Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Nat Cell Biol ; 26(1): 153-167, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38182825

ABSTRACT

In the mammalian liver, hepatocytes exhibit diverse metabolic and functional profiles based on their location within the liver lobule. However, it is unclear whether this spatial variation, called zonation, is governed by a well-defined gene regulatory code. Here, using a combination of single-cell multiomics, spatial omics, massively parallel reporter assays and deep learning, we mapped enhancer-gene regulatory networks across mouse liver cell types. We found that zonation affects gene expression and chromatin accessibility in hepatocytes, among other cell types. These states are driven by the repressors TCF7L1 and TBX3, alongside other core hepatocyte transcription factors, such as HNF4A, CEBPA, FOXA1 and ONECUT1. To examine the architecture of the enhancers driving these cell states, we trained a hierarchical deep learning model called DeepLiver. Our study provides a multimodal understanding of the regulatory code underlying hepatocyte identity and their zonation state that can be used to engineer enhancers with specific activity levels and zonation patterns.


Subject(s)
Deep Learning , Multiomics , Mice , Animals , Gene Regulatory Networks , Liver/metabolism , Hepatocytes , Mammals
2.
Mol Cancer Ther ; 23(1): 3-13, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-37748190

ABSTRACT

The Hippo pathway and its downstream effectors, the YAP and TAZ transcriptional coactivators, are deregulated in multiple different types of human cancer and are required for cancer cell phenotypes in vitro and in vivo, while largely dispensable for tissue homeostasis in adult mice. YAP/TAZ and their main partner transcription factors, the TEAD1-4 factors, are therefore promising anticancer targets. Because of frequent YAP/TAZ hyperactivation caused by mutations in the Hippo pathway components NF2 and LATS2, mesothelioma is one of the prime cancer types predicted to be responsive to YAP/TAZ-TEAD inhibitor treatment. Mesothelioma is a devastating disease for which currently no effective treatment options exist. Here, we describe a novel covalent YAP/TAZ-TEAD inhibitor, SWTX-143, that binds to the palmitoylation pocket of all four TEAD isoforms. SWTX-143 caused irreversible and specific inhibition of the transcriptional activity of YAP/TAZ-TEAD in Hippo-mutant tumor cell lines. More importantly, YAP/TAZ-TEAD inhibitor treatment caused strong mesothelioma regression in subcutaneous xenograft models with human cells and in an orthotopic mesothelioma mouse model. Finally, SWTX-143 also selectively impaired the growth of NF2-mutant kidney cancer cell lines, suggesting that the sensitivity of mesothelioma models to these YAP/TAZ-TEAD inhibitors can be extended to other tumor types with aberrations in Hippo signaling. In brief, we describe a novel and specific YAP/TAZ-TEAD inhibitor that has potential to treat multiple Hippo-mutant solid tumor types.


Subject(s)
Mesothelioma, Malignant , Mesothelioma , Adult , Humans , Animals , Mice , Hippo Signaling Pathway , YAP-Signaling Proteins , Transcription Factors/genetics , Transcription Factors/metabolism , Mesothelioma/drug therapy , Mesothelioma/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism
3.
Gastroenterology ; 160(3): 847-862, 2021 02.
Article in English | MEDLINE | ID: mdl-33127392

ABSTRACT

BACKGROUND AND AIMS: The Hippo pathway and its downstream effectors YAP and TAZ (YAP/TAZ) are heralded as important regulators of organ growth and regeneration. However, different studies provided contradictory conclusions about their role during regeneration of different organs, ranging from promoting proliferation to inhibiting it. Here we resolve the function of YAP/TAZ during regeneration of the liver, where Hippo's role in growth control has been studied most intensely. METHODS: We evaluated liver regeneration after carbon tetrachloride toxic liver injury in mice with conditional deletion of Yap/Taz in hepatocytes and/or biliary epithelial cells, and measured the behavior of different cell types during regeneration by histology, RNA sequencing, and flow cytometry. RESULTS: We found that YAP/TAZ were activated in hepatocytes in response to carbon tetrachloride toxic injury. However, their targeted deletion in adult hepatocytes did not noticeably impair liver regeneration. In contrast, Yap/Taz deletion in adult bile ducts caused severe defects and delay in liver regeneration. Mechanistically, we showed that Yap/Taz mutant bile ducts degenerated, causing cholestasis, which stalled the recruitment of phagocytic macrophages and the removal of cellular corpses from injury sites. Elevated bile acids activated pregnane X receptor, which was sufficient to recapitulate the phenotype observed in mutant mice. CONCLUSIONS: Our data show that YAP/TAZ are practically dispensable in hepatocytes for liver development and regeneration. Rather, YAP/TAZ play an indirect role in liver regeneration by preserving bile duct integrity and securing immune cell recruitment and function.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Chemical and Drug Induced Liver Injury/pathology , Cholestasis/pathology , Liver Regeneration/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Bile Ducts/pathology , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Cell Proliferation/genetics , Chemical and Drug Induced Liver Injury/complications , Cholestasis/etiology , Disease Models, Animal , Hepatocytes/drug effects , Hepatocytes/pathology , Hippo Signaling Pathway , Humans , Liver/drug effects , Liver/pathology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , YAP-Signaling Proteins
4.
Science ; 366(6468): 1029-1034, 2019 11 22.
Article in English | MEDLINE | ID: mdl-31754005

ABSTRACT

The Hippo signaling pathway and its two downstream effectors, the YAP and TAZ transcriptional coactivators, are drivers of tumor growth in experimental models. Studying mouse models, we show that YAP and TAZ can also exert a tumor-suppressive function. We found that normal hepatocytes surrounding liver tumors displayed activation of YAP and TAZ and that deletion of Yap and Taz in these peritumoral hepatocytes accelerated tumor growth. Conversely, experimental hyperactivation of YAP in peritumoral hepatocytes triggered regression of primary liver tumors and melanoma-derived liver metastases. Furthermore, whereas tumor cells growing in wild-type livers required YAP and TAZ for their survival, those surrounded by Yap- and Taz-deficient hepatocytes were not dependent on YAP and TAZ. Tumor cell survival thus depends on the relative activity of YAP and TAZ in tumor cells and their surrounding tissue, suggesting that YAP and TAZ act through a mechanism of cell competition to eliminate tumor cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Cholangiocarcinoma/metabolism , Hepatocytes/metabolism , Liver Neoplasms, Experimental/metabolism , Trans-Activators/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Survival , Cholangiocarcinoma/pathology , Hippo Signaling Pathway , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Liver Neoplasms, Experimental/pathology , Melanoma/metabolism , Melanoma/secondary , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Trans-Activators/economics , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Tumor Burden , YAP-Signaling Proteins
5.
Proc Natl Acad Sci U S A ; 115(42): 10684-10689, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30257938

ABSTRACT

The Hippo tumor-suppressor pathway regulates organ growth, cell proliferation, and stem cell biology. Defects in Hippo signaling and hyperactivation of its downstream effectors-Yorkie (Yki) in Drosophila and YAP/TAZ in mammals-result in progenitor cell expansion and overgrowth of multiple organs and contribute to cancer development. Deciphering the mechanisms that regulate the activity of the Hippo pathway is key to understanding its function and for therapeutic targeting. However, although the Hippo kinase cascade and several other upstream inputs have been identified, the mechanisms that regulate Yki/YAP/TAZ activity are still incompletely understood. To identify new regulators of Yki activity, we screened in Drosophila for suppressors of tissue overgrowth and Yki activation caused by overexpression of atypical protein kinase C (aPKC), a member of the apical cell polarity complex. In this screen, we identified mutations in the heterogeneous nuclear ribonucleoprotein Hrb27C that strongly suppressed the tissue defects induced by ectopic expression of aPKC. Hrb27C was required for aPKC-induced tissue growth and Yki target gene expression but did not affect general gene expression. Genetic and biochemical experiments showed that Hrb27C affects Yki phosphorylation. Other RNA-binding proteins known to interact with Hrb27C for mRNA transport in oocytes were also required for normal Yki activity, although they suppressed Yki output. Based on the known functions of Hrb27C, we conclude that Hrb27C-mediated control of mRNA splicing, localization, or translation is essential for coordinated activity of the Hippo pathway.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Nuclear Proteins/genetics , Poly(A)-Binding Protein II/genetics , Poly(A)-Binding Protein II/metabolism , RNA-Binding Proteins/genetics , Signal Transduction , Trans-Activators/genetics , YAP-Signaling Proteins
6.
Dev Cell ; 42(6): 667-680.e4, 2017 09 25.
Article in English | MEDLINE | ID: mdl-28950103

ABSTRACT

Hyperactivating mutations in Ras signaling are hallmarks of carcinomas. Ras signaling mediates cell fate decisions as well as proliferation during development. It is not known what dictates whether Ras signaling drives differentiation versus proliferation. Here we show that the Hippo pathway is critical for this decision. Loss of Hippo switches Ras activation from promoting cellular differentiation to aggressive cellular proliferation. Transcriptome analysis combined with genetic tests show that this excessive proliferation depends on the synergistic induction of Ras target genes. Using ChIP-nexus, we find that Hippo signaling keeps Ras targets in check by directly regulating the expression of two key downstream transcription factors of Ras signaling: the ETS-domain transcription factor Pointed and the repressor Capicua. Our results highlight how independent signaling pathways can impinge on each other at the level of transcription factors, thereby providing a safety mechanism to keep proliferation in check under normal developmental conditions.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Signal Transduction , Transcription, Genetic , ras Proteins/metabolism , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Developmental , Genes, Insect , Models, Biological , Mutation/genetics , Pupa/metabolism , Regulon/genetics , Sequence Analysis, RNA , Transcription Factors/metabolism
7.
PLoS Genet ; 9(8): e1003731, 2013 Aug.
Article in English | MEDLINE | ID: mdl-24009524

ABSTRACT

Organ development is directed by selector gene networks. Eye development in the fruit fly Drosophila melanogaster is driven by the highly conserved selector gene network referred to as the "retinal determination gene network," composed of approximately 20 factors, whose core comprises twin of eyeless (toy), eyeless (ey), sine oculis (so), dachshund (dac), and eyes absent (eya). These genes encode transcriptional regulators that are each necessary for normal eye development, and sufficient to direct ectopic eye development when misexpressed. While it is well documented that the downstream genes so, eya, and dac are necessary not only during early growth and determination stages but also during the differentiation phase of retinal development, it remains unknown how the retinal determination gene network terminates its functions in determination and begins to promote differentiation. Here, we identify a switch in the regulation of ey by the downstream retinal determination genes, which is essential for the transition from determination to differentiation. We found that central to the transition is a switch from positive regulation of ey transcription to negative regulation and that both types of regulation require so. Our results suggest a model in which the retinal determination gene network is rewired to end the growth and determination stage of eye development and trigger terminal differentiation. We conclude that changes in the regulatory relationships among members of the retinal determination gene network are a driving force for key transitions in retinal development.


Subject(s)
Cell Differentiation/genetics , Drosophila melanogaster/growth & development , Eye/growth & development , Organogenesis/genetics , Retina/growth & development , Animals , Conserved Sequence , DNA-Binding Proteins , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Retinaldehyde
8.
Curr Biol ; 23(3): 229-35, 2013 Feb 04.
Article in English | MEDLINE | ID: mdl-23333314

ABSTRACT

The Drosophila Yorkie (Yki) protein and its mammalian homolog Yes-associated protein (YAP) are potent growth promoters, and YAP overexpression is associated with multiple types of cancer. Yki and YAP are transcriptional coactivators and function as downstream effectors of the Hippo tumor suppressor pathway. The regulation of Yki and YAP by the Hippo signaling pathway has been extensively investigated; however, how they regulate gene expression is poorly understood. To identify additional regulators of Yki activity, we performed a genome-wide RNAi screen in Drosophila S2 cells. In this screen, we identified the conserved protein Mask (Multiple ankyrin repeats single KH domain) as a novel promoter of Yki activity in vitro and validated this function in vivo in Drosophila. We found that Mask is required downstream of the Hippo pathway for Yki to induce target-gene expression and that Mask forms complexes with Yki. The human Mask homolog MASK1 complexes with YAP and is required for the full activity of YAP. Additionally, elevated MASK1 expression is associated with worsened outcomes for breast cancer patients. We conclude that Mask is a novel cofactor for Yki/YAP required for optimal Yki/YAP activity during development and oncogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Gene Expression Regulation , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Trans-Activators/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line , DNA-Binding Proteins/genetics , Drosophila/genetics , Drosophila Proteins/genetics , Female , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA-Binding Proteins/metabolism , Signal Transduction , Transcription Factors , YAP-Signaling Proteins
9.
EMBO J ; 30(12): 2325-35, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21556047

ABSTRACT

The Hippo tumour suppressor pathway is a conserved signalling pathway that controls organ size. The core of the Hpo pathway is a kinase cascade, which in Drosophila involves the Hpo and Warts kinases that negatively regulate the activity of the transcriptional coactivator Yorkie. Although several additional components of the Hippo pathway have been discovered, the inputs that regulate Hippo signalling are not fully understood. Here, we report that induction of extra F-actin formation, by loss of Capping proteins A or B, or caused by overexpression of an activated version of the formin Diaphanous, induced strong overgrowth in Drosophila imaginal discs through modulating the activity of the Hippo pathway. Importantly, loss of Capping proteins and Diaphanous overexpression did not significantly affect cell polarity and other signalling pathways, including Hedgehog and Decapentaplegic signalling. The interaction between F-actin and Hpo signalling is evolutionarily conserved, as the activity of the mammalian Yorkie-orthologue Yap is modulated by changes in F-actin. Thus, regulators of F-actin, and in particular Capping proteins, are essential for proper growth control by affecting Hippo signalling.


Subject(s)
Actins/genetics , Drosophila Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Wings, Animal/cytology , Wings, Animal/growth & development , Actins/biosynthesis , Actins/chemistry , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Proliferation , Cells, Cultured , Cytoskeleton/chemistry , Cytoskeleton/genetics , Drosophila Proteins/biosynthesis , Drosophila Proteins/chemistry , Drosophila melanogaster/chemistry , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Formins , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Organ Specificity/genetics , Phenotype , Protein Serine-Threonine Kinases/chemistry , RNA Caps/antagonists & inhibitors , RNA Caps/chemistry , RNA Caps/genetics , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Wings, Animal/chemistry
10.
Proc Natl Acad Sci U S A ; 107(36): 15810-5, 2010 Sep 07.
Article in English | MEDLINE | ID: mdl-20798049

ABSTRACT

Defects in apical-basal cell polarity and abnormal expression of cell polarity determinants are often associated with cancer in vertebrates. In Drosophila, abnormal expression of apical-basal determinants can cause neoplastic phenotypes, including loss of cell polarity and overproliferation. However, the pathways through which apical-basal polarity determinants affect growth are poorly understood. Here, we investigated the mechanism by which the apical determinant Crumbs (Crb) affects growth in Drosophila imaginal discs. Overexpression of Crb causes severe overproliferation, and we found that loss of Crb similarly results in overgrowth of imaginal discs. Crb gain and loss of function caused defects in Hippo signaling, a key signaling pathway that controls tissue growth in Drosophila and mammals. Manipulation of Crb levels caused the up-regulation of Hippo target genes, genetically interacted with known Hippo pathway components, and required Yorkie, a transcriptional coactivator that acts downstream in the Hippo pathway, for target gene induction and overgrowth. Interestingly, Crb regulates growth and cell polarity through different motifs in its intracellular domain. A juxtamembrane FERM domain-binding motif is responsible for growth regulation and induction of Hippo target gene expression, whereas Crb uses a PDZ-binding motif to form a complex with other polarity factors. The Hippo pathway component Expanded, an apically localized adaptor protein, is mislocalized in both crb mutant cells and Crb overexpressing tissues, whereas the other Hippo pathway components, Fat and Merlin, are unaffected. Taken together, our data show that Crb regulates growth through Hippo signaling, and thus identify Crb as a previously undescribed upstream input into the Hippo pathway.


Subject(s)
Cell Polarity/physiology , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/physiology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Animals , Cell Proliferation , Drosophila
11.
J Cell Sci ; 122(Pt 14): 2351-9, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19531584

ABSTRACT

The Hippo tumor-suppressor pathway controls tissue growth in Drosophila and mammals by regulating cell proliferation and apoptosis. The Hippo pathway includes the Fat cadherin, a transmembrane protein, which acts upstream of several other components that form a kinase cascade that culminates in the regulation of gene expression through the transcriptional coactivator Yorkie (Yki). Our previous work in Drosophila indicated that Merlin (Mer) and Expanded (Ex) are members of the Hippo pathway and act upstream of the Hippo kinase. In contrast to this model, it was suggested that Mer and Ex primarily regulate membrane dynamics and receptor trafficking, thereby affecting Hippo pathway activity only indirectly. Here, we examined the effects of Mer, Ex and the Hippo pathway on the size of the apical membrane and on apical-basal polarity complexes. We found that mer;ex double mutant imaginal disc cells have significantly increased levels of apical membrane determinants, such as Crb, aPKC and Patj. These phenotypes were shared with mutations in other Hippo pathway components and required Yki, indicating that Mer and Ex signal through the Hippo pathway. Interestingly, however, whereas Crb was required for the accumulation of other apical proteins and for the expansion of the apical domain observed in Hippo pathway mutants, its elimination did not significantly reverse the overgrowth phenotype of warts mutant cells. Therefore, Hippo signaling regulates cell polarity complexes in addition to and independently of its growth control function in imaginal disc cells.


Subject(s)
Cell Proliferation , Drosophila Proteins/metabolism , Drosophila/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neurofibromin 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis , Cadherins/metabolism , Cell Membrane/metabolism , Cell Polarity , Drosophila/genetics , Drosophila/growth & development , Drosophila/ultrastructure , Drosophila Proteins/genetics , Eye/metabolism , Eye Proteins/metabolism , Gene Expression Regulation, Developmental , Genotype , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mutation , Neurofibromin 2/genetics , Nuclear Proteins/metabolism , Phenotype , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Trans-Activators/metabolism , Tumor Suppressor Proteins/genetics , Wings, Animal/metabolism , YAP-Signaling Proteins
12.
J Biol Chem ; 284(29): 19445-51, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19473977

ABSTRACT

Mast cell degranulation is a highly regulated, calcium-dependent process, which is important for the acute release of inflammatory mediators during the course of many pathological conditions. We previously found that Synaptotagmin-2, a calcium sensor in neuronal exocytosis, was expressed in a mast cell line. We postulated that this protein may be involved in the control of mast cell-regulated exocytosis, and we generated Synaptotagmin-2 knock-out mice to test our hypothesis. Mast cells from this mutant animal conferred an abnormally decreased passive cutaneous anaphylaxis reaction on mast cell-deficient mice that correlated with a specific defect in mast cell-regulated exocytosis, leaving constitutive exocytosis and nonexocytic mast cell effector responses intact. This defect was not secondary to abnormalities in the development, maturation, migration, morphology, synthesis, and storage of inflammatory mediators, or intracellular calcium transients of the mast cells. Unlike neurons, the lack of Synaptotagmin-2 in mast cells was not associated with increased spontaneous exocytosis.


Subject(s)
Exocytosis , Mast Cells/metabolism , Synaptotagmin II/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Calcium/metabolism , Cell Count , Cell Differentiation , Cells, Cultured , Cytoplasmic Granules/metabolism , Female , Hypersensitivity/genetics , Hypersensitivity/metabolism , Immunoblotting , Immunohistochemistry , Male , Mast Cells/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Synaptotagmin II/genetics
13.
Proc Natl Acad Sci U S A ; 105(39): 14897-902, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18809931

ABSTRACT

The conserved Hippo tumor suppressor pathway is a key signaling pathway that controls organ size in Drosophila. To date a signal transduction cascade from the Cadherin Fat at the plasma membrane into the nucleus has been discovered. However, how the Hippo pathway is regulated by extracellular signals is poorly understood. Fat not only regulates growth but also planar cell polarity, for which it interacts with the Dachsous (Ds) Cadherin, and Four-jointed (Fj), a transmembrane kinase that modulates the interaction between Ds and Fat. Ds and Fj are expressed in gradients and manipulation of their expression causes abnormal growth. However, how Ds and Fj regulate growth and whether they act through the Hippo pathway is not known. Here, we report that Ds and Fj regulate Hippo signaling to control growth. Interestingly, we found that Ds/Fj regulate the Hippo pathway through a remarkable logic. Induction of Hippo target genes is not proportional to the amount of Ds or Fj presented to a cell, as would be expected if Ds and Fj acted as traditional ligands. Rather, Hippo target genes are up-regulated when neighboring cells express different amounts of Ds or Fj. Consistent with a model that differences in Ds/Fj levels between cells regulate the Hippo pathway, we found that artificial Ds/Fj boundaries induce extra cell proliferation, whereas flattening the endogenous Ds and Fj gradients results in growth defects. The Ds/Fj signaling system thus defines a cell-to-cell signaling mechanism that regulates the Hippo pathway, thereby contributing to the control of organ size.


Subject(s)
Cadherins/metabolism , Cell Proliferation , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Membrane/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Signal Transduction
14.
J Biol Chem ; 282(29): 20809-15, 2007 Jul 20.
Article in English | MEDLINE | ID: mdl-17456473

ABSTRACT

Although it has been shown that mast cell-deficient mice have diminished innate immune responses against bacteria, the most important immunoprotective factors secreted from activated mast cells have not been identified. Mouse mast cell protease 6 is a tetramer-forming tryptase. This serine protease is abundant in the secretory granules and is exocytosed upon bacterial challenge. Here we have described the generation of a mast cell protease-6-null mouse. Our discovery that mice lacking this neutral protease cannot efficiently clear Klebsiella pneumoniae from their peritoneal cavities reveals an essential role for this serine protease, and presumably its human ortholog, in innate immunity.


Subject(s)
Mast Cells/cytology , Tryptases/metabolism , Animals , Interleukin-6/metabolism , Klebsiella Infections/metabolism , Klebsiella pneumoniae/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Peptide Hydrolases/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism
15.
Rev. invest. clín ; 45(5): 463-7, sept.-oct. 1993. tab
Article in Spanish | LILACS | ID: lil-138967

ABSTRACT

La terapia anticoagulante oral es un recurso ampliamente utilizado en la actualidad en la profilaxis y tratamiento de gran cantidad de procedimientos. Para una mayor efectividad, se han propuesto diversos esquemas para su vigilancia pero ninguno ha probado ser útil. Se han sugerido que el cociente de normalización internacional (en inglés INR) es un método seguro de control durante el empleo de anticoagulantes orales (AO), por lo que el objetivo de este trabajo fue determinar su utilidad clínica. La determinación del tiempo de protrombina (TP) se hizo mediante las técnicas cromogénicas y coagulométricas, y el valor del INR se obtuvo mediante el TP del paciente sobre el del control, elevado a la potencia de la sensibilidad internacional de la tromboplastina empleada (ISI). En nuestra clínica, el objetivo fue mantener siempre al enfermo en un rango terapéutico de INR entre dos y tres. Prestamos nuestra experiencia con 77 pacientes y 810 determinaciones en 18 meses. Se observaron 26 casos de hemorragia y tres de trombosis. En ninguno de esto eventos, en INR se encontraba en el rango deseado. No se presentaron eventos letales. El análisis mostró discrepancias significativas entre los resultados de INR y el cociente del tiempo de protrombina (CTP), los cuales son potencialmente favorecedores del INR. Nuestra experiencia apoya el uso del INR en la vigilancia de la anticoagulación oral como un método más útil que el CTP solo


Subject(s)
Humans , Administration, Oral , Anticoagulants/administration & dosage , Anticoagulants/standards , Hematology , Hemorrhage/complications , Thrombosis/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...