Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Exp Ther Med ; 18(4): 2401-2412, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31555351

ABSTRACT

Breast cancer is the most common cancer in women worldwide. Despite recent developments in breast cancer detection and treatment, 1.38 million women each year are still affected. Breast cancer heterogeneity at the population and single-cell level, complexity and developing different metastases are setting several challenges to develop efficient breast cancer therapies. RNA interference (RNAi) represents an opportunity to silence gene expression and inhibit specific pathways in cancer cells. In order to reap the full advantages of RNAi-based therapy, different pathways that sustain cancer cells growth have been targeted using specific siRNAs. The present study investigated the ability of a set of cytotoxic siRNAs to inhibit growth of breast cancer cells. These siRNAs are targeting eukaryotic elongation factor 2 (EEF2), polo-like kinase 1 (PLK1), G protein-coupled receptor kinase 4 (GRK4) and sphingosine kinase interacting protein (SKIP5). To facilitate their targeted delivery, the human epidermal growth factor receptor-3 (HER3)-specific aptamer A30 was used. The in vitro results described in this work indicate that combining the highly specific HER3 aptamer with cytotoxic siRNAs targeting (EEF2, PLK1, GRK4 and SKIP5) can inhibit its activity and ultimately suppress proliferation of HER3 positive breast cancer cells.

2.
Cell Rep ; 20(10): 2384-2395, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28877472

ABSTRACT

Activation of transcription factor NF-κB is a hallmark of infection with the gastric pathogen Helicobacter pylori, associated with inflammation and carcinogenesis. Genome-wide RNAi screening revealed numerous host factors involved in H. pylori-, but not IL-1ß- and TNF-α-dependent NF-κB regulation. Pathway analysis including CRISPR/Cas9-knockout and recombinant protein technology, immunofluorescence microscopy, immunoblotting, mass spectrometry, and mutant H. pylori strains identified the H. pylori metabolite D-glycero-ß-D-manno-heptose 1,7-bisphosphate (ßHBP) as a cagPAI type IV secretion system (T4SS)-dependent effector of NF-κB activation in infected cells. Upon pathogen-host cell contact, TIFA forms large complexes (TIFAsomes) including interacting host factors, such as TRAF2. NF-κB activation, TIFA phosphorylation, and TIFAsome formation depend on a functional ALPK1 kinase, highlighting the ALPK1-TIFA axis as a core innate immune pathway. ALPK1-TIFA-mediated NF-κB activation was independent of CagA protein translocation, indicating that CagA translocation and HBP delivery to host cells are distinct features of the pathogen's T4SS.


Subject(s)
Signal Transduction/physiology , Type IV Secretion Systems/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , CRISPR-Cas Systems/genetics , CRISPR-Cas Systems/physiology , Helicobacter Infections/immunology , Helicobacter Infections/metabolism , Helicobacter pylori/immunology , Helicobacter pylori/pathogenicity , Humans , Immunity, Innate/genetics , Immunity, Innate/physiology , Microscopy, Fluorescence , NF-kappa B/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , RNA Interference , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/metabolism , Type IV Secretion Systems/genetics
3.
PLoS Pathog ; 10(9): e1004351, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25187968

ABSTRACT

NOD1 is an intracellular pathogen recognition receptor that contributes to anti-bacterial innate immune responses, adaptive immunity and tissue homeostasis. NOD1-induced signaling relies on actin remodeling, however, the details of the connection of NOD1 and the actin cytoskeleton remained elusive. Here, we identified in a druggable-genome wide siRNA screen the cofilin phosphatase SSH1 as a specific and essential component of the NOD1 pathway. We show that depletion of SSH1 impaired pathogen induced NOD1 signaling evident from diminished NF-κB activation and cytokine release. Chemical inhibition of actin polymerization using cytochalasin D rescued the loss of SSH1. We further demonstrate that NOD1 directly interacted with SSH1 at F-actin rich sites. Finally, we show that enhanced cofilin activity is intimately linked to NOD1 signaling. Our data thus provide evidence that NOD1 requires the SSH1/cofilin network for signaling and to detect bacterial induced changes in actin dynamics leading to NF-κB activation and innate immune responses.


Subject(s)
Actins/metabolism , Cofilin 1/metabolism , Dysentery, Bacillary/microbiology , Nod1 Signaling Adaptor Protein/metabolism , Phosphoprotein Phosphatases/metabolism , Shigella flexneri/physiology , Actins/chemistry , Blotting, Western , Cells, Cultured , Cofilin 1/genetics , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation , HeLa Cells , High-Throughput Screening Assays , Humans , Immunoenzyme Techniques , Immunoprecipitation , Inflammation , Inflammation Mediators/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/antagonists & inhibitors , Nod1 Signaling Adaptor Protein/genetics , Phosphoprotein Phosphatases/genetics , Phosphorylation , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
4.
Mol Microbiol ; 94(1): 186-201, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25116793

ABSTRACT

Chlamydia trachomatis is an obligate intracellular pathogen responsible for a high burden of human disease. Here, a loss-of-function screen using a set of lentivirally transduced shRNAs identified 14 human host cell factors that modulate C. trachomatis infectivity. Notably, knockdown of dynamin, a host GTPase, decreased C. trachomatis infectivity. Dynamin functions in multiple cytoplasmic locations, including vesicle formation at the plasma membrane and the trans-Golgi network. However, its role in C. trachomatis infection remains unclear. Here we report that dynamin is essential for homotypic fusion of C. trachomatis inclusions but not for C. trachomatis internalization into the host cell. Further, dynamin activity is necessary for lipid transport into C. trachomatis inclusions and for normal re-differentiation from reticulate to elementary bodies. Fragmentation of the Golgi apparatus is proposed to be an important strategy used by C. trachomatis for efficient lipid acquisition and replication within the host. Here we show that a subset of C. trachomatis-infected cells displayed Golgi fragmentation, which was concurrent with increased mitotic accumulation. Golgi fragmentation was dispensable for dynamin-mediated lipid acquisition into C. trachomatis inclusions, irrespective of the cell cycle phase. Thus, our study reveals a critical role of dynamin in host-derived lipid acquisition for C. trachomatis development.


Subject(s)
Chlamydia Infections/enzymology , Chlamydia Infections/microbiology , Chlamydia trachomatis/growth & development , Chlamydia trachomatis/metabolism , Dynamin I/metabolism , Dynamins/metabolism , Lipid Metabolism , Chlamydia Infections/genetics , Chlamydia trachomatis/cytology , Chlamydia trachomatis/genetics , Dynamin I/genetics , Dynamin II , Dynamins/genetics , Golgi Apparatus/metabolism , Golgi Apparatus/microbiology , Humans
5.
Cell Microbiol ; 15(11): 1896-912, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23782461

ABSTRACT

Helicobacter pylori is a bacterial pathogen that colonizes the gastric niche of ∼ 50% of the human population worldwide and is known to cause peptic ulceration and gastric cancer. Pathology of infection strongly depends on a cag pathogenicity island (cagPAI)-encoded type IV secretion system (T4SS). Here, we aimed to identify as yet unknown bacterial factors involved in cagPAI effector function and performed a large-scale screen of an H. pylori transposon mutant library using activation of the pro-inflammatory transcription factor NF-κB in human gastric epithelial cells as a measure of T4SS function. Analysis of ∼ 3000 H. pylori mutants revealed three non-cagPAI genes that affected NF-κB nuclear translocation. Of these, the outer membrane protein HopQ from H. pylori strain P12 was essential for CagA translocation and for CagA-mediated host cell responses such as formation of the hummingbird phenotype and cell scattering. Besides that, deletion of hopQ reduced T4SS-dependent activation of NF-κB, induction of MAPK signalling and secretion of interleukin 8 (IL-8) in the host cells, but did not affect motility or the quantity of bacteria attached to host cells. Hence, we identified HopQ as a non-cagPAI-encoded cofactor of T4SS function.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Bacterial Secretion Systems , Helicobacter pylori/metabolism , Virulence Factors/metabolism , Bacterial Outer Membrane Proteins/genetics , Cell Line , DNA Transposable Elements , Epithelial Cells/microbiology , Gene Deletion , Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Humans , Interleukin-8/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Sequence Data , Mutagenesis, Insertional , NF-kappa B/metabolism , Sequence Analysis, DNA , Signal Transduction , Virulence Factors/genetics
6.
PLoS One ; 7(2): e31193, 2012.
Article in English | MEDLINE | ID: mdl-22384002

ABSTRACT

Since the worldwide increase in obesity represents a growing challenge for health care systems, new approaches are needed to effectively treat obesity and its associated diseases. One prerequisite for advances in this field is the identification of genes involved in adipogenesis and/or lipid storage. To provide a systematic analysis of genes that regulate adipose tissue biology and to establish a target-oriented compound screening, we performed a high throughput siRNA screen with primary (pre)adipocytes, using a druggable siRNA library targeting 7,784 human genes. The primary screen showed that 459 genes affected adipogenesis and/or lipid accumulation after knock-down. Out of these hits, 333 could be validated in a secondary screen using independent siRNAs and 110 genes were further regulated on the gene expression level during adipogenesis. Assuming that these genes are involved in neutral lipid storage and/or adipocyte differentiation, we performed InCell-Western analysis for the most striking hits to distinguish between the two phenotypes. Beside well known regulators of adipogenesis and neutral lipid storage (i.e. PPARγ, RXR, Perilipin A) the screening revealed a large number of genes which have not been previously described in the context of fatty tissue biology such as axonemal dyneins. Five out of ten axonemal dyneins were identified in our screen and quantitative RT-PCR-analysis revealed that these genes are expressed in preadipocytes and/or maturing adipocytes. Finally, to show that the genes identified in our screen are per se druggable we performed a proof of principle experiment using an antagonist for HTR2B. The results showed a very similar phenotype compared to knock-down experiments proofing the "druggability". Thus, we identified new adipogenesis-associated genes and those involved in neutral lipid storage. Moreover, by using a druggable siRNA library the screen data provides a very attractive starting point to identify anti-obesity compounds targeting the adipose tissue.


Subject(s)
Adipogenesis , Adipose Tissue/metabolism , Gene Expression Profiling , RNA, Small Interfering/metabolism , Adipocytes/cytology , Cell Differentiation , DNA/chemistry , Dyneins/metabolism , Gene Expression Regulation , Humans , Lipids/chemistry , Models, Biological , Obesity/genetics , Oligonucleotide Array Sequence Analysis , Phenotype , Quality Control , Serotonin 5-HT2 Receptor Antagonists/metabolism
7.
Cell Microbiol ; 13(10): 1573-85, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21824245

ABSTRACT

Chlamydiae are obligate intracellular Gram-negative bacteria that cause widespread diseases in humans. Due to the intimate association between bacterium and host, Chlamydia evolved various strategies to protect their host cell against death-inducing stimuli, allowing the bacterium to complete its development cycle. An RNA interference (RNAi)-based screen was used to identify host cell factors required for apoptosis resistance of human epithelial cells infected with Chlamydia trachomatis serovar L2. Among the 32 validated hits, the anti-apoptotic Bcl-2 family member Mcl-1 was identified as a target. Protein network analyses implicated the transcription factor hypoxia-induced factor 1 alpha (HIF-1α) to be central to the regulation of many of the identified targets. Further mechanistic investigations showed that HIF-1α was stabilized within the host cell cytoplasm during early infection time points, followed by its translocation to the nucleus and eventual transcriptional activation of Mcl-1. siRNA-mediated depletion of HIF-1α led to a drastic decrease in Mcl-1, rendering the cell sensitive to apoptosis induction. Taken together, our findings identify HIF-1α as responsible for upregulation of Mcl-1 and the maintenance of apoptosis resistance during Chlamydia infection.


Subject(s)
Apoptosis , Chlamydia trachomatis/pathogenicity , Host-Pathogen Interactions , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Cell Line , Epithelial Cells/microbiology , Gene Knockdown Techniques , Genetic Testing , Humans , Myeloid Cell Leukemia Sequence 1 Protein , RNA Interference , Systems Biology/methods
8.
Autophagy ; 7(8): 814-28, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21464618

ABSTRACT

Microtubule-associated protein 1 (MAP1) light chain 3 (LC3) has proven useful as autophagosomal marker in studies on the interaction between pathogens and the host autophagic machinery. However, the function of LC3 is known to extend above and beyond its role in autophagosome formation. We previously reported that intrinsic LC3 is associated with the intracellular Chlamydia trachomatis inclusion in human epithelial cells. Here we show that LC3, most likely the cytoplasmic nonlipidated form, interacts with the C. trachomatis inclusion as a microtubule-associated protein rather than an autophagosome-associated component. In contrast, N-terminally GFP-tagged LC3 exclusively targets autophagosomes rather than chlamydial inclusions. Immunofluorescence analysis revealed an association of LC3 and MAP1 subunits A and B with the inclusion as early as 18 h post infection. Inclusion-bound LC3 was connected with the microtubular network. Depolymerization of the microtubular architecture disrupted the association of LC3/MAP1s with the inclusion. Furthermore, siRNA-mediated silencing of the MAP1 and LC3 proteins revealed their essential function in the intracellular growth of C. trachomatis. Interestingly, defective autophagy remarkably enhanced chlamydial growth, suggesting a suppressive effect of the autophagic machinery on bacterial development. However, depletion of LC3 in autophagy-deficient cells noticeably reduced chlamydial propagation. Thus, our findings demonstrate a new function for LC3, distinct from autophagy, in intracellular bacterial pathogenesis.


Subject(s)
Autophagy , Chlamydia trachomatis/growth & development , Intracellular Space/microbiology , Microtubule-Associated Proteins/metabolism , Animals , Autophagy-Related Protein 5 , Chlamydia trachomatis/metabolism , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Inclusion Bodies/metabolism , Inclusion Bodies/ultrastructure , Intracellular Membranes/metabolism , Intracellular Membranes/ultrastructure , Lipids/chemistry , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Phagosomes/metabolism , Protein Biosynthesis , Protein Subunits/metabolism , Protein Transport , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/metabolism
9.
BMC Cell Biol ; 11: 21, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20233427

ABSTRACT

BACKGROUND: The nuclear factor-kappaB (NF-kappaB) family of transcription factors plays a role in a wide range of cellular processes including the immune response and cellular growth. In addition, deregulation of the NF-kappaB system has been associated with a number of disease states, including cancer. Therefore, insight into the regulation of NF-kappaB activation has crucial medical relevance, holding promise for novel drug target discovery. Transcription of NF-kappaB-induced genes is regulated by differential dynamics of single NF-kappaB subunits, but only a few methods are currently being applied to study dynamics. In particular, while oscillations of NF-kappaB activation have been observed in response to the cytokine tumor necrosis factor alpha (TNFalpha), little is known about the occurrence of oscillations in response to bacterial infections. RESULTS: To quantitatively assess NF-kappaB dynamics we generated human and murine monoclonal cell lines that stably express the NF-kappaB subunit p65 fused to GFP. Furthermore, a high-throughput assay based on automated microscopy coupled to image analysis to quantify p65-nuclear translocation was established. Using this assay, we demonstrate a stimulus- and cell line-specific temporal control of p65 translocation, revealing, for the first time, oscillations of p65 translocation in response to bacterial infection. Oscillations were detected at the single-cell level using real-time microscopy as well as at the population level using high-throughput image analysis. In addition, mathematical modeling of NF-kappaB dynamics during bacterial infections predicted masking of oscillations on the population level in asynchronous activations, which was experimentally confirmed. CONCLUSIONS: Taken together, this simple and cost effective assay constitutes an integrated approach to infer the dynamics of NF-kappaB kinetics in single cells and cell populations. Using a single system, novel factors modulating NF-kappaB can be identified and analyzed, providing new possibilities for a wide range of applications from therapeutic discovery and understanding of disease to host-pathogen interactions.


Subject(s)
NF-kappa B/metabolism , Animals , Cell Line , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori , High-Throughput Screening Assays , Humans , Mice , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
10.
Sci Signal ; 3(113): ra21, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20234004

ABSTRACT

Chlamydiae are obligate intracellular bacterial pathogens that have a major effect on human health. Because of their intimate association with their host, chlamydiae depend on various host cell functions for their survival. Here, we present an RNA-interference-based screen in human epithelial cells that identified 59 host factors that either positively or negatively influenced the replication of Chlamydia trachomatis (Ctr). Two factors, K-Ras and Raf-1, which are members of the canonical Ras-Raf-MEK (mitogen-activated or extracellular signal-regulated protein kinase kinase)-ERK (extracellular signal-regulated kinase) pathway, were identified as central components of signaling networks associated with hits from the screen. Depletion of Ras or Raf in HeLa cells increased pathogen growth. Mechanistic analyses revealed that ERK was activated independently of K-Ras and Raf-1. Infection with Ctr led to the Akt-dependent, increased phosphorylation (and inactivation) of Raf-1 at serine-259. Furthermore, phosphorylated Raf-1 relocalized from the cytoplasm to the intracellular bacterial inclusion in an Akt- and 14-3-3beta-dependent manner. Together, these findings not only show that Chlamydia regulates components of an important host cell signaling pathway, but also provide mechanistic insights into how this is achieved.


Subject(s)
Chlamydia Infections/metabolism , Chlamydia trachomatis , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Signal Transduction/physiology , Butadienes , Fluorescent Antibody Technique, Indirect , Gene Regulatory Networks/genetics , HeLa Cells , Humans , Nitriles , Phosphorylation , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , RNA Interference
11.
Nature ; 463(7282): 818-22, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-20081832

ABSTRACT

Influenza A virus, being responsible for seasonal epidemics and reoccurring pandemics, represents a worldwide threat to public health. High mutation rates facilitate the generation of viral escape mutants, rendering vaccines and drugs directed against virus-encoded targets potentially ineffective. In contrast, targeting host cell determinants temporarily dispensable for the host but crucial for virus replication could prevent viral escape. Here we report the discovery of 287 human host cell genes influencing influenza A virus replication in a genome-wide RNA interference (RNAi) screen. Using an independent assay we confirmed 168 hits (59%) inhibiting either the endemic H1N1 (119 hits) or the current pandemic swine-origin (121 hits) influenza A virus strains, with an overlap of 60%. Notably, a subset of these common hits was also essential for replication of a highly pathogenic avian H5N1 strain. In-depth analyses of several factors provided insights into their infection stage relevance. Notably, SON DNA binding protein (SON) was found to be important for normal trafficking of influenza virions to late endosomes early in infection. We also show that a small molecule inhibitor of CDC-like kinase 1 (CLK1) reduces influenza virus replication by more than two orders of magnitude, an effect connected with impaired splicing of the viral M2 messenger RNA. Furthermore, influenza-virus-infected p27(-/-) (cyclin-dependent kinase inhibitor 1B; Cdkn1b) mice accumulated significantly lower viral titres in the lung, providing in vivo evidence for the importance of this gene. Thus, our results highlight the potency of genome-wide RNAi screening for the dissection of virus-host interactions and the identification of drug targets for a broad range of influenza viruses.


Subject(s)
Biological Factors , Host-Pathogen Interactions , Influenza A Virus, H1N1 Subtype/growth & development , Influenza, Human/genetics , Influenza, Human/virology , RNA Interference , Virus Replication/physiology , Animals , Biological Factors/genetics , Biological Factors/metabolism , Cell Line , Cells, Cultured , Chick Embryo , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Epithelial Cells/virology , Genome, Human/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Influenza A Virus, H1N1 Subtype/classification , Lung/cytology , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics
12.
PLoS Pathog ; 5(3): e1000348, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19300516

ABSTRACT

Bcl-2 family proteins including the pro-apoptotic BH3-only proteins are central regulators of apoptotic cell death. Here we show by a focused siRNA miniscreen that the synergistic action of the BH3-only proteins Bim and Bmf is required for apoptosis induced by infection with Neisseria gonorrhoeae (Ngo). While Bim and Bmf were associated with the cytoskeleton of healthy cells, they both were released upon Ngo infection. Loss of Bim and Bmf from the cytoskeleton fraction required the activation of Jun-N-terminal kinase-1 (JNK-1), which in turn depended on Rac-1. Depletion and inhibition of Rac-1, JNK-1, Bim, or Bmf prevented the activation of Bak and Bax and the subsequent activation of caspases. Apoptosis could be reconstituted in Bim-depleted and Bmf-depleted cells by additional silencing of antiapoptotic Mcl-1 and Bcl-X(L), respectively. Our data indicate a synergistic role for both cytoskeletal-associated BH3-only proteins, Bim, and Bmf, in an apoptotic pathway leading to the clearance of Ngo-infected cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , Gonorrhea/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , Bcl-2-Like Protein 11 , Blotting, Western , Cell Adhesion/physiology , Cytoskeleton/ultrastructure , Flow Cytometry , Fluorescent Antibody Technique , Gonorrhea/pathology , HeLa Cells , Humans , Immunoprecipitation , Microscopy, Electron, Transmission , Mitogen-Activated Protein Kinase 8/metabolism , Neisseria gonorrhoeae/physiology , RNA, Small Interfering , rac GTP-Binding Proteins/metabolism
13.
Nature ; 457(7230): 731-5, 2009 Feb 05.
Article in English | MEDLINE | ID: mdl-19060882

ABSTRACT

The obligate intracellular bacterium Chlamydia trachomatis survives and replicates within a membrane-bound vacuole, termed the inclusion, which intercepts host exocytic pathways to obtain nutrients. Like many other intracellular pathogens, C. trachomatis has a marked requirement for host cell lipids, such as sphingolipids and cholesterol, produced in the endoplasmic reticulum and the Golgi apparatus. However, the mechanisms by which intracellular pathogens acquire host cell lipids are not well understood. In particular, no host cell protein responsible for transporting Golgi-derived lipids to the chlamydial inclusions has yet been identified. Here we show that Chlamydia infection in human epithelial cells induces Golgi fragmentation to generate Golgi ministacks surrounding the bacterial inclusion. Ministack formation is triggered by the proteolytic cleavage of the Golgi matrix protein golgin-84. Inhibition of golgin-84 truncation prevents Golgi fragmentation, causing a block in lipid acquisition and maturation of C. trachomatis. Golgi fragmentation by means of RNA-interference-mediated knockdown of distinct Golgi matrix proteins before infection enhances bacterial maturation. Our data functionally connect bacteria-induced golgin-84 cleavage, Golgi ministack formation, lipid acquisition and intracellular pathogen growth. We show that C. trachomatis subverts the structure and function of an entire host cell organelle for its own advantage.


Subject(s)
Chlamydia trachomatis/growth & development , Chlamydia trachomatis/pathogenicity , Golgi Apparatus/microbiology , Golgi Apparatus/pathology , Chlamydia muridarum/growth & development , Epithelial Cells/microbiology , Epithelial Cells/pathology , Gene Knockdown Techniques , Golgi Apparatus/metabolism , Golgi Matrix Proteins , HeLa Cells , Humans , Lipid Metabolism , Membrane Proteins/metabolism , Protein Processing, Post-Translational , RNA Interference , Vesicular Transport Proteins
14.
J Mol Biol ; 370(4): 620-32, 2007 Jul 20.
Article in English | MEDLINE | ID: mdl-17543336

ABSTRACT

p21-Activated kinases (PAKs) are serine/threonine kinases involved in multiple cellular functions including cytoskeleton regulation, proliferation and apoptosis. We performed a screen for proteins interacting with PAK-2, a ubiquitously expressed kinase involved in apoptotic signaling. Among the PAK-2 interacting proteins were different members of the Abl-binding protein family. Abl-binding proteins bound to a proline-rich region of PAK-2 located in the regulatory N terminus. Moreover, active PAK-2 phosphorylated Abl-binding proteins in vitro. Interestingly, we show that PAK-2 also interacted with c-Abl but via a different domain than with the Abl-binding proteins. PAK-2 and Abi-1 co-localized in the cytoplasm and to membrane dorsal ruffles induced by PDGF treatment. Expression of mutant PAK-2 deficient in binding to Abl-binding proteins or silencing of PAK-2 expression prevented the formation of membrane dorsal ruffles in response to PDGF. Our findings define a new class of PAK-interacting proteins, which play an important role in actin cytoskeletal reorganization.


Subject(s)
Cell Membrane/drug effects , Platelet-Derived Growth Factor/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Humans , Mice , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-abl/genetics , p21-Activated Kinases
15.
PLoS Pathog ; 2(10): e114, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17069460

ABSTRACT

Host cells infected with obligate intracellular bacteria Chlamydia trachomatis are profoundly resistant to diverse apoptotic stimuli. The molecular mechanisms underlying the block in apoptotic signaling of infected cells is not well understood. Here we investigated the molecular mechanism by which apoptosis induced via the tumor necrosis factor (TNF) receptor is prevented in infected epithelial cells. Infection with C. trachomatis leads to the up-regulation of cellular inhibitor of apoptosis (cIAP)-2, and interfering with cIAP-2 up-regulation sensitized infected cells for TNF-induced apoptosis. Interestingly, besides cIAP-2, cIAP-1 and X-linked IAP, although not differentially regulated by infection, are required to maintain apoptosis resistance in infected cells. We detected that IAPs are constitutively organized in heteromeric complexes and small interfering RNA-mediated silencing of one of these IAPs affects the stability of another IAP. In particular, the stability of cIAP-2 is modulated by the presence of X-linked IAP and their interaction is stabilized in infected cells. Our observations suggest that IAPs are functional and stable as heteromers, a thus far undiscovered mechanism of IAP regulation and its role in modulation of apoptosis.


Subject(s)
Apoptosis/physiology , Chlamydia trachomatis/growth & development , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Inhibitor of Apoptosis Proteins/metabolism , Apoptosis/genetics , Caspase 3/genetics , Caspase 3/metabolism , Cell Line , Chlamydia Infections/physiopathology , Chlamydia trachomatis/pathogenicity , Chlamydia trachomatis/physiology , Epithelial Cells/pathology , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/physiology , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins/genetics , Jurkat Cells , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/genetics , Signal Transduction/physiology , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism
16.
Cell Microbiol ; 8(10): 1643-55, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16984419

ABSTRACT

Infection with Chlamydophila pneumoniae (Cpn) renders host cells resistant to apoptosis induced by a variety of stimuli. While modulation of apoptosis has been extensively studied in cells acutely infected with Cpn, very little is known on how persistent chlamydial infection influences host cell survival. Here we show that epithelial cells persistently infected with Cpn resist apoptosis induced with TNFalpha or staurosporine. Cpn induced the activation of nuclear factor kappa B (NF-kappaB) and inhibition of NF-kappaB with a chemical inhibitor or by silencing expression of the p65 subunit sensitized infected cells for apoptosis induction by staurosporine or TNFalpha. Persistent infection resulted in the upregulation of the NF-kappaB regulated inhibitor of apoptosis protein 2 (cIAP-2) but not inhibitor of apoptosis protein 1 (cIAP-1). Interestingly, silencing of either cIAP-1 or cIAP-2 sensitized infected cells, suggesting that IAPs play an important role in the apoptosis resistance of persistently infected cells.


Subject(s)
Chlamydophila pneumoniae/physiology , Inhibitor of Apoptosis Proteins/physiology , NF-kappa B/physiology , Apoptosis , Caspase 3/metabolism , Caspase 8/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Epithelial Cells , Humans , Inhibitor of Apoptosis Proteins/genetics , Staurosporine/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation
17.
Cell Res ; 16(9): 759-70, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16940963

ABSTRACT

Members of the Rho family of GTPases are key regulators of the actin cytoskeleton. In particular, activated Rac1 stimulates membrane dorsal ruffle formation in response to platelet-derived growth factor (PDGF). Abl-interactor (Abi)-1 and betaPIX, a guanine nucleotide exchange factor for Rac1, localise at these Rac1-induced actin structures and play important roles in the induction of membrane dorsal ruffling in response to PDGF in fibroblasts. Here, we demonstrate a novel interaction between Abi-1 and betaPIX using the yeast two-hybrid system, in vitro pull-down assays, and in vivo co-immunoprecipitation experiments. In vitro, the C-terminal fragment of betaPIX interacted with Abi-1, while in vivo the N-terminal fragment of betaPIX interacted with Abi-1. The biological function of this interaction was investigated in mouse fibroblasts in response to PDGF stimulation. Abi-1 and betaPIX co-localised in the cytoplasm and to membrane dorsal ruffles after PDGF treatment. We show that the co-expression of Abi-1 and truncated forms of betaPIX in mouse fibroblasts blocked PDGF-induced membrane dorsal ruffles. Together, these results show that the interaction between Abi-1 and betaPIX is involved in the formation of growth factor-induced membrane dorsal ruffles.


Subject(s)
Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Platelet-Derived Growth Factor/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins/genetics , Cell Membrane Structures/metabolism , Cell Membrane Structures/ultrastructure , Cytoskeletal Proteins/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Guanine Nucleotide Exchange Factors/genetics , Mice , NIH 3T3 Cells , Platelet-Derived Growth Factor/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors , Two-Hybrid System Techniques
18.
Mol Biol Cell ; 17(6): 2811-23, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16554367

ABSTRACT

Microtubules are important for the turnover of podosomes, dynamic, actin-rich adhesions implicated in migration and invasion of monocytic cells. The molecular basis for this functional dependency, however, remained unclear. Here, we show that contact by microtubule plus ends critically influences the cellular fate of podosomes in primary human macrophages. In particular, we identify the kinesin KIF1C, a member of the Kinesin-3 family, as a plus-end-enriched motor that targets regions of podosome turnover. Expression of mutation constructs or small interfering RNA-/short hairpin RNA-based depletion of KIF1C resulted in decreased podosome dynamics and ultimately in podosome deficiency. Importantly, protein interaction studies showed that KIF1C binds to nonmuscle myosin IIA via its PTPD-binding domain, thus providing an interface between the actin and tubulin cytoskeletons, which may facilitate the subcellular targeting of podosomes by microtubules. This is the first report to implicate a kinesin in podosome regulation and also the first to describe a function for KIF1C in human cells.


Subject(s)
Cell Membrane Structures/physiology , Kinesins/physiology , Macrophages/physiology , Microtubules/physiology , Cell Differentiation , Cell Membrane Structures/ultrastructure , Cells, Cultured , Cloning, Molecular , Escherichia coli , Humans , Kinesins/deficiency , Kinesins/genetics , Macrophages/cytology , Microinjections , Mutagenesis , Plasmids , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
19.
Mol Cell Proteomics ; 4(1): 44-55, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15567892

ABSTRACT

Global approaches like proteome or transcriptome analyses have been performed extensively to identify candidate genes or proteins involved in biological and pathological processes. Here we describe the identification of proteins implicated in the regulation of apoptosis using proteome analysis and the functional validation of targets by RNA interference. A high-throughput platform for the validation of synthetic small interfering RNAs (siRNAs) by quantitative real-time PCR was established. Genes of the identified factors were silenced by automated siRNA transfection, and their role in apoptotic signaling was investigated. Using this strategy, nine new modulators of apoptosis were identified. A subsequent detailed study demonstrated that hepatoma-derived growth factor (HDGF) is required for TNFalpha-induced release of pro-apoptotic factors from mitochondria. The strategy described here may be used for hypothesis-free, global gene function analysis.


Subject(s)
Apoptosis/physiology , Proteome/analysis , RNA Interference , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/physiology , Jurkat Cells , Mitochondria/metabolism , Phenotype , Proteome/genetics , Proteome/physiology , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/metabolism
20.
EMBO J ; 21(8): 1916-29, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11953311

ABSTRACT

The human pathogen Neisseria gonorrhoeae induces host cell apoptosis during infection by delivering the outer membrane protein PorB to the host cell's mitochondria. PorB is a pore-forming beta-barrel protein sharing several features with the mitochondrial voltage-dependent anion channel (VDAC), which is involved in the regulation of apoptosis. Here we show that PorB of pathogenic Neisseria species produced by host cells is efficiently targeted to mitochondria. Imported PorB resides in the mitochondrial outer membrane and forms multimers with similar sizes as in the outer bacterial membrane. The mitochondria completely lose their membrane potential, a characteristic previously observed in cells infected with gonococci or treated with purified PorB. Closely related bacterial porins of non-pathogenic Neisseria mucosa or Escherichia coli remain in the cytosol. Import of PorB into mitochondria in vivo is independent of a linear signal sequence. Insertion of PorB into the mitochondrial outer membrane in vitro depends on the activity of Tom5, Tom20 and Tom40, but is independent of Tom70. Our data show that human VDAC and bacterial PorB are imported into mitochondria by a similar mechanism.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Ion Channels/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins , Neisseria gonorrhoeae/metabolism , Porins/metabolism , Receptors, Cell Surface , Receptors, Cytoplasmic and Nuclear , Bacterial Outer Membrane Proteins/genetics , Biological Transport , Carrier Proteins/genetics , Carrier Proteins/metabolism , Gene Expression , HeLa Cells , Humans , Intracellular Membranes/metabolism , Membrane Potentials , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Neisseria gonorrhoeae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction , Voltage-Dependent Anion Channels
SELECTION OF CITATIONS
SEARCH DETAIL
...