Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
3.
Nat Chem Biol ; 18(12): 1388-1398, 2022 12.
Article in English | MEDLINE | ID: mdl-36097295

ABSTRACT

The Janus tyrosine kinase (JAK) family of non-receptor tyrosine kinases includes four isoforms (JAK1, JAK2, JAK3, and TYK2) and is responsible for signal transduction downstream of diverse cytokine receptors. JAK inhibitors have emerged as important therapies for immun(onc)ological disorders, but their use is limited by undesirable side effects presumed to arise from poor isoform selectivity, a common challenge for inhibitors targeting the ATP-binding pocket of kinases. Here we describe the chemical proteomic discovery of a druggable allosteric cysteine present in the non-catalytic pseudokinase domain of JAK1 (C817) and TYK2 (C838), but absent from JAK2 or JAK3. Electrophilic compounds selectively engaging this site block JAK1-dependent trans-phosphorylation and cytokine signaling, while appearing to act largely as 'silent' ligands for TYK2. Importantly, the allosteric JAK1 inhibitors do not impair JAK2-dependent cytokine signaling and are inactive in cells expressing a C817A JAK1 mutant. Our findings thus reveal an allosteric approach for inhibiting JAK1 with unprecedented isoform selectivity.


Subject(s)
Cysteine , Proteomics , Signal Transduction , Cytokines , Protein Isoforms
4.
Diabetes ; 68(9): 1806-1818, 2019 09.
Article in English | MEDLINE | ID: mdl-31201281

ABSTRACT

Transcription factors positively and/or negatively impact gene expression by recruiting coregulatory factors, which interact through protein-protein binding. Here we demonstrate that mouse pancreas size and islet ß-cell function are controlled by the ATP-dependent Swi/Snf chromatin remodeling coregulatory complex that physically associates with Pdx1, a diabetes-linked transcription factor essential to pancreatic morphogenesis and adult islet cell function and maintenance. Early embryonic deletion of just the Swi/Snf Brg1 ATPase subunit reduced multipotent pancreatic progenitor cell proliferation and resulted in pancreas hypoplasia. In contrast, removal of both Swi/Snf ATPase subunits, Brg1 and Brm, was necessary to compromise adult islet ß-cell activity, which included whole-animal glucose intolerance, hyperglycemia, and impaired insulin secretion. Notably, lineage-tracing analysis revealed Swi/Snf-deficient ß-cells lost the ability to produce the mRNAs for Ins and other key metabolic genes without effecting the expression of many essential islet-enriched transcription factors. Swi/Snf was necessary for Pdx1 to bind to the Ins gene enhancer, demonstrating the importance of this association in mediating chromatin accessibility. These results illustrate how fundamental the Pdx1:Swi/Snf coregulator complex is in the pancreas, and we discuss how disrupting their association could influence type 1 and type 2 diabetes susceptibility.


Subject(s)
Cell Proliferation/physiology , Chromatin Assembly and Disassembly/physiology , DNA Helicases/metabolism , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/metabolism , Nuclear Proteins/metabolism , Pancreas/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , DNA Helicases/genetics , Gene Expression Regulation , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Homeodomain Proteins/genetics , Insulin/blood , Insulin-Secreting Cells/cytology , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Pancreas/cytology , Trans-Activators/genetics , Transcription Factors/genetics
5.
J Clin Invest ; 128(8): 3475-3489, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30010625

ABSTRACT

Chromatin remodeler Brahma related gene 1 (BRG1) is silenced in approximately 10% of human pancreatic ductal adenocarcinomas (PDAs). We previously showed that BRG1 inhibits the formation of intraductal pancreatic mucinous neoplasm (IPMN) and that IPMN-derived PDA originated from ductal cells. However, the role of BRG1 in pancreatic intraepithelial neoplasia-derived (PanIN-derived) PDA that originated from acinar cells remains elusive. Here, we found that exclusive elimination of Brg1 in acinar cells of Ptf1a-CreER; KrasG12D; Brg1fl/fl mice impaired the formation of acinar-to-ductal metaplasia (ADM) and PanIN independently of p53 mutation, while PDA formation was inhibited in the presence of p53 mutation. BRG1 bound to regions of the Sox9 promoter to regulate its expression and was critical for recruitment of upstream regulators, including PDX1, to the Sox9 promoter and enhancer in acinar cells. SOX9 expression was downregulated in BRG1-depleted ADMs/PanINs. Notably, Sox9 overexpression canceled this PanIN-attenuated phenotype in KBC mice. Furthermore, Brg1 deletion in established PanIN by using a dual recombinase system resulted in regression of the lesions in mice. Finally, BRG1 expression correlated with SOX9 expression in human PDAs. In summary, BRG1 is critical for PanIN initiation and progression through positive regulation of SOX9. Thus, the BRG1/SOX9 axis is a potential target for PanIN-derived PDA.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cell Transformation, Neoplastic/metabolism , DNA Helicases/biosynthesis , Nuclear Proteins/biosynthesis , Pancreatic Neoplasms/metabolism , SOX9 Transcription Factor/metabolism , Signal Transduction , Transcription Factors/biosynthesis , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , DNA Helicases/genetics , Female , Gene Expression Regulation , Humans , Male , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Response Elements , SOX9 Transcription Factor/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Pancreatic Neoplasms
6.
Epigenetics ; 13(4): 449-457, 2018.
Article in English | MEDLINE | ID: mdl-30056798

ABSTRACT

International experts gathered at the Mayo Clinic (Rochester MN, USA) on February 27th-28th, 2017 for a meeting entitled 'Basic and Translational Facets of the Epigenetics of GI Diseases'. This workshop summarized recent advances on the role of epigenetics in the pathobiology of gastrointestinal (GI) diseases. Highlights of the meeting included recent advances on the involvement of different epigenetic mechanisms in malignant and nonmalignant GI disorders and the epigenetic heterogeneity exhibited in these diseases. The translational value of epigenetic drugs, as well as the current and future use of epigenetic changes (i.e., DNA methylation patterns) as biomarkers for early detection tools or disease stratification were also important topics of discussion.


Subject(s)
Epigenesis, Genetic , Gastrointestinal Diseases/genetics , DNA Methylation , Genetic Heterogeneity , Genetic Markers , Humans , Translational Research, Biomedical
7.
Nat Commun ; 9(1): 485, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29396395

ABSTRACT

Pancreatic ß cells are highly specialized to regulate systemic glucose levels by secreting insulin. In adults, increase in ß-cell mass is limited due to brakes on cell replication. In contrast, proliferation is robust in neonatal ß cells that are functionally immature as defined by a lower set point for glucose-stimulated insulin secretion. Here we show that ß-cell proliferation and immaturity are linked by tuning expression of physiologically relevant, non-oncogenic levels of c-Myc. Adult ß cells induced to replicate adopt gene expression and metabolic profiles resembling those of immature neonatal ß that proliferate readily. We directly demonstrate that priming insulin-producing cells to enter the cell cycle promotes a functionally immature phenotype. We suggest that there exists a balance between mature functionality and the ability to expand, as the phenotypic state of the ß cell reverts to a less functional one in response to proliferative cues.


Subject(s)
Cell Proliferation/genetics , Insulin-Secreting Cells/cytology , Proto-Oncogene Proteins c-myc/genetics , Animals , Cell Cycle , Cell Differentiation/genetics , Cell Division/genetics , Gene Expression , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Mice , Mice, Transgenic , Phenotype
8.
Pancreatology ; 17(3): 350-353, 2017.
Article in English | MEDLINE | ID: mdl-28473229

ABSTRACT

OBJECTIVES: Pancreatic ductal adenocarcinoma (PDAC) is thought to derive from different precursor lesions including the recently identified atypical flat lesions (AFL). While all precursor lesions and PDAC share ductal characteristics, there is an ongoing debate about the cellular origin of the different PDAC precursor lesions. In particular, pancreatic acinar cells have previously been shown to display a remarkable plasticity being able to undergo ductal dedifferentiation in the context of oncogenic stimuli. METHODS: Histological analyses were performed in a murine PDAC model that specifically expresses oncogenic Kras in adult pancreatic acinar cells. Occurrence, characterization, and lineage tracing of AFLs were investigated. RESULTS: Upon expression of oncogenic Kras in adult pancreatic acinar cells, AFLs with typical morphology and expression profile arise. Lineage tracing confirmed that the AFLs were of acinar origin. CONCLUSIONS: Using a murine PDAC model, this study identifies pancreatic acinar cells as a cellular source for AFLs.


Subject(s)
Acinar Cells/pathology , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Animals , Carcinoma, Pancreatic Ductal/chemically induced , Carcinoma, Pancreatic Ductal/metabolism , Cell Differentiation , Cell Transformation, Neoplastic/pathology , Estrogen Antagonists , Immunohistochemistry , Mice , Pancreatic Neoplasms/chemically induced , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/biosynthesis , Proto-Oncogene Proteins p21(ras)/genetics , Tamoxifen
10.
Clin Cancer Res ; 23(16): 4865-4874, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28424202

ABSTRACT

Purpose: Pancreatic cysts are estimated to be present in 2%-3% of the adult population. Unfortunately, current diagnostics do not accurately distinguish benign cysts from those that can progress into invasive cancer. Misregulated pericellular proteolysis is a hallmark of malignancy, and therefore, we used a global approach to discover protease activities that differentiate benign nonmucinous cysts from premalignant mucinous cysts.Experimental Design: We employed an unbiased and global protease profiling approach to discover protease activities in 23 cyst fluid samples. The distinguishing activities of select proteases was confirmed in 110 samples using specific fluorogenic substrates and required less than 5 µL of cyst fluid.Results: We determined that the activities of the aspartyl proteases gastricsin and cathepsin E are highly increased in fluid from mucinous cysts. IHC analysis revealed that gastricsin expression was associated with regions of low-grade dysplasia, whereas cathepsin E expression was independent of dysplasia grade. Gastricsin activity differentiated mucinous from nonmucinous cysts with a specificity of 100% and a sensitivity of 93%, whereas cathepsin E activity was 92% specific and 70% sensitive. Gastricsin significantly outperformed the most widely used molecular biomarker, carcinoembryonic antigen (CEA), which demonstrated 94% specificity and 65% sensitivity. Combined analysis of gastricsin and CEA resulted in a near perfect classifier with 100% specificity and 98% sensitivity.Conclusions: Quantitation of gastricsin and cathepsin E activities accurately distinguished mucinous from nonmucinous pancreatic cysts and has the potential to replace current diagnostics for analysis of these highly prevalent lesions. Clin Cancer Res; 23(16); 4865-74. ©2017 AACR.


Subject(s)
Cyst Fluid/enzymology , Pancreatic Cyst/enzymology , Pancreatic Neoplasms/enzymology , Peptide Hydrolases/metabolism , Animals , Biomarkers, Tumor/metabolism , Carcinoembryonic Antigen/metabolism , Cathepsin E/metabolism , Diagnosis, Differential , Fluorescent Dyes/metabolism , Humans , Mice, Knockout , Mice, Transgenic , Neoplasms, Cystic, Mucinous, and Serous/diagnosis , Neoplasms, Cystic, Mucinous, and Serous/enzymology , Pancreatic Cyst/diagnosis , Pancreatic Neoplasms/diagnosis , Pancreatic Pseudocyst/diagnosis , Pancreatic Pseudocyst/enzymology , Pepsin A/metabolism , Retrospective Studies , Sensitivity and Specificity
11.
Cancer Res ; 76(11): 3351-63, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27032419

ABSTRACT

Aberrant regulation of cellular extrusion can promote invasion and metastasis. Here, we identify molecular requirements for early cellular invasion using a premalignant mouse model of pancreatic cancer with conditional knockout of p120 catenin (Ctnnd1). Mice with biallelic loss of p120 catenin progressively develop high-grade pancreatic intraepithelial neoplasia (PanIN) lesions and neoplasia accompanied by prominent acute and chronic inflammatory processes, which is mediated, in part, through NF-κB signaling. Loss of p120 catenin in the context of oncogenic Kras also promotes remarkable apical and basal epithelial cell extrusion. Abundant single epithelial cells exit PanIN epithelium basally, retain epithelial morphology, survive, and display features of malignancy. Similar extrusion defects are observed following p120 catenin knockdown in vitro, and these effects are completely abrogated by the activation of S1P/S1pr2 signaling. In the context of oncogenic Kras, p120 catenin loss significantly reduces expression of genes mediating S1P/S1pr2 signaling in vivo and in vitro, and this effect is mediated at least, in part, through activation of NF-κB. These results provide insight into mechanisms controlling early events in the metastatic process and suggest that p120 catenin and S1P/S1pr2 signaling enhance cancer progression by regulating epithelial cell invasion. Cancer Res; 76(11); 3351-63. ©2016 AACR.


Subject(s)
Carcinoma in Situ/pathology , Carcinoma, Pancreatic Ductal/pathology , Catenins/metabolism , Epithelial Cells/pathology , Metaplasia/pathology , Pancreatic Neoplasms/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma in Situ/genetics , Carcinoma in Situ/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Catenins/genetics , Cell Proliferation , Epithelial Cells/metabolism , Humans , Metaplasia/genetics , Metaplasia/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Invasiveness , Neoplasm Staging , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Cells, Cultured , Delta Catenin
12.
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
13.
Genes Dev ; 30(24): 2669-2683, 2016 12 15.
Article in English | MEDLINE | ID: mdl-28087712

ABSTRACT

Aberrant activation of embryonic signaling pathways is frequent in pancreatic ductal adenocarcinoma (PDA), making developmental regulators therapeutically attractive. Here we demonstrate diverse functions for pancreatic and duodenal homeobox 1 (PDX1), a transcription factor indispensable for pancreas development, in the progression from normal exocrine cells to metastatic PDA. We identify a critical role for PDX1 in maintaining acinar cell identity, thus resisting the formation of pancreatic intraepithelial neoplasia (PanIN)-derived PDA. Upon neoplastic transformation, the role of PDX1 changes from tumor-suppressive to oncogenic. Interestingly, subsets of malignant cells lose PDX1 expression while undergoing epithelial-to-mesenchymal transition (EMT), and PDX1 loss is associated with poor outcome. This stage-specific functionality arises from profound shifts in PDX1 chromatin occupancy from acinar cells to PDA. In summary, we report distinct roles of PDX1 at different stages of PDA, suggesting that therapeutic approaches against this potential target need to account for its changing functions at different stages of carcinogenesis. These findings provide insight into the complexity of PDA pathogenesis and advocate a rigorous investigation of therapeutically tractable targets at distinct phases of PDA development and progression.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Pancreatic Neoplasms/genetics , Trans-Activators/metabolism , Acinar Cells/pathology , Animals , Carcinoma, Pancreatic Ductal/physiopathology , Gene Deletion , Homeodomain Proteins/genetics , Humans , Mice , Pancreatic Neoplasms/physiopathology , Tissue Array Analysis , Trans-Activators/genetics , Tumor Cells, Cultured
14.
Dev Cell ; 35(6): 674-84, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26702828

ABSTRACT

Neoplastic transformation requires changes in cellular identity. Emerging evidence increasingly points to cellular reprogramming, a process during which fully differentiated and functional cells lose aspects of their identity while gaining progenitor characteristics, as a critical early step during cancer initiation. This cell identity crisis persists even at the malignant stage in certain cancers, suggesting that reactivation of progenitor functions supports tumorigenicity. Here, we review recent findings that establish the essential role of cellular reprogramming during neoplastic transformation and the major players involved in it with a special emphasis on pancreatic cancer.


Subject(s)
Cell Differentiation/physiology , Cell Transformation, Neoplastic/pathology , Cellular Reprogramming/genetics , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/pathology , Animals , Cell Lineage/physiology , Humans
15.
Sci Rep ; 5: 9841, 2015 Apr 29.
Article in English | MEDLINE | ID: mdl-25923013

ABSTRACT

Metastasis is responsible for over 90% of cancer-associated mortality. In epithelial carcinomas, a key process in metastatic progression is the epigenetic reprogramming of an epithelial-to-mesenchymal transition-like (EMT) change towards invasive cellular phenotypes. In non-epithelial cancers, different mechanisms must underlie metastatic change, but relatively little is known about the factors involved. Here, we identify the chromatin regulatory Sirtuin factor SIRT7 as a key regulator of metastatic phenotypes in both epithelial and mesenchymal cancer cells. In epithelial prostate carcinomas, high SIRT7 levels are associated with aggressive cancer phenotypes, metastatic disease, and poor patient prognosis, and depletion of SIRT7 can reprogram these cells to a less aggressive phenotype. Interestingly, SIRT7 is also important for maintaining the invasiveness and metastatic potential of non-epithelial sarcoma cells. Moreover, SIRT7 inactivation dramatically suppresses cancer cell metastasis in vivo, independent of changes in primary tumor growth. Mechanistically, we also uncover a novel link between SIRT7 and its family member SIRT1, providing the first demonstration of direct interaction and functional interplay between two mammalian sirtuins. Together with previous work, our findings highlight the broad role of SIRT7 in maintaining the metastatic cellular phenotype in diverse cancers.


Subject(s)
Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Sarcoma/genetics , Sirtuins/genetics , Cell Line, Tumor , Chromatin/genetics , Disease Progression , Epigenesis, Genetic/genetics , Humans , Phenotype , Prognosis , Sarcoma/pathology
16.
Genes Dev ; 29(6): 658-71, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25792600

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) develops predominantly through pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasm (IPMN) precursor lesions. Pancreatic acinar cells are reprogrammed to a "ductal-like" state during PanIN-PDA formation. Here, we demonstrate a parallel mechanism operative in mature duct cells during which functional cells undergo "ductal retrogression" to form IPMN-PDA. We further identify critical antagonistic roles for Brahma-related gene 1 (Brg1), a catalytic subunit of the SWI/SNF complexes, during IPMN-PDA development. In mature duct cells, Brg1 inhibits the dedifferentiation that precedes neoplastic transformation, thus attenuating tumor initiation. In contrast, Brg1 promotes tumorigenesis in full-blown PDA by supporting a mesenchymal-like transcriptional landscape. We further show that JQ1, a drug that is currently being tested in clinical trials for hematological malignancies, impairs PDA tumorigenesis by both mimicking some and inhibiting other Brg1-mediated functions. In summary, our study demonstrates the context-dependent roles of Brg1 and points to potential therapeutic treatment options based on epigenetic regulation in PDA.


Subject(s)
Carcinoma, Pancreatic Ductal/physiopathology , Cell Transformation, Neoplastic/genetics , DNA Helicases/metabolism , Nuclear Proteins/metabolism , Pancreatic Neoplasms/physiopathology , Transcription Factors/metabolism , Animals , Azepines/pharmacology , Azepines/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Cell Transformation, Neoplastic/drug effects , DNA Helicases/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice , Nuclear Proteins/genetics , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins p21(ras)/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Transcription Factors/genetics , Triazoles/pharmacology , Triazoles/therapeutic use , Tumor Cells, Cultured
17.
Nat Cell Biol ; 16(3): 255-67, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24561622

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) develops through distinct precursor lesions, including pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasia (IPMN). However, genetic features resulting in IPMN-associated PDA (IPMN-PDA) versus PanIN-associated PDA (PanIN-PDA) are largely unknown. Here we find that loss of Brg1, a core subunit of SWI/SNF chromatin remodelling complexes, cooperates with oncogenic Kras to form cystic neoplastic lesions that resemble human IPMN and progress to PDA. Although Brg1-null IPMN-PDA develops rapidly, it possesses a distinct transcriptional profile compared with PanIN-PDA driven by mutant Kras and hemizygous p53 deletion. IPMN-PDA also is less lethal, mirroring prognostic trends in PDA patients. In addition, Brg1 deletion inhibits Kras-dependent PanIN development from adult acinar cells, but promotes Kras-driven preneoplastic transformation in adult duct cells. Therefore, this study implicates Brg1 as a determinant of context-dependent Kras-driven pancreatic tumorigenesis and suggests that chromatin remodelling may underlie the development of distinct PDA subsets.


Subject(s)
Adenocarcinoma, Mucinous/metabolism , Carcinoma, Pancreatic Ductal/metabolism , DNA Helicases/physiology , Nuclear Proteins/physiology , Pancreatic Neoplasms/metabolism , Transcription Factors/physiology , Adenocarcinoma, Mucinous/pathology , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation , Chromatin Assembly and Disassembly , Female , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Pancreas/pathology , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics
18.
Cancer Res ; 73(12): 3771-82, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23610444

ABSTRACT

Colon cancer is one of the deadliest cancers worldwide because of its metastasis to other essential organs. Metastasis of colon cancer involves a complex set of events, including epithelial-to-mesenchymal transition (EMT) that increases invasiveness of the tumor cells. Here, we show that the xeroderma pigmentosum group E (XPE) gene product, damaged DNA-binding protein (DDB)-2, is downregulated in high-grade colon cancers, and it plays a dominant role in the suppression of EMT of the colon cancer cells. Depletion of DDB2 promotes mesenchymal phenotype, whereas expression of DDB2 promotes epithelial phenotype. DDB2 constitutively represses genes that are the key activators of EMT, indicating that DDB2 is a master regulator of EMT of the colon cancer cells. Moreover, we observed evidence that DDB2 functions as a barrier for EMT induced by hypoxia and TGF-ß. Also, we provide evidence that DDB2 inhibits metastasis of colon cancer. The results presented here identify a transcriptional regulatory pathway of DDB2 that is directly linked to the mechanisms that suppress metastasis of colon cancer.


Subject(s)
Colonic Neoplasms/genetics , DNA-Binding Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Animals , Blotting, Western , Cadherins/metabolism , Cell Hypoxia , Cell Line, Tumor , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA-Binding Proteins/metabolism , HCT116 Cells , Humans , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Male , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Mice, SCID , Neoplasm Invasiveness , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
19.
Cancer Biol Ther ; 14(2): 108-16, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23114715

ABSTRACT

Phenethyl isothiocyanate (PEITC) is a promising cancer chemopreventive agent commonly found in edible cruciferous vegetables. It has been implicated also for therapy, and is in clinical trial for lung cancer. Here, we provide evidence that the tumor suppressive effect of PEITC is related to its ability to induce expression of damaged DNA binding protein 2 (DDB2), a DNA repair protein involved also in apoptosis and premature senescence. DDB2 expression is attenuated in a wide variety of cancers including the aggressive colon cancers. We show that, in colon cancer cells, reactive oxygen species, which are induced by PEITC, augment expression of DDB2 through the p38MAPK/JNK pathway, independently of p53. PEITC-induced expression of DDB2 is critical for inhibition of tumor progression by PEITC. Tumors derived from DDB2-deficient colon cancer cells are refractory to PEITC-treatments, resulting from deficiencies in apoptosis and senescence. The DDB2-proficient tumors, on the other hand, respond effectively to PEITC. The results show that PEITC can be used to induce expression of DDB2, and that expression of DDB2 is critical for effective response of tumors to PEITC.


Subject(s)
Anticarcinogenic Agents/therapeutic use , DNA-Binding Proteins/genetics , Isothiocyanates/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Animals , Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isothiocyanates/pharmacology , Male , Mice , Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
20.
Int J Mol Sci ; 13(9): 11012-11026, 2012.
Article in English | MEDLINE | ID: mdl-23109835

ABSTRACT

Premature senescence induced by DNA damage or oncogene is a critical mechanism of tumor suppression. Reactive oxygen species (ROS) have been implicated in the induction of premature senescence response. Several pathological disorders such as cancer, aging and age related neurological abnormalities have been linked to ROS deregulation. Here, we discuss how Damaged DNA binding Protein-2 (DDB2), a nucleotide excision repair protein, plays an important role in ROS regulation by epigenetically repressing the antioxidant genes MnSOD and Catalase. We further revisit a model in which DDB2 plays an instrumental role in DNA damage induced ROS accumulation, ROS induced premature senescence and inhibition of skin tumorigenesis.


Subject(s)
Aging, Premature/genetics , Cellular Senescence/genetics , DNA Damage/genetics , DNA-Binding Proteins/genetics , Reactive Oxygen Species/metabolism , Animals , Apoptosis/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Catalase/biosynthesis , Catalase/genetics , Cell Transformation, Neoplastic , Cullin Proteins/metabolism , DNA Repair/genetics , DNA-Binding Proteins/metabolism , Humans , Mice , Signal Transduction/genetics , Skin Neoplasms/pathology , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , Xeroderma Pigmentosum/genetics
SELECTION OF CITATIONS
SEARCH DETAIL