Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
J Med Chem ; 66(24): 16939-16952, 2023 12 28.
Article in English | MEDLINE | ID: mdl-38096359

ABSTRACT

In this study, a one-pot synthesis via photoinduced C(sp2)-C(sp3) coupling followed by amide formation to access proteolysis targeting chimeras (PROTACs) was developed. The described protocol was studied on cereblon (CRBN)-based E3-ligase binders and (+)-JQ-1, a bromodomain inhibitor, to generate BET (bromodomain and extra terminal domain) targeting protein degraders. The generated PROTACs were profiled in vitro and tested for their degradation ability with several potent candidates identified. Upfront, the individual reactions of the one-pot transformation were carefully optimized for CRBN binder functionalization and multiple heterobifunctional linker moieties were designed and synthesized. Separate scopes detailing the C(sp2)-C(sp3) coupling and one-pot PROTAC synthesis are described in this report as well as a library miniaturization study showing the high-throughput compatibility. Overall, the developed protocol provides rapid access to PROTACs in a single process, thereby allowing efficient generation of CRBN-based PROTAC libraries.


Subject(s)
Proteolysis Targeting Chimera , Ubiquitin-Protein Ligases , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Amides
2.
ChemMedChem ; 18(6): e202200686, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36649575

ABSTRACT

The bromodomain and PHD-finger containing transcription factor (BPTF) is part of the nucleosome remodeling factor (NURF) complex and has been implicated in multiple cancer types. Here, we report the discovery of a potent and selective chemical probe targeting the bromodomain of BPTF with an attractive pharmacokinetic profile enabling cellular and in vivo experiments in mice. Microarray-based transcriptomics in presence of the probe in two lung cancer cell lines revealed only minor effects on the transcriptome. Profiling against a panel of cancer cell lines revealed that the antiproliferative effect does not correlate with BPTF dependency score in depletion screens. Both observations and the multi-domain architecture of BPTF suggest that depleting the protein by proteolysis targeting chimeras (PROTACs) could be a promising strategy to target cancer cell proliferation. We envision that the presented chemical probe and the related negative control will enable the research community to further explore scientific hypotheses with respect to BPTF bromodomain inhibition.


Subject(s)
Lung Neoplasms , Transcription Factors , Animals , Mice , Cell Proliferation , Gene Expression Regulation , Nuclear Proteins/metabolism , Transcription Factors/metabolism
3.
Gut ; 72(2): 345-359, 2023 02.
Article in English | MEDLINE | ID: mdl-35428659

ABSTRACT

OBJECTIVE: The lysyl oxidase-like protein 2 (LOXL2) contributes to tumour progression and metastasis in different tumour entities, but its role in pancreatic ductal adenocarcinoma (PDAC) has not been evaluated in immunocompetent in vivo PDAC models. DESIGN: Towards this end, we used PDAC patient data sets, patient-derived xenograft in vivo and in vitro models, and four conditional genetically-engineered mouse models (GEMMS) to dissect the role of LOXL2 in PDAC. For GEMM-based studies, K-Ras +/LSL-G12D;Trp53 LSL-R172H;Pdx1-Cre mice (KPC) and the K-Ras +/LSL-G12D;Pdx1-Cre mice (KC) were crossed with Loxl2 allele floxed mice (Loxl2Exon2 fl/fl) or conditional Loxl2 overexpressing mice (R26Loxl2 KI/KI) to generate KPCL2KO or KCL2KO and KPCL2KI or KCL2KI mice, which were used to study overall survival; tumour incidence, burden and differentiation; metastases; epithelial to mesenchymal transition (EMT); stemness and extracellular collagen matrix (ECM) organisation. RESULTS: Using these PDAC mouse models, we show that while Loxl2 ablation had little effect on primary tumour development and growth, its loss significantly decreased metastasis and increased overall survival. We attribute this effect to non-cell autonomous factors, primarily ECM remodelling. Loxl2 overexpression, on the other hand, promoted primary and metastatic tumour growth and decreased overall survival, which could be linked to increased EMT and stemness. We also identified tumour-associated macrophage-secreted oncostatin M (OSM) as an inducer of LOXL2 expression, and show that targeting macrophages in vivo affects Osm and Loxl2 expression and collagen fibre alignment. CONCLUSION: Taken together, our findings establish novel pathophysiological roles and functions for LOXL2 in PDAC, which could be potentially exploited to treat metastatic disease.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Mice , Animals , Epithelial-Mesenchymal Transition/genetics , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Disease Models, Animal , Macrophages/metabolism , Amino Acid Oxidoreductases/genetics , Pancreatic Neoplasms
4.
Gut ; 72(3): 535-548, 2023 03.
Article in English | MEDLINE | ID: mdl-36109153

ABSTRACT

OBJECTIVE: GATA6 is a key regulator of the classical phenotype in pancreatic ductal adenocarcinoma (PDAC). Low GATA6 expression associates with poor patient outcome. GATA4 is the second most expressed GATA factor in the pancreas. We assessed whether, and how, GATA4 contributes to PDAC phenotype and analysed the association of expression with outcome and response to chemotherapy. DESIGN: We analysed PDAC transcriptomic data, stratifying cases according to GATA4 and GATA6 expression and identified differentially expressed genes and pathways. The genome-wide distribution of GATA4 was assessed, as well as the effects of GATA4 knockdown. A multicentre tissue microarray study to assess GATA4 and GATA6 expression in samples (n=745) from patients with resectable was performed. GATA4 and GATA6 levels were dichotomised into high/low categorical variables; association with outcome was assessed using univariable and multivariable Cox regression models. RESULTS: GATA4 messenger RNA is enriched in classical, compared with basal-like tumours. We classified samples in 4 groups as high/low for GATA4 and GATA6. Reduced expression of GATA4 had a minor transcriptional impact but low expression of GATA4 enhanced the effects of GATA6 low expression. GATA4 and GATA6 display a partially overlapping genome-wide distribution, mainly at promoters. Reduced expression of both proteins in tumours was associated with the worst patient survival. GATA4 and GATA6 expression significantly decreased in metastases and negatively correlated with basal markers. CONCLUSIONS: GATA4 and GATA6 cooperate to maintain the classical phenotype. Our findings provide compelling rationale to assess their expression as biomarkers of poor prognosis and therapeutic response.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/pathology , Pancreas/pathology , Carcinoma, Pancreatic Ductal/pathology , Gene Expression Profiling , GATA6 Transcription Factor/genetics , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism
5.
Gut ; 71(4): 766-777, 2022 04.
Article in English | MEDLINE | ID: mdl-33846140

ABSTRACT

OBJECTIVE: Molecular taxonomy of tumours is the foundation of personalised medicine and is becoming of paramount importance for therapeutic purposes. Four transcriptomics-based classification systems of pancreatic ductal adenocarcinoma (PDAC) exist, which consistently identified a subtype of highly aggressive PDACs with basal-like features, including ΔNp63 expression and loss of the epithelial master regulator GATA6. We investigated the precise molecular events driving PDAC progression and the emergence of the basal programme. DESIGN: We combined the analysis of patient-derived transcriptomics datasets and tissue samples with mechanistic experiments using a novel dual-recombinase mouse model for Gata6 deletion at late stages of KRasG12D-driven pancreatic tumorigenesis (Gata6LateKO). RESULTS: This comprehensive human-to-mouse approach showed that GATA6 loss is necessary, but not sufficient, for the expression of ΔNp63 and the basal programme in patients and in mice. The concomitant loss of HNF1A and HNF4A, likely through epigenetic silencing, is required for the full phenotype switch. Moreover, Gata6 deletion in mice dramatically increased the metastatic rate, with a propensity for lung metastases. Through RNA-Seq analysis of primary cells isolated from mouse tumours, we show that Gata6 inhibits tumour cell plasticity and immune evasion, consistent with patient-derived data, suggesting that GATA6 works as a barrier for acquiring the fully developed basal and metastatic phenotype. CONCLUSIONS: Our work provides both a mechanistic molecular link between the basal phenotype and metastasis and a valuable preclinical tool to investigate the most aggressive subtype of PDAC. These data, therefore, are important for understanding the pathobiological features underlying the heterogeneity of pancreatic cancer in both mice and human.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Carcinoma, Pancreatic Ductal/pathology , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Mice , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms
6.
Cancers (Basel) ; 12(8)2020 Jul 22.
Article in English | MEDLINE | ID: mdl-32707920

ABSTRACT

Hypoxia is a common phenomenon that occurs in most solid tumors. Regardless of tumor origin, the evolution of a hypoxia-adapted phenotype is critical for invasive cancer development. Pancreatic ductal adenocarcinoma is also characterized by hypoxia, desmoplasia, and the presence of necrosis, predicting poor outcome. Carbonic anhydrase IX (CAIX) is one of the most strict hypoxia regulated genes which plays a key role in the adaptation of cancer cells to hypoxia and acidosis. Here, we summarize clinical data showing that CAIX expression is associated with tumor necrosis, vascularization, expression of Frizzled-1, mucins, or proteins involved in glycolysis, and inevitably, poor prognosis of pancreatic cancer patients. We also describe the transcriptional regulation of CAIX in relation to signaling pathways activated in pancreatic cancers. A large part deals with the preclinical evidence supporting the relevance of CAIX in processes leading to the aggressive behavior of pancreatic tumors. Furthermore, we focus on CAIX occurrence in pre-cancerous lesions, and for the first time, we describe CAIX expression within intraductal papillary mucinous neoplasia. Our review concludes with a detailed account of clinical trials implicating that treatment consisting of conventionally used therapies combined with CAIX targeting could result in an improved anti-cancer response in pancreatic cancer patients.

7.
Oncogene ; 39(32): 5455-5467, 2020 08.
Article in English | MEDLINE | ID: mdl-32587399

ABSTRACT

As the catalog of oncogenic driver mutations is expanding, it becomes clear that alterations in a given gene might have different functions and should not be lumped into one class. The transcription factor GATA3 is a paradigm of this. We investigated the functions of the most common GATA3 mutation (X308_Splice) and five additional mutations, which converge into a neoprotein that we called "neoGATA3," associated with excellent prognosis in patients. Analysis of available molecular data from >3000 breast cancer patients revealed a dysregulation of the ER-dependent transcriptional response in tumors carrying neoGATA3-generating mutations. Mechanistic studies in vitro showed that neoGATA3 interferes with the transcriptional programs controlled by estrogen and progesterone receptors, without fully abrogating them. ChIP-Seq analysis indicated that ER binding is reduced in neoGATA3-expressing cells, especially at distal regions, suggesting that neoGATA3 interferes with the fine tuning of ER-dependent gene expression. This has opposite outputs in distinct hormonal context, having pro- or anti-proliferative effects, depending on the estrogen/progesterone ratio. Our data call for functional analyses of putative cancer drivers to guide clinical application.


Subject(s)
Breast Neoplasms/genetics , GATA3 Transcription Factor/genetics , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/physiology , Female , GATA3 Transcription Factor/immunology , GATA3 Transcription Factor/metabolism , Humans , Mutation , Oncogenes , RNA Splicing , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Messenger/metabolism , Receptors, Estrogen/immunology , Receptors, Estrogen/metabolism , Receptors, Progesterone/immunology , Receptors, Progesterone/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/pathology
8.
Nat Commun ; 11(1): 2682, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32472071

ABSTRACT

Pancreatic cancer stem cells (PaCSCs) drive pancreatic cancer tumorigenesis, chemoresistance and metastasis. While eliminating this subpopulation of cells would theoretically result in tumor eradication, PaCSCs are extremely plastic and can successfully adapt to targeted therapies. In this study, we demonstrate that PaCSCs increase expression of interferon-stimulated gene 15 (ISG15) and protein ISGylation, which are essential for maintaining their metabolic plasticity. CRISPR-mediated ISG15 genomic editing reduces overall ISGylation, impairing PaCSCs self-renewal and their in vivo tumorigenic capacity. At the molecular level, ISG15 loss results in decreased mitochondrial ISGylation concomitant with increased accumulation of dysfunctional mitochondria, reduced oxidative phosphorylation (OXPHOS) and impaired mitophagy. Importantly, disruption in mitochondrial metabolism affects PaCSC metabolic plasticity, making them susceptible to prolonged inhibition with metformin in vivo. Thus, ISGylation is critical for optimal and efficient OXPHOS by ensuring the recycling of dysfunctional mitochondria, and when absent, a dysregulation in mitophagy occurs that negatively impacts PaCSC stemness.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/genetics , Cytokines/metabolism , Mitophagy/genetics , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/pathology , Ubiquitins/metabolism , Cell Line , Cell Plasticity/physiology , Cell Transformation, Neoplastic/pathology , Cytokines/genetics , Humans , Metformin/pharmacology , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Phosphorylation , Pancreatic Neoplasms/mortality , RNA Editing/genetics , Ubiquitins/genetics
9.
J Med Chem ; 62(17): 7976-7997, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31365252

ABSTRACT

Phosphoglycerate dehydrogenase (PHGDH) is known to be the rate-limiting enzyme in the serine synthesis pathway in humans. It converts glycolysis-derived 3-phosphoglycerate to 3-phosphopyruvate in a co-factor-dependent oxidation reaction. Herein, we report the discovery of BI-4916, a prodrug of the co-factor nicotinamide adenine dinucleotide (NADH/NAD+)-competitive PHGDH inhibitor BI-4924, which has shown high selectivity against the majority of other dehydrogenase targets. Starting with a fragment-based screening, a subsequent hit optimization using structure-based drug design was conducted to deliver a single-digit nanomolar lead series and to improve potency by 6 orders of magnitude. To this end, an intracellular ester cleavage mechanism of the ester prodrug was utilized to achieve intracellular enrichment of the actual carboxylic acid based drug and thus overcome high cytosolic levels of the competitive cofactors NADH/NAD+.


Subject(s)
Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Phosphoglycerate Dehydrogenase/antagonists & inhibitors , Serine/antagonists & inhibitors , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Indoles/chemical synthesis , Indoles/chemistry , Models, Molecular , Molecular Structure , Phosphoglycerate Dehydrogenase/metabolism , Serine/biosynthesis , Structure-Activity Relationship
10.
Stem Cells Int ; 2019: 1378639, 2019.
Article in English | MEDLINE | ID: mdl-31191663

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is currently the fourth leading cause of cancer-related mortality. Cancer stem cells (CSCs) have been shown to be the drivers of pancreatic tumor growth, metastasis, and chemoresistance, but our understanding of these cells is still limited by our inability to efficiently identify and isolate them. While a number of markers capable of identifying pancreatic CSCs (PaCSCs) have been discovered since 2007, there is no doubt that more markers are still needed. The anthrax toxin receptor 1 (ANTXR1) was identified as a functional biomarker of triple-negative breast CSCs, and PDAC patients stratified based on ANTXR1 expression levels showed increased mortality and enrichment of pathways known to be necessary for CSC biology, including TGF-ß, NOTCH, Wnt/ß-catenin, and IL-6/JAK/STAT3 signaling and epithelial to mesenchymal transition, suggesting that ANTXR1 may represent a putative PaCSC marker. In this study, we show that ANTXR1+ cells are not only detectable across a panel of 7 PDAC patient-derived xenograft primary cultures but ANTXR1 expression significantly increased in CSC-enriched 3D sphere cultures. Importantly, ANTXR1+ cells also coexpressed other known PaCSC markers such as CD44, CD133, and autofluorescence, and ANTXR1+ cells displayed enhanced CSC functional and molecular properties, including increased self-renewal and expression of pluripotency-associated genes, compared to ANTXR1- cells. Thus, this study validates ANTXR1 as a new PaCSC marker and we propose its use in identifying CSCs in this tumor type and its exploitation in the development of CSC-targeted therapies for PDAC.

11.
Oncogene ; 38(27): 5469-5485, 2019 07.
Article in English | MEDLINE | ID: mdl-30936462

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an inherently chemoresistant tumor. Chemotherapy leads to apoptosis of cancer cells, and in previous studies we have shown that tumor-associated macrophage (TAM) infiltration increases following chemotherapy in PDAC. Since one of the main functions of macrophages is to eliminate apoptotic cells, we hypothesized that TAMs phagocytose chemotherapy-induced apoptotic cells and secrete factors, which favor PDAC chemoresistance. To test this hypothesis, primary human PDAC cultures were treated with conditioned media (CM) from monocyte-derived macrophage cultures incubated with apoptotic PDAC cells (MØApopCM). MØApopCM pretreatment rendered naïve PDAC cells resistant to Gemcitabine- or Abraxane-induced apoptosis. Proteomic analysis of MØApopCM identified YWHAZ/14-3-3 protein zeta/delta (14-3-3ζ), a major regulator of apoptotic cellular pathways, as a potential mediator of chemoresistance, which was subsequently validated in patient transcriptional datasets, serum samples from PDAC patients and using recombinant 14-3-3ζ and inhibitors thereof. Moreover, in mice bearing orthotopic PDAC tumors, the antitumor potential of Gemcitabine was significantly enhanced by elimination of TAMs using clodronate liposomes or by pharmacological inhibition of the Axl receptor tyrosine kinase, a 14-3-3ζ interacting partner. These data highlight a unique regulatory mechanism by which chemotherapy-induced apoptosis acts as a switch to initiate a protumor/antiapoptotic mechanism in PDAC via 14-3-3ζ/Axl signaling, leading to phosphorylation of Akt and activation of cellular prosurvival mechanisms. The data presented therefore challenge the idea that apoptosis of tumor cells is therapeutically beneficial, at least when immune sensor cells, such as macrophages, are present.


Subject(s)
14-3-3 Proteins/metabolism , Antimetabolites, Antineoplastic/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Macrophages/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Animals , Apoptosis , Carcinoma, Pancreatic Ductal/pathology , Cell Polarity , Cell Proliferation , Culture Media, Conditioned , Deoxycytidine/therapeutic use , Humans , Macrophages/cytology , Mice , Pancreatic Neoplasms/pathology , Gemcitabine , Axl Receptor Tyrosine Kinase
12.
Trends Cancer ; 5(3): 145-148, 2019 03.
Article in English | MEDLINE | ID: mdl-30898261

ABSTRACT

The SLIT/ROBO pathway is classically known for its role in axon guidance, and is frequently lost in pancreatic cancer, but the biology behind this is poorly understood. Two recent articles (Nat. Commun. 2018;9:5082; Nat. Commun. 2018;9:5083) using mouse models show that SLIT/ROBO signaling is crucial to determine pancreatic cell identity during development and suppresses the stromal activation in disease, through divergent mechanisms. SLIT/ROBO proteins thus link development and cancer, but appear to have distinct functions in health and disease.


Subject(s)
Pancreatic Neoplasms , Transcription Factors , Animals , Mice , Signal Transduction
13.
J Pathol ; 246(3): 352-365, 2018 11.
Article in English | MEDLINE | ID: mdl-30058725

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC), which is the primary cause of pancreatic cancer mortality, is poorly responsive to currently available interventions. Identifying new targets that drive PDAC formation and progression is critical for developing alternative therapeutic strategies to treat this lethal malignancy. Using genetic and pharmacological approaches, we investigated in vivo and in vitro whether uptake of the monoamine serotonin [5-hydroxytryptamine (5-HT)] is required for PDAC development. We demonstrated that pancreatic acinar cells have the ability to readily take up 5-HT in a transport-mediated manner. 5-HT uptake promoted activation of the small GTPase Ras-related C3 botulinum toxin substrate 1 (Rac1), which is required for transdifferentiation of acinar cells into acinar-to-ductal metaplasia (ADM), a key determinant in PDAC development. Consistent with the central role played by Rac1 in ADM formation, inhibition of the 5-HT transporter Sert (Slc6a4) with fluoxetine reduced ADM formation both in vitro and in vivo in a cell-autonomous manner. In addition, fluoxetine treatment profoundly compromised the stromal reaction and affected the proliferation and lipid metabolism of malignant PDAC cells. We propose that Sert is a promising therapeutic target to counteract the early event of ADM, with the potential to stall the initiation and progression of pancreatic carcinogenesis. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Cell Proliferation , Genes, ras , Neuropeptides/metabolism , Pancreas/enzymology , Pancreatic Neoplasms/enzymology , Serotonin/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/prevention & control , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transdifferentiation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chick Embryo , Disease Models, Animal , Enzyme Activation , Fluoxetine/pharmacology , Genetic Predisposition to Disease , Humans , Metaplasia , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Pathologic , Pancreas/drug effects , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/prevention & control , Phenotype , Rats , Serotonin Plasma Membrane Transport Proteins/drug effects , Serotonin Plasma Membrane Transport Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Signal Transduction , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism
14.
Nature ; 554(7693): 533-537, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29443959

ABSTRACT

Chronic inflammation increases the risk of developing one of several types of cancer. Inflammatory responses are currently thought to be controlled by mechanisms that rely on transcriptional networks that are distinct from those involved in cell differentiation. The orphan nuclear receptor NR5A2 participates in a wide variety of processes, including cholesterol and glucose metabolism in the liver, resolution of endoplasmic reticulum stress, intestinal glucocorticoid production, pancreatic development and acinar differentiation. In genome-wide association studies, single nucleotide polymorphisms in the vicinity of NR5A2 have previously been associated with the risk of pancreatic adenocarcinoma. In mice, Nr5a2 heterozygosity sensitizes the pancreas to damage, impairs regeneration and cooperates with mutant Kras in tumour progression. Here, using a global transcriptomic analysis, we describe an epithelial-cell-autonomous basal pre-inflammatory state in the pancreas of Nr5a2+/- mice that is reminiscent of the early stages of pancreatitis-induced inflammation and is conserved in histologically normal human pancreases with reduced expression of NR5A2 mRNA. In Nr5a2+/-mice, NR5A2 undergoes a marked transcriptional switch, relocating from differentiation-specific to inflammatory genes and thereby promoting gene transcription that is dependent on the AP-1 transcription factor. Pancreatic deletion of Jun rescues the pre-inflammatory phenotype, as well as binding of NR5A2 to inflammatory gene promoters and the defective regenerative response to damage. These findings support the notion that, in the pancreas, the transcriptional networks involved in differentiation-specific functions also suppress inflammatory programmes. Under conditions of genetic or environmental constraint, these networks can be subverted to foster inflammation.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation , Inflammation/genetics , Pancreas/metabolism , Pancreas/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Transcriptome , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Chromatin/genetics , Chromatin/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Regulatory Networks/genetics , Genes, jun/genetics , Heterozygote , Humans , Mice , Organ Specificity/genetics , Pancreatitis/genetics , Promoter Regions, Genetic/genetics , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factor AP-1/metabolism
15.
Diabetes ; 67(3): 448-460, 2018 03.
Article in English | MEDLINE | ID: mdl-29263149

ABSTRACT

GATA4 and GATA6 play essential, but redundant, roles in pancreas formation in mice, and GATA6 mutations cause pancreatic agenesis in humans. GATA6 mutations have also recently been linked to adult-onset diabetes, with subclinical or no exocrine insufficiency, suggesting an important role for GATA6 in human ß-cell physiology. To investigate the role of GATA6 in the adult endocrine pancreas, we generated mice in which Gata6 is specifically inactivated in the pancreas. These mice develop glucose intolerance. Islets deficient in GATA6 activity display decreased insulin content and impaired insulin secretion. Gata6-deficient ß-cells exhibit ultrastructural abnormalities, including increased immature insulin granules, swollen mitochondria, and disorganized endoplasmic reticulum. We also demonstrate that Pdx1 expression in adult ß-cells depends on GATA sites in transgenic reporter mice and that loss of GATA6 greatly affects ß-cell-specific gene expression. These findings demonstrate the essential role of GATA6 in ß-cell function.


Subject(s)
Endoplasmic Reticulum Stress , GATA6 Transcription Factor/metabolism , Insulin-Secreting Cells/metabolism , Insulin/biosynthesis , Mitochondria/metabolism , Secretory Vesicles/metabolism , Animals , Blood Glucose/analysis , Female , GATA6 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Genes, Reporter , Glucose Intolerance/blood , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Glucose Intolerance/physiopathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/ultrastructure , Male , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission , Mitochondria/pathology , Mitochondria/ultrastructure , Mutation , Organelle Biogenesis , Secretory Vesicles/pathology , Secretory Vesicles/ultrastructure , Tissue Culture Techniques , Trans-Activators/genetics , Trans-Activators/metabolism
16.
Gut ; 66(9): 1665-1676, 2017 09.
Article in English | MEDLINE | ID: mdl-27325420

ABSTRACT

BACKGROUND AND AIMS: The role of GATA factors in cancer has gained increasing attention recently, but the function of GATA6 in pancreatic ductal adenocarcinoma (PDAC) is controversial. GATA6 is amplified in a subset of tumours and was proposed to be oncogenic, but high GATA6 levels are found in well-differentiated tumours and are associated with better patient outcome. By contrast, a tumour-suppressive function of GATA6 was demonstrated using genetic mouse models. We aimed at clarifying GATA6 function in PDAC. DESIGN: We combined GATA6 silencing and overexpression in PDAC cell lines with GATA6 ChIP-Seq and RNA-Seq data, in order to understand the mechanism of GATA6 functions. We then confirmed some of our observations in primary patient samples, some of which were included in the ESPAC-3 randomised clinical trial for adjuvant therapy. RESULTS: GATA6 inhibits the epithelial-mesenchymal transition (EMT) in vitro and cell dissemination in vivo. GATA6 has a unique proepithelial and antimesenchymal function, and its transcriptional regulation is direct and implies, indirectly, the regulation of other transcription factors involved in EMT. GATA6 is lost in tumours, in association with altered differentiation and the acquisition of a basal-like molecular phenotype, consistent with an epithelial-to-epithelial (ET2) transition. Patients with basal-like GATA6low tumours have a shorter survival and have a distinctly poor response to adjuvant 5-fluorouracil (5-FU)/leucovorin. However, modulation of GATA6 expression in cultured cells does not directly regulate response to 5-FU. CONCLUSIONS: We provide mechanistic insight into GATA6 tumour-suppressive function, its role as a regulator of canonical epithelial differentiation, and propose that loss of GATA6 expression is both prognostic and predictive of response to adjuvant therapy.


Subject(s)
Carcinoma, Pancreatic Ductal , Epithelial-Mesenchymal Transition/genetics , Fluorouracil/pharmacology , GATA6 Transcription Factor , Pancreatic Neoplasms , Animals , Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/genetics , Chemotherapy, Adjuvant/methods , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Statistics as Topic
17.
PLoS Genet ; 12(12): e1006463, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27911893

ABSTRACT

The m-AAA protease preserves proteostasis of the inner mitochondrial membrane. It ensures a functional respiratory chain, by controlling the turnover of respiratory complex subunits and allowing mitochondrial translation, but other functions in mitochondria are conceivable. Mutations in genes encoding subunits of the m-AAA protease have been linked to various neurodegenerative diseases in humans, such as hereditary spastic paraplegia and spinocerebellar ataxia. While essential functions of the m-AAA protease for neuronal survival have been established, its role in adult glial cells remains enigmatic. Here, we show that deletion of the highly expressed subunit AFG3L2 in mature mouse oligodendrocytes provokes early-on mitochondrial fragmentation and swelling, as previously shown in neurons, but causes only late-onset motor defects and myelin abnormalities. In contrast, total ablation of the m-AAA protease, by deleting both Afg3l2 and its paralogue Afg3l1, triggers progressive motor dysfunction and demyelination, owing to rapid oligodendrocyte cell death. Surprisingly, the mice showed premature hair greying, caused by progressive loss of melanoblasts that share a common developmental origin with Schwann cells and are targeted in our experiments. Thus, while both neurons and glial cells are dependant on the m-AAA protease for survival in vivo, complete ablation of the complex is necessary to trigger death of oligodendrocytes, hinting to cell-autonomous thresholds of vulnerability to m-AAA protease deficiency.


Subject(s)
ATP-Dependent Proteases/genetics , Demyelinating Diseases/genetics , Hair/metabolism , Metalloendopeptidases/genetics , Mitochondria/genetics , ATP-Dependent Proteases/biosynthesis , ATPases Associated with Diverse Cellular Activities , Animals , Cell Death/genetics , Cell Survival/genetics , Hair/growth & development , Humans , Metalloendopeptidases/biosynthesis , Mice , Mitochondria/metabolism , Mitochondria/pathology , Mutation , Myelin Sheath/metabolism , Neuroglia/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Schwann Cells/metabolism
18.
Environ Technol ; 37(19): 2428-34, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26852791

ABSTRACT

The removal of simazine from both pure water and solute-bearing well water was studied by adsorption on two solids: zeolite H-Y from the commercial Na form and porous silica tailored by the sol-gel technique. The pH dependence of the amount adsorbed in a closed system at constant total simazine content as well as the apparent isotherms of adsorption was measured in all four cases. The low ion content of natural water suffices to alter the adsorption features in the case of silica, but not with zeolite H-Y. Iteration of the adsorption process onto constant amounts of solid allowed bringing the residual simazine concentration below 0.05 mg/L, the value allowed by Italian laws in wastewaters.


Subject(s)
Simazine/isolation & purification , Wastewater/chemistry , Water Pollutants, Chemical/isolation & purification , Water Purification/methods , Italy , Porosity , Silicon Dioxide/chemistry , Simazine/analysis , Simazine/chemistry , Water Pollutants, Chemical/analysis , Water Pollutants, Chemical/chemistry , Zeolites/chemistry
19.
Gut ; 65(3): 476-86, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25596178

ABSTRACT

BACKGROUND AND AIMS: Gata6 is required to complete and maintain acinar differentiation in the mouse pancreas. Pancreas-specific Gata6 ablation during development causes extensive and persistent acinar-ductal metaplasia, which is considered an initial step of mutant KRas-driven carcinogenesis. Therefore, the Gata6-null pancreas might represent a tumour-prone environment. We investigated whether Gata6 plays a role during pancreatic tumorigenesis. DESIGN: We analysed genetically engineered mouse models and human pancreatic ductal adenocarcinoma (PDAC) cell lines, using a combination of histopathological studies, genome-wide expression and chromatin immunoprecipitation experiments to understand the role of Gata6 in the initiation and progression of KRas(G12V)-driven tumours RESULTS: We show that Gata6 maintains the acinar differentiation programme, both directly and indirectly, and it concomitantly suppresses ectopic programmes in the pancreas. Gata6 ablation renders acinar cells more sensitive to KRas(G12V), thereby accelerating tumour development. Gata6 expression is spontaneously lost in a mouse model of KRas(G12V)-driven PDAC, in association with altered cell differentiation. Using a combination of ChIP-Seq and RNA-Seq, we show that Gata6 exerts its tumour-suppressive effect through the promotion of cell differentiation, the suppression of inflammatory pathways, and the direct repression of cancer-related pathways. Among them is the epidermal growth factor receptor (EGFR) pathway, the activity of which is upregulated in the normal and preneoplastic Gata6-null pancreas. Accordingly, GATA6-silencing in human PDAC cells leads to an upregulation of EGFR. CONCLUSIONS: We propose that, in the pancreas, Gata6 acts as a tumour suppressor by enforcing acinar cell differentiation, by directly and indirectly repressing ectopic differentiation programmes, and by regulating crucial cancer-related gene expression pathways.


Subject(s)
Acinar Cells/physiology , Biomarkers, Tumor/metabolism , Carcinogenesis/metabolism , Carcinoma, Pancreatic Ductal/metabolism , GATA6 Transcription Factor/metabolism , Pancreatic Neoplasms/metabolism , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Differentiation , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Transgenic , Mutation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics
20.
J Clin Invest ; 125(2): 824-30, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25574842

ABSTRACT

NOTCH signaling suppresses tumor growth and proliferation in several types of stratified epithelia. Here, we show that missense mutations in NOTCH1 and NOTCH2 found in human bladder cancers result in loss of function. In murine models, genetic ablation of the NOTCH pathway accelerated bladder tumorigenesis and promoted the formation of squamous cell carcinomas, with areas of mesenchymal features. Using bladder cancer cells, we determined that the NOTCH pathway stabilizes the epithelial phenotype through its effector HES1 and, consequently, loss of NOTCH activity favors the process of epithelial-mesenchymal transition. Evaluation of human bladder cancer samples revealed that tumors with low levels of HES1 present mesenchymal features and are more aggressive. Together, our results indicate that NOTCH serves as a tumor suppressor in the bladder and that loss of this pathway promotes mesenchymal and invasive features.


Subject(s)
Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Urinary Bladder Neoplasms/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice , Receptor, Notch1/genetics , Receptor, Notch2/genetics , Transcription Factor HES-1 , Tumor Suppressor Proteins/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...