Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Nature ; 554(7690): 62-68, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29364867

ABSTRACT

The poor correlation of mutational landscapes with phenotypes limits our understanding of the pathogenesis and metastasis of pancreatic ductal adenocarcinoma (PDAC). Here we show that oncogenic dosage-variation has a critical role in PDAC biology and phenotypic diversification. We find an increase in gene dosage of mutant KRAS in human PDAC precursors, which drives both early tumorigenesis and metastasis and thus rationalizes early PDAC dissemination. To overcome the limitations posed to gene dosage studies by the stromal richness of PDAC, we have developed large cell culture resources of metastatic mouse PDAC. Integration of cell culture genomes, transcriptomes and tumour phenotypes with functional studies and human data reveals additional widespread effects of oncogenic dosage variation on cell morphology and plasticity, histopathology and clinical outcome, with the highest KrasMUT levels underlying aggressive undifferentiated phenotypes. We also identify alternative oncogenic gains (Myc, Yap1 or Nfkb2), which collaborate with heterozygous KrasMUT in driving tumorigenesis, but have lower metastatic potential. Mechanistically, different oncogenic gains and dosages evolve along distinct evolutionary routes, licensed by defined allelic states and/or combinations of hallmark tumour suppressor alterations (Cdkn2a, Trp53, Tgfß-pathway). Thus, evolutionary constraints and contingencies direct oncogenic dosage gain and variation along defined routes to drive the early progression of PDAC and shape its downstream biology. Our study uncovers universal principles of Ras-driven oncogenesis that have potential relevance beyond pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Evolution, Molecular , Gene Dosage , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Adaptor Proteins, Signal Transducing/genetics , Alleles , Animals , Carcinogenesis/genetics , Cell Cycle Proteins , Cyclin-Dependent Kinase Inhibitor p16/genetics , Disease Progression , Female , Genes, myc , Genes, p53 , Humans , Male , Mice , Mutation , NF-kappa B p52 Subunit/genetics , Neoplasm Metastasis/genetics , Nuclear Proteins/genetics , Phenotype , Phosphoproteins/genetics , Transcription Factors/genetics , Transcriptome/genetics , Transforming Growth Factor beta1/genetics , YAP-Signaling Proteins
2.
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
3.
BMC Dev Biol ; 21(1): 4, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33517884

ABSTRACT

BACKGROUND: Our previous study have shown that the PSMD11 protein was an important survival factor for cancer cells except for its key role in regulation of assembly and activity of the 26S proteasome. To further investigate the role of PSMD11 in carcinogenesis, we constructed a conditional exon 5 floxed allele of PSMD11 (PSMD11flx) in mice. RESULTS: It was found that homozygous PSMD11 flx/flx mice showed normal and exhibited a normal life span and fertility, and showed roughly equivalent expression of PSMD11 in various tissues, suggesting that the floxed allele maintained the wild-type function. Cre recombinase could induce efficient knockout of the floxed PSMD11 allele both in vitro and in vivo. Mice with constitutive single allele deletion of PSMD11 derived from intercrossing between PSMD11flx/flx and CMV-Cre mice were all viable and fertile, and showed apparent growth retardation, suggesting that PSMD11 played a significant role in the development of mice pre- or postnatally. No whole-body PSMD11 deficient embryos (PSMD11-/-) were identified in E7.5-8.5 embryos in uteros, indicating that double allele knockout of PSMD11 leads to early embryonic lethality. To avoid embryonic lethality produced by whole-body PSMD11 deletion, we further developed conditional PSMD11 global knockout mice with genotype Flp;FSF-R26CAG - CreERT2/+; PSMD11 flx/flx, and demonstrated that PSMD11 could be depleted in a temporal and tissue-specific manner. Meanwhile, it was found that depletion of PSMD11 could induce massive apoptosis in MEFs. CONCLUSIONS: In summary, our data demonstrated that we have successfully generated a conditional knockout allele of PSMD11 in mice, and found that PSMD11 played a key role in early and postnatal development in mice, the PSMD11 flx/flx mice will be an invaluable tool to explore the functions of PSMD11 in development and diseases.


Subject(s)
Alleles , Proteasome Endopeptidase Complex/genetics , Animals , Homozygote , Mice , Mice, Knockout
4.
Development ; 143(23): 4394-4404, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27789621

ABSTRACT

Olfactory epithelium (OE) has a lifelong capacity for neurogenesis due to the presence of basal stem cells. Despite the ability to generate short-term cultures, the successful in vitro expansion of purified stem cells from adult OE has not been reported. We sought to establish expansion-competent OE stem cell cultures to facilitate further study of the mechanisms and cell populations important in OE renewal. Successful cultures were prepared using adult mouse basal cells selected for expression of c-KIT. We show that c-KIT signaling regulates self-renewal capacity and prevents neurodifferentiation in culture. Inhibition of TGFß family signaling, a known negative regulator of embryonic basal cells, is also necessary for maintenance of the proliferative, undifferentiated state in vitro Characterizing successful cultures, we identified expression of BMI1 and other Polycomb proteins not previously identified in olfactory basal cells but known to be essential for self-renewal in other stem cell populations. Inducible fate mapping demonstrates that BMI1 is expressed in vivo by multipotent OE progenitors, validating our culture model. These findings provide mechanistic insights into the renewal and potency of olfactory stem cells.


Subject(s)
Cell Self Renewal/physiology , Neurogenesis/physiology , Olfactory Mucosa/cytology , Proto-Oncogene Proteins c-kit/metabolism , Stem Cells/cytology , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Cell Differentiation/physiology , Cell Lineage , Cell Proliferation/physiology , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Transgenic , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction
5.
J Cell Biochem ; 119(8): 6644-6656, 2018 08.
Article in English | MEDLINE | ID: mdl-29665121

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) remains one of the most devastating disease with the 5-year survival rate less than 6%. In this study, we investigated if inhibiting protein synthesis directly with homoharringtonine (HHT) could induce acute apoptosis in pancreatic cancer cells through quick depletion of multiple short-lived critical members of the central proteome, example, PSMD11(26S proteasome non-ATPase regulatory subunit 11). It was shown that although HHT could inhibit proliferation and growth of MiaPaCa-2 and PANC-1 cells in a time- and dose-dependent manner, only part of pancreatic cancer cells could be induced to die through acute apoptosis. Mechanistic studies showed that HHT could induce quick protein synthesis of PSMD11 through activating MEK1/ERK1/2 signaling pathway in pancreatic cancer cells. Inhibiting MEK1/ERK1/2 pathway with sorafenib could improve the cytotoxity of HHT in vitro and in a genetically engineered mouse model of pancreatic cancer. These results suggest that quick induction of PSMD11 or other acute apoptosis inhibitors through activation of the MEK1/ERK1/2 signaling pathway may be one of the important surviving mechanism which can help pancreatic cancer cells avoid acute apoptosis, it may have significant implications for the targeted therapy of pancreatic ductal adenocarcinoma.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Homoharringtonine/pharmacology , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , Proteasome Endopeptidase Complex/biosynthesis , Protein Biosynthesis/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Humans , MAP Kinase Kinase 1/genetics , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Neoplasm Proteins/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proteasome Endopeptidase Complex/genetics
6.
Circ Res ; 119(8): 921-30, 2016 Sep 30.
Article in English | MEDLINE | ID: mdl-27481956

ABSTRACT

RATIONALE: Culture-expanded cells originating from cardiac tissue that express the cell surface receptor cKit are undergoing clinical testing as a cell source for heart failure and congenital heart disease. Although accumulating data support that mesenchymal stem cells (MSCs) enhance the efficacy of cardiac cKit(+) cells (CSCs), the underlying mechanism for this synergistic effect remains incompletely understood. OBJECTIVE: To test the hypothesis that MSCs stimulate endogenous CSCs to proliferate, migrate, and differentiate via the SDF1/CXCR4 and stem cell factor/cKit pathways. METHODS AND RESULTS: Using genetic lineage-tracing approaches, we show that in the postnatal murine heart, cKit(+) cells proliferate, migrate, and form cardiomyocytes, but not endothelial cells. CSCs exhibit marked chemotactic and proliferative responses when cocultured with MSCs but not with cardiac stromal cells. Antagonism of the CXCR4 pathway with AMD3100 (an SDF1/CXCR4 antagonist) inhibited MSC-induced CSC chemotaxis but stimulated CSC cardiomyogenesis (P<0.0001). Furthermore, MSCs enhanced CSC proliferation via the stem cell factor/cKit and SDF1/CXCR4 pathways (P<0.0001). CONCLUSIONS: Together these findings show that MSCs exhibit profound, yet differential, effects on CSC migration, proliferation, and differentiation and suggest a mechanism underlying the improved cardiac regeneration associated with combination therapy using CSCs and MSCs. These findings have important therapeutic implications for cell-based therapy strategies that use mixtures of CSCs and MSCs.


Subject(s)
Chemokine CXCL12/biosynthesis , Mesenchymal Stem Cells/physiology , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-kit/biosynthesis , Receptors, CXCR4/biosynthesis , Stem Cell Factor/biosynthesis , Animals , Animals, Newborn , Cell Movement/physiology , Cells, Cultured , Coculture Techniques , Humans , Mice , Mice, Transgenic , Pilot Projects , Signal Transduction/physiology , Swine
7.
Proc Natl Acad Sci U S A ; 112(42): 13051-6, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26438843

ABSTRACT

The degree to which cKit-expressing progenitors generate cardiomyocytes in the heart is controversial. Genetic fate-mapping studies suggest minimal contribution; however, whether or not minimal contribution reflects minimal cardiomyogenic capacity is unclear because the embryonic origin and role in cardiogenesis of these progenitors remain elusive. Using high-resolution genetic fate-mapping approaches with cKit(CreERT2/+) and Wnt1::Flpe mouse lines, we show that cKit delineates cardiac neural crest progenitors (CNC(kit)). CNC(kit) possess full cardiomyogenic capacity and contribute to all CNC derivatives, including cardiac conduction system cells. Furthermore, by modeling cardiogenesis in cKit(CreERT2)-induced pluripotent stem cells, we show that, paradoxically, the cardiogenic fate of CNC(kit) is regulated by bone morphogenetic protein antagonism, a signaling pathway activated transiently during establishment of the cardiac crescent, and extinguished from the heart before CNC invasion. Together, these findings elucidate the origin of cKit(+) cardiac progenitors and suggest that a nonpermissive cardiac milieu, rather than minimal cardiomyogenic capacity, controls the degree of CNC(kit) contribution to myocardium.


Subject(s)
Myocytes, Cardiac/metabolism , Neural Crest/cytology , Proto-Oncogene Proteins c-kit/genetics , Stem Cells/cytology , Animals , Bone Morphogenetic Proteins/antagonists & inhibitors , Mice , Mice, Transgenic , Myocytes, Cardiac/cytology , Neural Crest/metabolism
8.
J Physiol ; 593(2): 403-14, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25630261

ABSTRACT

Oesophageal achalasia is a disease known to result from reduced relaxation of the lower oesophageal sphincter (LES). Nitric oxide (NO) is one of the main inhibitory transmitters. NO-sensitive guanylyl cyclase (NO-GC) acts as the key target of NO and, by the generation of cGMP, mediates nitrergic relaxation in the LES. To date, the exact mechanism of nitrergic LES relaxation is still insufficiently elucidated. To clarify the role of NO-GC in LES relaxation, we used cell-specific knockout (KO) mouse lines for NO-GC. These include mice lacking NO-GC in smooth muscle cells (SMC-GCKO), in interstitial cells of Cajal (ICC-GCKO) and in both SMC/ICC (SMC/ICC-GCKO). We applied oesophageal manometry to study the functionality of LES in vivo. Isometric force studies were performed to monitor LES responsiveness to exogenous NO and electric field stimulation of intrinsic nerves in vitro. Cell-specific expression/deletion of NO-GC was monitored by immunohistochemistry. Swallowing-induced LES relaxation is strongly reduced by deletion of NO-GC in ICC. Basal LES tone is affected by NO-GC deletion in either SMC or ICC. Lack of NO-GC in both cells leads to a complete interruption of NO-induced relaxation and, therefore, to an achalasia-like phenotype similar to that seen in global GCKO mice. Our data indicate that regulation of basal LES tone is based on a dual mechanism mediated by NO-GC in SMC and ICC whereas swallow-induced LES relaxation is mainly regulated by nitrergic mechanisms in ICC.


Subject(s)
Esophageal Sphincter, Lower/metabolism , Guanylate Cyclase/metabolism , Interstitial Cells of Cajal/metabolism , Muscle Relaxation , Nitric Oxide/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Esophageal Sphincter, Lower/cytology , Esophageal Sphincter, Lower/physiology , Guanylate Cyclase/genetics , Interstitial Cells of Cajal/physiology , Isometric Contraction , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Receptors, Cytoplasmic and Nuclear/genetics , Soluble Guanylyl Cyclase
9.
Eur J Immunol ; 44(1): 296-306, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24127407

ABSTRACT

Mast cells are abundantly situated at contact sites between the body and its environment, such as the skin and, especially during certain immune responses, at mucosal surfaces. They mediate allergic reactions and degrade toxins as well as venoms. However, their roles during innate and adaptive immune responses remain controversial and it is likely that major functions remain to be discovered. Recent developments in mast cell-specific conditional gene targeting in the mouse promise to enhance our understanding of these fascinating cells. To complete the genetic toolbox to study mast cell development, homeostasis and function, it is imperative to inducibly manipulate their gene expression. Here, we report the generation of a novel knock-in mouse line expressing a tamoxifen-inducible version of the Cre recombinase from within the endogenous c-Kit locus. We demonstrate highly efficient and specific inducible expression of a fluorescent reporter protein in mast cells both in vivo and in vitro. Furthermore, induction of diphtheria toxin A expression allowed selective and efficient ablation of mast cells at various anatomical locations, while other hematopoietic cells remain unaffected. This novel mouse strain will hence be very valuable to study mast cell homeostasis and how specific genes influence their functions in physiology and pathology.


Subject(s)
Diphtheria Toxin/metabolism , Gene Targeting/methods , Integrases/metabolism , Mast Cells/immunology , Mice, Transgenic/immunology , Peptide Fragments/metabolism , Animals , Cell Death/drug effects , Cell Death/genetics , Diphtheria Toxin/genetics , Gene Expression Regulation/drug effects , Gene Knock-In Techniques , Genetic Loci/genetics , Integrases/genetics , Mast Cells/drug effects , Mast Cells/pathology , Mice , Organ Specificity , Peptide Fragments/genetics , Proto-Oncogene Proteins c-kit/genetics , Tamoxifen/administration & dosage , Transgenes/genetics
10.
Am J Physiol Gastrointest Liver Physiol ; 307(1): G98-106, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24833707

ABSTRACT

Nitric oxide (NO) is a major inhibitory neurotransmitter in the gastrointestinal (GI) tract. Its main effector, NO-sensitive guanylyl cyclase (NO-GC), is expressed in several GI cell types, including smooth muscle cells (SMC), interstitial cells of Cajal (ICC), and fibroblast-like cells. Up to date, the interplay between neurons and these cells to initiate a nitrergic inhibitory junction potential (IJP) is unclear. Here, we investigate the origin of the nitrergic IJP in murine fundus and colon. IJPs were determined in fundus and colon SMC of mice lacking NO-GC globally (GCKO) and specifically in SMC (SM-GCKO), ICC (ICC-GCKO), and both SMC/ICC (SM/ICC-GCKO). Nitrergic IJP was abolished in ICC-GCKO fundus and reduced in SM-GCKO fundus. In the colon, the amplitude of nitrergic IJP was reduced in ICC-GCKO, whereas nitrergic IJP in SM-GCKO was reduced in duration. These results were corroborated by loss of the nitrergic IJP in global GCKO. In conclusion, our results prove the obligatory role of NO-GC in ICC for the initiation of an IJP. NO-GC in SMC appears to enhance the nitrergic IJP, resulting in a stronger and prolonged hyperpolarization in fundus and colon SMC, respectively. Thus NO-GC in both cell types is mandatory to induce a full nitrergic IJP. Our data from the colon clearly reveal the nitrergic IJP to be biphasic, resulting from individual inputs of ICC and SMC.


Subject(s)
Colon/innervation , Gastric Fundus/innervation , Interstitial Cells of Cajal/metabolism , Neural Inhibition , Nitrergic Neurons/metabolism , Nitric Oxide/metabolism , Synaptic Transmission , Animals , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Inhibitory Postsynaptic Potentials , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Time Factors
11.
Gastroenterology ; 145(1): 188-196, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23528627

ABSTRACT

BACKGROUND & AIMS: It is not clear how nitric oxide (NO) released from enteric neurons relaxes gastrointestinal (GI) smooth muscle. In analogy to the vascular system, NO might directly induce relaxation of smooth muscle cells (SMCs) by acting on its receptor, NO-sensitive guanylyl cyclase (NO-GC). Alternatively, intermediate cells, such as the interstitial cells of Cajal (ICCs), might detect nitrergic signals to indirectly regulate smooth muscle tone, and thereby regulate the motor function of the GI tract. We investigated the role of ICCs and SMCs in nitrergic relaxation using mice with cell-specific disruption of the gene encoding the ß1 subunit of NO-GC (GUCY1B3). METHODS: We created mice that lack NO-GC specifically in SMCs (SM-guanylyl cyclase knockout [GCKO]), ICCs (ICC-GCKO), or both (SM/ICC-GCKO). We investigated the effects of exogenous and endogenous NO on murine fundus using isometric force studies. Total gut transit time was measured to monitor the functional consequences of NO-GC deletion on GI motility in vivo. RESULTS: NO-GC is expressed in ICC and SMC. Deletion of the NO receptor from SMCs incompletely reduced NO-induced fundus relaxation, which was hardly affected after ICC-specific deletion. Gut transit time did not change in SM-GCKO or ICC-GCKO mice compared with control mice. However, nitrergic relaxation was not observed in SM/ICC-GCKO mice, which had increased gut transit time compared with controls. CONCLUSIONS: In mice, NO-GC is the only NO receptor to relax the fundus; deletion of NO-GC from the combination of SMCs and ICCs blocks nitrergic signaling. Therefore, ICCs and SMCs jointly mediate the relaxant effect of enteric NO.


Subject(s)
Gastric Fundus/physiology , Guanylate Cyclase/physiology , Interstitial Cells of Cajal/physiology , Myocytes, Smooth Muscle/physiology , Nitric Oxide/physiology , Signal Transduction/physiology , Animals , Electric Stimulation , Gastrointestinal Motility , Mice , Muscle Relaxation/drug effects , Nifedipine/pharmacology
12.
Proc Natl Acad Sci U S A ; 108(24): 9945-50, 2011 Jun 14.
Article in English | MEDLINE | ID: mdl-21628592

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a fatal disease with poor patient outcome often resulting from late diagnosis in advanced stages. To date methods to diagnose early-stage PDAC are limited and in vivo detection of pancreatic intraepithelial neoplasia (PanIN), a preinvasive precursor of PDAC, is impossible. Using a cathepsin-activatable near-infrared probe in combination with flexible confocal fluorescence lasermicroscopy (CFL) in a genetically defined mouse model of PDAC we were able to detect and grade murine PanIN lesions in real time in vivo. Our diagnostic approach is highly sensitive and specific and proved superior to clinically established fluorescein-enhanced imaging. Translation of this endoscopic technique into the clinic should tremendously improve detection of pancreatic neoplasia, thus reforming management of patients at risk for PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/diagnosis , Molecular Imaging/methods , Pancreatic Neoplasms/diagnosis , Precancerous Conditions/diagnosis , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cathepsins/genetics , Cathepsins/metabolism , Female , Fluorescent Dyes/metabolism , Gene Expression Profiling , Humans , Immunohistochemistry , Male , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Reproducibility of Results , Sensitivity and Specificity
13.
J Am Soc Nephrol ; 24(8): 1263-73, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23744888

ABSTRACT

The renin-angiotensin-aldosterone system (RAAS) regulates BP and salt-volume homeostasis. Juxtaglomerular (JG) cells synthesize and release renin, which is the first and rate-limiting step in the RAAS. Intense pathologic stresses cause a dramatic increase in the number of renin-producing cells in the kidney, termed JG cell recruitment, but how this occurs is not fully understood. Here, we isolated renal CD44(+) mesenchymal stem cell (MSC)-like cells and found that they differentiated into JG-like renin-expressing cells both in vitro and in vivo. Sodium depletion and captopril led to activation and differentiation of these cells into renin-expressing cells in the adult kidney. In summary, CD44(+) MSC-like cells exist in the adult kidney and can differentiate into JG-like renin-producing cells under conditions that promote JG cell recruitment.


Subject(s)
Adult Stem Cells/metabolism , Captopril/pharmacology , Cell Differentiation/physiology , Juxtaglomerular Apparatus/cytology , Kidney/cytology , Mesenchymal Stem Cells/metabolism , Renin-Angiotensin System/physiology , Renin/metabolism , Animals , Cell Differentiation/drug effects , Juxtaglomerular Apparatus/metabolism , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Renin-Angiotensin System/drug effects
14.
Nat Commun ; 14(1): 1201, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36882420

ABSTRACT

SNAIL is a key transcriptional regulator in embryonic development and cancer. Its effects in physiology and disease are believed to be linked to its role as a master regulator of epithelial-to-mesenchymal transition (EMT). Here, we report EMT-independent oncogenic SNAIL functions in cancer. Using genetic models, we systematically interrogated SNAIL effects in various oncogenic backgrounds and tissue types. SNAIL-related phenotypes displayed remarkable tissue- and genetic context-dependencies, ranging from protective effects as observed in KRAS- or WNT-driven intestinal cancers, to dramatic acceleration of tumorigenesis, as shown in KRAS-induced pancreatic cancer. Unexpectedly, SNAIL-driven oncogenesis was not associated with E-cadherin downregulation or induction of an overt EMT program. Instead, we show that SNAIL induces bypass of senescence and cell cycle progression through p16INK4A-independent inactivation of the Retinoblastoma (RB)-restriction checkpoint. Collectively, our work identifies non-canonical EMT-independent functions of SNAIL and unravel its complex context-dependent role in cancer.


Subject(s)
Pancreatic Neoplasms , Snail Family Transcription Factors , Carcinogenesis , Cell Transformation, Neoplastic , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras) , Animals , Snail Family Transcription Factors/genetics
15.
J Cell Sci ; 123(Pt 24): 4231-9, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21081648

ABSTRACT

The epithelial to mesenchymal transition (EMT) is a crucial step in tumor progression, and the TGFß-SMAD signaling pathway is an inductor of EMT in many tumor types. One hallmark of EMT is downregulation of the adherens junction protein E-cadherin, a process mediated by transcription factors such as the zinc fingers SNAIL and SLUG. Here, we report that the catalytic IκB kinase (IKK) subunit IKKα is necessary for the silencing of E-cadherin in a Panc1 cell model of TGFß-SMAD-mediated EMT, independently of NFκB. IKKα regulates canonical TGFß-SMAD signaling by interacting with SMAD3 and controlling SMAD complex formation on DNA. Furthermore, we demonstrate that the TGFß-IKKα-SMAD signaling pathway induces transcription of the genes encoding SNAIL and SLUG. In addition, we demonstrate that IKKα also modulates canonical TGFß-SMAD signaling in human MDA-MB231 breast cancer cells, arguing for a more general impact of IKKα on the control of TGFß-SMAD signaling. Taken together, these findings indicate that IKKα contributes to the tumor-promoting function of the TGFß-SMAD signaling pathway in particular cancers.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , I-kappa B Kinase/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Transcription Factors/genetics , Transforming Growth Factor beta/pharmacology , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Down-Regulation/drug effects , Epithelial-Mesenchymal Transition/genetics , Humans , Models, Biological , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Transport/drug effects , Snail Family Transcription Factors , Transcription Factors/metabolism
16.
Am J Physiol Cell Physiol ; 301(6): C1445-57, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21865585

ABSTRACT

Nitric oxide (NO) induces relaxation of colonic smooth muscle cells predominantly by cGMP/cGMP-dependent protein kinase I (cGKI)-induced phosphorylation of the inositol 1,4,5-trisphosphate receptor (IP(3)R)-associated cGMP kinase substrate (IRAG), to block store-dependent calcium signaling. In the present study we analyzed the structure and function of the human IRAG/MRVI1 gene. We describe four unique first exon variants transcribed from individual promoters in diverse human tissues. Tissue-specific alternative splicing with exon skipping and alternative splice donor and acceptor site usage further increases diversity of IRAG mRNA variants that encode for NH(2)- and COOH-terminally truncated proteins. At the functional level, COOH-terminally truncated IRAG variants lacking both the cGKI phosphorylation and the IP(3)RI interaction site counteract cGMP-mediated inhibition of calcium transients and relaxation of human colonic smooth muscle cells. Since COOH-terminally truncated IRAG mRNA isoforms are widely expressed in human tissues, our results point to an important role of IRAG variants as negative modulators of nitric oxide/cGKI-dependent signaling. The complexity of alternative splicing of the IRAG gene impressively demonstrates how posttranscriptional processing generates functionally distinct proteins from a single gene.


Subject(s)
Colon/metabolism , Gene Expression Profiling , Membrane Proteins/genetics , Muscle Contraction/genetics , Muscle, Smooth/metabolism , Phosphoproteins/genetics , Amino Acid Sequence , Base Sequence , Blotting, Western , Cell Line , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Humans , Immunoprecipitation , Membrane Proteins/metabolism , Molecular Sequence Data , Phosphoproteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Isoforms/genetics , RNA Isoforms/metabolism , RNA Processing, Post-Transcriptional , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transfection
17.
Proc Natl Acad Sci U S A ; 105(29): 10137-42, 2008 Jul 22.
Article in English | MEDLINE | ID: mdl-18621715

ABSTRACT

Site- and time-specific somatic gene transfer by using the avian sarcoma-leukosis retrovirus RCAS (replication-competent avian sarcoma-leukosis virus long terminal repeat with splice acceptor) has been shown to be a powerful tool to analyze gene function in vivo. RCAS retroviruses that express the avian subgroup A envelope transduce only mammalian cells genetically engineered to express the avian retroviral receptor, tumor virus A (TVA). Here, we generated a knockin mouse line termed LSL-R26(Tva-lacZ) with concomitant conditional expression of TVA and lacZ by targeting the Rosa26 locus. A loxP-flanked transcriptional stop cassette was used for conditional activation of TVA and LacZ expression in a Cre-recombinase-dependent manner. To demonstrate the ability of this system for conditional somatic gene transfer in vivo, we directed TVA expression to the pancreas. Introduction of an RCAS vector with Bryan-RSV polymerase and subgroup A envelope [RCASBP(A)] carrying oncogenic Kras(G12D) induced focal ductal pancreatic lesions that recapitulate human pancreatic intraepithelial neoplasias that progress to pancreatic ductal adenocarcinomas. TVA-mediated infection of genetically engineered mice with endogenous expression of Kras(G12D) in pancreatic progenitor cells by using RCASBP(A) virus carrying a short hairpin RNA directed against murine TP53, resulted in dramatically enhanced progression to invasive adenocarcinomas. These results show that conditional expression of TVA enables spatiotemporal gene expression and knockdown in a small subset of somatic cells in vivo. Therefore, it closely models carcinogenesis in humans where tumors evolve from somatic gene mutations in developmentally normal cells. Combined with the growing number of Cre expression models, RCAS-TVA-based gene expression and knockdown systems open up promising perspectives for analysis of gene function in a time-controlled and tissue-specific fashion in vitro and in vivo.


Subject(s)
Alpharetrovirus/genetics , Avian Proteins/genetics , Carcinoma, Pancreatic Ductal/genetics , Genetic Vectors , Pancreatic Neoplasms/genetics , Receptors, Virus/genetics , Animals , Carcinoma, Pancreatic Ductal/etiology , Carcinoma, Pancreatic Ductal/pathology , Disease Models, Animal , Gene Expression , Genes, p53 , Humans , Lac Operon , Mice , Mice, Transgenic , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/pathology , RNA Interference , RNA, Small Interfering/genetics
18.
Cancer Discov ; 11(12): 3158-3177, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34282029

ABSTRACT

Biliary tract cancer ranks among the most lethal human malignancies, representing an unmet clinical need. Its abysmal prognosis is tied to an increasing incidence and a fundamental lack of mechanistic knowledge regarding the molecular basis of the disease. Here, we show that the Pdx1-positive extrahepatic biliary epithelium is highly susceptible toward transformation by activated PIK3CAH1047R but refractory to oncogenic KrasG12D. Using genome-wide transposon screens and genetic loss-of-function experiments, we discover context-dependent genetic interactions that drive extrahepatic cholangiocarcinoma (ECC) and show that PI3K signaling output strength and repression of the tumor suppressor p27Kip1 are critical context-specific determinants of tumor formation. This contrasts with the pancreas, where oncogenic Kras in concert with p53 loss is a key cancer driver. Notably, inactivation of p27Kip1 permits KrasG12D-driven ECC development. These studies provide a mechanistic link between PI3K signaling, tissue-specific tumor suppressor barriers, and ECC pathogenesis, and present a novel genetic model of autochthonous ECC and genes driving this highly lethal tumor subtype. SIGNIFICANCE: We used the first genetically engineered mouse model for extrahepatic bile duct carcinoma to identify cancer genes by genome-wide transposon-based mutagenesis screening. Thereby, we show that PI3K signaling output strength and p27Kip1 function are critical determinants for context-specific ECC formation. This article is highlighted in the In This Issue feature, p. 2945.


Subject(s)
Bile Duct Neoplasms , Biliary Tract Neoplasms , Animals , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Biliary Tract Neoplasms/genetics , Genes, Tumor Suppressor , Humans , Mice , Phosphatidylinositol 3-Kinases/genetics
19.
Gastroenterology ; 137(1): 361-71, 371.e1-5, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19362090

ABSTRACT

BACKGROUND & AIMS: Early metastasis is a hallmark of pancreatic ductal adenocarcinoma and responsible for >90% of pancreatic cancer death. Because little is known about the biology and genetics of the metastatic process, we desired to elucidate molecular pathways mediating pancreatic cancer metastasis in vivo by an unbiased forward genetic approach. METHODS: Highly metastatic pancreatic cancer cell populations were selected by serial in vivo passaging of parental cells with low metastatic potential and characterized by global gene expression profiling, chromatin immunoprecipitation, and in vivo metastatic assay. RESULTS: In vivo selection of highly metastatic pancreatic cancer cells induced epithelial-mesenchymal transition (EMT), loss of E-cadherin expression, and up-regulation of mesenchymal genes such as Snail. Genetic inactivation of E-cadherin in parental cells induced EMT and increased metastasis in vivo. Silencing of E-cadherin in highly metastatic cells is mediated by a transcriptional repressor complex containing Snail and histone deacetylase 1 (HDAC1) and HDAC2. In line, mesenchymal pancreatic cancer specimens and primary cell lines from genetically engineered Kras(G12D) mice showed HDAC-dependent down-regulation of E-cadherin and high metastatic potential. Finally, transforming growth factor beta-driven E-cadherin silencing and EMT of human pancreatic cancer cells depends on HDAC activity. CONCLUSIONS: We provide the first in vivo evidence that HDACs and Snail play an essential role in silencing E-cadherin during the metastatic process of pancreatic cancer cells. These data link the epigenetic HDAC machinery to EMT and metastasis and provide preclinical evidence that HDACs are promising targets for antimetastatic therapy.


Subject(s)
Cadherins/metabolism , Histone Deacetylases/metabolism , Lung Neoplasms/enzymology , Pancreatic Neoplasms/enzymology , Transcription Factors/metabolism , Animals , Antigens, CD , Antineoplastic Agents/pharmacology , Cadherins/genetics , Cell Line, Tumor , Cell Transdifferentiation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Silencing , Histone Deacetylase 1 , Histone Deacetylase 2 , Histone Deacetylase Inhibitors , Histone Deacetylases/genetics , Humans , Hydroxamic Acids/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Mice , Mice, Nude , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic , RNA Interference , Repressor Proteins/metabolism , Snail Family Transcription Factors , Transcription Factors/genetics , Transfection
20.
Am J Physiol Cell Physiol ; 297(5): C1146-56, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19726747

ABSTRACT

Neuronal nitric oxide synthase (nNOS)-derived nitric oxide (NO) acts as a neurotransmitter and intracellular signaling molecule in the central and peripheral nervous system. NO regulates multiple processes like neuronal development, plasticity, and differentiation and is a mediator of neurotoxicity. The nNOS gene is highly complex with 12 alternative first exons, exon 1a-1l, transcribed from distinct promoters, leading to nNOS variants with different 5'-untranslated regions. Transcriptional control of the nNOS gene is not understood in detail. To investigate regulation of nNOS gene expression by retinoic acid (RA), we used the human neuroblastoma cell line TGW-nu-I as a model system. We show that RA induces nNOS transcription in a protein synthesis-dependent fashion. We identify the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway and the atypical orphan nuclear receptor DAX1 (NR0B1) as critical mediators involved in RA-induced nNOS gene transcription. RA treatment increases DAX1 expression via PI3K/Akt signaling. Upregulation of DAX1 expression in turn induces nNOS transcription in response to RA. These results identify nNOS as a target gene of a novel RA/PI3K/Akt/DAX1-dependent pathway in human neuroblastoma cells and stress the functional importance of the transcriptional regulator DAX1 for nNOS gene expression in response to RA treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Neuroblastoma/metabolism , Nitric Oxide Synthase Type I/drug effects , Signal Transduction/drug effects , Tretinoin/pharmacology , Blotting, Western , Cell Line, Tumor , DAX-1 Orphan Nuclear Receptor , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Gene Expression , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Immunoprecipitation , Nitric Oxide Synthase Type I/biosynthesis , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Retinoic Acid/drug effects , Receptors, Retinoic Acid/metabolism , Repressor Proteins/drug effects , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL