but differed as compared to their splenic counterparts in the production of IL-12 following co-stimulation with toll-like receptor (TLR) agonists. Hepatic DCs from IL-12 treated mice acquired enhanced T cell proliferative capabilities similar to levels observed using splenic DCs. Furthermore, IL-12 administration preferentially increased hepatic T cell activation and IFNγ expression in the RENCA mouse model of renal cell carcinoma. Collectively, the data shows systemic IL-12 administration enables hepatic DCs to overcome at least some aspects of the inherently suppressive milieu of the hepatic environment that could have important implications for the design of IL-12-based immunotherapeutic strategies targeting hepatic malignancies and infections.


Subject(s)
Carcinoma, Renal Cell/immunology , Dendritic Cells/drug effects , Interleukin-12/pharmacology , Liver/immunology , Animals , Dendritic Cells/immunology , Flow Cytometry , Interferon-gamma/metabolism , Interleukin-12/administration & dosage , Liver/cytology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Statistics, Nonparametric , T-Lymphocytes/immunology
12.
Cancer Res ; 71(12): 4074-84, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21540234

ABSTRACT

mTOR is a central mediator of cancer cell growth, but it also directs immune cell differentiation and function. On this basis, we had explored the hypothesis that mTOR inhibition can enhance cancer immunotherapy. Here, we report that a combination of αCD40 agonistic antibody and the ATP-competitive mTOR kinase inhibitory drug AZD8055 elicited synergistic antitumor responses in a model of metastatic renal cell carcinoma. In contrast to the well-established mTOR inhibitor rapamycin, AZD8055 increased the infiltration, activation, and proliferation of CD8(+) T cells and natural killer cells in liver metastatic foci when combined with the CD40 agonist. AZD8055/αCD40-treated mice also display an increased incidence of matured macrophages and dendritic cells compared with that achieved in mice by αCD40 or AZD8055 treatment alone. We found that the combination treatment also increased macrophage production of TNFα, which played an indispensable role in activation of the observed antitumor immune response. Levels of Th1 cytokines, including interleukin 12, IFN-γ, TNFα, and the Th1-associated chemokines RANTES, MIG, and IP-10 were each elevated significantly in the livers of mice treated with the combinatorial therapy versus individual treatments. Notably, the AZD8055/αCD40-induced antitumor response was abolished in IFN-γ(-/-) and CD40(-/-) mice, establishing the reliance of the combination therapy on host IFN-γ and CD40 expression. Our findings offer a preclinical proof of concept that, unlike rapamycin, the ATP-competitive mTOR kinase inhibitor AZD8055 can contribute with αCD40 treatment to trigger a restructuring of the tumor immune microenvironment to trigger regressions of an established metastatic cancer.


Subject(s)
Antibodies/pharmacology , Antineoplastic Agents/pharmacology , CD40 Antigens/agonists , Morpholines/pharmacology , Neoplasms, Experimental/drug therapy , Neoplasms/therapy , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , CD40 Antigens/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/drug effects , Humans , Immunotherapy , Interferon-gamma/physiology , Interleukin-12/biosynthesis , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Macrophage Activation/drug effects , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Sirolimus/pharmacology
13.
Cancer Res ; 71(7): 2718-27, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21324921

ABSTRACT

Obesity is a risk factor for development of certain cancers but the basis for this risk is unclear. In this study, we developed a novel mouse model that demonstrates directly how lipogenic phenotypes commonly associated with diet-induced metabolic syndromes can influence hepatic cancer development. Activated AKT and ß-catenin (AKT/CAT) genes were hydrodynamically codelivered using the Sleeping Beauty transposon to initiate liver tumorigenesis. AKT/CAT and MET/CAT combination induced microscopic tumor foci by 4 weeks, whereas no tumorigenesis resulted from delivery of AKT, MET, or CAT alone. Primary AKT/CAT tumor cells were steatotic (fatty) hepatocellular adenomas which progressed to hepatocellular carcinomas (HCC) upon in vivo passage, whereas primary MET/CAT tumors emerged directly as frank HCC. Conversion of AKT/CAT tumor cells to frank HCC during passage was associated with induction of the human HCC marker α-fetoprotein and the stem cell marker CD133. Using hierarchical clustering and gene set enrichment analysis, we compared the primary murine AKT/CAT and MET/CAT tumors to a panel of 53 human HCCs and determined that these two mouse models could be stratified as distinct subtypes associated in humans with poor clinical prognosis. The chief molecular networks identified in primary and passaged AKT/CAT tumors were steatosis and lipid metabolic pathways, respectively. Our findings show how coactivation of the AKT and CAT pathways in hepatocytes can efficiently model development of a lipogenic tumor phenotype. Furthermore, we believe that our approach could speed the dissection of microenvironmental factors responsible for driving steatotic-neoplastic transformation to frank carcinoma, through genetic modification of existing immunodefined transgenic models.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic/metabolism , Liver Neoplasms/metabolism , Oncogene Protein v-akt/metabolism , beta Catenin/metabolism , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , DNA Transposable Elements , Enzyme Activation , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice , Oncogenes , Proto-Oncogene Proteins c-met/metabolism
14.
J Immunol ; 186(2): 838-47, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21148802

ABSTRACT

The fate of invariant NKT (iNKT) cells following activation remains controversial and unclear. We systemically examined how iNKT cells are regulated following TCR-dependent and -independent activation with α-galactosylceramide (αGC) or IL-18 plus IL-12, respectively. Our studies reveal activation by αGC or IL-18 plus IL-12 induced transient depletion of iNKT cells exclusively in the liver that was independent of caspase 3-mediated apoptosis. The loss of iNKT cells was followed by repopulation and expansion of phenotypically distinct cells via different mechanisms. Liver iNKT cell expansion following αGC, but not IL-18 plus IL-12, treatment required an intact spleen and IFN-γ. Additionally, IL-18 plus IL-12 induced a more prolonged expansion of liver iNKT cells compared with αGC. iNKT cells that repopulate the liver following αGC had higher levels of suppressive receptors PD-1 and Lag3, whereas those that repopulate the liver following IL-18 plus IL-12 had increased levels of TCR and ICOS. In contrast to acute treatment that caused a transient loss of iNKT cells, chronic αGC or IL-18 plus IL-12 treatment caused long-term systemic loss requiring an intact thymus for repopulation of the liver. This report reveals a previously undefined role for the liver in the depletion of activated iNKT cells. Additionally, TCR-dependent and -independent activation differentially regulate iNKT cell distribution and phenotype. These results provide new insights for understanding how iNKT cells are systemically regulated following activation.


Subject(s)
Cell Differentiation/immunology , Liver/immunology , Liver/metabolism , Lymphocyte Activation/immunology , Lymphocyte Depletion , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell/physiology , Animals , Caspase 3/metabolism , Galactosylceramides/physiology , Immunophenotyping , Interleukin-12/physiology , Interleukin-18/physiology , Liver/cytology , Lymphocyte Depletion/methods , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , Spleen/cytology , Spleen/immunology , Spleen/metabolism
15.
Proc Natl Acad Sci U S A ; 106(46): 19455-60, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19892741

ABSTRACT

Treatment of mice bearing orthotopic, metastatic tumors with anti-CD40 antibody resulted in only partial, transient anti-tumor effects whereas combined treatment with IL-2/anti-CD40, induced tumor regression. The mechanisms for these divergent anti-tumor responses were examined by profiling tumor-infiltrating leukocyte subsets and chemokine expression within the tumor microenvironment after immunotherapy. IL-2/anti-CD40, but not anti-CD40 alone, induced significant infiltration of established tumors by NK and CD8(+) T cells. To further define the role of chemokines in leukocyte recruitment into tumors, we evaluated anti-tumor responses in mice lacking the chemokine receptor, CCR2. The anti-tumor effects and leukocyte recruitment mediated by anti-CD40 alone, were completely abolished in CCR2(-/-) mice. In contrast, IL-2/anti-CD40-mediated leukocyte recruitment and reductions in primary tumors and metastases were maintained in CCR2(-/-) mice. Treatment of mice with IL-2/anti-CD40, but not anti-CD40 alone, also caused an IFN-gamma-dependent increase in the expression of multiple Th1 chemokines within the tumor microenvironment. Interestingly, although IL-2/anti-CD40 treatment increased Tregs in the spleen, it also caused a coincident IFN-gamma-dependent reduction in CD4(+)/FoxP3(+) Tregs, myeloid-derived suppressor cells and Th2 chemokine expression specifically within the tumor microenvironment that was not observed after treatment with anti-CD40 alone. Similar effects were observed using IL-15 in combination with anti-CD40. Taken together, our data demonstrate that IL-2/anti-CD40, but not anti-CD40 alone, can preferentially reduce the overall immunosuppressive milieu within the tumor microenvironment. These results suggest that the use of anti-CD40 in combination with IL-2 or IL-15 may hold substantially more promise for clinical cancer treatment than anti-CD40 alone.


Subject(s)
Antibodies/therapeutic use , CD40 Antigens/agonists , Immunosuppression Therapy/methods , Interleukin-2/therapeutic use , Neoplasms/therapy , Animals , Arginase/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Chemokine CCL5/biosynthesis , Chemokine CXCL9/biosynthesis , Chemokines/biosynthesis , Chemokines, CC/biosynthesis , Drug Synergism , Lymphocytes, Tumor-Infiltrating/immunology , Macrophage Inflammatory Proteins/biosynthesis , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Receptors, CCR2/biosynthesis , Receptors, CCR2/genetics , Receptors, Cytokine/biosynthesis
16.
J Immunol ; 182(7): 4328-38, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19299733

ABSTRACT

IL-27 exerts antitumor activity in murine orthotopic neuroblastoma, but only partial antitumor effect in disseminated disease. This study demonstrates that combined treatment with IL-2 and IL-27 induces potent antitumor activity in disseminated neuroblastoma metastasis. Complete durable tumor regression was achieved in 90% of mice bearing metastatic TBJ-IL-27 tumors treated with IL-2 compared with only 40% of mice bearing TBJ-IL-27 tumors alone and 0% of mice bearing TBJ-FLAG tumors with or without IL-2 treatment. Comparable antitumor effects were achieved by IL-27 protein produced upon hydrodynamic IL-27 plasmid DNA delivery when combined with IL-2. Although delivery of IL-27 alone, or in combination with IL-2, mediated pronounced regression of neuroblastoma metastases in the liver, combined delivery of IL-27 and IL-2 was far more effective than IL-27 alone against bone marrow metastases. Combined exposure to IL-27 produced by tumor and IL-2 synergistically enhances the generation of tumor-specific CTL reactivity. Potentiation of CTL reactivity by IL-27 occurs via mechanisms that appear to be engaged during both the initial sensitization and effector phase. Potent immunologic memory responses are generated in mice cured of their disseminated disease by combined delivery of IL-27 and IL-2, and depletion of CD8(+) ablates the antitumor efficacy of this combination. Moreover, IL-27 delivery can inhibit the expansion of CD4(+)CD25(+)Foxp3(+) regulatory and IL-17-expressing CD4(+) cells that are otherwise observed among tumor-infiltrating lymphocytes from mice treated with IL-2. These studies demonstrate that IL-27 and IL-2 synergistically induce complete tumor regression and long-term survival in mice bearing widely metastatic neuroblastoma tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/immunology , Interleukin-2/immunology , Interleukins/immunology , Lymphocyte Activation/drug effects , Neuroblastoma/immunology , T-Lymphocytes, Cytotoxic/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bone Marrow Neoplasms/drug therapy , Bone Marrow Neoplasms/secondary , Drug Synergism , Flow Cytometry , Interferon-gamma/immunology , Interleukin-2/administration & dosage , Interleukins/administration & dosage , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Neuroblastoma/drug therapy , Neuroblastoma/secondary , T-Lymphocytes, Cytotoxic/immunology
17.
Cancer Res ; 68(17): 7130-6, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18757428

ABSTRACT

Nitric oxide (NO(*)), an important signaling molecule and a component of inflammatory response, is involved in tumorigenesis. However, the quantity of NO(*) and the cellular microenvironment influences the role of NO(*) in tumor development. We used a genetic strategy to test the hypothesis that an inflammatory microenvironment with an enhanced level of NO(*) accelerates spontaneous tumor development. C. parvum-induced inflammation and increased NO(*) synthase-2 (NOS2) expression coincided with accelerated spontaneous tumor development, mostly lymphomas, in p53-/-NOS2+/+ C57BL6 mice when compared with the controls (P = 0.001). However, p53-/-NOS2-/- mice did not show any difference in tumor latency between C. parvum-treated and control groups. In C. parvum-treated p53-/-NOS2+/+ mice, tumor development was preceded by a higher expression of NOS2 and phosphorylated Akt-Ser(473) (pAkt-Ser473) in spleen, increased cell proliferation measured by Ki-67 IHC in spleen and thymus, and a lower apoptotic index and CD95-L expression in spleen and thymus. C. parvum-treated p53-/-NOS2+/+ mice showed an increase in the number of Foxp3(+) T-reg cells, dendritic cells (DC), as well as increased CD80(+), CD86(+), CD40(+), and CD83(+) on DC in the spleen. Regulatory T-cells (T-reg) and the maturation of DC may modulate tumorigenesis. An increase in the FoxP3(+)T-reg cells in C. parvum-treated p53-/-NOS2+/+ mice indicates a role of NO(*) in the regulation of T-reg cells that may contribute to a protumor shift of the immune environment favoring an accelerated tumor development. These data provide genetic and mechanistic evidence that an inflammatory microenvironment and an increased level of NO(*) can accelerate tumor development.


Subject(s)
Inflammation/pathology , Neoplasms, Experimental/pathology , Nitric Oxide/physiology , Animals , Apoptosis , Flow Cytometry , Immunohistochemistry , Interferon-gamma/metabolism , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
J Immunol ; 180(7): 4495-506, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18354171

ABSTRACT

Innate immune responses provide the host with its first line of defense against infections. Signals generated by subsets of lymphocytes, including NK cells, NKT cells, and APC during this early host response determine the nature of downstream adaptive immune responses. In the present study, we have examined the role of innate NK cells in an autoimmune model through the use of primary immunization with the myelin oligodendrocyte glycoprotein peptide to induce experimental autoimmune encephalomyelitis (EAE). Our studies have shown that in vivo depletion of NK cells can affect the adaptive immune responses, because NK cells were found to regulate the degree of clinical paralysis and to alter immune adaptive responses to the myelin oligodendrocyte glycoprotein peptide. The requirement for NK cells was reflected by changes in the T cell responses and diminished clinical disease seen in mice treated with anti-NK1.1, anti-asialo GM1, and selected Ly49 subtype-depleted mice. In addition to alteration in T cell responses, the maturational status of dendritic cells in lymph nodes was altered both quantitatively and qualitatively. Finally, examination of TCR Vbeta usage of the brain lymphocytes from EAE mice indicated a spectra-type change in receptor expression in NK- depleted mice as compared with non-NK-depleted EAE mice. These findings further establish a recently postulated link between NK cells and the generation of autoreactive T cells.


Subject(s)
Adaptation, Physiological/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Killer Cells, Natural/immunology , Animals , Antigens, Ly/immunology , Cells, Cultured , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Lectins, C-Type/immunology , Mice , Mice, Inbred C57BL , Myelin Proteins , Myelin-Associated Glycoprotein/immunology , Myelin-Associated Glycoprotein/pharmacology , Myelin-Oligodendrocyte Glycoprotein , Receptors, Antigen, T-Cell/immunology , Receptors, NK Cell Lectin-Like
19.
Cell Immunol ; 249(1): 8-19, 2007 Sep.
Article in English | MEDLINE | ID: mdl-18039542

ABSTRACT

Analysis of the NK cell developmental pathway suggests that CD2 expression may be important in regulating NK maturation. To test this hypothesis, we developed mice containing only an inhibitory CD2 molecule by linking the extracellular domain of CD2 to an intracellular immunoreceptor tyrosine-based inhibitory motif (ITIM) motif. Mice containing the CD2 Tg(ITIM) transgene, introduced into a CD2 KO background, have no morphologically detectable lymph nodes, although development of the thymus appears normal. In addition, these mice had major loss of both NK and NKT subsets in peripheral organs, while T and B cell frequencies were intact. Expression of CD2 was low on T cells and lacking on B cells and functional defects were observed in these populations. NKT cells expressing CD4 were absent, while the CD8+ and double negative NKT cells were retained. Small subsets of NK cells were detected but expression of CD2 on these cells was very low or absent, and their maturation was impaired. Based on the phenotype described here, we believe that these mice represent a unique model to study lymphoid organ and lymphocyte development.


Subject(s)
CD2 Antigens/immunology , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Thymus Gland/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD2 Antigens/chemistry , CD2 Antigens/genetics , CD2 Antigens/metabolism , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Cell Differentiation , Humans , Killer Cells, Natural/cytology , Lymphocyte Count , Lymphocyte Subsets/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Structure, Tertiary , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tyrosine
20.
Nat Med ; 13(3): 354-60, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17334371

ABSTRACT

Protective cell-mediated immune responses in cancer are critically dependent on T-helper type 1 (T(H)1) cytokines such as interferon-gamma (IFN-gamma). We have previously shown that the combination of CD40 stimulation and interleukin-2 (IL-2) leads to synergistic antitumor responses in several models of advanced metastatic disease. We now report that after this treatment and other immunotherapy regimens, the CD4+ T-cell population, in contrast to CD8+ T cells, did not significantly increase but rather exhibited a substantial level of apoptosis that was dependent on IFN-gamma. Mice immunized with tumor cells and treated with an immunotherapy regimen that was initially protective were later unable to mount effective memory responses compared with immunized mice not receiving immunotherapy. Immunotherapy given to tumor-bearing Ifngr-/- mice resulted in restoration of secondary responses. Thus, although immunotherapeutic regimens inducing strong IFN-gamma responses can lead to successful early antitumor efficacy, they may also impair the development of durable antitumor responses.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Carcinoma, Renal Cell/immunology , Immunologic Memory , Immunotherapy, Active , Interferon-gamma/physiology , Kidney Neoplasms/immunology , Melanoma, Experimental/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/prevention & control , Cell Line , Cells, Cultured , Clonal Deletion/genetics , Clonal Deletion/immunology , Female , Immunologic Memory/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/prevention & control , Melanoma, Experimental/pathology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL