Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
PLoS Pathog ; 10(9): e1004351, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25187968

RESUMO

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.


Assuntos
Actinas/metabolismo , Cofilina 1/metabolismo , Disenteria Bacilar/microbiologia , Proteína Adaptadora de Sinalização NOD1/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Shigella flexneri/fisiologia , Actinas/química , Western Blotting , Células Cultivadas , Cofilina 1/genética , Ensaio de Imunoadsorção Enzimática , Técnica Indireta de Fluorescência para Anticorpo , Regulação da Expressão Gênica , Células HeLa , Ensaios de Triagem em Larga Escala , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Inflamação , Mediadores da Inflamação/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD1/antagonistas & inibidores , Proteína Adaptadora de Sinalização NOD1/genética , Fosfoproteínas Fosfatases/genética , Fosforilação , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
2.
Nature ; 463(7282): 818-22, 2010 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-20081832

RESUMO

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.


Assuntos
Fatores Biológicos , Interações Hospedeiro-Patógeno , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Influenza Humana/genética , Influenza Humana/virologia , Interferência de RNA , Replicação Viral/fisiologia , Animais , Fatores Biológicos/genética , Fatores Biológicos/metabolismo , Linhagem Celular , Células Cultivadas , Embrião de Galinha , Inibidor de Quinase Dependente de Ciclina p27/deficiência , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Células Epiteliais/virologia , Genoma Humano/genética , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Vírus da Influenza A Subtipo H1N1/classificação , Pulmão/citologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética
3.
Mol Microbiol ; 94(1): 186-201, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25116793

RESUMO

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.


Assuntos
Infecções por Chlamydia/enzimologia , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/crescimento & desenvolvimento , Chlamydia trachomatis/metabolismo , Dinamina I/metabolismo , Dinaminas/metabolismo , Metabolismo dos Lipídeos , Infecções por Chlamydia/genética , Chlamydia trachomatis/citologia , Chlamydia trachomatis/genética , Dinamina I/genética , Dinamina II , Dinaminas/genética , Complexo de Golgi/metabolismo , Complexo de Golgi/microbiologia , Humanos
4.
Nature ; 457(7230): 731-5, 2009 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19060882

RESUMO

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.


Assuntos
Chlamydia trachomatis/crescimento & desenvolvimento , Chlamydia trachomatis/patogenicidade , Complexo de Golgi/microbiologia , Complexo de Golgi/patologia , Chlamydia muridarum/crescimento & desenvolvimento , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Técnicas de Silenciamento de Genes , Complexo de Golgi/metabolismo , Proteínas da Matriz do Complexo de Golgi , Células HeLa , Humanos , Metabolismo dos Lipídeos , Proteínas de Membrana/metabolismo , Processamento de Proteína Pós-Traducional , Interferência de RNA , Proteínas de Transporte Vesicular
5.
Cell Microbiol ; 15(11): 1896-912, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23782461

RESUMO

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.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Sistemas de Secreção Bacterianos , Helicobacter pylori/metabolismo , Fatores de Virulência/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Linhagem Celular , Elementos de DNA Transponíveis , Células Epiteliais/microbiologia , Deleção de Genes , Helicobacter pylori/genética , Helicobacter pylori/patogenicidade , Humanos , Interleucina-8/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Dados de Sequência Molecular , Mutagênese Insercional , NF-kappa B/metabolismo , Análise de Sequência de DNA , Transdução de Sinais , Fatores de Virulência/genética
6.
Cell Microbiol ; 13(10): 1573-85, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21824245

RESUMO

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.


Assuntos
Apoptose , Chlamydia trachomatis/patogenicidade , Interações Hospedeiro-Patógeno , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Linhagem Celular , Células Epiteliais/microbiologia , Técnicas de Silenciamento de Genes , Testes Genéticos , Humanos , Proteína de Sequência 1 de Leucemia de Células Mieloides , Interferência de RNA , Biologia de Sistemas/métodos
7.
PLoS Pathog ; 5(3): e1000348, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19300516

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/fisiologia , Gonorreia/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/fisiologia , Proteína 11 Semelhante a Bcl-2 , Western Blotting , Adesão Celular/fisiologia , Citoesqueleto/ultraestrutura , Citometria de Fluxo , Imunofluorescência , Gonorreia/patologia , Células HeLa , Humanos , Imunoprecipitação , Microscopia Eletrônica de Transmissão , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Neisseria gonorrhoeae/fisiologia , RNA Interferente Pequeno , Proteínas rac de Ligação ao GTP/metabolismo
8.
BMC Cell Biol ; 11: 21, 2010 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-20233427

RESUMO

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.


Assuntos
NF-kappa B/metabolismo , Animais , Linhagem Celular , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter pylori , Ensaios de Triagem em Larga Escala , Humanos , Camundongos , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo
9.
Mol Biol Cell ; 17(6): 2811-23, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16554367

RESUMO

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.


Assuntos
Estruturas da Membrana Celular/fisiologia , Cinesinas/fisiologia , Macrófagos/fisiologia , Microtúbulos/fisiologia , Diferenciação Celular , Estruturas da Membrana Celular/ultraestrutura , Células Cultivadas , Clonagem Molecular , Escherichia coli , Humanos , Cinesinas/deficiência , Cinesinas/genética , Macrófagos/citologia , Microinjeções , Mutagênese , Plasmídeos , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
10.
Exp Ther Med ; 18(4): 2401-2412, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31555351

RESUMO

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.

11.
J Mol Biol ; 370(4): 620-32, 2007 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-17543336

RESUMO

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.


Assuntos
Membrana Celular/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular , Membrana Celular/metabolismo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Humanos , Camundongos , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-abl/genética , Quinases Ativadas por p21
12.
PLoS Pathog ; 2(10): e114, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17069460

RESUMO

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.


Assuntos
Apoptose/fisiologia , Chlamydia trachomatis/crescimento & desenvolvimento , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Proteínas Inibidoras de Apoptose/metabolismo , Apoptose/genética , Caspase 3/genética , Caspase 3/metabolismo , Linhagem Celular , Infecções por Chlamydia/fisiopatologia , Chlamydia trachomatis/patogenicidade , Chlamydia trachomatis/fisiologia , Células Epiteliais/patologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Regulação Enzimológica da Expressão Gênica/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Células HeLa , Humanos , Proteínas Inibidoras de Apoptose/genética , Células Jurkat , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores do Fator de Necrose Tumoral/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
13.
Cell Rep ; 20(10): 2384-2395, 2017 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-28877472

RESUMO

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.


Assuntos
Transdução de Sinais/fisiologia , Sistemas de Secreção Tipo IV/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/fisiologia , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/metabolismo , Helicobacter pylori/imunologia , Helicobacter pylori/patogenicidade , Humanos , Imunidade Inata/genética , Imunidade Inata/fisiologia , Microscopia de Fluorescência , NF-kappa B/metabolismo , Moléculas com Motivos Associados a Patógenos/metabolismo , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Interferência de RNA , Transdução de Sinais/genética , Fator de Necrose Tumoral alfa/metabolismo , Sistemas de Secreção Tipo IV/genética
14.
PLoS One ; 7(2): e31193, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22384002

RESUMO

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.


Assuntos
Adipogenia , Tecido Adiposo/metabolismo , Perfilação da Expressão Gênica , RNA Interferente Pequeno/metabolismo , Adipócitos/citologia , Diferenciação Celular , DNA/química , Dineínas/metabolismo , Regulação da Expressão Gênica , Humanos , Lipídeos/química , Modelos Biológicos , Obesidade/genética , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Controle de Qualidade , Antagonistas do Receptor 5-HT2 de Serotonina/metabolismo
15.
Autophagy ; 7(8): 814-28, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21464618

RESUMO

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.


Assuntos
Autofagia , Chlamydia trachomatis/crescimento & desenvolvimento , Espaço Intracelular/microbiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Animais , Proteína 5 Relacionada à Autofagia , Chlamydia trachomatis/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Corpos de Inclusão/metabolismo , Corpos de Inclusão/ultraestrutura , Membranas Intracelulares/metabolismo , Membranas Intracelulares/ultraestrutura , Lipídeos/química , Camundongos , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/metabolismo , Fagossomos/metabolismo , Biossíntese de Proteínas , Subunidades Proteicas/metabolismo , Transporte Proteico , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes de Fusão/metabolismo
16.
Sci Signal ; 3(113): ra21, 2010 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-20234004

RESUMO

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.


Assuntos
Infecções por Chlamydia/metabolismo , Chlamydia trachomatis , Células Epiteliais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Transdução de Sinais/fisiologia , Butadienos , Técnica Indireta de Fluorescência para Anticorpo , Redes Reguladoras de Genes/genética , Células HeLa , Humanos , Nitrilas , Fosforilação , Proteínas Proto-Oncogênicas c-raf/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Interferência de RNA
18.
Cell Res ; 16(9): 759-70, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16940963

RESUMO

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.


Assuntos
Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Ciclo Celular/genética , Estruturas da Membrana Celular/metabolismo , Estruturas da Membrana Celular/ultraestrutura , Proteínas do Citoesqueleto/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Camundongos , Células NIH 3T3 , Fator de Crescimento Derivado de Plaquetas/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho , Técnicas do Sistema de Duplo-Híbrido
19.
Cell Microbiol ; 8(10): 1643-55, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16984419

RESUMO

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.


Assuntos
Chlamydophila pneumoniae/fisiologia , Proteínas Inibidoras de Apoptose/fisiologia , NF-kappa B/fisiologia , Apoptose , Caspase 3/metabolismo , Caspase 8/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células Epiteliais , Humanos , Proteínas Inibidoras de Apoptose/genética , Estaurosporina/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima
20.
Mol Cell Proteomics ; 4(1): 44-55, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15567892

RESUMO

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.


Assuntos
Apoptose/fisiologia , Proteoma/análise , Interferência de RNA , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Células Jurkat , Mitocôndrias/metabolismo , Fenótipo , Proteoma/genética , Proteoma/fisiologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Transcrição Gênica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA