Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Genes Dev ; 32(15-16): 1020-1034, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30068703

ABSTRACT

RNA-binding proteins (RBPs) are expressed broadly during both development and malignant transformation, yet their mechanistic roles in epithelial homeostasis or as drivers of tumor initiation and progression are incompletely understood. Here we describe a novel interplay between RBPs LIN28B and IMP1 in intestinal epithelial cells. Ribosome profiling and RNA sequencing identified IMP1 as a principle node for gene expression regulation downstream from LIN28B In vitro and in vivo data demonstrate that epithelial IMP1 loss increases expression of WNT target genes and enhances LIN28B-mediated intestinal tumorigenesis, which was reversed when we overexpressed IMP1 independently in vivo. Furthermore, IMP1 loss in wild-type or LIN28B-overexpressing mice enhances the regenerative response to irradiation. Together, our data provide new evidence for the opposing effects of the LIN28B-IMP1 axis on post-transcriptional regulation of canonical WNT signaling, with implications in intestinal homeostasis, regeneration and tumorigenesis.


Subject(s)
Carcinogenesis , Gene Expression Regulation , Intestinal Mucosa/metabolism , RNA-Binding Proteins/metabolism , Regulon , Wnt Signaling Pathway , Animals , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Humans , Intestinal Mucosa/physiology , Mice , Mice, Transgenic , Oncogenes , Protein Biosynthesis , RNA-Binding Proteins/physiology , Regeneration , Stem Cells/metabolism
2.
Genes Dev ; 30(2): 233-47, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26773005

ABSTRACT

The two major isoforms of the paired-related homeodomain transcription factor 1 (Prrx1), Prrx1a and Prrx1b, are involved in pancreatic development, pancreatitis, and carcinogenesis, although the biological role that these isoforms serve in the systemic dissemination of pancreatic ductal adenocarcinoma (PDAC) has not been investigated. An epithelial-mesenchymal transition (EMT) is believed to be important for primary tumor progression and dissemination, whereas a mesenchymal-epithelial transition (MET) appears crucial for metastatic colonization. Here, we describe novel roles for both isoforms in the metastatic cascade using complementary in vitro and in vivo models. Prrx1b promotes invasion, tumor dedifferentiation, and EMT. In contrast, Prrx1a stimulates metastatic outgrowth in the liver, tumor differentiation, and MET. We further demonstrate that the switch from Prrx1b to Prrx1a governs EMT plasticity in both mouse models of PDAC and human PDAC. Last, we identify hepatocyte growth factor ( HGF) as a novel transcriptional target of Prrx1b. Targeted therapy of HGF in combination with gemcitabine in a preclinical model of PDAC reduces primary tumor volume and eliminates metastatic disease. Overall, we provide new insights into the isoform-specific roles of Prrx1a and Prrx1b in primary PDAC formation, dissemination, and metastatic colonization, allowing for novel therapeutic strategies targeting EMT plasticity.


Subject(s)
Carcinoma, Pancreatic Ductal/physiopathology , Homeodomain Proteins/metabolism , Neoplasm Invasiveness/physiopathology , Pancreatic Neoplasms/physiopathology , Animals , Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/genetics , Cells, Cultured , Gene Expression Regulation, Neoplastic , Hepatocyte Growth Factor/genetics , Homeodomain Proteins/genetics , Humans , Mice , Neoplasm Metastasis/genetics , Pancreatic Neoplasms/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tumor Cells, Cultured
3.
Cancer Res ; 75(19): 4074-85, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26294209

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are an immunosuppressive population of immature myeloid cells found in advanced-stage cancer patients and mouse tumor models. Production of inducible nitric oxide synthase (iNOS) and arginase, as well as other suppressive mechanisms, allows MDSCs to suppress T-cell-mediated tumor clearance and foster tumor progression. Using an unbiased global gene expression approach in conditional p120-catenin knockout mice (L2-cre;p120ctn(f/f)), a model of oral-esophageal cancer, we have identified CD38 as playing a vital role in MDSC biology, previously unknown. CD38 belongs to the ADP-ribosyl cyclase family and possesses both ectoenzyme and receptor functions. It has been described to function in lymphoid and early myeloid cell differentiation, cell activation, and neutrophil chemotaxis. We find that CD38 expression in MDSCs is evident in other mouse tumor models of esophageal carcinogenesis, and CD38(high) MDSCs are more immature than MDSCs lacking CD38 expression, suggesting a potential role for CD38 in the maturation halt found in MDSC populations. CD38(high) MDSCs also possess a greater capacity to suppress activated T cells, and promote tumor growth to a greater degree than CD38(low) MDSCs, likely as a result of increased iNOS production. In addition, we have identified novel tumor-derived factors, specifically IL6, IGFBP3, and CXCL16, which induce CD38 expression by MDSCs ex vivo. Finally, we have detected an expansion of CD38(+) MDSCs in peripheral blood of advanced-stage cancer patients and validated targeting CD38 in vivo as a novel approach to cancer therapy.


Subject(s)
ADP-ribosyl Cyclase 1/physiology , Carcinoma, Squamous Cell/immunology , Esophageal Neoplasms/immunology , Membrane Glycoproteins/physiology , Myeloid Cells/immunology , Neoplasm Proteins/physiology , Tumor Escape/immunology , ADP-ribosyl Cyclase 1/biosynthesis , ADP-ribosyl Cyclase 1/blood , ADP-ribosyl Cyclase 1/genetics , Animals , Arginase/physiology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cytokines/physiology , Disease Models, Animal , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/immunology , Humans , Immune Tolerance/physiology , Lymphocyte Activation , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/blood , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/chemistry , Myelopoiesis/physiology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nitric Oxide Synthase Type II/physiology , T-Lymphocytes/immunology , Tumor Stem Cell Assay
4.
Mol Cancer Res ; 13(11): 1478-86, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26194191

ABSTRACT

UNLABELLED: The colon tumor microenvironment is becoming increasingly recognized as a complex but central player in the development of many cancers. Previously, we identified an oncogenic role for the mRNA-binding protein IMP1 (IGF2BP1) in the epithelium during colon tumorigenesis. In the current study, we reveal the contribution of stromal IMP1 in the context of colitis-associated colon tumorigenesis. Interestingly, stromal deletion of Imp1 (Dermo1Cre;Imp1(LoxP/LoxP), or Imp1(ΔMes)) in the azoxymethane/dextran sodium sulfate (AOM/DSS) model of colitis-associated cancer resulted in increased tumor numbers of larger size and more advanced histologic grade than controls. In addition, Imp1(ΔMes) mice exhibited a global increase in protumorigenic microenvironment factors, including enhanced inflammation and stromal components. Evaluation of purified mesenchyme from AOM/DSS-treated Imp1(ΔMes) mice demonstrated an increase in hepatocyte growth factor (HGF), which has not been associated with regulation via IMP1. Genetic knockdown of Imp1 in human primary fibroblasts confirmed an increase in HGF with Imp1 loss, demonstrating a specific, cell-autonomous role for Imp1 loss to increase HGF expression. Taken together, these data demonstrate a novel tumor-suppressive role for IMP1 in colon stromal cells and underscore an exquisite, context-specific function for mRNA-binding proteins, such as IMP1, in disease states. IMPLICATIONS: The tumor-suppressive role of stromal IMP1 and its ability to modulate protumorigenic factors suggest that IMP1 status is important for the initiation and growth of epithelial tumors.


Subject(s)
RNA-Binding Proteins/metabolism , Tumor Microenvironment , Animals , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Gene Deletion , Hepatocyte Growth Factor/metabolism , Humans , Mesoderm/metabolism , Mice , RNA-Binding Proteins/genetics , Stromal Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...