Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Cell ; 162(3): 564-79, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26232226

ABSTRACT

During differentiation, human embryonic stem cells (hESCs) shut down the regulatory network conferring pluripotency in a process we designated pluripotent state dissolution (PSD). In a high-throughput RNAi screen using an inclusive set of differentiation conditions, we identify centrally important and context-dependent processes regulating PSD in hESCs, including histone acetylation, chromatin remodeling, RNA splicing, and signaling pathways. Strikingly, we detected a strong and specific enrichment of cell-cycle genes involved in DNA replication and G2 phase progression. Genetic and chemical perturbation studies demonstrate that the S and G2 phases attenuate PSD because they possess an intrinsic propensity toward the pluripotent state that is independent of G1 phase. Our data therefore functionally establish that pluripotency control is hardwired to the cell-cycle machinery, where S and G2 phase-specific pathways deterministically restrict PSD, whereas the absence of such pathways in G1 phase potentially permits the initiation of differentiation.


Subject(s)
Cell Cycle , Embryonic Stem Cells/cytology , Gene Regulatory Networks , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Differentiation , Cyclin B2/metabolism , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Humans , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism
2.
Cell ; 163(1): 230-45, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26365490

ABSTRACT

Embryonic stem cells (ESCs) repress the expression of exogenous proviruses and endogenous retroviruses (ERVs). Here, we systematically dissected the cellular factors involved in provirus repression in embryonic carcinomas (ECs) and ESCs by a genome-wide siRNA screen. Histone chaperones (Chaf1a/b), sumoylation factors (Sumo2/Ube2i/Sae1/Uba2/Senp6), and chromatin modifiers (Trim28/Eset/Atf7ip) are key determinants that establish provirus silencing. RNA-seq analysis uncovered the roles of Chaf1a/b and sumoylation modifiers in the repression of ERVs. ChIP-seq analysis demonstrates direct recruitment of Chaf1a and Sumo2 to ERVs. Chaf1a reinforces transcriptional repression via its interaction with members of the NuRD complex (Kdm1a, Hdac1/2) and Eset, while Sumo2 orchestrates the provirus repressive function of the canonical Zfp809/Trim28/Eset machinery by sumoylation of Trim28. Our study reports a genome-wide atlas of functional nodes that mediate proviral silencing in ESCs and illuminates the comprehensive, interconnected, and multi-layered genetic and epigenetic mechanisms by which ESCs repress retroviruses within the genome.


Subject(s)
Embryonic Stem Cells/virology , Endogenous Retroviruses/genetics , Proviruses/genetics , Animals , Chromatin Assembly Factor-1/genetics , Chromatin Assembly Factor-1/metabolism , Embryonal Carcinoma Stem Cells/virology , Epigenesis, Genetic , Mice , Small Ubiquitin-Related Modifier Proteins/metabolism
3.
PLoS Biol ; 22(1): e3002406, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38227562

ABSTRACT

Breast tumours are embedded in a collagen I-rich extracellular matrix (ECM) network, where nutrients are scarce due to limited blood flow and elevated tumour growth. Metabolic adaptation is required for cancer cells to endure these conditions. Here, we demonstrated that the presence of ECM supported the growth of invasive breast cancer cells, but not non-transformed mammary epithelial cells, under amino acid starvation, through a mechanism that required macropinocytosis-dependent ECM uptake. Importantly, we showed that this behaviour was acquired during carcinoma progression. ECM internalisation, followed by lysosomal degradation, contributed to the up-regulation of the intracellular levels of several amino acids, most notably tyrosine and phenylalanine. This resulted in elevated tyrosine catabolism on ECM under starvation, leading to increased fumarate levels, potentially feeding into the tricarboxylic acid (TCA) cycle. Interestingly, this pathway was required for ECM-dependent cell growth and invasive cell migration under amino acid starvation, as the knockdown of p-hydroxyphenylpyruvate hydroxylase-like protein (HPDL), the third enzyme of the pathway, opposed cell growth and motility on ECM in both 2D and 3D systems, without affecting cell proliferation on plastic. Finally, high HPDL expression correlated with poor prognosis in breast cancer patients. Collectively, our results highlight that the ECM in the tumour microenvironment (TME) represents an alternative source of nutrients to support cancer cell growth by regulating phenylalanine and tyrosine metabolism.


Subject(s)
Amino Acids , Breast Neoplasms , Humans , Female , Amino Acids/metabolism , Breast Neoplasms/metabolism , Extracellular Matrix/metabolism , Tyrosine/metabolism , Phenylalanine , Tumor Microenvironment
4.
PLoS Genet ; 13(4): e1006698, 2017 04.
Article in English | MEDLINE | ID: mdl-28403141

ABSTRACT

The cellular machinery required for the fusion of constitutive secretory vesicles with the plasma membrane in metazoans remains poorly defined. To address this problem we have developed a powerful, quantitative assay for measuring secretion and used it in combination with combinatorial gene depletion studies in Drosophila cells. This has allowed us to identify at least three SNARE complexes mediating Golgi to PM transport (STX1, SNAP24/29 and Syb; STX1, SNAP24/29 and YKT6; STX4, SNAP24 and Syb). RNAi mediated depletion of YKT6 and VAMP3 in mammalian cells also blocks constitutive secretion suggesting that YKT6 has an evolutionarily conserved role in this process. The unexpected role of YKT6 in plasma membrane fusion may in part explain why RNAi and gene disruption studies have failed to produce the expected phenotypes in higher eukaryotes.


Subject(s)
Cell Membrane/genetics , Drosophila Proteins/genetics , R-SNARE Proteins/genetics , SNARE Proteins/genetics , Vesicle-Associated Membrane Protein 3/genetics , Animals , Cell Membrane/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Heterozygote , Humans , Membrane Fusion/genetics , Protein Transport/genetics , R-SNARE Proteins/metabolism , RNA Interference , SNARE Proteins/metabolism , Shiga Toxin 1/genetics , Shiga Toxin 1/metabolism , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/genetics , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/metabolism , Vesicle-Associated Membrane Protein 3/metabolism
5.
Glycobiology ; 28(8): 580-591, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29757379

ABSTRACT

Quiescin sulfhydryl oxidase 1 (QSOX1) catalyzes the formation of disulfide bonds in protein substrates. Unlike other enzymes with related activities, which are commonly found in the endoplasmic reticulum, QSOX1 is localized to the Golgi apparatus or secreted. QSOX1 is upregulated in quiescent fibroblast cells and secreted into the extracellular environment, where it contributes to extracellular matrix assembly. QSOX1 is also upregulated in adenocarcinomas, though the extent to which it is secreted in this context is currently unknown. To achieve a better understanding of factors that dictate QSOX1 localization and function, we aimed to determine how post-translational modifications affect QSOX1 trafficking and activity. We found a highly conserved N-linked glycosylation site to be required for QSOX1 secretion from fibroblasts and other cell types. Notably, QSOX1 lacking a glycan at this site arrives at the Golgi, suggesting that it passes endoplasmic reticulum quality control but is not further transported to the cell surface for secretion. The QSOX1 transmembrane segment is dispensable for Golgi localization and secretion, as fully luminal and transmembrane variants displayed the same trafficking behavior. This study provides a key example of the effect of glycosylation on Golgi exit and contributes to an understanding of late secretory sorting and quality control.


Subject(s)
Fibroblasts/metabolism , Golgi Apparatus/metabolism , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Cell Line , Fibroblasts/cytology , Glycosylation , Golgi Apparatus/genetics , Humans , Oxidoreductases Acting on Sulfur Group Donors/genetics , Protein Transport/physiology
6.
Breast Cancer Res ; 20(1): 22, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29566768

ABSTRACT

BACKGROUND: Phosphatase and tensin homolog (PTEN) is one of the most frequently inactivated tumor suppressors in breast cancer. While PTEN itself is not considered a druggable target, PTEN synthetic-sick or synthetic-lethal (PTEN-SSL) genes are potential drug targets in PTEN-deficient breast cancers. Therefore, with the aim of identifying potential targets for precision breast cancer therapy, we sought to discover PTEN-SSL genes present in a broad spectrum of breast cancers. METHODS: To discover broad-spectrum PTEN-SSL genes in breast cancer, we used a multi-step approach that started with (1) a genome-wide short interfering RNA (siRNA) screen of ~ 21,000 genes in a pair of isogenic human mammary epithelial cell lines, followed by (2) a short hairpin RNA (shRNA) screen of ~ 1200 genes focused on hits from the first screen in a panel of 11 breast cancer cell lines; we then determined reproducibility of hits by (3) identification of overlaps between our results and reanalyzed data from 3 independent gene-essentiality screens, and finally, for selected candidate PTEN-SSL genes we (4) confirmed PTEN-SSL activity using either drug sensitivity experiments in a panel of 19 cell lines or mutual exclusivity analysis of publicly available pan-cancer somatic mutation data. RESULTS: The screens (steps 1 and 2) and the reproducibility analysis (step 3) identified six candidate broad-spectrum PTEN-SSL genes (PIK3CB, ADAMTS20, AP1M2, HMMR, STK11, and NUAK1). PIK3CB was previously identified as PTEN-SSL, while the other five genes represent novel PTEN-SSL candidates. Confirmation studies (step 4) provided additional evidence that NUAK1 and STK11 have PTEN-SSL patterns of activity. Consistent with PTEN-SSL status, inhibition of the NUAK1 protein kinase by the small molecule drug HTH-01-015 selectively impaired viability in multiple PTEN-deficient breast cancer cell lines, while mutations affecting STK11 and PTEN were largely mutually exclusive across large pan-cancer data sets. CONCLUSIONS: Six genes showed PTEN-SSL patterns of activity in a large proportion of PTEN-deficient breast cancer cell lines and are potential specific vulnerabilities in PTEN-deficient breast cancer. Furthermore, the NUAK1 PTEN-SSL vulnerability identified by RNA interference techniques can be recapitulated and exploited using the small molecule kinase inhibitor HTH-01-015. Thus, NUAK1 inhibition may be an effective strategy for precision treatment of PTEN-deficient breast tumors.


Subject(s)
Breast Neoplasms/genetics , PTEN Phosphohydrolase/genetics , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Repressor Proteins/genetics , AMP-Activated Protein Kinase Kinases , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/genetics , Genome, Human/genetics , Genomics/methods , Humans , Mammary Glands, Human/metabolism , Neoplasm Proteins/genetics , PTEN Phosphohydrolase/deficiency , RNA, Small Interfering/genetics , Synthetic Lethal Mutations/genetics
7.
EMBO J ; 33(22): 2659-75, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25190516

ABSTRACT

The small GTPase Arf1 plays critical roles in membrane traffic by initiating the recruitment of coat proteins and by modulating the activity of lipid-modifying enzymes. Here, we report an unexpected but evolutionarily conserved role for Arf1 and the ArfGEF GBF1 at mitochondria. Loss of function of ARF-1 or GBF-1 impaired mitochondrial morphology and activity in Caenorhabditis elegans. Similarly, mitochondrial defects were observed in mammalian and yeast cells. In Saccharomyces cerevisiae, aberrant clusters of the mitofusin Fzo1 accumulated in arf1-11 mutants and were resolved by overexpression of Cdc48, an AAA-ATPase involved in ER and mitochondria-associated degradation processes. Yeast Arf1 co-fractionated with ER and mitochondrial membranes and interacted genetically with the contact site component Gem1. Furthermore, similar mitochondrial abnormalities resulted from knockdown of either GBF-1 or contact site components in worms, suggesting that the role of Arf1 in mitochondrial functioning is linked to ER-mitochondrial contacts. Thus, Arf1 is involved in mitochondrial homeostasis and dynamics, independent of its role in vesicular traffic.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Mitochondria/enzymology , Saccharomyces cerevisiae/enzymology , ADP-Ribosylation Factor 1/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/genetics , Mitochondrial Membranes/enzymology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
8.
Biochim Biophys Acta ; 1860(8): 1623-39, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26968459

ABSTRACT

BACKGROUND: While the underlying causes of cancer are genetic modifications, changes in cellular states mediate cancer development. Tumor cells display markedly changed glycosylation states, of which the O-GalNAc glycans called the Tn and TF antigens are particularly common. How these antigens get over-expressed is not clear. The expression levels of glycosylation enzymes fail to explain it. SCOPE OF REVIEW: We describe the regulation of O-GalNAc glycosylation initiation and extension with emphasis on the initiating enzymes ppGalNAcTs (GALNTs), and introduce the GALA pathway--a change in GALNTs compartmentation within the secretory pathway that regulates Tn levels. We discuss the roles of O-GalNAc glycans and GALNTs in tumorigenic processes and finally consider diagnostic and therapeutic perspectives. MAJOR CONCLUSIONS: Contrary to a common hypothesis, short O-glycans in tumors are not the result of an incomplete glycosylation process but rather reveal the activation of regulatory pathways. Surprisingly, high Tn levels reveal a major shift in the O-glycoproteome rather than a shortening of O-glycans. These changes are driven by membrane trafficking events. GENERAL SIGNIFICANCE: Many attempts to use O-glycans for biomarker, antibody and therapeutic vaccine development have been made, but suffer limitations including poor sensitivity and/or specificity that may in part derive from lack of a mechanistic understanding. Deciphering how short O-GalNAc glycans are regulated would open new perspectives to exploit this biology for therapeutic usage. This article is part of a Special Issue entitled "Glycans in personalised medicine" Guest Editor: Professor Gordan Lauc.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate , Galactosamine , Glycoproteins , Neoplasm Proteins , Neoplasms , Oligosaccharides , Animals , Antigens, Tumor-Associated, Carbohydrate/genetics , Antigens, Tumor-Associated, Carbohydrate/metabolism , Cancer Vaccines/therapeutic use , Galactosamine/genetics , Galactosamine/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Glycosylation , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , Oligosaccharides/genetics , Oligosaccharides/metabolism
9.
J Virol ; 89(21): 11116-28, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26311884

ABSTRACT

UNLABELLED: Coronaviruses are RNA viruses with a large zoonotic reservoir and propensity for host switching, representing a real threat for public health, as evidenced by severe acute respiratory syndrome (SARS) and the emerging Middle East respiratory syndrome (MERS). Cellular factors required for their replication are poorly understood. Using genome-wide small interfering RNA (siRNA) screening, we identified 83 novel genes supporting infectious bronchitis virus (IBV) replication in human cells. Thirty of these hits can be placed in a network of interactions with viral proteins and are involved in RNA splicing, membrane trafficking, and ubiquitin conjugation. In addition, our screen reveals an unexpected role for valosin-containing protein (VCP/p97) in early steps of infection. Loss of VCP inhibits a previously uncharacterized degradation of the nucleocapsid N protein. This inhibition derives from virus accumulation in early endosomes, suggesting a role for VCP in the maturation of virus-loaded endosomes. The several host factors identified in this study may provide avenues for targeted therapeutics. IMPORTANCE: Coronaviruses are RNA viruses representing a real threat for public health, as evidenced by SARS and the emerging MERS. However, cellular factors required for their replication are poorly understood. Using genome-wide siRNA screening, we identified novel genes supporting infectious bronchitis virus (IBV) replication in human cells. The several host factors identified in this study may provide directions for future research on targeted therapeutics.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Coronavirus Infections/transmission , Endosomes/virology , Infectious bronchitis virus/physiology , Virus Release/physiology , Adenosine Triphosphatases/genetics , Animals , Blotting, Western , Cell Cycle Proteins/genetics , Cell Line , Chlorocebus aethiops , Fluorescent Antibody Technique , Genome-Wide Association Study , Humans , Molecular Sequence Annotation , RNA, Small Interfering/genetics , Valosin Containing Protein , Vero Cells
10.
Nature ; 468(7321): 316-20, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20953172

ABSTRACT

The derivation of human ES cells (hESCs) from human blastocysts represents one of the milestones in stem cell biology. The full potential of hESCs in research and clinical applications requires a detailed understanding of the genetic network that governs the unique properties of hESCs. Here, we report a genome-wide RNA interference screen to identify genes which regulate self-renewal and pluripotency properties in hESCs. Interestingly, functionally distinct complexes involved in transcriptional regulation and chromatin remodelling are among the factors identified in the screen. To understand the roles of these potential regulators of hESCs, we studied transcription factor PRDM14 to gain new insights into its functional roles in the regulation of pluripotency. We showed that PRDM14 regulates directly the expression of key pluripotency gene POU5F1 through its proximal enhancer. Genome-wide location profiling experiments revealed that PRDM14 colocalized extensively with other key transcription factors such as OCT4, NANOG and SOX2, indicating that PRDM14 is integrated into the core transcriptional regulatory network. More importantly, in a gain-of-function assay, we showed that PRDM14 is able to enhance the efficiency of reprogramming of human fibroblasts in conjunction with OCT4, SOX2 and KLF4. Altogether, our study uncovers a wealth of novel hESC regulators wherein PRDM14 exemplifies a key transcription factor required for the maintenance of hESC identity and the reacquisition of pluripotency in human somatic cells.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genome, Human/genetics , RNA Interference , Repressor Proteins/metabolism , Animals , Base Sequence , Cell Line , Cellular Reprogramming/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , RNA-Binding Proteins , Repressor Proteins/genetics , SOXB1 Transcription Factors/metabolism , Transcription Factors
11.
Proc Natl Acad Sci U S A ; 110(31): E2885-94, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23858473

ABSTRACT

Ecotropic viral integration site-1 (EVI1) is an oncogenic zinc finger transcription factor whose expression is frequently up-regulated in myeloid leukemia and epithelial cancers. To better understand the mechanisms underlying EVI1-associated disease, we sought to define the EVI1 interactome in cancer cells. By using stable isotope labeling by amino acids in cell culture (SILAC)-based quantitative proteomics, we could confidently assign 78 proteins as EVI1-interacting partners for FLAG-tagged EVI1. Subsequently, we showed that 22 of 27 tested interacting proteins could coimmunoprecipitate with endogenous EVI1 protein, which represented an 81.5% validation rate. Additionally, by comparing the stable isotope labeling by amino acids in cell culture (SILAC) data with high-throughput yeast two hybrid results, we showed that five of these proteins interacted directly with EVI1. Functional classification of EVI1-interacting proteins revealed associations with cellular transcription machinery; modulators of transcription; components of WNT, TGF-ß, and RAS pathways; and proteins regulating DNA repair, recombination, and mitosis. We also identified EVI1 phosphorylation sites by MS analysis and showed that Ser538 and Ser858 can be phosphorylated and dephosphorylated by two EVI1 interactome proteins, casein kinase II and protein phosphatase-1α. Finally, mutations that impair EVI1 phosphorylation at these sites reduced EVI1 DNA binding through its C-terminal zinc finger domain and induced cancer cell proliferation. Collectively, these combinatorial proteomic approaches demonstrate that EVI1 interacts with large and complex networks of proteins, which integrate signals from various different signaling pathways important for oncogenesis. Comprehensive analysis of the EVI1 interactome has thus provided an important resource for dissecting the molecular mechanisms of EVI1-associated disease.


Subject(s)
DNA-Binding Proteins/metabolism , Mitosis , Neoplasms/metabolism , Oncogene Proteins/metabolism , Recombinational DNA Repair , Transcription Factors/metabolism , Wnt Signaling Pathway , DNA-Binding Proteins/genetics , HeLa Cells , Humans , MDS1 and EVI1 Complex Locus Protein , Neoplasms/genetics , Neoplasms/pathology , Oncogene Proteins/genetics , Phosphorylation/genetics , Proto-Oncogenes/genetics , Transcription Factors/genetics
12.
Proc Natl Acad Sci U S A ; 110(34): E3152-61, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23912186

ABSTRACT

Invasiveness underlies cancer aggressiveness and is a hallmark of malignancy. Most malignant tumors have elevated levels of Tn, an O-GalNAc glycan. Mechanisms underlying Tn up-regulation and its effects remain unclear. Here we show that Golgi-to-endoplasmic reticulum relocation of polypeptide N-acetylgalactosamine-transferases (GalNAc-Ts) drives high Tn levels in cancer cell lines and in 70% of malignant breast tumors. This process stimulates cell adhesion to the extracellular matrix, as well as migration and invasiveness. The GalNAc-Ts lectin domain, mediating high-density glycosylation, is critical for these effects. Interfering with the lectin domain function inhibited carcinoma cell migration in vitro and metastatic potential in mice. We also show that stimulation of cell migration is dependent on Tn-bearing proteins present in lamellipodia of migrating cells. Our findings suggest that relocation of GalNAc-Ts to the endoplasmic reticulum frequently occurs upon cancerous transformation to enhance tumor cell migration and invasiveness through modification of cell surface proteins.


Subject(s)
Acetylgalactosamine/metabolism , Endoplasmic Reticulum/metabolism , Gene Expression Regulation, Neoplastic/physiology , Glycosyltransferases/metabolism , Neoplasm Invasiveness/physiopathology , Neoplasms/physiopathology , Animals , Antigens, Tumor-Associated, Carbohydrate/metabolism , Blotting, Western , Cell Line , Cell Movement/physiology , Cloning, Molecular , Fluorescent Antibody Technique , Glycosylation , Golgi Apparatus/metabolism , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Neoplasms/metabolism
13.
BMC Bioinformatics ; 14: 290, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-24088301

ABSTRACT

BACKGROUND: RNAi screening is a powerful method to study the genetics of intracellular processes in metazoans. Technically, the approach has been largely inspired by techniques and tools developed for compound screening, including those for data analysis. However, by contrast with compounds, RNAi inducing agents can be linked to a large body of gene-centric, publically available data. However, the currently available software applications to analyze RNAi screen data usually lack the ability to visualize associated gene information in an interactive fashion. RESULTS: Here, we present ScreenSifter, an open-source desktop application developed to facilitate storing, statistical analysis and rapid and intuitive biological data mining of RNAi screening datasets. The interface facilitates meta-data acquisition and long-term safe-storage, while the graphical user interface helps the definition of a hit list and the visualization of biological modules among the hits, through Gene Ontology and protein-protein interaction analyses. The application also allows the visualization of screen-to-screen comparisons. CONCLUSIONS: Our software package, ScreenSifter, can accelerate and facilitate screen data analysis and enable discovery by providing unique biological data visualization capabilities.


Subject(s)
Computational Biology/methods , Data Mining/methods , Databases, Nucleic Acid , RNA Interference , Software , Internet , User-Computer Interface
14.
Mol Syst Biol ; 8: 629, 2012.
Article in English | MEDLINE | ID: mdl-23212246

ABSTRACT

The Golgi apparatus has many important physiological functions, including sorting of secretory cargo and biosynthesis of complex glycans. These functions depend on the intricate and compartmentalized organization of the Golgi apparatus. To investigate the mechanisms that regulate Golgi architecture, we developed a quantitative morphological assay using three different Golgi compartment markers and quantitative image analysis, and performed a kinome- and phosphatome-wide RNAi screen in HeLa cells. Depletion of 159 signaling genes, nearly 20% of genes assayed, induced strong and varied perturbations in Golgi morphology. Using bioinformatics data, a large regulatory network could be constructed. Specific subnetworks are involved in phosphoinositides regulation, acto-myosin dynamics and mitogen activated protein kinase signaling. Most gene depletion also affected Golgi functions, in particular glycan biosynthesis, suggesting that signaling cascades can control glycosylation directly at the Golgi level. Our results provide a genetic overview of the signaling pathways that control the Golgi apparatus in human cells.


Subject(s)
Golgi Apparatus/metabolism , RNA Interference , Signal Transduction , Actomyosin/genetics , Actomyosin/metabolism , Cell Cycle , Computational Biology , Fluorescent Antibody Technique , Gene Expression Regulation , Glycosylation , HeLa Cells , Humans , Image Processing, Computer-Assisted , Lectins/chemistry , Lectins/genetics , Microscopy, Fluorescence , Phenotype , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Pilot Projects , Polysaccharides/biosynthesis , Protein Kinases/genetics , Protein Kinases/metabolism , Reproducibility of Results
15.
Inflamm Res ; 62(2): 133-43, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23052185

ABSTRACT

OBJECTIVE AND DESIGN: We investigated the role and regulation of zinc transporters in the activation of the inflammatory response in macrophages. Our exploratory computational study found that Zip14 (SLC39A14) was consistently up-regulated in activated macrophages; we therefore focused subsequently on that gene in the mechanistic study. MATERIAL: The expression and function of Zip14 was assessed in primary macrophages obtained by in-vitro differentiation of monocytes from human blood. METHODS: Primary macrophages were subjected to treatments with lipopolysaccharides, cytokines, chemicals, and pharmacological agents. SLC39A14 and inflammatory cytokine gene expressions were assessed by RT-qPCR. Zip14 siRNA knockdown was performed to explore the gene function. RESULTS: Lipopolysaccharide's inflammatory stimulus was a strong inducer of SLC39A14 mRNA expression in macrophages. This induction was dependent on calcium signaling, GC-rich DNA-binding, and NF-κB down-regulation. Impregnation of lipopolysaccharide-stimulated macrophages with the glucocorticoid dexamethasone further enhanced Zip14 expression while reducing interleukin-6 and tumor necrosis factor-α production. Zip14 knockdown in macrophages attenuated the expression and secretion of cytokines, indicating a buffering function for this zinc transporter. CONCLUSIONS: Collectively, our results identified the zinc transporter Zip14 as expressed downstream of lipopolysaccharide signals in macrophages. Zip14 induction had a regulatory function in cytokine production.


Subject(s)
Cation Transport Proteins/immunology , Macrophages/immunology , Animals , Calcium/metabolism , Cation Transport Proteins/genetics , Cell Differentiation , Dexamethasone/pharmacology , Gene Expression Regulation/drug effects , Humans , Immunosuppressive Agents/pharmacology , Inflammation/immunology , Interferon-gamma/pharmacology , Lipopolysaccharides , Lung/immunology , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/immunology , RNA, Messenger/metabolism , Spleen/immunology
16.
Matrix Biol ; 118: 110-128, 2023 04.
Article in English | MEDLINE | ID: mdl-36924903

ABSTRACT

Imbalance of collagen I expression results in severe pathologies. Apart from activation by the TGFß-receptor/Smad pathway, control of collagen I expression remains poorly understood. Here, we used human dermal fibroblasts expressing a mCherry fluorescent protein driven by endogenous COL1A1 promoter to functionally screen the kinome and phosphatome. We identify 8 negative regulators, revealing that collagen is under tonic repression. The cell surface receptor BDKRB2 represses collagen I and other pro-fibrotic genes. Interestingly, it also promotes other basal membrane ECM genes. This function is independent of the natural ligand, bradykinin, and of SMAD2/3 factors, instead requiring constant ERK1/2 repression. TGFß stimulation induces rapid BDKRB2 transcriptional downregulation. Human fibrotic fibroblasts have reduced BDKRB2 levels and enhancing its expression in keloid fibroblasts represses COL1A1. We propose that tonic signalling by BDKRB2 prevents collagen overproduction in skin fibroblasts.


Subject(s)
Collagen Type I , Skin , Humans , Collagen Type I/genetics , Collagen Type I/metabolism , Skin/metabolism , Collagen/metabolism , Transforming Growth Factor beta/metabolism , Cells, Cultured , Fibroblasts/metabolism , Receptors, Bradykinin/metabolism
18.
Nature ; 439(7076): 604-7, 2006 Feb 02.
Article in English | MEDLINE | ID: mdl-16452979

ABSTRACT

Yeast genetics and in vitro biochemical analysis have identified numerous genes involved in protein secretion. As compared with yeast, however, the metazoan secretory pathway is more complex and many mechanisms that regulate organization of the Golgi apparatus remain poorly characterized. We performed a genome-wide RNA-mediated interference screen in a Drosophila cell line to identify genes required for constitutive protein secretion. We then classified the genes on the basis of the effect of their depletion on organization of the Golgi membranes. Here we show that depletion of class A genes redistributes Golgi membranes into the endoplasmic reticulum, depletion of class B genes leads to Golgi fragmentation, depletion of class C genes leads to aggregation of Golgi membranes, and depletion of class D genes causes no obvious change. Of the 20 new gene products characterized so far, several localize to the Golgi membranes and the endoplasmic reticulum.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila/genetics , Drosophila/metabolism , Genomics , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Animals , Cell Line , Drosophila/cytology , Endoplasmic Reticulum/metabolism , Genes, Insect/genetics , Genes, Reporter , Horseradish Peroxidase/genetics , Horseradish Peroxidase/metabolism , Intracellular Membranes/metabolism , Protein Sorting Signals/genetics , Protein Sorting Signals/physiology , RNA Interference
20.
Nat Cell Biol ; 6(2): 106-12, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14743217

ABSTRACT

Protein kinase D (PKD) binds to diacylglycerol (DAG) in the trans-Golgi network (TGN) and is activated by trimeric G-protein subunits beta gamma. This complex then regulates the formation of transport carriers in the TGN that traffic to the plasma membrane in non-polarized cells. Here we report specificity of different PKD isoforms in regulating protein trafficking from the TGN. Kinase-inactive forms of PKD1, PKD2 and PKD3 localize to the TGN in polarized and non-polarized cells. PKD activity is required only for the transport of proteins containing basolateral sorting information, and seems to be cargo specific.


Subject(s)
Protein Kinase C/metabolism , Protein Kinases/metabolism , trans-Golgi Network/metabolism , Animals , Cell Line , Cell Polarity , Diglycerides/metabolism , Dogs , Epithelial Cells/cytology , Epithelial Cells/metabolism , HeLa Cells , Humans , Isoenzymes/metabolism , Protein Kinase C/genetics , Protein Kinase D2 , Protein Kinases/genetics , Protein Transport/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL