Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Nat Commun ; 14(1): 7791, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38057326

ABSTRACT

Oncogenic lesions in pancreatic ductal adenocarcinoma (PDAC) hijack the epigenetic machinery in stromal components to establish a desmoplastic and therapeutic resistant tumor microenvironment (TME). Here we identify Class I histone deacetylases (HDACs) as key epigenetic factors facilitating the induction of pro-desmoplastic and pro-tumorigenic transcriptional programs in pancreatic stromal fibroblasts. Mechanistically, HDAC-mediated changes in chromatin architecture enable the activation of pro-desmoplastic programs directed by serum response factor (SRF) and forkhead box M1 (FOXM1). HDACs also coordinate fibroblast pro-inflammatory programs inducing leukemia inhibitory factor (LIF) expression, supporting paracrine pro-tumorigenic crosstalk. HDAC depletion in cancer-associated fibroblasts (CAFs) and treatment with the HDAC inhibitor entinostat (Ent) in PDAC mouse models reduce stromal activation and curb tumor progression. Notably, HDAC inhibition (HDACi) enriches a lipogenic fibroblast subpopulation, a potential precursor for myofibroblasts in the PDAC stroma. Overall, our study reveals the stromal targeting potential of HDACi, highlighting the utility of this epigenetic modulating approach in PDAC therapeutics.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Mice , Cell Line, Tumor , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreas/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Fibroblasts/metabolism , Carcinogenesis/pathology , Tumor Microenvironment
2.
Nat Commun ; 14(1): 5195, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37673892

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy in need of new therapeutic options. Using unbiased analyses of super-enhancers (SEs) as sentinels of core genes involved in cell-specific function, here we uncover a druggable SE-mediated RNA-binding protein (RBP) cascade that supports PDAC growth through enhanced mRNA translation. This cascade is driven by a SE associated with the RBP heterogeneous nuclear ribonucleoprotein F, which stabilizes protein arginine methyltransferase 1 (PRMT1) to, in turn, control the translational mediator ubiquitin-associated protein 2-like. All three of these genes and the regulatory SE are essential for PDAC growth and coordinately regulated by the Myc oncogene. In line with this, modulation of the RBP network by PRMT1 inhibition reveals a unique vulnerability in Myc-high PDAC patient organoids and markedly reduces tumor growth in male mice. Our study highlights a functional link between epigenetic regulation and mRNA translation and identifies components that comprise unexpected therapeutic targets for PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Male , Animals , Mice , RNA , Epigenesis, Genetic , Regulatory Sequences, Nucleic Acid , Pancreatic Neoplasms/genetics , Carcinoma, Pancreatic Ductal/genetics , Methyltransferases , RNA-Binding Proteins/genetics
3.
bioRxiv ; 2023 Sep 14.
Article in English | MEDLINE | ID: mdl-37745372

ABSTRACT

Oncogenic lesions in pancreatic ductal adenocarcinoma (PDAC) hijack the epigenetic machinery in stromal components to establish a desmoplastic and therapeutic resistant tumor microenvironment (TME). Here we identify Class I histone deacetylases (HDACs) as key epigenetic factors facilitating the induction of pro-desmoplastic and pro-tumorigenic transcriptional programs in pancreatic stromal fibroblasts. Mechanistically, HDAC-mediated changes in chromatin architecture enable the activation of pro-desmoplastic programs directed by serum response factor (SRF) and forkhead box M1 (FOXM1). HDACs also coordinate fibroblast pro-inflammatory programs inducing leukemia inhibitory factor (LIF) expression, supporting paracrine pro-tumorigenic crosstalk. HDAC depletion in cancer-associated fibroblasts (CAFs) and treatment with the HDAC inhibitor entinostat (Ent) in PDAC mouse models reduce stromal activation and curb tumor progression. Notably, HDAC inhibition (HDACi) enriches a lipogenic fibroblast subpopulation, a potential precursor for myofibroblasts in the PDAC stroma. Overall, our study reveals the stromal targeting potential of HDACi, highlighting the utility of this epigenetic modulating approach in PDAC therapeutics.

4.
Proc Natl Acad Sci U S A ; 119(51): e2213041119, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36508655

ABSTRACT

The pleiotropic actions of the Farnesoid X Receptor (FXR) are required for gut health, and reciprocally, reduced intestinal FXR signaling is seen in inflammatory bowel diseases (IBDs). Here, we show that activation of FXR selectively in the intestine is protective in inflammation-driven models of IBD. Prophylactic activation of FXR restored homeostatic levels of pro-inflammatory cytokines, most notably IL17. Importantly, these changes were attributed to FXR regulation of innate lymphoid cells (ILCs), with both the inflammation-driven increases in ILCs, and ILC3s in particular, and the induction of Il17a and Il17f in ILC3s blocked by FXR activation. Moreover, a population of ILC precursor-like cells increased with treatment, implicating FXR in the maturation/differentiation of ILC precursors. These findings identify FXR as an intrinsic regulator of intestinal ILCs and a potential therapeutic target in inflammatory intestinal diseases.


Subject(s)
Immunity, Innate , Inflammatory Bowel Diseases , Humans , Lymphocytes , Inflammatory Bowel Diseases/drug therapy , Cytokines , Inflammation
5.
J Addict Nurs ; 28(3): 124-130, 2017.
Article in English | MEDLINE | ID: mdl-28863053

ABSTRACT

Stigma and insufficient training for addressing alcohol or drug problems in clinical settings are two significant barriers to earlier identification and access to care for patients with alcohol or drug (AOD)-related problems. Nurses are crucial players in this strategy, yet nurses often report poor motivation and insufficient training for working with AOD-affected patients. This article describes and reports preliminary evaluation results of an educational experience that integrated AOD curriculum into a community/public health nursing clinical practicum. The purpose of the intervention was to meet the required objectives for our university's community/public health nursing clinical course, improve students' attitudes and therapeutic commitment for working with AOD-affected patients, and prepare students for providing high-quality nursing care for individuals with AOD-related problems.


Subject(s)
Attitude of Health Personnel , Curriculum , Students, Nursing , Substance-Related Disorders/nursing , Adolescent , Adult , Education, Nursing, Baccalaureate/methods , Female , Humans , Male , Program Evaluation , Surveys and Questionnaires , Texas , Young Adult
6.
Nat Rev Cancer ; 17(5): 332, 2017 04 24.
Article in English | MEDLINE | ID: mdl-28436470
7.
Proc Natl Acad Sci U S A ; 114(5): 1129-1134, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096419

ABSTRACT

A fibroinflammatory stromal reaction cooperates with oncogenic signaling to influence pancreatic ductal adenocarcinoma (PDAC) initiation, progression, and therapeutic outcome, yet the mechanistic underpinning of this crosstalk remains poorly understood. Here we show that stromal cues elicit an adaptive response in the cancer cell including the rapid mobilization of a transcriptional network implicated in accelerated growth, along with anabolic changes of an altered metabolome. The close overlap of stroma-induced changes in vitro with those previously shown to be regulated by oncogenic Kras in vivo suggests that oncogenic Kras signaling-a hallmark and key driver of PDAC-is contingent on stromal inputs. Mechanistically, stroma-activated cancer cells show widespread increases in histone acetylation at transcriptionally enhanced genes, implicating the PDAC epigenome as a presumptive point of convergence between these pathways and a potential therapeutic target. Notably, inhibition of the bromodomain and extraterminal (BET) family of epigenetic readers, and of Bromodomain-containing protein 2 (BRD2) in particular, blocks stroma-inducible transcriptional regulation in vitro and tumor progression in vivo. Our work suggests the existence of a molecular "AND-gate" such that tumor activation is the consequence of mutant Kras and stromal cues, providing insight into the role of the tumor microenvironment in the origin and treatment of Ras-driven tumors.


Subject(s)
Carcinoma, Pancreatic Ductal/physiopathology , Fibroblasts/physiology , Gene Expression Regulation, Neoplastic/genetics , Histone Code , Metabolome , Pancreatic Neoplasms/physiopathology , Stromal Cells/physiology , Tumor Microenvironment/physiology , Acetylation , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cytokines/metabolism , Energy Metabolism , Enhancer Elements, Genetic , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/physiology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/physiology , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/physiology , Transcription Factors , Tumor Cells, Cultured
8.
Sci Signal ; 9(430): ra57, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27245614

ABSTRACT

Rapamycin has been used as a clinical immunosuppressant for many years; however, the molecular basis for its selective effects on lymphocytes remains unclear. We investigated the role of two canonical effectors of the mammalian target of rapamycin (mTOR): ribosomal S6 kinases (S6Ks) and eukaryotic initiation factor 4E (eIF4E)-binding proteins (4E-BPs). S6Ks are thought to regulate cell growth (increase in cell size), and 4E-BPs are thought to control proliferation (increase in cell number), with mTORC1 signaling serving to integrate these processes. However, we found that the 4E-BP-eIF4E signaling axis controlled both the growth and proliferation of lymphocytes, processes for which the S6Ks were dispensable. Furthermore, rapamycin disrupted eIF4E function selectively in lymphocytes, which was due to the increased abundance of 4E-BP2 relative to that of 4E-BP1 in these cells and the greater sensitivity of 4E-BP2 to rapamycin. Together, our findings suggest that the 4E-BP-eIF4E axis is uniquely rapamycin-sensitive in lymphocytes and that this axis promotes clonal expansion of these cells by coordinating growth and proliferation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Lymphocytes/drug effects , Nucleocytoplasmic Transport Proteins/metabolism , Phosphoproteins/metabolism , Signal Transduction , Animals , Cell Cycle Proteins , Cell Enlargement , Cell Membrane/metabolism , Cell Proliferation , Crosses, Genetic , Eukaryotic Initiation Factors , Female , Gene Expression Regulation , Guanosine Triphosphate , Immunosuppressive Agents/pharmacology , Lymphocytes/cytology , Male , Mice , Mice, Inbred C57BL , Sirolimus/pharmacology
9.
Nat Rev Cancer ; 16(5): 288-304, 2016 04 26.
Article in English | MEDLINE | ID: mdl-27112207

ABSTRACT

The past several years have seen dramatic leaps in our understanding of how gene expression is rewired at the translation level during tumorigenesis to support the transformed phenotype. This work has been driven by an explosion in technological advances and is revealing previously unimagined regulatory mechanisms that dictate functional expression of the cancer genome. In this Review we discuss emerging trends and exciting new discoveries that reveal how this translational circuitry contributes to specific aspects of tumorigenesis and cancer cell function, with a particular focus on recent insights into the role of translational control in the adaptive response to oncogenic stress conditions.


Subject(s)
Genome , Neoplasms/genetics , Protein Biosynthesis , Humans , Oncogenes , RNA, Neoplasm/genetics
10.
Cancer Discov ; 6(3): 270-85, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26715645

ABSTRACT

UNLABELLED: Pancreas ductal adenocarcinoma (PDAC) has one of the worst 5-year survival rates of all solid tumors, and thus new treatment strategies are urgently needed. Here, we report that targeting Bruton tyrosine kinase (BTK), a key B-cell and macrophage kinase, restores T cell-dependent antitumor immune responses, thereby inhibiting PDAC growth and improving responsiveness to standard-of-care chemotherapy. We report that PDAC tumor growth depends on cross-talk between B cells and FcRγ(+) tumor-associated macrophages, resulting in T(H)2-type macrophage programming via BTK activation in a PI3Kγ-dependent manner. Treatment of PDAC-bearing mice with the BTK inhibitor PCI32765 (ibrutinib) or by PI3Kγ inhibition reprogrammed macrophages toward a T(H)1 phenotype that fostered CD8(+) T-cell cytotoxicity, and suppressed PDAC growth, indicating that BTK signaling mediates PDAC immunosuppression. These data indicate that pharmacologic inhibition of BTK in PDAC can reactivate adaptive immune responses, presenting a new therapeutic modality for this devastating tumor type. SIGNIFICANCE: We report that BTK regulates B-cell and macrophage-mediated T-cell suppression in pancreas adenocarcinomas. Inhibition of BTK with the FDA-approved inhibitor ibrutinib restores T cell-dependent antitumor immune responses to inhibit PDAC growth and improves responsiveness to chemotherapy, presenting a new therapeutic modality for pancreas cancer.


Subject(s)
Cell Communication/immunology , Immune System/cytology , Immune System/pathology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Protein-Tyrosine Kinases/metabolism , Agammaglobulinaemia Tyrosine Kinase , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Disease Models, Animal , Disease Progression , Humans , Leukocytes/immunology , Leukocytes/metabolism , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Pancreatic Neoplasms/genetics , Receptors, IgG/metabolism , Signal Transduction
11.
Cell ; 162(1): 59-71, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26095252

ABSTRACT

eIF4E, the major cap-binding protein, has long been considered limiting for translating the mammalian genome. However, the eIF4E dose requirement at an organismal level remains unexplored. By generating an Eif4e haploinsufficient mouse, we found that a 50% reduction in eIF4E expression, while compatible with normal development and global protein synthesis, significantly impeded cellular transformation. Genome-wide translational profiling uncovered a translational program induced by oncogenic transformation and revealed a critical role for the dose of eIF4E, specifically in translating a network of mRNAs enriched for a unique 5' UTR signature. In particular, we demonstrate that the dose of eIF4E is essential for translating mRNAs that regulate reactive oxygen species, fueling transformation and cancer cell survival in vivo. Our findings indicate eIF4E is maintained at levels in excess for normal development that are hijacked by cancer cells to drive a translational program supporting tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic , Embryo, Mammalian/metabolism , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Dosage , 5' Untranslated Regions , Animals , Carcinogenesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Biosynthesis , Reactive Oxygen Species/metabolism
12.
Cancer Res ; 75(14): 2851-62, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25977330

ABSTRACT

Oncogenic K-Ras mutation occurs frequently in several types of cancers, including pancreatic and lung cancers. Tumors with K-Ras mutation are resistant to chemotherapeutic drugs as well as molecular targeting agents. Although numerous approaches are ongoing to find effective ways to treat these tumors, there are still no effective therapies for K-Ras mutant cancer patients. Here we report that K-Ras mutant cancers are more dependent on K-Ras in anchorage-independent culture conditions than in monolayer culture conditions. In seeking to determine mechanisms that contribute to the K-Ras dependency in anchorage-independent culture conditions, we discovered the involvement of Met in K-Ras-dependent, anchorage-independent cell growth. The Met signaling pathway is enhanced and plays an indispensable role in anchorage-independent growth even in cells in which Met is not amplified. Indeed, Met expression is elevated under anchorage-independent growth conditions and is regulated by K-Ras in a MAPK/ERK kinase (MEK)-dependent manner. Remarkably, in spite of a global downregulation of mRNA translation during anchorage-independent growth, we find that Met mRNA translation is specifically enhanced under these conditions. Importantly, ectopic expression of an active Met mutant rescues K-Ras ablation-derived growth suppression, indicating that K-Ras-mediated Met expression drives "K-Ras addiction" in anchorage-independent conditions. Our results indicate that enhanced Met expression and signaling is essential for anchorage-independent growth of K-Ras mutant cancer cells and suggests that pharmacological inhibitors of Met could be effective for K-Ras mutant tumor patients.


Subject(s)
Cell Proliferation/genetics , Genes, ras/physiology , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Animals , Cell Adhesion/genetics , Cells, Cultured , HCT116 Cells , HEK293 Cells , Humans , Mice , Protein Biosynthesis , Signal Transduction/physiology , Tissue Culture Techniques
13.
Proc Natl Acad Sci U S A ; 110(29): 11988-93, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23803853

ABSTRACT

Myc is one of the most commonly deregulated oncogenes in human cancer, yet therapies directly targeting Myc hyperactivation are not presently available in the clinic. The evolutionarily conserved function of Myc in modulating protein synthesis control is critical to the Myc oncogenic program. Indeed, enhancing the protein synthesis capacity of cancer cells directly contributes to their survival, proliferation, and genome instability. Therefore, inhibiting enhanced protein synthesis may represent a highly relevant strategy for the treatment of Myc-dependent human cancers. However, components of the translation machinery that can be exploited as therapeutic targets for Myc-driven cancers remain poorly defined. Here, we uncover a surprising and important functional link between Myc and mammalian target of rapamycin (mTOR)-dependent phosphorylation of eukaryotic translation initiation factor 4E binding protein-1 (4EBP1), a master regulator of protein synthesis control. Using a pharmacogenetic approach, we find that mTOR-dependent phosphorylation of 4EBP1 is required for cancer cell survival in Myc-dependent tumor initiation and maintenance. We further show that a clinical mTOR active site inhibitor, which is capable of blocking mTOR-dependent 4EBP1 phosphorylation, has remarkable therapeutic efficacy in Myc-driven hematological cancers. Additionally, we demonstrate the clinical implications of these results by delineating a significant link between Myc and mTOR-dependent phosphorylation of 4EBP1 and therapeutic response in human lymphomas. Together, these findings reveal that an important mTOR substrate is found hyperactivated downstream of Myc oncogenic activity to promote tumor survival and confers synthetic lethality, thereby revealing a unique therapeutic approach to render Myc druggable in the clinic.


Subject(s)
B-Lymphocytes/physiology , Benzoxazoles/pharmacology , Carrier Proteins/metabolism , Cell Transformation, Neoplastic/metabolism , Phosphoproteins/metabolism , Protein Biosynthesis/physiology , Proto-Oncogene Proteins c-myc/metabolism , Pyrimidines/pharmacology , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Benzoxazoles/administration & dosage , Blotting, Western , Cell Cycle Proteins , Eukaryotic Initiation Factors , Everolimus , Humans , Mice , Mice, Transgenic , Microarray Analysis , Phosphorylation , Pyrimidines/administration & dosage , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/antagonists & inhibitors
14.
Nature ; 493(7432): 371-7, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23172145

ABSTRACT

Hyperconnectivity of neuronal circuits due to increased synaptic protein synthesis is thought to cause autism spectrum disorders (ASDs). The mammalian target of rapamycin (mTOR) is strongly implicated in ASDs by means of upstream signalling; however, downstream regulatory mechanisms are ill-defined. Here we show that knockout of the eukaryotic translation initiation factor 4E-binding protein 2 (4E-BP2)-an eIF4E repressor downstream of mTOR-or eIF4E overexpression leads to increased translation of neuroligins, which are postsynaptic proteins that are causally linked to ASDs. Mice that have the gene encoding 4E-BP2 (Eif4ebp2) knocked out exhibit an increased ratio of excitatory to inhibitory synaptic inputs and autistic-like behaviours (that is, social interaction deficits, altered communication and repetitive/stereotyped behaviours). Pharmacological inhibition of eIF4E activity or normalization of neuroligin 1, but not neuroligin 2, protein levels restores the normal excitation/inhibition ratio and rectifies the social behaviour deficits. Thus, translational control by eIF4E regulates the synthesis of neuroligins, maintaining the excitation-to-inhibition balance, and its dysregulation engenders ASD-like phenotypes.


Subject(s)
Autistic Disorder/genetics , Autistic Disorder/physiopathology , Eukaryotic Initiation Factor-4E/metabolism , Protein Biosynthesis , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Eukaryotic Initiation Factors/deficiency , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Male , Mice , Mice, Knockout , Phenotype , Synapses/metabolism
15.
Science ; 338(6108): 818-22, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23042294

ABSTRACT

The endoplasmic reticulum (ER) is the primary organelle for folding and maturation of secretory and transmembrane proteins. Inability to meet protein-folding demand leads to "ER stress," and activates IRE1α, an ER transmembrane kinase-endoribonuclease (RNase). IRE1α promotes adaptation through splicing Xbp1 mRNA or apoptosis through incompletely understood mechanisms. Here, we found that sustained IRE1α RNase activation caused rapid decay of select microRNAs (miRs -17, -34a, -96, and -125b) that normally repress translation of Caspase-2 mRNA, and thus sharply elevates protein levels of this initiator protease of the mitochondrial apoptotic pathway. In cell-free systems, recombinant IRE1α endonucleolytically cleaved microRNA precursors at sites distinct from DICER. Thus, IRE1α regulates translation of a proapoptotic protein through terminating microRNA biogenesis, and noncoding RNAs are part of the ER stress response.


Subject(s)
Caspase 2/genetics , Caspase 2/metabolism , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Endoplasmic Reticulum Stress , Endoribonucleases/metabolism , MicroRNAs/metabolism , Protein Serine-Threonine Kinases/metabolism , 3' Untranslated Regions , Animals , Apoptosis , Brefeldin A/pharmacology , Cell-Free System , Cells, Cultured , Down-Regulation , Endoplasmic Reticulum/metabolism , Endoribonucleases/chemistry , Endoribonucleases/genetics , Enzyme Activation , HEK293 Cells , Humans , Mice , Mice, Knockout , Mutant Proteins , Protein Biosynthesis , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation
16.
Nat Med ; 17(4): 500-3, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21460848

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is a lethal disease. Overall survival is typically 6 months from diagnosis. Numerous phase 3 trials of agents effective in other malignancies have failed to benefit unselected PDA populations, although patients do occasionally respond. Studies in other solid tumors have shown that heterogeneity in response is determined, in part, by molecular differences between tumors. Furthermore, treatment outcomes are improved by targeting drugs to tumor subtypes in which they are selectively effective, with breast and lung cancers providing recent examples. Identification of PDA molecular subtypes has been frustrated by a paucity of tumor specimens available for study. We have overcome this problem by combined analysis of transcriptional profiles of primary PDA samples from several studies, along with human and mouse PDA cell lines. We define three PDA subtypes: classical, quasimesenchymal and exocrine-like, and we present evidence for clinical outcome and therapeutic response differences between them. We further define gene signatures for these subtypes that may have utility in stratifying patients for treatment and present preclinical model systems that may be used to identify new subtype specific therapies.


Subject(s)
Carcinoma, Pancreatic Ductal/classification , Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/classification , Pancreatic Neoplasms/genetics , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Erlotinib Hydrochloride , Female , GATA6 Transcription Factor/genetics , Gene Expression Profiling , Humans , Male , Mice , Pancreatic Neoplasms/drug therapy , Pharmacogenetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Quinazolines/pharmacology , ras Proteins/genetics , Gemcitabine
17.
Genes Cancer ; 1(2): 125-41, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20975924

ABSTRACT

Factors associated with tumor sensitivity to epidermal growth factor receptor (EGFR) inhibitors in the context of wild-type EGFR remain elusive. This study investigates the mechanistic basis of responsiveness to EGFR inhibitors in the RIP1-Tag2 (RT2) mouse model of pancreatic neuroendocrine tumorigenesis (PNET). Upon treatment of RT2 mice with EGFR inhibitors, PNET tumors harboring wild-type, nonamplified alleles of Egfr grow at a markedly reduced rate and display a significant increase in tumor cell apoptosis, as well as reduced neovascularization. The authors identify Tgf-α and Hb-egf as key limiting mediators of separable pathological functions of Egfr in neuroendocrine tumor progression: Tgf-α mutant tumors present with an elevated apoptotic index, whereas Hb-egf mutant lesions exhibit decreased angiogenic switching and neovascularization. This study not only associates Tgf-α and Hb-egf expression with wild-type Egfr oncogenicity but also ascribes the proangiogenic activity of Egfr in this tumor model to a novel mesenchymal Hb-egf/Egfr signaling axis, whereby endothelial and pericyte-derived Hb-egf activates Egfr specifically in tumor-associated perivascular cells, leading to increased pericyte coverage of the tumor endothelium and enhanced angiogenesis.

18.
Cancer Cell ; 17(2): 135-47, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-20138012

ABSTRACT

Cancer-associated fibroblasts (CAFs) support tumorigenesis by stimulating angiogenesis, cancer cell proliferation, and invasion. We demonstrate that CAFs also mediate tumor-enhancing inflammation. Using a mouse model of squamous skin carcinogenesis, we found a proinflammatory gene signature in CAFs isolated from dysplastic skin. This signature was maintained in CAFs from subsequent skin carcinomas and was evident in mammary and pancreatic tumors in mice and in cognate human cancers. The inflammatory signature was already activated in CAFs isolated from the initial hyperplastic stage in multistep skin tumorigenesis. CAFs from this pathway promoted macrophage recruitment, neovascularization, and tumor growth, activities that are abolished when NF-kappaB signaling was inhibited. Additionally, we show that normal dermal fibroblasts can be "educated" by carcinoma cells to express proinflammatory genes.


Subject(s)
Fibroblasts/physiology , Inflammation Mediators/metabolism , NF-kappa B/physiology , Neoplasms/genetics , Animals , Carcinoma, Pancreatic Ductal/blood supply , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Female , Fibroblasts/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Neoplasms/blood supply , Neoplasms/immunology , Neoplasms/pathology , Neovascularization, Pathologic , Skin Neoplasms/blood supply , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Uterine Cervical Neoplasms/blood supply , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/pathology
19.
Genes Dev ; 23(1): 24-36, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19136624

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by the deregulation of the hedgehog signaling pathway. The Sonic Hedgehog ligand (Shh), absent in the normal pancreas, is highly expressed in pancreatic tumors and is sufficient to induce neoplastic precursor lesions in mouse models. We investigated the mechanism of Shh signaling in PDAC carcinogenesis by genetically ablating the canonical bottleneck of hedgehog signaling, the transmembrane protein Smoothened (Smo), in the pancreatic epithelium of PDAC-susceptible mice. We report that multistage development of PDAC tumors is not affected by the deletion of Smo in the pancreas, demonstrating that autocrine Shh-Ptch-Smo signaling is not required in pancreatic ductal cells for PDAC progression. However, the expression of Gli target genes is maintained in Smo-negative ducts, implicating alternative means of regulating Gli transcription in the neoplastic ductal epithelium. In PDAC tumor cells, we find that Gli transcription is decoupled from upstream Shh-Ptch-Smo signaling and is regulated by TGF-beta and KRAS, and we show that Gli1 is required both for survival and for the KRAS-mediated transformed phenotype of cultured PDAC cancer cells.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Kruppel-Like Transcription Factors/metabolism , Pancreatic Ducts/metabolism , Pancreatic Neoplasms/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line , Cell Survival , Cells, Cultured , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Pancreatic Ducts/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Smoothened Receptor , Transforming Growth Factor beta/metabolism , Zinc Finger Protein GLI1
20.
Dis Model Mech ; 1(2-3): 155-67; discussion 165, 2008.
Article in English | MEDLINE | ID: mdl-19048079

ABSTRACT

The tumor microenvironment consists of stromal cells and extracellular factors that evolve in parallel with carcinoma cells. To gain insights into the activities of stromal cell populations, we developed and applied multicolor imaging techniques to analyze the behavior of these cells within different tumor microenvironments in the same live mouse. We found that regulatory T-lymphocytes (Tregs) migrated in proximity to blood vessels. Dendritic-like cells, myeloid cells and carcinoma-associated fibroblasts all exhibited higher motility in the microenvironment at the tumor periphery than within the tumor mass. Since oxygen levels differ between tumor microenvironments, we tested if acute hypoxia could account for the differences in cell migration. Direct visualization revealed that Tregs ceased migration under acute systemic hypoxia, whereas myeloid cells continued migrating. In the same mouse and microenvironment, we experimentally subdivided the myeloid cell population and revealed that uptake of fluorescent dextran defined a low-motility subpopulation expressing markers of tumor-promoting, alternatively activated macrophages. In contrast, fluorescent anti-Gr1 antibodies marked myeloid cells patrolling inside tumor vessels and in the stroma. Our techniques allow real-time combinatorial analysis of cell populations based on spatial location, gene expression, behavior and cell surface molecules within intact tumors. The techniques are not limited to investigations in cancer, but could give new insights into cell behavior more broadly in development and disease.


Subject(s)
Microscopy, Confocal/methods , Neoplasms/pathology , Stromal Cells/pathology , Cell Hypoxia , Cell Movement , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...