Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
Add more filters

Publication year range
1.
EMBO Rep ; 23(11): e55399, 2022 11 07.
Article in English | MEDLINE | ID: mdl-36194675

ABSTRACT

Anticancer T cells acquire a dysfunctional state characterized by poor effector function and expression of inhibitory receptors, such as PD-1. Blockade of PD-1 leads to T cell reinvigoration and is increasingly applied as an effective anticancer treatment. Recent work challenged the commonly held view that the phosphatase PTPN11 (known as SHP-2) is essential for PD-1 signaling in T cells, suggesting functional redundancy with the homologous phosphatase PTPN6 (SHP-1). Therefore, we investigated the effect of concomitant Ptpn6 and Ptpn11 deletion in T cells on their ability to mount antitumour responses. In vivo data show that neither sustained nor acute Ptpn6/11 deletion improves T cell-mediated tumor control. Sustained loss of Ptpn6/11 also impairs the therapeutic effects of anti-PD1 treatment. In vitro results show that Ptpn6/11-deleted CD8+ T cells exhibit impaired expansion due to a survival defect and proteomics analyses reveal substantial alterations, including in apoptosis-related pathways. These data indicate that concomitant ablation of Ptpn6/11 in polyclonal T cells fails to improve their anticancer properties, implying that caution shall be taken when considering their inhibition for immunotherapeutic approaches.


Subject(s)
CD8-Positive T-Lymphocytes , Programmed Cell Death 1 Receptor , CD8-Positive T-Lymphocytes/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Signal Transduction
2.
PLoS Genet ; 17(12): e1009250, 2021 12.
Article in English | MEDLINE | ID: mdl-34860830

ABSTRACT

Epigenetic mechanisms are gatekeepers for the gene expression patterns that establish and maintain cellular identity in mammalian development, stem cells and adult homeostasis. Amongst many epigenetic marks, methylation of histone 3 lysine 4 (H3K4) is one of the most widely conserved and occupies a central position in gene expression. Mixed lineage leukemia 1 (MLL1/KMT2A) is the founding mammalian H3K4 methyltransferase. It was discovered as the causative mutation in early onset leukemia and subsequently found to be required for the establishment of definitive hematopoiesis and the maintenance of adult hematopoietic stem cells. Despite wide expression, the roles of MLL1 in non-hematopoietic tissues remain largely unexplored. To bypass hematopoietic lethality, we used bone marrow transplantation and conditional mutagenesis to discover that the most overt phenotype in adult Mll1-mutant mice is intestinal failure. MLL1 is expressed in intestinal stem cells (ISCs) and transit amplifying (TA) cells but not in the villus. Loss of MLL1 is accompanied by loss of ISCs and a differentiation bias towards the secretory lineage with increased numbers and enlargement of goblet cells. Expression profiling of sorted ISCs revealed that MLL1 is required to promote expression of several definitive intestinal transcription factors including Pitx1, Pitx2, Foxa1, Gata4, Zfp503 and Onecut2, as well as the H3K27me3 binder, Bahcc1. These results were recapitulated using conditional mutagenesis in intestinal organoids. The stem cell niche in the crypt includes ISCs in close association with Paneth cells. Loss of MLL1 from ISCs promoted transcriptional changes in Paneth cells involving metabolic and stress responses. Here we add ISCs to the MLL1 repertoire and observe that all known functions of MLL1 relate to the properties of somatic stem cells, thereby highlighting the suggestion that MLL1 is a master somatic stem cell regulator.


Subject(s)
Adult Stem Cells/physiology , Cell Differentiation/genetics , Histone-Lysine N-Methyltransferase/genetics , Intestinal Failure/genetics , Intestinal Mucosa/pathology , Myeloid-Lymphoid Leukemia Protein/genetics , Animals , Bone Marrow Transplantation , DNA Methylation , Disease Models, Animal , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/metabolism , Humans , Intestinal Failure/pathology , Intestinal Mucosa/cytology , Jejunum/cytology , Jejunum/pathology , Mice , Mice, Transgenic , Mutagenesis , Mutation , Myeloid-Lymphoid Leukemia Protein/metabolism , Stem Cell Niche
3.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Article in English | MEDLINE | ID: mdl-34039707

ABSTRACT

Specified intestinal epithelial cells reprogram and contribute to the regeneration and renewal of the epithelium upon injury. Mutations that deregulate such renewal processes may contribute to tumorigenesis. Using intestinal organoids, we show that concomitant activation of Notch signaling and ablation of p53 induce a highly proliferative and regenerative cell state, which is associated with increased levels of Yap and the histone methyltransferase Mll1. The induced signaling system orchestrates high proliferation, self-renewal, and niche-factor-independent growth, and elevates the trimethylation of histone 3 at lysine 4 (H3K4me3). We demonstrate that Yap and Mll1 are also elevated in patient-derived colorectal cancer (CRC) organoids and control growth and viability. Our data suggest that Notch activation and p53 ablation induce a signaling circuitry involving Yap and the epigenetic regulator Mll1, which locks cells in a proliferative and regenerative state that renders them susceptible for tumorigenesis.


Subject(s)
Cell Cycle Proteins/physiology , Histone-Lysine N-Methyltransferase/physiology , Myeloid-Lymphoid Leukemia Protein/physiology , Receptors, Notch/metabolism , Signal Transduction , Transcription Factors/physiology , Tumor Suppressor Protein p53/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Mutation , Organoids/metabolism , Transcription Factors/metabolism
4.
Breast Cancer Res ; 24(1): 23, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35365185

ABSTRACT

BACKGROUND: PI3K signaling is frequently activated in breast cancer and is targeted by PI3K inhibitors. However, resistance of tumor cells to PI3K inhibition, often mediated by activated receptor tyrosine kinases, is commonly observed and reduces the potency of PI3K inhibitors. Therefore, new treatment strategies to overcome resistance to PI3K inhibitors are urgently needed to boost their efficacy. The phosphatase SHP2, which plays a crucial role in mediating signal transduction between receptor tyrosine kinases and both the PI3K and MAPK pathways, is a potential target for combination treatment. METHODS: We tested combinations of PI3K and SHP2 inhibitors in several experimental breast cancer models that are resistant to PI3K inhibition. Using cell culturing, biochemical and genetic approaches, we evaluated tumor cell proliferation and signaling output in cells treated with PI3K and SHP2 inhibitors. RESULTS: Combination treatment with PI3K and SHP2 inhibitors counteracted both acquired and intrinsic breast cancer cell resistance to PI3K inhibition that is mediated by activated receptor tyrosine kinases. Dual PI3K and SHP2 inhibition blocked proliferation and led to sustained inactivation of PI3K and MAPK signaling, where resistant cells rapidly re-activated these pathways upon PI3K inhibitor monotreatment. In addition, we demonstrate that overexpression of SHP2 induced resistance to PI3K inhibition, and that SHP2 was frequently activated during the development of PI3K inhibitor resistance after prolonged treatment of sensitive cells. CONCLUSIONS: Our results highlight the importance of SHP2 as a player in resistance to PI3K inhibitors. Combination treatment with PI3K and SHP2 inhibitors could pave the way for significant improvements in therapies for breast cancer.


Subject(s)
Breast Neoplasms , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Female , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Signal Transduction
5.
Genes Dev ; 28(3): 290-303, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24493648

ABSTRACT

Myelination depends on the synthesis of large amounts of myelin transcripts and proteins and is controlled by Nrg1/ErbB/Shp2 signaling. We developed a novel pulse labeling strategy based on stable isotope labeling with amino acids in cell culture (SILAC) to measure the dynamics of myelin protein production in mice. We found that protein synthesis is dampened in the maturing postnatal peripheral nervous system, and myelination then slows down. Remarkably, sustained activation of MAPK signaling by expression of the Mek1DD allele in mice overcomes the signals that end myelination, resulting in continuous myelin growth. MAPK activation leads to minor changes in transcript levels but massively up-regulates protein production. Pharmacological interference in vivo demonstrates that the effects of activated MAPK signaling on translation are mediated by mTOR-independent mechanisms but in part also by mTOR-dependent mechanisms. Previous work demonstrated that loss of ErbB3/Shp2 signaling impairs Schwann cell development and disrupts the myelination program. We found that activated MAPK signaling strikingly compensates for the absence of ErbB3 or Shp2 during Schwann cell development and myelination.


Subject(s)
Cell Differentiation , Mitogen-Activated Protein Kinases/metabolism , Myelin Sheath/metabolism , Neuregulin-1/metabolism , Receptor, ErbB-3/metabolism , Schwann Cells/cytology , Alleles , Animals , Gene Expression Regulation/genetics , MAP Kinase Kinase 1/genetics , Mechanistic Target of Rapamycin Complex 1 , Mice , Microscopy, Electron, Transmission , Multiprotein Complexes , Mutation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Receptor, ErbB-3/genetics , Schwann Cells/ultrastructure , Signal Transduction , TOR Serine-Threonine Kinases
6.
Brain ; 142(12): 3806-3833, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31665242

ABSTRACT

Microglia of the developing brain have unique functional properties but how their activation states are regulated is poorly understood. Inflammatory activation of microglia in the still-developing brain of preterm-born infants is associated with permanent neurological sequelae in 9 million infants every year. Investigating the regulators of microglial activation in the developing brain across models of neuroinflammation-mediated injury (mouse, zebrafish) and primary human and mouse microglia we found using analysis of genes and proteins that a reduction in Wnt/ß-catenin signalling is necessary and sufficient to drive a microglial phenotype causing hypomyelination. We validated in a cohort of preterm-born infants that genomic variation in the Wnt pathway is associated with the levels of connectivity found in their brains. Using a Wnt agonist delivered by a blood-brain barrier penetrant microglia-specific targeting nanocarrier we prevented in our animal model the pro-inflammatory microglial activation, white matter injury and behavioural deficits. Collectively, these data validate that the Wnt pathway regulates microglial activation, is critical in the evolution of an important form of human brain injury and is a viable therapeutic target.


Subject(s)
Brain/metabolism , Inflammation/metabolism , Microglia/metabolism , Wnt Signaling Pathway/physiology , Animals , Animals, Genetically Modified , Blood-Brain Barrier/metabolism , Cells, Cultured , Computational Biology , Humans , Mice , Zebrafish
7.
Genes Dev ; 26(15): 1743-57, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22810622

ABSTRACT

Neocortical neurons have highly branched dendritic trees that are essential for their function. Indeed, defects in dendritic arborization are associated with human neurodevelopmental disorders. The molecular mechanisms regulating dendritic arbor complexity, however, are still poorly understood. Here, we uncover the molecular basis for the regulation of dendritic branching during cortical development. We show that during development, dendritic branching requires post-mitotic suppression of the RhoGTPase Cdc42. By generating genetically modified mice, we demonstrate that this is catalyzed in vivo by the novel Cdc42-GAP NOMA-GAP. Loss of NOMA-GAP leads to decreased neocortical volume, associated specifically with profound oversimplification of cortical dendritic arborization and hyperactivation of Cdc42. Remarkably, dendritic complexity and cortical thickness can be partially restored by genetic reduction of post-mitotic Cdc42 levels. Furthermore, we identify the actin regulator cofilin as a key regulator of dendritic complexity in vivo. Cofilin activation during late cortical development depends on NOMA-GAP expression and subsequent inhibition of Cdc42. Strikingly, in utero expression of active cofilin is sufficient to restore postnatal dendritic complexity in NOMA-GAP-deficient animals. Our findings define a novel cell-intrinsic mechanism to regulate dendritic branching and thus neuronal complexity in the cerebral cortex.


Subject(s)
Actin Depolymerizing Factors/metabolism , Dendrites/metabolism , GTPase-Activating Proteins/metabolism , Neocortex/growth & development , Neocortex/metabolism , cdc42 GTP-Binding Protein/metabolism , Animals , Cells, Cultured , Female , GTPase-Activating Proteins/genetics , Mice , Mice, Transgenic
8.
EMBO J ; 34(11): 1493-508, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-25736378

ABSTRACT

In this study, we have used techniques from cell biology, biochemistry, and genetics to investigate the role of the tyrosine phosphatase Shp2 in tumor cells of MMTV-PyMT mouse mammary glands. Genetic ablation or pharmacological inhibition of Shp2 induces senescence, as determined by the activation of senescence-associated ß-gal (SA-ß-gal), cyclin-dependent kinase inhibitor 1B (p27), p53, and histone 3 trimethylated lysine 9 (H3K9me3). Senescence induction leads to the inhibition of self-renewal of tumor cells and blockage of tumor formation and growth. A signaling cascade was identified that acts downstream of Shp2 to counter senescence: Src, focal adhesion kinase, and Map kinase inhibit senescence by activating the expression of S-phase kinase-associated protein 2 (Skp2), Aurora kinase A (Aurka), and the Notch ligand Delta-like 1 (Dll1), which block p27 and p53. Remarkably, the expression of Shp2 and of selected target genes predicts human breast cancer outcome. We conclude that therapies, which rely on senescence induction by inhibiting Shp2 or controlling its target gene products, may be useful in blocking breast cancer.


Subject(s)
Cellular Senescence , Mammary Neoplasms, Animal/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Animals , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Calcium-Binding Proteins , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Histones , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Methylation , Mice , Mice, Transgenic , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Cell Microbiol ; 20(11): e12891, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30009515

ABSTRACT

Chronic periodontitis is characterised by gingival inflammation and alveolar bone loss. A major aetiological agent is Porphyromonas gingivalis, which secretes proteases that activate protease-activated receptor 2 (PAR2 ). PAR2 expressed on oral keratinocytes is activated by proteases released by P. gingivalis, inducing secretion of interleukin 6 (IL-6), and global knockout of PAR2 prevents bone loss and inflammation in a periodontal disease model in mice. To test the hypothesis that PAR2 expressed on gingival keratinocytes is required for periodontal disease pathology, keratinocyte-specific PAR2 -null mice were generated using K14-Cre targeted deletion of the PAR2 gene (F2rl1). These mice were subjected to a model of periodontitis involving placement of a ligature around a tooth, combined with P. gingivalis infection ("Lig + Inf"). The intervention caused a significant 44% decrease in alveolar bone volume (assessed by microcomputed tomography) in wildtype (K14-Cre:F2rl1wt/wt ), but not littermate keratinocyte-specific PAR2 -null (K14-Cre:F2rl1fl/fl ) mice. Keratinocyte-specific ablation of PAR2 prevented the significant Lig + Inf-induced increase (2.8-fold) in the number of osteoclasts in alveolar bone and the significant up-regulation (2.4-4-fold) of the inflammatory markers IL-6, IL-1ß, interferon-γ, myeloperoxidase, and CD11b in gingival tissue. These data suggest that PAR2 expressed on oral epithelial cells is a critical regulator of periodontitis-induced bone loss and will help in designing novel therapies with which to treat the disease.


Subject(s)
Alveolar Bone Loss/etiology , Gingivitis/genetics , Keratinocytes/metabolism , Periodontal Diseases/etiology , Receptor, PAR-2/metabolism , Alveolar Bone Loss/genetics , Animals , Bacteroidaceae Infections/etiology , CD11b Antigen/metabolism , Disease Models, Animal , Gene Expression Regulation , Gingivitis/etiology , Interleukin-6/metabolism , Keratinocytes/pathology , Mice, Mutant Strains , Porphyromonas gingivalis/pathogenicity , Receptor, PAR-2/genetics
10.
J Biol Chem ; 291(37): 19618-30, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27484798

ABSTRACT

The A-kinase anchoring protein (AKAP) GSK3ß interaction protein (GSKIP) is a cytosolic scaffolding protein binding protein kinase A (PKA) and glycogen synthase kinase 3ß (GSK3ß). Here we show that both the AKAP function of GSKIP, i.e. its direct interaction with PKA, and its direct interaction with GSK3ß are required for the regulation of ß-catenin and thus Wnt signaling. A cytoplasmic destruction complex targets ß-catenin for degradation and thus prevents Wnt signaling. Wnt signals cause ß-catenin accumulation and translocation into the nucleus, where it induces Wnt target gene expression. GSKIP facilitates control of the ß-catenin stabilizing phosphorylation at Ser-675 by PKA. Its interaction with GSK3ß facilitates control of the destabilizing phosphorylation of ß-catenin at Ser-33/Ser-37/Thr-41. The influence of GSKIP on ß-catenin is explained by its scavenger function; it recruits the kinases away from the destruction complex without forming a complex with ß-catenin. The regulation of ß-catenin by GSKIP is specific for this AKAP as AKAP220, which also binds PKA and GSK3ß, did not affect Wnt signaling. We find that the binding domain of AKAP220 for GSK3ß is a conserved GSK3ß interaction domain (GID), which is also present in GSKIP. Our findings highlight an essential compartmentalization of both PKA and GSK3ß by GSKIP, and ascribe a function to a cytosolic AKAP-PKA interaction as a regulatory factor in the control of canonical Wnt signaling. Wnt signaling controls different biological processes, including embryonic development, cell cycle progression, glycogen metabolism, and immune regulation; deregulation is associated with diseases such as cancer, type 2 diabetes, inflammatory, and Alzheimer's and Parkinson's diseases.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Repressor Proteins/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , A Kinase Anchor Proteins , A549 Cells , Cyclic AMP-Dependent Protein Kinases/genetics , Glycogen Synthase Kinase 3 beta/genetics , HEK293 Cells , HeLa Cells , Humans , Protein Domains , Repressor Proteins/genetics , beta Catenin/genetics
11.
EMBO J ; 32(14): 1977-89, 2013 Jul 17.
Article in English | MEDLINE | ID: mdl-23736260

ABSTRACT

We show that activation of Wnt/ß-catenin and attenuation of Bmp signals, by combined gain- and loss-of-function mutations of ß-catenin and Bmpr1a, respectively, results in rapidly growing, aggressive squamous cell carcinomas (SCC) in the salivary glands of mice. Tumours contain transplantable and hyperproliferative tumour propagating cells, which can be enriched by fluorescence activated cell sorting (FACS). Single mutations stimulate stem cells, but tumours are not formed. We show that ß-catenin, CBP and Mll promote self-renewal and H3K4 tri-methylation in tumour propagating cells. Blocking ß-catenin-CBP interaction with the small molecule ICG-001 and small-interfering RNAs against ß-catenin, CBP or Mll abrogate hyperproliferation and H3K4 tri-methylation, and induce differentiation of cultured tumour propagating cells into acini-like structures. ICG-001 decreases H3K4me3 at promoters of stem cell-associated genes in vitro and reduces tumour growth in vivo. Remarkably, high Wnt/ß-catenin and low Bmp signalling also characterize human salivary gland SCC and head and neck SCC in general. Our work defines mechanisms by which ß-catenin signals remodel chromatin and control induction and maintenance of tumour propagating cells. Further, it supports new strategies for the therapy of solid tumours.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Salivary Gland Neoplasms/genetics , Salivary Gland Neoplasms/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , CREB-Binding Protein/antagonists & inhibitors , CREB-Binding Protein/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation/drug effects , Epigenesis, Genetic , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Histone Methyltransferases , Humans , Mice , Mice, Inbred NOD , Mice, Mutant Strains , Mice, SCID , Mice, Transgenic , Mutation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pyrimidinones/pharmacology , Salivary Gland Neoplasms/pathology , Transplantation, Heterologous , Wnt Signaling Pathway/drug effects , beta Catenin/antagonists & inhibitors
12.
Bioessays ; 37(5): 502-13, 2015 May.
Article in English | MEDLINE | ID: mdl-25707700

ABSTRACT

In this article we discuss the molecular signaling mechanisms that coordinate interactions between Schwann cells and the neurons of the peripheral nervous system. Such interactions take place perpetually during development and in adulthood, and are critical for the homeostasis of the peripheral nervous system (PNS). Neurons provide essential signals to control Schwann cell functions, whereas Schwann cells promote neuronal survival and allow efficient transduction of action potentials. Deregulation of neuron-Schwann cell interactions often results in developmental abnormalities and diseases. Recent investigations have shown that during development, neuronally provided signals, such as Neuregulin, Jagged, and Wnt interact to fine-tune the Schwann cell lineage progression. In adult, the signal exchange between neurons and Schwann cells ensures proper nerve function and regeneration. Identification of the mechanisms of neuron-Schwann cell interactions is therefore essential for our understanding of the development, function and pathology of the peripheral nervous system as a whole.


Subject(s)
Axons/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Peripheral Nervous System/cytology , Peripheral Nervous System/metabolism , Animals , Humans , Signal Transduction/physiology , Wnt Proteins/metabolism
13.
Proc Natl Acad Sci U S A ; 111(9): 3472-7, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24550486

ABSTRACT

In the development of the mammalian intestine, Notch and Wnt/ß-catenin signals control stem cell maintenance and their differentiation into absorptive and secretory cells. Mechanisms that regulate differentiation of progenitors into the three secretory lineages, goblet, paneth, or enteroendocrine cells, are not fully understood. Using conditional mutagenesis in mice, we observed that Shp2-mediated MAPK signaling determines the choice between paneth and goblet cell fates and also affects stem cells, which express the leucine-rich repeat-containing receptor 5 (Lgr5). Ablation of the tyrosine phosphatase Shp2 in the intestinal epithelium reduced MAPK signaling and led to a reduction of goblet cells while promoting paneth cell development. Conversely, conditional mitogen-activated protein kinase kinase 1 (Mek1) activation rescued the Shp2 phenotype, promoted goblet cell and inhibited paneth cell generation. The Shp2 mutation also expanded Lgr5+ stem cell niches, which could be restricted by activated Mek1 signaling. Changes of Lgr5+ stem cell quantities were accompanied by alterations of paneth cells, indicating that Shp2/MAPK signaling might affect stem cell niches directly or via paneth cells. Remarkably, inhibition of MAPK signaling in intestinal organoids and cultured cells changed the relative abundance of Tcf4 isoforms and by this, promoted Wnt/ß-catenin activity. The data thus show that Shp2-mediated MAPK signaling controls the choice between goblet and paneth cell fates by regulating Wnt/ß-catenin activity.


Subject(s)
Cell Differentiation/physiology , Goblet Cells/physiology , Intestinal Mucosa/cytology , Paneth Cells/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Stem Cells/cytology , Wnt Signaling Pathway/physiology , Animals , Blotting, Western , Goblet Cells/cytology , HT29 Cells , Humans , Immunoprecipitation , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Organ Culture Techniques , Paneth Cells/cytology , beta Catenin/metabolism
14.
Gut ; 65(10): 1690-701, 2016 10.
Article in English | MEDLINE | ID: mdl-26156959

ABSTRACT

OBJECTIVES: We have previously identified a 115-gene signature that characterises the metastatic potential of human primary colon cancers. The signature included the canonical Wnt target gene BAMBI, which promoted experimental metastasis in mice. Here, we identified three new direct Wnt target genes from the signature, and studied their functions in epithelial-mesenchymal transition (EMT), cell migration and experimental metastasis. DESIGN: We examined experimental liver metastases following injection of selected tumour cells into spleens of NOD/SCID mice. Molecular and cellular techniques were used to identify direct transcription target genes of Wnt/ß-catenin signals. Microarray analyses and experiments that interfered with cell migration through inhibitors were performed to characterise downstream signalling systems. RESULTS: Three new genes from the colorectal cancer (CRC) metastasis signature, BOP1, CKS2 and NFIL3, were identified as direct transcription targets of ß-catenin/TCF4. Overexpression and knocking down of these genes in CRC cells promoted and inhibited, respectively, experimental metastasis in mice, EMT and cell motility in culture. Cell migration was repressed by interfering with distinct signalling systems through inhibitors of PI3K, JNK, p38 mitogen-activated protein kinase and/or mTOR. Gene expression profiling identified a series of migration-promoting genes, which were induced by BOP1, CKS2 and NFIL3, and could be repressed by inhibitors that are specific to these pathways. CONCLUSIONS: We identified new direct Wnt/ß-catenin target genes, BOP1, CKS2 and NFIL3, which induced EMT, cell migration and experimental metastasis of CRC cells. These genes crosstalk with different downstream signalling systems, and activate migration-promoting genes. These pathways and downstream genes may serve as therapeutic targets in the treatment of CRC metastasis.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , CDC28 Protein Kinase, S cerevisiae/genetics , Cell Movement/genetics , Colorectal Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Liver Neoplasms , Nuclear Proteins/genetics , Wnt Signaling Pathway/genetics , Animals , CDC2-CDC28 Kinases , Cell Cycle Proteins , Disease Models, Animal , Humans , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Liver Neoplasms, Experimental , Mice , Neoplasm Metastasis , RNA-Binding Proteins , Tumor Cells, Cultured
15.
Proc Natl Acad Sci U S A ; 110(45): 18174-9, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24151333

ABSTRACT

During late Schwann cell development, immature Schwann cells segregate large axons from bundles, a process called "axonal radial sorting." Here we demonstrate that canonical Wnt signals play a critical role in radial sorting and assign a role to Wnt and Rspondin ligands in this process. Mice carrying ß-catenin loss-of-function mutations show a delay in axonal sorting; conversely, gain-of-function mutations result in accelerated sorting. Sorting deficits are accompanied by abnormal process extension, differentiation, and aberrant cell cycle exit of the Schwann cells. Using primary cultured Schwann cells, we analyze the upstream effectors, Wnt and Rspondin ligands that initiate signaling, and downstream genetic programs that mediate the Wnt response. Our analysis contributes to a better understanding of the mechanisms of Schwann cell development and fate decisions.


Subject(s)
Axons/physiology , Cell Lineage/physiology , Schwann Cells/physiology , Thrombospondins/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Animals , Blotting, Western , DNA Primers/genetics , Flow Cytometry , In Situ Hybridization , Mice , Mice, Transgenic , Microarray Analysis , Mutation/genetics , Paracrine Communication/physiology , Real-Time Polymerase Chain Reaction , Sciatic Nerve/physiology , Sciatic Nerve/ultrastructure , beta Catenin/genetics
16.
Genes Cells ; 19(11): 842-52, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25297509

ABSTRACT

Afadin is a filamentous actin-binding protein and a mediator of nectin signaling. Nectins are Ig-like cell adhesion molecules, and the nectin family is composed of four members, nectin-1 to nectin-4. Nectins show homophilic and heterophilic interactions with other nectins or proteins on adjacent cells. Nectin signaling induces formation of cell-cell junctions and is required for the development of epithelial tissues, including skin. This study investigated the role of afadin in epithelial tissue development and established epithelium-specific afadin-deficient (CKO) mice. Although showing no obvious abnormality in the skin development and homeostasis, the mice showed the reduced neutrophil infiltration into the epidermis during chemical-induced inflammation with 12-O-tetradecanoylphorbol 13-acetate (TPA). Immunohistochemical and quantitative real-time PCR analyses showed that the expression levels of cytokines including Cxcl2, Il-1ß and Tnf-α were reduced in CKO keratinocytes compared with control keratinocytes during TPA-induced inflammation. Primary-cultured skin keratinocytes from CKO mice also showed reduced expression of these cytokines and weak activation of Rap1 compared with those from control mice after the TPA treatment. These results suggested a remarkable function of afadin, which was able to enhance cytokine expression through Rap1 activation in keratinocytes during inflammation.


Subject(s)
Chemokine CXCL2/metabolism , Interleukin-1beta/metabolism , Keratinocytes/metabolism , Microfilament Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Epidermis/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Mice , Mice, Knockout , Microfilament Proteins/genetics , Neutrophils/metabolism , Primary Cell Culture , Tetradecanoylphorbol Acetate , rap1 GTP-Binding Proteins/metabolism
17.
Proc Natl Acad Sci U S A ; 109(27): 10921-6, 2012 Jul 03.
Article in English | MEDLINE | ID: mdl-22711842

ABSTRACT

Progenitor cells of the first and second heart fields depend on cardiac-specific transcription factors for their differentiation. Using conditional mutagenesis of mouse embryos, we define the hierarchy of signaling events that controls the expression of cardiac-specific transcription factors during differentiation of cardiac progenitors at embryonic day 9.0. Wnt/ß-catenin and Bmp act downstream of Notch/RBPJ at this developmental stage. Mutation of Axin2, the negative regulator of canonical Wnt signaling, enhances Wnt and Bmp4 signals and suffices to rescue the arrest of cardiac differentiation caused by loss of RBPJ. Using FACS enrichment of cardiac progenitors in RBPJ and RBPJ/Axin2 mutants, embryo cultures in the presence of the Bmp inhibitor Noggin, and by crossing a Bmp4 mutation into the RBPJ/Axin2 mutant background, we show that Wnt and Bmp4 signaling activate specific and nonoverlapping cardiac-specific genes in the cardiac progenitors: Nkx2-5, Isl1 and Baf60c are controlled by Wnt/ß-catenin, and Gata4, SRF, and Mef2c are controlled by Bmp signaling. Our study contributes to the understanding of the regulatory hierarchies of cardiac progenitor differentiation and outflow tract development and has implications for understanding and modeling heart development.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Embryonic Stem Cells/metabolism , Myocytes, Cardiac/cytology , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Animals , Axin Protein/genetics , Axin Protein/metabolism , Cell Differentiation/physiology , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Embryonic Stem Cells/cytology , Female , Flow Cytometry , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Lac Operon , Male , Mice , Mice, Mutant Strains , Muscle Proteins/genetics , Muscle Proteins/metabolism , Pregnancy , Receptors, Notch/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Nature ; 452(7187): 650-3, 2008 Apr 03.
Article in English | MEDLINE | ID: mdl-18385740

ABSTRACT

Continuous turnover of epithelia is ensured by the extensive self-renewal capacity of tissue-specific stem cells. Similarly, epithelial tumour maintenance relies on cancer stem cells (CSCs), which co-opt stem cell properties. For most tumours, the cellular origin of these CSCs and regulatory pathways essential for sustaining stemness have not been identified. In murine skin, follicular morphogenesis is driven by bulge stem cells that specifically express CD34. Here we identify a population of cells in early epidermal tumours characterized by phenotypic and functional similarities to normal bulge skin stem cells. This population contains CSCs, which are the only cells with tumour initiation properties. Transplants derived from these CSCs preserve the hierarchical organization of the primary tumour. We describe beta-catenin signalling as being essential in sustaining the CSC phenotype. Ablation of the beta-catenin gene results in the loss of CSCs and complete tumour regression. In addition, we provide evidence for the involvement of increased beta-catenin signalling in malignant human squamous cell carcinomas. Because Wnt/beta-catenin signalling is not essential for normal epidermal homeostasis, such a mechanistic difference may thus be targeted to eliminate CSCs and consequently eradicate squamous cell carcinomas.


Subject(s)
Cell Transformation, Neoplastic , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction , Skin Neoplasms/pathology , beta Catenin/metabolism , Animals , Antigens, CD34/metabolism , Cell Line, Tumor , Cells, Cultured , Epidermis/pathology , Humans , Mice , Mice, Nude , Neoplasm Transplantation
20.
PLoS One ; 19(3): e0282938, 2024.
Article in English | MEDLINE | ID: mdl-38512983

ABSTRACT

Previously, we found that Wnt and Notch signaling govern stem cells of clear cell kidney cancer (ccRCC) in patients. To mimic stem cell responses in the normal kidney in vitro in a marker-unbiased fashion, we have established tubular organoids (tubuloids) from total single adult mouse kidney epithelial cells in Matrigel and serum-free conditions. Deep proteomic and phosphoproteomic analyses revealed that tubuloids resembled renewal of adult kidney tubular epithelia, since tubuloid cells displayed activity of Wnt and Notch signaling, long-term proliferation and expression of markers of proximal and distal nephron lineages. In our wish to model stem cell-derived human ccRCC, we have generated two types of genetic double kidney mutants in mice: Wnt-ß-catenin-GOF together with Notch-GOF and Wnt-ß-catenin-GOF together with a most common alteration in ccRCC, Vhl-LOF. An inducible Pax8-rtTA-LC1-Cre was used to drive recombination specifically in adult kidney epithelial cells. We confirmed mutagenesis of ß-catenin, Notch and Vhl alleles on DNA, protein and mRNA target gene levels. Surprisingly, we observed symptoms of chronic kidney disease (CKD) in mutant mice, but no increased proliferation and tumorigenesis. Thus, the responses of kidney stem cells in the tubuloid and genetic systems produced different phenotypes, i.e. enhanced renewal versus CKD.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Renal Insufficiency, Chronic , Adult , Humans , Mice , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , beta Catenin/metabolism , Proteomics , Stem Cells/metabolism , Renal Insufficiency, Chronic/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL