Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Mol Cancer Ther ; 20(8): 1481-1494, 2021 08.
Article in English | MEDLINE | ID: mdl-34045231

ABSTRACT

Oncolytic vaccinia viruses have promising efficacy and safety profiles in cancer therapy. Although antitumor activity can be increased by manipulating viral genes, the relative efficacy of individual modifications has been difficult to assess without side-by-side comparisons. This study sought to compare the initial antitumor activity after intravenous administration of five vaccinia virus variants of the same Western Reserve backbone and thymidine kinase gene deletion in RIP-Tag2 transgenic mice with spontaneous pancreatic neuroendocrine tumors. Tumors had focal regions of infection at 5 days after all viruses. Natural killer (NK) cells were restricted to these sites of infection, but CD8+ T cells and tumor cell apoptosis were widespread and varied among the viruses. Antitumor activity of virus VV-A34, bearing amino acid substitution A34K151E to increase viral spreading, and virus VV-IL2v, expressing a mouse IL2 variant (mIL2v) with attenuated IL2 receptor alpha subunit binding, was similar to control virus VV-GFP. However, antitumor activity was significantly greater after virus VV-A34/IL2v, which expressed mIL2v together with A34K151E mutation and viral B18R gene deletion, and virus VV-GMCSF that expressed mouse GM-CSF. Both viruses greatly increased expression of CD8 antigens Cd8a/Cd8b1 and cytotoxicity genes granzyme A, granzyme B, Fas ligand, and perforin-1 in tumors. VV-A34/IL2v led to higher serum IL2 and greater tumor expression of death receptor ligand TRAIL, but VV-GMCSF led to higher serum GM-CSF, greater expression of leukocyte chemokines and adhesion molecules, and more neutrophil recruitment. Together, the results show that antitumor activity is similarly increased by viral expression of GM-CSF or IL2v combined with additional genetic modifications.


Subject(s)
Apoptosis , Cytokines/metabolism , Immunity , Neuroendocrine Tumors/therapy , Oncolytic Virotherapy/methods , Pancreatic Neoplasms/therapy , Vaccinia virus/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Female , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Interleukin-2/genetics , Interleukin-2/immunology , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/virology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/virology , Tumor Cells, Cultured
3.
Trends Cell Biol ; 26(7): 486-497, 2016 07.
Article in English | MEDLINE | ID: mdl-27056543

ABSTRACT

Atherosclerosis, cancer, and various chronic fibrotic conditions are characterized by an increase in the migratory behavior of resident cells and the enhanced invasion of assorted exogenous cells across a stiffened extracellular matrix (ECM). This stiffened scaffold aberrantly engages cellular mechanosignaling networks in cells, which promotes the assembly of invadosomes and lamellae for cell invasion and migration. Accordingly, deciphering the conserved molecular mechanisms whereby matrix stiffness fosters invadosome and lamella formation could identify therapeutic targets to treat fibrotic conditions, and reducing ECM stiffness could ameliorate disease progression.


Subject(s)
Cell Movement , Disease , Neoplasm Invasiveness/pathology , Animals , Biomechanical Phenomena , Extracellular Matrix/metabolism , Humans , Podosomes/pathology
4.
Nat Med ; 22(5): 497-505, 2016 05.
Article in English | MEDLINE | ID: mdl-27089513

ABSTRACT

Fibrosis compromises pancreatic ductal carcinoma (PDAC) treatment and contributes to patient mortality, yet antistromal therapies are controversial. We found that human PDACs with impaired epithelial transforming growth factor-ß (TGF-ß) signaling have high epithelial STAT3 activity and develop stiff, matricellular-enriched fibrosis associated with high epithelial tension and shorter patient survival. In several KRAS-driven mouse models, both the loss of TGF-ß signaling and elevated ß1-integrin mechanosignaling engaged a positive feedback loop whereby STAT3 signaling promotes tumor progression by increasing matricellular fibrosis and tissue tension. In contrast, epithelial STAT3 ablation attenuated tumor progression by reducing the stromal stiffening and epithelial contractility induced by loss of TGF-ß signaling. In PDAC patient biopsies, higher matricellular protein and activated STAT3 were associated with SMAD4 mutation and shorter survival. The findings implicate epithelial tension and matricellular fibrosis in the aggressiveness of SMAD4 mutant pancreatic tumors and highlight STAT3 and mechanics as key drivers of this phenotype.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Extracellular Matrix/metabolism , Integrin beta Chains/metabolism , Pancreatic Neoplasms/genetics , STAT3 Transcription Factor/metabolism , Transforming Growth Factor beta/metabolism , Animals , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Chromatography, Liquid , Collagen/metabolism , Disease Models, Animal , Disease Progression , Extracellular Matrix/pathology , Fibrosis , Genotype , Humans , Mice , Microscopy, Atomic Force , Mutation , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prognosis , Proteomics , Proto-Oncogene Proteins p21(ras)/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction , Smad4 Protein/genetics , Survival Rate , Tandem Mass Spectrometry , Tumor Microenvironment
6.
Cancer Cell ; 25(6): 719-34, 2014 Jun 16.
Article in English | MEDLINE | ID: mdl-24856586

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is associated with marked fibrosis and stromal myofibroblasts, but their functional contribution remains unknown. Transgenic mice with the ability to delete αSMA(+) myofibroblasts in pancreatic cancer were generated. Depletion starting at either noninvasive precursor (pancreatic intraepithelial neoplasia) or the PDAC stage led to invasive, undifferentiated tumors with enhanced hypoxia, epithelial-to-mesenchymal transition, and cancer stem cells, with diminished animal survival. In PDAC patients, fewer myofibroblasts in their tumors also correlated with reduced survival. Suppressed immune surveillance with increased CD4(+)Foxp3(+) Tregs was observed in myofibroblast-depleted mouse tumors. Although myofibroblast-depleted tumors did not respond to gemcitabine, anti-CTLA4 immunotherapy reversed disease acceleration and prolonged animal survival. This study underscores the need for caution in targeting carcinoma-associated fibroblasts in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Fibroblasts/pathology , Fibrosis/pathology , Pancreatic Neoplasms/pathology , Animals , Carcinoma, Pancreatic Ductal/immunology , Disease Models, Animal , Fibroblasts/immunology , Fibrosis/immunology , Humans , Immune Tolerance , Mice , Mice, Transgenic , Pancreatic Neoplasms/immunology , Survival Analysis
7.
Cancer Res ; 73(17): 5336-46, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23856251

ABSTRACT

The tumor stromal environment can dictate many aspects of tumor progression. A complete understanding of factors driving stromal activation and their role in tumor metastasis is critical to furthering research with the goal of therapeutic intervention. Polyoma middle T-induced mammary carcinomas lacking the type II TGF-ß receptor (PyMT(mgko)) are highly metastatic compared with control PyMT-induced carcinomas (PyMT(fl/fl)). We hypothesized that the PyMT(mgko)-activated stroma interacts with carcinoma cells to promote invasion and metastasis. We show that the extracellular matrix associated with PyMT(mgko) tumors is stiffer and has more fibrillar collagen and increased expression of the collagen crosslinking enzyme lysyl oxidase (LOX) compared with PyMT(fl/fl) mammary carcinomas. Inhibition of LOX activity in PyMT(mgko) mice had no effect on tumor latency and size, but significantly decreased tumor metastasis through inhibition of tumor cell intravasation. This phenotype was associated with a decrease in keratin 14-positive myoepithelial cells in PyMT(mgko) tumors following LOX inhibition as well as a decrease in focal adhesion formation. Interestingly, the primary source of LOX was found to be activated fibroblasts. LOX expression in these fibroblasts can be driven by myeloid cell-derived TGF-ß, which is significantly linked to human breast cancer. Overall, stromal expansion in PyMT(mgko) tumors is likely caused through the modulation of immune cell infiltrates to promote fibroblast activation. This feeds back to the epithelium to promote metastasis by modulating phenotypic characteristics of basal cells. Our data indicate that epithelial induction of microenvironmental changes can play a significant role in tumorigenesis and attenuating these changes can inhibit metastasis. Cancer Res; 73(17); 5336-46. ©2013 AACR.


Subject(s)
Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Protein Serine-Threonine Kinases/physiology , Protein-Lysine 6-Oxidase/metabolism , Receptors, Transforming Growth Factor beta/physiology , Stromal Cells/enzymology , Transforming Growth Factor beta/physiology , Animals , Carcinogenesis , Collagen/metabolism , Enzyme Inhibitors/pharmacology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Focal Adhesion Kinase 1/metabolism , Humans , In Situ Hybridization , Keratin-14/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Transgenic , Microscopy, Atomic Force , Myeloid Cells/metabolism , Myeloid Cells/pathology , Phosphorylation , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type II , Signal Transduction , Stromal Cells/pathology
8.
PLoS One ; 8(2): e55982, 2013.
Article in English | MEDLINE | ID: mdl-23418490

ABSTRACT

Side population (SP) cells have been reported to have properties of cancer stem-like cells (CSCs) in non-small cell lung carcinoma (NSCLC), yet their molecular features have not been fully elucidated. Here we show that, NSCLC-SP cells were enriched in G(0)/G-(1) phase of cell cycle, had higher aldehyde dehydrogenase activity as well as higher clonogenic and self-renewing ability compared to main population (MP) cells. Interestingly, SP cells were also able to trans-differentiate into angiogenic tubules in vitro and were highly tumorigenic as compared to MP cells. SP-derived tumors demonstrated the intratumoral heterogeneity comprising of both SP and MP cells, suggesting the self-renewal and differentiation ability of SP cells are manifested in vivo as well. ßArrestin-1 (ßArr1) is involved in the progression of various cancers including NSCLCs and we find that depletion of ßArr1 significantly blocked the SP phenotype; whereas depletion of ßArr2 had relatively minor effects. Ectopic expression of ßArr1 resulted in increased SP frequency and ABCG2 expression while abrogation of ßArr1 expression suppressed the self-renewal growth and expansion of A549 cells. Anti-apoptotic protein Mcl-1 is known to be one of the key regulators of self-renewal of tissue stem cells and is thought to contribute to survival of NSCLC cells. Our experiments show that higher levels of Mcl-1 were expressed in SP cells compared to MP cells at both transcriptional and translational levels. In addition, Obatoclax, a pharmacological inhibitor of Mcl-1, could effectively prevent the self-renewal of both EGFR-inhibitor sensitive and resistant NSCLC cells. In conclusion, our findings suggest that ßArr1 and Mcl-1 are involved in the self-renewal and expansion of NSCLC-CSCs and are potential targets for anti-cancer therapy.


Subject(s)
Arrestins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Lung/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Indoles , Lung/drug effects , Lung/pathology , Lung Neoplasms/pathology , Mice , Myeloid Cell Leukemia Sequence 1 Protein , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pyrroles/pharmacology , beta-Arrestins
9.
Gastroenterology ; 138(4): 1595-606, 1606.e1-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20080097

ABSTRACT

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers. It is characterized by substantial tumor cell invasion and early-stage metastasis. We developed an in vivo model to analyze interactions between cancer and stromal cells during early stages of PDAC. METHODS: Human pancreatic adenocarcinoma cells were grafted onto the chick chorioallantoic membrane (CAM). Human and chicken GeneChips were used simultaneously to study gene regulation during PDAC cell invasion. Bioinformatic analysis was used to identify human orthologs and cell specificity of gene expression. The effects of netrin-1 encoded by NTN1 were investigated in adhesion, invasion, and apoptosis assays. The effects of NTN1 silencing with small interfering RNAs were investigated in PDAC cells in vivo. NTN1 expression was measured in human PDAC samples. RESULTS: PDAC cells rapidly invade the CAM stroma and remodel the CAM vasculature. Around 800 stromal genes were up-regulated by >2-fold; the angiogenesis regulators vascular endothelial growth factor D, thrombospondin 1, and CD151 were among the most highly regulated genes. Silencing of tumor cell NTN1, which is up-regulated 4-fold in the PDAC model, inhibited tumor cell invasion in vivo. Netrin-1 conferred apoptosis resistance to tumor and endothelial cells in vitro, induced their invasion, and provided an adhesive substrate for tumor cells. NTN1 and its gene product are strongly overexpressed in human PDAC samples. CONCLUSIONS: We developed a useful tool to study the invasive mechanisms of early-stage PDAC. Netrin-1 might be an important regulator of pancreatic tumor growth that functions in tumor and endothelial cells.


Subject(s)
Adenocarcinoma/pathology , Endothelial Cells/physiology , Nerve Growth Factors/physiology , Pancreatic Neoplasms/pathology , Tumor Suppressor Proteins/physiology , Animals , Apoptosis , CA-19-9 Antigen/blood , Cell Line, Tumor , Chick Embryo , Disease Progression , Gene Expression Profiling , Humans , Neoplasm Invasiveness , Netrin-1 , Pancreatic Neoplasms/metabolism
10.
Proc Natl Acad Sci U S A ; 106(42): 17769-74, 2009 Oct 20.
Article in English | MEDLINE | ID: mdl-19805200

ABSTRACT

The somatostatin receptor subtype 2 (sst2) behaves as a tumor suppressor when expressed and stimulated by its ligand somatostatin in pancreatic cancer. We reveal a mechanism underlying oncosuppressive action of sst2, whereby this inhibitory receptor upregulates the expression of the secreted angioinhibitory factor thrombospondin-1 (TSP-1), as demonstrated in exocrine BxPC-3 and endocrine BON pancreatic cancer cells. The sst2-dependent upregulation of TSP-1 occurs through the inhibition of the PI3K pathway. It depends on transcriptional and translational events, involving a previously undescribed IRES in the 5'-UTR of TSP-1 mRNA. Chick chorioallantoic membrane was used as an in vivo model to demonstrate that TSP-1 is a critical effector of the inhibitory role of sst2 on the neoangiogenesis and oncogenesis induced by pancreatic cancer cells. TSP-1 reduced in vitro tubulogenesis of endothelial cells when grown in conditioned medium from pancreatic cancer cells expressing sst2, as compared to those expressing the control vector. TSP-1 inhibited tumor cell-induced neoangiogenesis by directly sequestering the proangiogenic factor VEGF, and inactivating the angiogenesis initiated by VEGFR2 phosphorylation in endothelial cells. Using human pancreatic tissue-microarrays, the expression of both sst2 and TSP-1 was shown to be correlated during the pancreatic neoplastic program. Both proteins are nearly undetectable in normal exocrine pancreas and in most invasive cancer lesions, but their expression is strikingly upregulated in most preinvasive cancer-adjacent lesions. The upregulation of both sst2 and TSP-1 tumor suppressors may function as an early negative feedback to restrain pancreatic carcinogenesis.


Subject(s)
Pancreatic Neoplasms/physiopathology , Receptors, Somatostatin/physiology , Thrombospondin 1/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Line, Tumor , Chick Embryo , Gene Expression Profiling , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/prevention & control , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/genetics , Phosphatidylinositol 3-Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptors, Somatostatin/genetics , Thrombospondin 1/genetics , Transplantation, Heterologous , Tumor Suppressor Proteins/genetics , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
11.
Mol Cell Endocrinol ; 286(1-2): 230-7, 2008 May 14.
Article in English | MEDLINE | ID: mdl-18359151

ABSTRACT

Since its discovery three decades ago as an inhibitor of GH release from the pituitary gland, somatostatin has attracted much attention because of its functional role in the regulation of a wide variety of physiological functions in the brain, pituitary, pancreas, gastrointestinal tract, adrenals, thyroid, kidney and immune system. In addition to its negative role in the control of endocrine and exocrine secretions, somatostatin and analogs also exert inhibitory effects on the proliferation and survival of normal and tumor cells. Over the past 15 years, studies have begun to reveal some of the molecular mechanisms underlying the antitumor activity of somatostatin. This review covers the present knowledge in the antitumor effect of somatostatin and analogs and discusses the perspectives of novel clinical strategies based on somatostatin receptor sst2 gene transfer therapy.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Neoplasms/metabolism , Receptors, Somatostatin/metabolism , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Human Growth Hormone/metabolism , Humans , Neoplasms/drug therapy , Receptors, Somatostatin/agonists , Signal Transduction/drug effects , Somatostatin/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...