Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
bioRxiv ; 2023 Sep 27.
Article in English | MEDLINE | ID: mdl-37745543

ABSTRACT

The apical-basal polarity of pancreatic acinar cells is essential for maintaining tissue architecture. However, the mechanisms by which polarity proteins regulate acinar pancreas tissue homeostasis are poorly understood. Here, we evaluate the role of Par3 in acinar pancreas injury and homeostasis. While Par3 loss in the mouse pancreas disrupts tight junctions, Par3 loss is dispensable for pancreatogenesis. However, with aging, Par3 loss results in low-grade inflammation, acinar degeneration, and pancreatic lipomatosis. Par3 loss also exacerbates pancreatitis-induced acinar cell loss, resulting in pronounced pancreatic lipomatosis and failure to regenerate. Moreover, Par3 loss in mice harboring mutant Kras causes extensive pancreatic intraepithelial neoplastic (PanIN) lesions and large pancreatic cysts. We also show that Par3 loss restricts injury-induced primary ciliogenesis. Significantly, targeting BET proteins enhances primary ciliogenesis during pancreatitis-induced injury and, in mice with Par3 loss, limits pancreatitis-induced acinar loss and facilitates acinar cell regeneration. Combined, this study demonstrates how Par3 restrains pancreatitis- and Kras-induced changes in the pancreas and identifies a potential role for BET inhibitors to attenuate pancreas injury and facilitate pancreas tissue regeneration.

2.
JCI Insight ; 7(9)2022 05 09.
Article in English | MEDLINE | ID: mdl-35380995

ABSTRACT

To elicit effective antitumor responses, CD8+ T cells need to infiltrate tumors and sustain their effector function within the immunosuppressive tumor microenvironment (TME). Here, we evaluate the role of MNK activity in regulating CD8+ T cell infiltration and antitumor activity in pancreatic and thyroid tumors. We first show that human pancreatic and thyroid tumors with increased MNK activity are associated with decreased infiltration by CD8+ T cells. We then show that, while MNK inhibitors increase CD8+ T cells in these tumors, they induce a T cell exhaustion phenotype in the tumor microenvironment. Mechanistically, we show that the exhaustion phenotype is not caused by upregulation of programmed cell death ligand 1 (PD-L1) but is caused by tumor-associated macrophages (TAMs) becoming more immunosuppressive following MNK inhibitor treatment. Reversal of CD8+ T cell exhaustion by an anti-PD-1 antibody or TAM depletion synergizes with MNK inhibitors to control tumor growth and prolong animal survival. Importantly, we show in ex vivo human pancreatic tumor slice cultures that MNK inhibitors increase the expression of markers associated with immunosuppressive TAMs. Together, these findings demonstrate a role of MNKs modulating a protumoral phenotype in macrophages and identify combination regimens involving MNK inhibitors to enhance antitumor immune responses.


Subject(s)
B7-H1 Antigen , Thyroid Neoplasms , Animals , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Macrophages/metabolism , Phenotype , Thyroid Neoplasms/metabolism , Tumor Microenvironment
3.
Cell Rep ; 38(9): 110441, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35235808

ABSTRACT

Gα13 transduces signals from G-protein-coupled receptors. While Gα13 functions as a tumor suppressor in lymphomas, it is not known whether Gα13 is pro-tumorigenic or tumor suppressive in genetically engineered mouse (GEM) models of epithelial cancers. Here, we show that loss of Gα13 in the Kras/Tp53 (KPC) GEM model promotes well-differentiated tumors and reduces survival. Mechanistically, tumors developing in KPC mice with Gα13 loss exhibit increased E-cadherin expression and mTOR signaling. Importantly, human pancreatic ductal adenocarcinoma (PDAC) tumors with low Gα13 expression also exhibit increased E-cadherin expression and mTOR signaling. Treatment with the mTOR inhibitor rapamycin decreases the growth of syngeneic KPC tumors with Gα13 loss by promoting cell death. This work establishes a tumor-suppressive role of Gα13 in pancreatic tumorigenesis in the KPC GEM model and suggests targeting mTOR in human PDAC tumors with Gα13 loss.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Cadherins/metabolism , Carcinogenesis , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Disease Models, Animal , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Pancreatic Neoplasms
4.
Cancers (Basel) ; 13(3)2021 Jan 25.
Article in English | MEDLINE | ID: mdl-33503832

ABSTRACT

The advent of immunotherapy has transformed the treatment landscape for several human malignancies. Antibodies against immune checkpoints, such as anti-PD-1/PD-L1 and anti-CTLA-4, demonstrate durable clinical benefits in several cancer types. However, checkpoint blockade has failed to elicit effective anti-tumor responses in pancreatic ductal adenocarcinoma (PDAC), which remains one of the most lethal malignancies with a dismal prognosis. As a result, there are significant efforts to identify novel immune-based combination regimens for PDAC, which are typically first tested in preclinical models. Here, we discuss the utility and limitations of syngeneic and genetically-engineered mouse models that are currently available for testing immunotherapy regimens. We also discuss patient-derived xenograft mouse models, human PDAC organoids, and ex vivo slice cultures of human PDAC tumors that can complement murine models for a more comprehensive approach to predict response and resistance to immunotherapy regimens.

5.
Nat Commun ; 11(1): 4027, 2020 08 12.
Article in English | MEDLINE | ID: mdl-32788676

ABSTRACT

Programmed cell death or apoptosis is a central biological process that is dysregulated in many diseases, including inflammatory conditions and cancer. The detection and quantification of apoptotic cells in vivo is hampered by the need for fixatives or washing steps for non-fluorogenic reagents, and by the low levels of free calcium in diseased tissues that restrict the use of annexins. In this manuscript, we report the rational design of a highly stable fluorogenic peptide (termed Apo-15) that selectively stains apoptotic cells in vitro and in vivo in a calcium-independent manner and under wash-free conditions. Furthermore, using a combination of chemical and biophysical methods, we identify phosphatidylserine as a molecular target of Apo-15. We demonstrate that Apo-15 can be used for the quantification and imaging of drug-induced apoptosis in preclinical mouse models, thus creating opportunities for assessing the in vivo efficacy of anti-inflammatory and anti-cancer therapeutics.


Subject(s)
Apoptosis , Imaging, Three-Dimensional , Peptides, Cyclic/pharmacology , Amino Acid Sequence , Animals , Apoptosis/drug effects , Cell Line , Female , Humans , Mice, Inbred C57BL , Microscopy, Fluorescence , Neutrophils/cytology , Neutrophils/drug effects , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/chemistry , Phagocytosis/drug effects , Phosphatidylserines/metabolism
6.
Int J Mol Sci ; 20(17)2019 Sep 02.
Article in English | MEDLINE | ID: mdl-31480735

ABSTRACT

Bromodomain and extraterminal domain (BET) proteins, which are important epigenetic readers, are often dysregulated in cancer. While a number of BET inhibitors are currently in early phase clinical trials, BET inhibitors show limited single-agent activity. The purpose of this study is to determine if Quercetin, a naturally occurring polyphenolic flavonoid often found abundant in fruits and vegetables, can enhance the anti-tumor effects of BET inhibitors. The efficacy of the combination was evaluated in vitro and in a xenograft model of pancreatic cancer. Co-treatment with BET inhibitors and Quercetin promoted apoptosis, decreased sphere-forming ability by cancer cells, and decreased cell proliferation. We found that hnRNPA1, a nuclear protein known to control mRNA export and mRNA translation of anti-apoptotic proteins, mediates some anti-tumor effects by Quercetin. Additionally, we show that combining BET inhibitors with Quercetin or hnRNPA1 knockdown decreased the anti-apoptotic protein Survivin. Significantly, Quercetin decreased hnRNPA1 in vivo and enhanced the effects of BET inhibitors at suppressing tumor growth. Together, these results demonstrate that Quercetin enhances the efficacy of BET inhibitors by suppressing hnRNPA1, and identify combination therapy with Quercetin and BET inhibitors for the treatment of cancer patients.


Subject(s)
Acetanilides/pharmacology , Antineoplastic Agents/pharmacology , Azepines/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Quercetin/pharmacology , Triazoles/pharmacology , Animals , Cell Line, Tumor , Female , Humans , Mice, Nude , Rats , Survivin/metabolism
7.
Sci Rep ; 8(1): 13225, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30185888

ABSTRACT

The fibrotic reaction is a characteristic feature of human pancreatic ductal adenocarcinoma (PDAC) tumors. It is associated with activation and proliferation of pancreatic stellate cells (PSCs), which are key regulators of fibrosis in vivo. While there is increasing interest in the regulation of PD-L1 expression in cancer and immune cells, the expression and regulation of PD-L1 in other stromal cells, such as PSCs, has not been fully evaluated. Here we show that PSCs in vitro express higher PD-L1 mRNA and protein levels compared to the levels present in PDAC cells. We show that inhibitors targeting bromodomain and extra-terminal (BET) proteins and BRD4 knockdown decrease interferon-γ (IFN-γ)-induced PD-L1 expression in PSCs. We also show that c-MYC, one of the well-established targets of BET inhibitors, does not mediate IFN-γ-regulated PD-L1 expression in PSCs. Instead we show that interferon regulatory factor 1 (IRF1) mediates IFN-γ-induced PD-L1 expression in PSCs. Finally, while we show that BET inhibitors do not regulate IFN-γ-induced IRF1 expression in PSCs, BET inhibitors decrease binding of IRF1 and BRD4 to the PD-L1 promoter. Together, these results demonstrate the interplay between IRF1 and BRD4 in the regulation of PD-L1 in PSCs.


Subject(s)
B7-H1 Antigen/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Interferon Regulatory Factor-1/metabolism , Nuclear Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/metabolism , Transcription Factors/metabolism , B7-H1 Antigen/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Cycle Proteins , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Interferon Regulatory Factor-1/genetics , Nuclear Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/pathology , Transcription Factors/genetics , Pancreatic Neoplasms
8.
Science ; 361(6409)2018 09 28.
Article in English | MEDLINE | ID: mdl-30262472

ABSTRACT

Cancer cells from a primary tumor can disseminate to other tissues, remaining dormant and clinically undetectable for many years. Little is known about the cues that cause these dormant cells to awaken, resume proliferating, and develop into metastases. Studying mouse models, we found that sustained lung inflammation caused by tobacco smoke exposure or nasal instillation of lipopolysaccharide converted disseminated, dormant cancer cells to aggressively growing metastases. Sustained inflammation induced the formation of neutrophil extracellular traps (NETs), and these were required for awakening dormant cancer. Mechanistic analysis revealed that two NET-associated proteases, neutrophil elastase and matrix metalloproteinase 9, sequentially cleaved laminin. The proteolytically remodeled laminin induced proliferation of dormant cancer cells by activating integrin α3ß1 signaling. Antibodies against NET-remodeled laminin prevented awakening of dormant cells. Therapies aimed at preventing dormant cell awakening could potentially prolong the survival of cancer patients.


Subject(s)
Carcinogenesis/metabolism , Extracellular Traps/enzymology , Lamins/metabolism , Lung Neoplasms/pathology , Neutrophils/enzymology , Pneumonia/pathology , Animals , DNA/metabolism , Humans , Inflammation/chemically induced , Inflammation/microbiology , Integrin alpha3beta1/metabolism , Leukocyte Elastase/metabolism , Lipopolysaccharides , Lung/pathology , MCF-7 Cells , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/pathology , Pneumonia/chemically induced , Pneumonia/microbiology , Pneumonia, Bacterial/etiology , Pneumonia, Bacterial/pathology , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases/antagonists & inhibitors , Protein-Arginine Deiminases/metabolism , Proteolysis , Rats , Signal Transduction , Smoking , Nicotiana
9.
Mol Cancer Res ; 11(9): 1078-87, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23761168

ABSTRACT

UNLABELLED: Patients with pancreatic cancer, which is characterized by an extensive collagen-rich fibrotic reaction, often present with metastases. A critical step in cancer metastasis is epithelial-to-mesenchymal transition (EMT), which can be orchestrated by the Snail family of transcription factors. To understand the role of Snail (SNAI1) in pancreatic cancer development, we generated transgenic mice expressing Snail in the pancreas. Because chronic pancreatitis can contribute to pancreatic cancer development, Snail-expressing mice were treated with cerulein to induce pancreatitis. Although significant tissue injury was observed, a minimal difference in pancreatitis was seen between control and Snail-expressing mice. However, because Kras mutation is necessary for tumor development in mouse models of pancreatic cancer, we generated mice expressing both mutant Kras(G12D) and Snail (Kras(+)/Snail(+)). Compared with control mice (Kras(+)/Snai(-)), Kras(+)/Snail(+) mice developed acinar ectasia and more advanced acinar-to-ductal metaplasia. The Kras(+)/Snail(+) mice exhibited increased fibrosis, increased phosphorylated Smad2, increased TGF-ß2 expression, and activation of pancreatic stellate cells. To further understand the mechanism by which Snail promoted fibrosis, we established an in vitro model to examine the effect of Snail expression in pancreatic cancer cells on stellate cell collagen production. Snail expression in pancreatic cancer cells increased TGF-ß2 levels, and conditioned media from Snail-expressing pancreatic cancer cells increased collagen production by stellate cells. Additionally, inhibiting TGF-ß signaling in stellate cells attenuated the conditioned media-induced collagen production by stellate cells. Together, these results suggest that Snail contributes to pancreatic tumor development by promoting fibrotic reaction through increased TGF-ß signaling. IMPLICATIONS: Expression of the EMT regulator Snail in the context of mutant Kras provides new insight into pancreatic cancer progression.


Subject(s)
Epithelial-Mesenchymal Transition , Proto-Oncogene Proteins p21(ras)/metabolism , Transcription Factors/metabolism , Animals , Cell Proliferation , Ceruletide , Collagen/metabolism , Fibrosis/metabolism , Mice , Mice, Transgenic , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/metabolism , Pancreatitis, Chronic/chemically induced , Pancreatitis, Chronic/metabolism , Pancreatitis, Chronic/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction , Smad2 Protein/metabolism , Snail Family Transcription Factors , Transcription Factors/genetics , Transforming Growth Factor beta2/metabolism , Tumor Cells, Cultured
10.
J Biol Chem ; 287(9): 6218-29, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22232555

ABSTRACT

The Snail family of transcription factors has been implicated in pancreatic cancer progression. We recently showed that Snail (Snai1) promotes membrane-type 1 matrix metalloproteinase (MT1-MMP)- and ERK1/2-dependent scattering of pancreatic cancer cells in three-dimensional type I collagen. In this study, we examine the role of Slug (Snai2) in regulating pancreatic cancer cell scattering in three-dimensional type I collagen. Although Slug increased MT1-MMP expression and ERK1/2 activity, Slug-expressing cells failed to scatter in three-dimensional collagen. Moreover, in contrast to Snail-expressing cells, Slug-expressing cells did not demonstrate increased collagen I binding, collagen I-driven motility, or α2ß1-integrin expression. Significantly, inhibiting ß1-integrin function decreased migration and scattering of Snail-expressing cells in three-dimensional collagen. As Rho GTPases have been implicated in invasion and migration, we also analyzed the contribution of Rac1 and Rho signaling to the differential migration and scattering of pancreatic cancer cells. Snail-induced migration and scattering were attenuated by Rac1 inhibition. In contrast, inhibiting Rho-associated kinase ROCK1/2 increased migration and scattering of Slug-expressing cells in three-dimensional collagen and thus phenocopied the effects of Snail in pancreatic cancer cells. Additionally, the increased migration and scattering in three-dimensional collagen of Slug-expressing cells following ROCK1/2 inhibition was dependent on ß1-integrin function. Overall, these results demonstrate differential effects of Snail and Slug in pancreatic cancer and identify the interplay between Rho signaling and ß1-integrin that functions to regulate the differential scattering and migration of Snail- and Slug-expressing pancreatic cancer cells.


Subject(s)
Cell Movement/physiology , Integrin beta1/metabolism , Pancreatic Neoplasms/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , rho-Associated Kinases/metabolism , Collagen Type I/metabolism , Humans , Matrix Metalloproteinase 14/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation/physiology , Snail Family Transcription Factors , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tumor Cells, Cultured , rac1 GTP-Binding Protein/metabolism
11.
J Surg Res ; 173(1): 105-12, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22099597

ABSTRACT

Pancreatic adenocarcinoma remains among the most lethal of human malignancies. Overall 5-y survival is less than 5%, and only 20% of patients presenting with localized disease amenable to surgical resection. Even in patients who undergo resection, long-term survival remains extremely poor. A major contributor to the aggressiveness of multiple cancers, and pancreatic cancer in particular, is the process of epithelial-to-mesenchymal transition (EMT). This review highlights the growing evidence of EMT in pancreatic cancer progression, focusing on the contribution of EMT to the development of cancer stem cells and on interaction of EMT with other pathways central to cancer progression, such as Hedgehog signaling, the K-ras oncogene, and transforming growth factor-beta (TGF-ß). We will also discuss EMT-targeting agents currently in development and in clinical trials that may help to reduce the morbidity and mortality associated with pancreatic cancer.


Subject(s)
Disease Progression , Epithelial-Mesenchymal Transition/physiology , Neoplastic Stem Cells/physiology , Pancreatic Neoplasms/physiopathology , Adenocarcinoma/physiopathology , Hedgehog Proteins/physiology , Humans , Signal Transduction/physiology , Transforming Growth Factor beta/physiology
12.
Biochem J ; 441(2): 541-52, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22187935

ABSTRACT

PDAC (pancreatic ductal adenocarcinoma) is among the most deadly of human malignances. A hallmark of the disease is a pronounced collagen-rich fibrotic extracellular matrix known as the desmoplastic reaction. Intriguingly, it is precisely these areas of fibrosis in which human PDAC tumours demonstrate increased expression of a key collagenase, MT1-MMP [membrane-type 1 MMP (matrix metalloproteinase); also known as MMP-14]. Furthermore, a cytokine known to mediate fibrosis in vivo, TGF-ß1 (transforming growth factor-ß1), is up-regulated in human PDAC tumours and can promote MT1-MMP expression. In the present review, we examine the regulation of PDAC progression through the interplay between type I collagen (the most common extracellular matrix present in human PDAC tumours), MT1-MMP and TGF-ß1. Specifically, we examine the way in which signalling events through these pathways mediates invasion, regulates microRNAs and contributes to chemoresistance.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Collagen Type I/metabolism , Pancreatic Neoplasms/metabolism , Carcinoma, Pancreatic Ductal/pathology , Disease Progression , Drug Resistance, Neoplasm/physiology , Fibrosis , Humans , Matrix Metalloproteinase 14/physiology , MicroRNAs/metabolism , Myofibroblasts/metabolism , Pancreatic Neoplasms/pathology , Snail Family Transcription Factors , Transcription Factors/metabolism , Transforming Growth Factor beta/physiology
13.
Mol Cancer Res ; 9(10): 1294-304, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21856775

ABSTRACT

Pancreatic cancer is associated with a pronounced fibrotic reaction that was recently shown to limit delivery of chemotherapy. To identify potential therapeutic targets to overcome this fibrosis, we examined the interplay between fibrosis and the key proteinase membrane type 1-matrix metalloproteinase (MT1-MMP, MMP-14), which is required for growth and invasion in the collagen-rich microenvironment. In this article, we show that compared with control mice (Kras(+)/MT1-MMP(-)) that express an activating Kras(G12D) mutation necessary for pancreatic cancer development, littermate mice that express both MT1-MMP and Kras(G12D) (Kras(+)/MT1-MMP(+)) developed a greater number of large, dysplastic mucin-containing papillary lesions. These lesions were associated with a significant amount of surrounding fibrosis, increased α-smooth muscle actin (+) cells in the stroma, indicative of activated myofibroblasts, and increased Smad2 phosphorylation. To further understand how MT1-MMP promotes fibrosis, we established an in vitro model to examine the effect of expressing MT1-MMP in pancreatic ductal adenocarcinoma (PDAC) cells on stellate cell collagen deposition. Conditioned media from MT1-MMP-expressing PDAC cells grown in three-dimensional collagen enhanced Smad2 nuclear translocation, promoted Smad2 phosphorylation, and increased collagen production by stellate cells. Inhibiting the activity or expression of the TGF-ß type I receptor in stellate cells attenuated MT1-MMP conditioned medium-induced collagen expression by stellate cells. In addition, a function-blocking anti-TGF-ß antibody also inhibited MT1-MMP conditioned medium-induced collagen expression in stellate cells. Overall, we show that the bona fide collagenase MT1-MMP paradoxically contributes to fibrosis by increasing TGF-ß signaling and that targeting MT1-MMP may thus help to mitigate fibrosis.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Matrix Metalloproteinase 14/metabolism , Pancreas/pathology , Pancreatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Transforming Growth Factor beta/metabolism , ras Proteins/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Fibrosis , Humans , Immunohistochemistry , Matrix Metalloproteinase 14/biosynthesis , Matrix Metalloproteinase 14/genetics , Mice , Mice, Transgenic , Pancreas/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras)/biosynthesis , Proto-Oncogene Proteins p21(ras)/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , ras Proteins/biosynthesis , ras Proteins/genetics
14.
J Cell Biochem ; 112(10): 2966-73, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21678462

ABSTRACT

Pancreatic cancer is one of the deadliest of cancers with a dismal 5-year survival rate. Epidemiological studies have identified chronic pancreatitis as a risk factor for pancreatic cancer. Pancreatic cancer cells also demonstrate increased expression of the transcription factor Snail, a key regulator of epithelial-mesenchymal transition. As ethanol is one of the major causes of pancreatitis, we examined the effect of ethanol on Snail family members in immortalized human pancreatic ductal epithelial (HPDE) cells and in pancreatic cancer cells. Ethanol induced Snail mRNA levels 2.5-fold in HPDE cells, with only 1.5-fold mRNA induction of the Snail-related protein slug. In contrast, ethanol increased Slug mRNA levels 1.5- to 2-fold in pancreatic cancer cells, with minimal effect on Snail. Because Snail increases invasion of cancer cells, we examined the effect of ethanol on invasion of HPDE and pancreatic cancer cells. Surprisingly, ethanol decreased invasion of HPDE cells, but had no effect on invasion of pancreatic cancer cells. Mechanistically, ethanol increased adhesion of HPDE cells to collagen and increased expression of the collagen binding α2- and ß1-integrins. In contrast, ethanol did not affect collagen adhesion or integrin expression in pancreatic cancer cells. Also in contrast to HPDE cells, ethanol did not attenuate ERK1/2 phosphorylation in pancreatic cancer cells; however, inhibiting ERK1/2 decreased pancreatic cancer cell invasion. Overall, our results identify the differential effects of ethanol on premalignant and malignant pancreatic cells, and demonstrate the pleiotropic effects of ethanol on pancreatic cancer progression.


Subject(s)
Ethanol/toxicity , Pancreatic Ducts/cytology , Pancreatic Neoplasms/metabolism , Transcription Factors/metabolism , Cell Line , Cell Line, Tumor , Gene Expression/drug effects , Gene Expression/genetics , Humans , Integrin alpha2/metabolism , Integrin beta1/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Snail Family Transcription Factors , Transcription Factors/genetics
15.
J Biol Chem ; 286(12): 10495-504, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21288898

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by pronounced fibrotic reaction composed primarily of type I collagen. Although type I collagen functions as a barrier to invasion, pancreatic cancer cells have been shown to respond to type I collagen by becoming more motile and invasive. Because epithelial-mesenchymal transition is also associated with cancer invasion, we examined the extent to which collagen modulated the expression of Snail, a well known regulator of epithelial-mesenchymal transition. Relative to cells grown on tissue culture plastic, PDAC cells grown in three-dimensional collagen gels induced Snail. Inhibiting the activity or expression of the TGF-ß type I receptor abrogated collagen-induced Snail. Downstream of the receptor, we showed that Smad3 and Smad4 were critical for the induction of Snail by collagen. In contrast, Smad2 or ERK1/2 was not involved in collagen-mediated Snail expression. Overexpression of Snail in PDAC cells resulted in a robust membrane type 1-matrix metalloproteinase (MT1-MMP, MMP-14)-dependent invasion through collagen-coated transwell chambers. Snail-expressing PDAC cells also demonstrated MT1-MMP-dependent scattering in three-dimensional collagen gels. Mechanistically, Snail increased the expression of MT1-MMP through activation of ERK-MAPK signaling, and inhibiting ERK signaling in Snail-expressing cells blocked two-dimensional collagen invasion and attenuated scattering in three-dimensional collagen. To provide in vivo support for our findings that Snail can regulate MT1-MMP, we examined the expression of Snail and MT1-MMP in human PDAC tumors and found a statistically significant positive correlation between MT1-MMP and Snail in these tumors. Overall, our data demonstrate that pancreatic cancer cells increase Snail on encountering collagen-rich milieu and suggest that the desmoplastic reaction actively contributes to PDAC progression.


Subject(s)
Adenocarcinoma/enzymology , Carcinoma, Pancreatic Ductal/enzymology , Collagen Type I/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Matrix Metalloproteinase 14/biosynthesis , Neoplasm Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Transformed , Collagen Type I/genetics , Humans , MAP Kinase Signaling System/genetics , Matrix Metalloproteinase 14/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Smad Proteins/genetics , Smad Proteins/metabolism , Snail Family Transcription Factors , Transcription Factors
16.
Cancer Res ; 71(3): 1019-28, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21148071

ABSTRACT

One of the hallmarks of human pancreatic ductal adenocarcinoma (PDAC) is its pronounced type I collagen-rich fibrotic reaction. Although recent reports have shown that the fibrotic reaction can limit the efficacy of gemcitabine chemotherapy, the underlying mechanisms remain poorly understood. In this article, we show that the type I collagen allows PDAC cells to override checkpoint arrest induced by gemcitabine. Relative to cells grown on tissue culture plastic, PDAC cells grown in 3-dimensional collagen microenvironment have minimal Chk1 phosphorylation and continue to proliferate in the presence of gemcitabine. Collagen increases membrane type 1 matrix metalloproteinase (MT1-MMP)-dependent ERK1/2 phosphorylation to limit the effect of gemcitabine. Collagen also increases MT1-MMP-dependent high mobility group A2 (HMGA2) expression, a nonhistone DNA-binding nuclear protein involved in chromatin remodeling and gene transcription, to attenuate the effect of gemcitabine. Overexpression of MT1-MMP in the collagen microenvironment increases ERK1/2 phosphorylation and HMGA2 expression, and thereby further attenuates gemcitabine-induced checkpoint arrest. MT1-MMP also allows PDAC cells to continue to proliferate in the presence of gemcitabine in a xenograft mouse model. Clinically, human tumors with increased MT1-MMP show increased HMGA2 expression. Overall, our data show that collagen upregulation of MT1-MMP contributes to gemcitabine resistance in vitro and in a xenograft mouse model, and suggest that targeting MT1-MMP could be a novel approach to sensitize pancreatic tumors to gemcitabine.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Collagen Type I/chemistry , Deoxycytidine/analogs & derivatives , HMGA2 Protein/biosynthesis , Matrix Metalloproteinase 14/biosynthesis , Pancreatic Neoplasms/drug therapy , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm , Drug Synergism , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , HMGA2 Protein/genetics , Humans , Matrix Metalloproteinase 14/genetics , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Targeted Therapy/methods , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Up-Regulation/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
17.
J Biol Chem ; 285(51): 40333-41, 2010 Dec 17.
Article in English | MEDLINE | ID: mdl-20956525

ABSTRACT

IFNα exerts potent inhibitory activities against malignant melanoma cells in vitro and in vivo, but the mechanisms by which it generates its antitumor effects remain unknown. We examined the effects of interferon α (IFNα) on the expression of human members of the Schlafen (SLFN) family of genes, a group of cell cycle regulators that mediate growth-inhibitory responses. Using quantitative RT-real time PCR, we found detectable basal expression of all the different human SLFN genes examined (SLFN5, SLFN11, SLFN12, SLFN13, and SLFN14), in malignant melanoma cells and primary normal human melanocytes, but SLFN5 basal expression was suppressed in all analyzed melanoma cell lines. Treatment of melanoma cells with IFNα resulted in induction of expression of SLFN5 in malignant cells, suggesting a potential involvement of this gene in the antitumor effects of IFNα. Importantly, stable knockdown of SLFN5 in malignant melanoma cells resulted in increased anchorage-independent growth, as evidenced by enhanced colony formation in soft agar assays. Moreover, SLFN5 knockdown also resulted in increased invasion in three-dimensional collagen, suggesting a dual role for SLFN5 in the regulation of invasion and anchorage-independent growth of melanoma cells. Altogether, our findings suggest an important role for the SLFN family of proteins in the generation of the anti-melanoma effects of IFNα and for the first time directly implicate a member of the human SLFN family in the regulation of cell invasion.


Subject(s)
Cell Cycle Proteins/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Immunologic Factors/pharmacology , Interferon-alpha/pharmacology , Melanocytes/metabolism , Melanoma/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Melanocytes/pathology , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Neoplasm Invasiveness
18.
J Biol Chem ; 285(36): 28363-72, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20605790

ABSTRACT

Membrane type 1-matrix metalloproteinase (MT1-MMP, MMP14), which is associated with extracellular matrix (ECM) breakdown in squamous cell carcinoma (SCC), promotes tumor formation and epithelial-mesenchymal transition. However, in this report we demonstrate that MT1-MMP, by cleaving the underlying ECM, causes cellular aggregation of keratinocytes and SCC cells. Treatment with an MMP inhibitor abrogated MT1-MMP-induced phenotypic changes, but decreasing E-cadherin expression did not affect MT1-MMP-induced cellular aggregation. As ROCK1/2 can regulate cell-cell and cell-ECM interaction, we examined its role in mediating MT1-MMP-induced phenotypic changes. Blocking ROCK1/2 expression or activity abrogated the cellular aggregation resulting from MT1-MMP expression. Additionally, blocking Rho and non-muscle myosin attenuated MT1-MMP-induced phenotypic changes. Moreover, SCC cells expressing only the catalytically active MT1-MMP protein demonstrated increased cellular aggregation and increased myosin II activity in vivo when injected subcutaneously into nude mice. Together, these results demonstrate that expression of MT1-MMP may be anti-tumorigenic in keratinocytes by promoting cellular aggregation.


Subject(s)
Keratinocytes/cytology , Keratinocytes/metabolism , Matrix Metalloproteinase 14/metabolism , Myosins/metabolism , Signal Transduction , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , Animals , Biocatalysis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Aggregation , Cell Line, Tumor , Cell Size , Gene Expression Regulation, Neoplastic , Humans , Keratinocytes/enzymology , Matrix Metalloproteinase 14/genetics , Mice , Myosin Type II/metabolism , Phenotype
19.
Cancers (Basel) ; 2(4): 2084-97, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-24281219

ABSTRACT

Pancreatic adenocarcinoma (PDAC) is one of the most lethal human malignancies, with median survival of less than one year and overall five-year survival of less than 5%. There is increasing evidence demonstrating that epithelial-mesenchymal transition (EMT) contributes to pancreatic cancer metastasis and to treatment resistance. In this review, we will examine the data demonstrating the role and regulation of EMT in pancreatic cancer progression, focusing particularly on the transcription factors and microRNAs involved in EMT. We will examine how EMT is involved in the generation and maintenance of stem cells, and the role of EMT in modulating resistance of PDAC cells to drug therapies. We will also identify putative EMT-targeting agents that may help to reduce the morbidity and mortality associated with pancreatic cancer.

20.
J Cell Biochem ; 108(3): 726-36, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19681038

ABSTRACT

Members of Snail family of transcription factors play an important role in oral cancer progression by inducing epithelial-mesenchymal transition, by promoting invasion and by increasing matrix metalloproteinase (MMP) expression. Although Snail (Snai1) is the best characterized and the most extensively studied member of this family, the role and regulation of Slug (Snai2) in oral cancer progression is less well understood. In this report, we show that transforming growth factor-beta1 (TGF-beta1) increases Slug levels in tert-immortalized oral keratinocytes and in malignant oral squamous cell carcinoma (OSCC) cells. Inhibiting ERK1/2 signaling, but not PI3-kinase signaling, blocked TGF-beta1-induced Slug expression in the malignant UMSCC1 cells. To further examine the role of Slug in OSCC progression, we generated UMSCC1 cells with inducible expression of Slug protein. Induction of Slug in UMSCC1 cells did not repress E-cadherin levels or regulate individual movement of UMSCC1 cells. Instead, Slug enhanced cohort migration and Matrigel invasion by UMSCC1 cells. Slug increased MMP-9 levels and MMP-9-specific siRNA blocked Slug-induced Matrigel invasion. Interestingly, Slug-specific siRNA attenuated TGF-beta1-induced MMP-9 expression and Matrigel invasion. These data demonstrate that TGF-beta1 increases Slug via ERK1/2 signaling, and thereby contributes to OSCC progression.


Subject(s)
Matrix Metalloproteinase 9/metabolism , Mouth Neoplasms/enzymology , Transcription Factors/metabolism , Transforming Growth Factor beta1/pharmacology , Animals , Cadherins/metabolism , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/drug effects , Collagen/metabolism , Drug Combinations , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Laminin/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mouth Neoplasms/pathology , Neoplasm Invasiveness , Proteoglycans/metabolism , Snail Family Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...