77 results on '"Agaisse H"'
Search Results
2. Host Pathways Important for Coxiella burnetii Infection Revealed by Genome-Wide RNA Interference Screening
- Author
-
Shuman, HA, McDonough, JA, Newton, HJ, Klum, S, Swiss, R, Agaisse, H, Roy, CR, Shuman, HA, McDonough, JA, Newton, HJ, Klum, S, Swiss, R, Agaisse, H, and Roy, CR
- Abstract
UNLABELLED: Coxiella burnetii is an intracellular pathogen that replicates within a lysosome-like vacuole. A Dot/Icm type IVB secretion system is used by C. burnetii to translocate effector proteins into the host cytosol that likely modulate host factor function. To identify host determinants required for C. burnetii intracellular growth, a genome-wide screen was performed using gene silencing by small interfering RNA (siRNA). Replication of C. burnetii was measured by immunofluorescence microscopy in siRNA-transfected HeLa cells. Newly identified host factors included components of the retromer complex, which mediates cargo cycling between the endocytic pathway and the Golgi apparatus. Reducing the levels of the retromer cargo-adapter VPS26-VPS29-VPS35 complex or retromer-associated sorting nexins abrogated C. burnetii replication. Several genes, when silenced, resulted in enlarged vacuoles or an increased number of vacuoles within C. burnetii-infected cells. Silencing of the STX17 gene encoding syntaxin-17 resulted in a striking defect in homotypic fusion of vacuoles containing C. burnetii, suggesting a role for syntaxin-17 in regulating this process. Lastly, silencing host genes needed for C. burnetii replication correlated with defects in the translocation of Dot/Icm effectors, whereas, silencing of genes that affected vacuole morphology, but did not impact replication, did not affect Dot/Icm translocation. These data demonstrate that C. burnetii vacuole maturation is important for creating a niche that permits Dot/Icm function. Thus, genome-wide screening has revealed host determinants involved in sequential events that occur during C. burnetii infection as defined by bacterial uptake, vacuole transport and acidification, activation of the Dot/Icm system, homotypic fusion of vacuoles, and intracellular replication. IMPORTANCE: Q fever in humans is caused by the bacterium Coxiella burnetii. Infection with C. burnetii is marked by its unique ability to replicate
- Published
- 2013
3. A recombinase-mediated system for elimination of antibiotic resistance gene markers from genetically engineered Bacillus thuringiensis strains
- Author
-
Sanchis, V, primary, Agaisse, H, additional, Chaufaux, J, additional, and Lereclus, D, additional
- Published
- 1997
- Full Text
- View/download PDF
4. Genetic analysis of cryIIIA gene expression in Bacillus thuringiensis
- Author
-
Salamitou, S., primary, Agaisse, H., additional, Bravo, A., additional, and Lereclus, D., additional
- Published
- 1996
- Full Text
- View/download PDF
5. Identification of a Bacillus thuringiensis gene that positively regulates transcription of the phosphatidylinositol-specific phospholipase C gene at the onset of the stationary phase
- Author
-
Lereclus, D, primary, Agaisse, H, additional, Gominet, M, additional, Salamitou, S, additional, and Sanchis, V, additional
- Published
- 1996
- Full Text
- View/download PDF
6. How does Bacillus thuringiensis produce so much insecticidal crystal protein?
- Author
-
Agaisse, H, primary and Lereclus, D, additional
- Published
- 1995
- Full Text
- View/download PDF
7. Expression in Bacillus subtilis of the Bacillus thuringiensis cryIIIA toxin gene is not dependent on a sporulation-specific sigma factor and is increased in a spo0A mutant
- Author
-
Agaisse, H, primary and Lereclus, D, additional
- Published
- 1994
- Full Text
- View/download PDF
8. A genetic system that reports transient activation of genes in Bacillus
- Author
-
Salamitou, S., Agaisse, H., and Lereclus, D.
- Published
- 1997
- Full Text
- View/download PDF
9. Bacillus popilliae cry18Aa operon is transcribed by sigmaE and sigmaK forms of RNA polymerase from a single initiation site.
- Author
-
Zhang, J, Schairer, H U, Schnetter, W, Lereclus, D, and Agaisse, H
- Abstract
Bacillus popilliae is an obligate pathogen for larvae of the insect family Scarabaeidae (Coleoptera). It forms parasporal crystals upon sporulation. The gene cry18Aa coding for the parasporal crystal protein and an upstream open reading frame, orf1, were previously isolated from B.popilliae. Here we report an analysis of cry18Aa transcription in Bacillus thuringiensis. The only transcriptional start site of cry18Aa was found 29 bp upstream of the open reading frame orf1, suggesting that orf1 and cry18Aa are transcribed as an operon. lacZ fusion to the cry18Aa promoter was used to follow the time-course of cry18Aa transcription in wild type B.thuringiensis and in various B.thuringiensis sporulation-deficient mutants (spo0A, sigE or sigK). In wild type B.thuringiensis, the cry18Aa promoter was activated 2 h after the end of exponential growth and the expression lasted to the late sporulation phase. The results of promoter activity in Spo+or Spo-backgrounds together with the results of primer extension experiments suggest that the transcription from this promoter can be driven by both sigmaE and sigmaK types of RNA polymerase at a single start site. The promoter region of cry18Aa operon fits the consensus sequences of both sigmaE and sigmaK dependent promoters of Bacillus.
- Published
- 1998
- Full Text
- View/download PDF
10. Construction of new insecticidal Bacillus thuringiensis recombinant strains by using the sporulation non-dependent expression system of cryIIIA and a site specific recombination vector
- Author
-
Sanchis, V., Agaisse, H., Chaufaux, J., and Lereclus, D.
- Published
- 1996
- Full Text
- View/download PDF
11. High-level transcription of the cryIIIA toxin gene of Bacillus thuringiensis depends on a second promoter located 600 bp upstream of the translational start site
- Author
-
Marlene Teixeira De-Souza, Agaisse, H., Lereclus, D., ProdInra, Migration, Unité de recherche Génétique Microbienne (UGM), and Institut National de la Recherche Agronomique (INRA)
- Subjects
[SDV] Life Sciences [q-bio] ,[SDV]Life Sciences [q-bio] - Abstract
20 ref.
12. Characterization of MxiE- and H-NS-Dependent Expression of ipaH7.8 , ospC1 , yccE , and yfdF in Shigella flexneri.
- Author
-
Hall CP, Jadeja NB, Sebeck N, and Agaisse H
- Subjects
- Shigella flexneri genetics, Shigella flexneri metabolism, Type III Secretion Systems genetics, Type III Secretion Systems metabolism, Transcription Factors genetics, Transcription Factors metabolism, DNA-Binding Proteins genetics, Bacterial Proteins genetics, Bacterial Proteins metabolism
- Abstract
Shigella flexneri uses a type 3 secretion system (T3SS) apparatus to inject virulence effector proteins into the host cell cytosol. Upon host cell contact, MxiE, an S. flexneri AraC-like transcriptional regulator, is required for the expression of a subset of T3SS effector genes encoded on the large virulence plasmid. Here, we defined the MxiE regulon using RNA-seq. We identified virulence plasmid- and chromosome-encoded genes that are activated in response to type 3 secretion in a MxiE-dependent manner. Bioinformatic analysis revealed that similar to previously known MxiE-dependent genes, chromosome-encoded genes yccE and yfdF contain a regulatory element known as the MxiE box, which is required for their MxiE-dependent expression. The significant AT enrichment of MxiE-dependent genes suggested the involvement of H-NS. Using a dominant negative H-NS system, we demonstrate that H-NS silences the expression of MxiE-dependent genes located on the virulence plasmid ( ipaH7 .8 and ospC1 ) and the chromosome ( yccE and yfdF ). Furthermore, we show that MxiE is no longer required for the expression of ipaH7.8 , ospC1, yccE , and yfdF when H-NS silencing is relieved. Finally, we show that the H-NS anti-silencer VirB is not required for ipaH7.8 and yccE expression upon MxiE/IpgC overexpression. Based on these genetic studies, we propose a model of MxiE-dependent gene regulation in which MxiE counteracts H-NS-mediated silencing. IMPORTANCE The expression of horizontally acquired genes, including virulence genes, is subject to complex regulation involving xenogeneic silencing proteins, and counter-silencing mechanisms. The pathogenic properties of Shigella flexneri mainly rely on the acquisition of the type 3 secretion system (T3SS) and cognate effector proteins, whose expression is repressed by the xenogeneic silencing protein H-NS. Based on previous studies, releasing H-NS-mediated silencing mainly relies on two mechanisms involving (i) a temperature shift leading to the release of H-NS at the virF promoter, and (ii) the virulence factor VirB, which dislodges H-NS upon binding to specific motifs upstream of virulence genes, including those encoding the T3SS. In this study, we provide genetic evidence supporting the notion that, in addition to VirB, the AraC family member MxiE also contributes to releasing H-NS-mediated silencing in S. flexneri.
- Published
- 2022
- Full Text
- View/download PDF
13. The type 3 secretion effector IpgD promotes S. flexneri dissemination.
- Author
-
Köseoğlu VK, Jones MK, and Agaisse H
- Subjects
- Actins metabolism, Animals, Bacterial Proteins genetics, Cell Surface Extensions microbiology, Colon microbiology, Disease Models, Animal, Dysentery, Bacillary microbiology, HT29 Cells, Host-Pathogen Interactions, Humans, Phosphoric Monoester Hydrolases genetics, Rabbits, Shigella flexneri genetics, Bacterial Proteins metabolism, Cell Surface Extensions metabolism, Phosphatidylinositol 4,5-Diphosphate metabolism, Phosphoric Monoester Hydrolases metabolism, Shigella flexneri metabolism, Type III Secretion Systems metabolism
- Abstract
The bacterial pathogen Shigella flexneri causes 270 million cases of bacillary dysentery worldwide every year, resulting in more than 200,000 deaths. S. flexneri pathogenic properties rely on its ability to invade epithelial cells and spread from cell to cell within the colonic epithelium. This dissemination process relies on actin-based motility in the cytosol of infected cells and formation of membrane protrusions that project into adjacent cells and resolve into double-membrane vacuoles (DMVs) from which the pathogen escapes, thereby achieving cell-to-cell spread. S. flexneri dissemination is facilitated by the type 3 secretion system (T3SS) through poorly understood mechanisms. Here, we show that the T3SS effector IpgD facilitates the resolution of membrane protrusions into DMVs during S. flexneri dissemination. The phosphatidylinositol 4-phosphatase activity of IpgD decreases PtdIns(4,5)P2 levels in membrane protrusions, thereby counteracting de novo cortical actin formation in protrusions, a process that restricts the resolution of protrusions into DMVs. Finally, using an infant rabbit model of shigellosis, we show that IpgD is required for efficient cell-to-cell spread in vivo and contributes to the severity of dysentery., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
14. Efferocytosis by Paneth cells within the intestine.
- Author
-
Shankman LS, Fleury ST, Evans WB, Penberthy KK, Arandjelovic S, Blumberg RS, Agaisse H, and Ravichandran KS
- Subjects
- Animals, Apoptosis, Humans, Inflammation, Intestines, Mice, Phagocytes, Paneth Cells, Phagocytosis
- Abstract
Apoptotic cells are quickly and efficiently engulfed and removed via the process of efferocytosis by either professional phagocytes, such as macrophages, or non-professional phagocytes, including epithelial cells.
1 , 2 In addition to debris removal, a key benefit of efferocytosis is that phagocytes engulfing apoptotic cells release anti-inflammatory mediators3 , 4 that help reduce local tissue inflammation;5 conversely, accumulation of uncleared apoptotic cells predisposes to a pro-inflammatory tissue milieu.6-8 Due to their high proliferative capacity, intestinal epithelial cells (iECs) are sensitive to inflammation, irradiation, and chemotherapy-induced DNA damage, leading to apoptosis. Mechanisms of iEC death in the context of irradiation has been studied,9 , 10 but phagocytosis of dying iECs is poorly understood. Here, we identify an unexpected efferocytic role for Paneth cells, which reside in intestinal crypts and are linked to innate immunity and maintenance of the stem cell niche in the crypt.11 , 12 Through a series of studies spanning in vitro efferocytosis, ex vivo intestinal organoids ("enteroids"), and in vivo Cre-mediated deletion of Paneth cells, we show that Paneth cells mediate apoptotic cell uptake of dying neighbors. The relevance of Paneth-cell-mediated efferocytosis was revealed ex vivo and in mice after low-dose cesium-137 (137 Cs) irradiation, mimicking radiation therapies given to cancer patients often causing significant apoptosis of iECs. These data advance a new concept that Paneth cells can act as phagocytes and identify another way in which Paneth cells contribute to the overall health of the intestine. These observations also have implications for individuals undergoing chemotherapy or chronic inflammatory bowel disease., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
15. Mechanisms of bacillary dysentery: lessons learnt from infant rabbits.
- Author
-
Yum LK and Agaisse H
- Subjects
- Animals, Animals, Newborn microbiology, Colon immunology, Diarrhea immunology, Diarrhea microbiology, Disease Models, Animal, Dysentery, Bacillary immunology, Guinea Pigs, Humans, Intestinal Mucosa immunology, Intestinal Mucosa microbiology, Intestinal Mucosa pathology, Rabbits, Colon microbiology, Colon pathology, Dysentery, Bacillary microbiology, Dysentery, Bacillary pathology, Shigella flexneri pathogenicity
- Abstract
The bacterial pathogen Shigella flexneri causes more than 250 million cases of bacillary dysentery (blood in stool) every year across the world. This human-specific disease is characterized by profuse bloody diarrhea, dramatic ulceration of the colonic epithelium and immune cell infiltration of the colonic tissue. A major challenge in understanding the mechanisms supporting bacillary dysentery is the reliance on animal models that do not fully recapitulate the symptoms observed in humans, including bloody diarrhea. Here we outline advances provided by a recently developed infant rabbit model of bacillary dysentery. The infant rabbit model defines bacillary dysentery as a critical combination of massive vascular lesions and dramatic epithelial fenestration due to intracellular infection and cell-to-cell spread, respectively. The infant rabbit model provides an unprecedented framework for understanding how the cell biology of Shigella flexneri infection relates to pathogenesis.
- Published
- 2020
- Full Text
- View/download PDF
16. WIPF2 promotes Shigella flexneri actin-based motility and cell-to-cell spread.
- Author
-
Michard C, Yum LK, and Agaisse H
- Subjects
- Cell Line, Tumor, Dysentery, Bacillary parasitology, Epithelial Cells metabolism, Epithelial Cells parasitology, HT29 Cells, HeLa Cells, Humans, Shigella flexneri physiology, Vacuoles metabolism, Actins metabolism, Cell Movement physiology, Dysentery, Bacillary metabolism, Microfilament Proteins metabolism, Shigella flexneri metabolism
- Abstract
Shigella flexneri is an intracellular pathogen that disseminates in colonic epithelial cells through actin-based motility and formation of membrane protrusions at cell-cell contacts, that project into adjacent cells and resolve into vacuoles, from which the pathogen escapes, thereby achieving cell-to-cell spread. Actin nucleation at the bacterial pole relies on the recruitment of the nucleation-promoting factor N-WASP, which activates the actin nucleator ARP2/3. In cells, the vast majority of N-WASP exists as a complex with WIP. The involvement of WIP in N-WASP-dependent actin-based motility of various pathogens, including vaccinia virus and S. flexneri, has been highly controversial. Here, we show that WIPF2 was the only WIP family member expressed in the human colonic epithelial cell line HT-29, and its depletion impaired S. flexneri dissemination. WIPF2 depletion increased the number of cytosolic bacteria lacking actin tails (non-motile) and decreased the velocity of motile bacteria. This correlated with a decrease in the recruitment of N-WASP to the bacterial pole, and among N-WASP-positive bacteria, a decrease in actin tail-positive bacteria, suggesting that WIPF2 is required for N-WASP recruitment and activation at the bacterial pole. In addition, when motile bacteria formed protrusions, WIPF2 depletion decreased the number of membrane protrusions that successfully resolved into vacuoles., (© 2019 John Wiley & Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
17. Evolutionary Perspectives on the Moonlighting Functions of Bacterial Factors That Support Actin-Based Motility.
- Author
-
Köseoğlu VK and Agaisse H
- Subjects
- Bacterial Adhesion, Bacterial Proteins metabolism, Host-Pathogen Interactions, Humans, Listeria pathogenicity, Listeria physiology, Shigella pathogenicity, Shigella physiology, Actins metabolism, Bacteria pathogenicity, Bacterial Physiological Phenomena, Biofilms growth & development
- Abstract
Various bacterial pathogens display an intracellular lifestyle and spread from cell to cell through actin-based motility (ABM). ABM requires actin polymerization at the bacterial pole and is mediated by the expression of bacterial factors that hijack the host cell actin nucleation machinery or exhibit intrinsic actin nucleation properties. It is increasingly recognized that bacterial ABM factors, in addition to having a crucial task during the intracellular phase of infection, display "moonlighting" adhesin functions, such as bacterial aggregation, biofilm formation, and host cell adhesion/invasion. Here, we review our current knowledge of ABM factors and their additional functions, and we propose that intracellular ABM functions have evolved from ancestral, extracellular adhesin functions., (Copyright © 2019 Köseoğlu and Agaisse.)
- Published
- 2019
- Full Text
- View/download PDF
18. The Autotransporter IcsA Promotes Shigella flexneri Biofilm Formation in the Presence of Bile Salts.
- Author
-
Köseoğlu VK, Hall CP, Rodríguez-López EM, and Agaisse H
- Subjects
- Bacterial Outer Membrane Proteins metabolism, Bacterial Proteins genetics, Bile Acids and Salts metabolism, DNA-Binding Proteins genetics, Shigella flexneri drug effects, Shigella flexneri metabolism, Transcription Factors genetics, Bacterial Proteins physiology, Bile Acids and Salts pharmacology, Biofilms drug effects, Biofilms growth & development, DNA-Binding Proteins physiology, Shigella flexneri physiology, Transcription Factors physiology, Type V Secretion Systems metabolism
- Abstract
Shigella flexneri is an intracellular bacterial pathogen that invades epithelial cells in the colonic mucosa, leading to bloody diarrhea. A previous study showed that S. flexneri forms biofilms in the presence of bile salts, through an unknown mechanism. Here, we investigated the potential role of adhesin-like autotransporter proteins in S. flexneri biofilm formation. BLAST search analysis revealed that the S. flexneri 2457T genome harbors 4 genes, S1242 , S1289 , S2406 , and icsA , encoding adhesin-like autotransporter proteins. Deletion mutants of the S1242 , S1289 , S2406 and icsA genes were generated and tested for biofilm formation. Phenotypic analysis of the mutant strains revealed that disruption of icsA abolished bile salt-induced biofilm formation. IcsA is an outer membrane protein secreted at the bacterial pole that is required for S. flexneri actin-based motility during intracellular infection. In extracellular biofilms, IcsA was also secreted at the bacterial pole and mediated bacterial cell-cell contacts and aggregative growth in the presence of bile salts. Dissecting individual roles of bile salts showed that deoxycholate is a robust biofilm inducer compared to cholate. The release of the extracellular domain of IcsA through IcsP-mediated cleavage was greater in the presence of cholate, suggesting that the robustness of biofilm formation was inversely correlated with IcsA processing. Accordingly, deletion of icsP abrogated IcsA processing in biofilms and enhanced biofilm formation., (Copyright © 2019 American Society for Microbiology.)
- Published
- 2019
- Full Text
- View/download PDF
19. Critical role of bacterial dissemination in an infant rabbit model of bacillary dysentery.
- Author
-
Yum LK, Byndloss MX, Feldman SH, and Agaisse H
- Subjects
- Animals, Animals, Newborn microbiology, Colon microbiology, Colon pathology, Diarrhea microbiology, Disease Models, Animal, Dysentery, Bacillary microbiology, Epithelial Cells microbiology, Female, Gastrointestinal Hemorrhage microbiology, HT29 Cells, Humans, Intestinal Mucosa microbiology, Intestinal Mucosa pathology, Pregnancy, Rabbits, Diarrhea pathology, Dysentery, Bacillary pathology, Gastrointestinal Hemorrhage pathology, Shigella flexneri pathogenicity, Type III Secretion Systems immunology
- Abstract
The bacterial pathogen Shigella flexneri causes 270 million cases of bacillary dysentery (blood in stool) worldwide every year, resulting in more than 200,000 deaths. A major challenge in combating bacillary dysentery is the lack of a small-animal model that recapitulates the symptoms observed in infected individuals, including bloody diarrhea. Here, we show that similar to humans, infant rabbits infected with S. flexneri experience severe inflammation, massive ulceration of the colonic mucosa, and bloody diarrhea. T3SS-dependent invasion of epithelial cells is necessary and sufficient for mediating immune cell infiltration and vascular lesions. However, massive ulceration of the colonic mucosa, bloody diarrhea, and dramatic weight loss are strictly contingent on the ability of the bacteria to spread from cell to cell. The infant rabbit model features bacterial dissemination as a critical determinant of S. flexneri pathogenesis and provides a unique small-animal model for research and development of therapeutic interventions.
- Published
- 2019
- Full Text
- View/download PDF
20. A murine model of diarrhea, growth impairment and metabolic disturbances with Shigella flexneri infection and the role of zinc deficiency.
- Author
-
Q S Medeiros PH, Ledwaba SE, Bolick DT, Giallourou N, Yum LK, Costa DVS, Oriá RB, Barry EM, Swann JR, Lima AÂM, Agaisse H, and Guerrant RL
- Subjects
- Animals, Anti-Bacterial Agents administration & dosage, Body Weight, Colon metabolism, Colon microbiology, Colon pathology, Diarrhea drug therapy, Diarrhea metabolism, Diarrhea microbiology, Dysentery, Bacillary drug therapy, Dysentery, Bacillary metabolism, Dysentery, Bacillary microbiology, Feces enzymology, Feces microbiology, Intestinal Mucosa microbiology, Intestinal Mucosa pathology, Metabolome, Mice, Inbred C57BL, Mutation, Shigella flexneri genetics, Shigella flexneri growth & development, Type III Secretion Systems genetics, Diarrhea pathology, Disease Models, Animal, Dysentery, Bacillary pathology, Shigella flexneri pathogenicity, Zinc deficiency
- Abstract
Shigella is one of the major enteric pathogens worldwide. We present a murine model of S. flexneri infection and investigate the role of zinc deficiency (ZD). C57BL/6 mice fed either standard chow (HC) or ZD diets were pretreated with an antibiotic cocktail and received S. flexneri strain 2457T orally. Antibiotic pre-treated ZD mice showed higher S. flexneri colonization than non-treated mice. ZD mice showed persistent colonization for at least 50 days post-infection (pi). S. flexneri -infected mice showed significant weight loss, diarrhea and increased levels of fecal MPO and LCN in both HC and ZD fed mice. S. flexneri preferentially colonized the colon, caused epithelial disruption and inflammatory cell infiltrate, and promoted cytokine production which correlated with weight loss and histopathological changes. Infection with S. flexneri ΔmxiG (critical for type 3 secretion system) did not cause weight loss or diarrhea, and had decreased stool shedding duration and tissue burden. Several biochemical changes related to energy, inflammation and gut-microbial metabolism were observed. Zinc supplementation increased weight gains and reduced intestinal inflammation and stool shedding in ZD infected mice. In conclusion, young antibiotic-treated mice provide a new model of oral S. flexneri infection, with ZD promoting prolonged infection outcomes.
- Published
- 2019
- Full Text
- View/download PDF
21. Principles of intracellular bacterial pathogen spread from cell to cell.
- Author
-
Weddle E and Agaisse H
- Subjects
- Animals, Humans, Bacteria pathogenicity, Bacterial Infections microbiology, Host-Pathogen Interactions physiology, Virulence physiology
- Abstract
Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2018
- Full Text
- View/download PDF
22. Spatial, Temporal, and Functional Assessment of LC3-Dependent Autophagy in Shigella flexneri Dissemination.
- Author
-
Weddle E and Agaisse H
- Subjects
- Bacterial Proteins genetics, Cell Line, Dysentery, Bacillary physiopathology, Humans, Intravital Microscopy, Microscopy, Confocal, Mutation, Protein Binding, Spatio-Temporal Analysis, Virulence Factors genetics, Virulence Factors metabolism, Autophagy, Bacterial Proteins metabolism, Epithelial Cells microbiology, Host-Pathogen Interactions, Microtubule-Associated Proteins metabolism, Shigella flexneri growth & development, Vacuoles microbiology
- Abstract
Shigella flexneri disseminates within the colonic mucosa by displaying actin-based motility in the cytosol of epithelial cells. Motile bacteria form membrane protrusions that project into adjacent cells and resolve into double-membrane vacuoles (DMVs) from which the bacteria escape, thereby achieving cell-to-cell spread. During dissemination, S. flexneri is targeted by LC3-dependent autophagy, a host cell defense mechanism against intracellular pathogens. The S. flexneri type III secretion system effector protein IcsB was initially proposed to counteract the recruitment of the LC3-dependent autophagy machinery to cytosolic bacteria. However, a recent study proposed that LC3 was recruited to bacteria in DMVs formed during cell-to-cell spread. To resolve the controversy and clarify the role of autophagy in S. flexneri infection, we tracked dissemination using live confocal microscopy and determined the spatial and temporal recruitment of LC3 to bacteria. This approach demonstrated that (i) LC3 was exclusively recruited to wild-type or icsB bacteria located in DMVs and (ii) the icsB mutant was defective in cell-to-cell spread due to failure to escape LC3-positive as well as LC3-negative DMVs. Failure of S. flexneri to escape DMVs correlated with late LC3 recruitment, suggesting that LC3 recruitment is the consequence and not the cause of DMV escape failure. Inhibition of autophagy had no positive impact on the spreading of wild-type or icsB mutant bacteria. Our results unambiguously demonstrate that IcsB is required for DMV escape during cell-to-cell spread, regardless of LC3 recruitment, and do not support the previously proposed notion that autophagy counters S. flexneri dissemination., (Copyright © 2018 American Society for Microbiology.)
- Published
- 2018
- Full Text
- View/download PDF
23. The Ethanolamine Permease EutH Promotes Vacuole Adaptation of Salmonella enterica and Listeria monocytogenes during Macrophage Infection.
- Author
-
Anderson CJ, Satkovich J, Köseoğlu VK, Agaisse H, and Kendall MM
- Subjects
- Bacterial Proteins physiology, Biological Transport physiology, Ethanolamines metabolism, Listeria monocytogenes pathogenicity, Macrophages pathology, Salmonella enterica pathogenicity, Vacuoles microbiology
- Abstract
Ethanolamine is a ubiquitous and essential molecule within a host. Significantly, bacterial pathogens exploit ethanolamine during infection to promote growth and regulate virulence. The ethanolamine permease EutH is dispensable for growth in vitro under standard conditions, whereas EutH is required for ethanolamine utilization at low pH. These findings suggested a model in which EutH facilitates diffusion of ethanolamine into the bacterial cell in acidic environments. To date, the ecological significance of this model has not been thoroughly investigated, and the importance of EutH to bacterial growth under physiologically relevant conditions is not known. During infection, immune cells internalize invading bacteria within an acidic, nutrient-depleted vacuole called the phagosome. Here, we investigated the hypothesis that EutH promotes bacterial survival following phagocytosis. Our findings indicate that EutH is important for survival and replication of the facultative intracellular pathogens Salmonella enterica serovar Typhimurium and Listeria monocytogenes during prolonged or transient exposure to the phagosome, respectively. Furthermore, in agreement with EutH being important in the acidic environment, neutralization of the vacuole abolished the requirement for EutH. Significantly, consistent with a role for EutH in promoting intramacrophage survival, EutH was not required during S Typhimurium local intestinal infection but specifically conferred an advantage upon dissemination to peripheral organs. These findings reveal a physiologically relevant and conserved role for EutH in spatiotemporal niche adaptation during infection., (Copyright © 2018 American Society for Microbiology.)
- Published
- 2018
- Full Text
- View/download PDF
24. Cardiolipin Synthesis and Outer Membrane Localization Are Required for Shigella flexneri Virulence.
- Author
-
Rossi RM, Yum L, Agaisse H, and Payne SM
- Subjects
- Actins metabolism, Bacterial Outer Membrane Proteins chemistry, Bacterial Proteins genetics, Bacterial Proteins metabolism, Cardiolipins metabolism, Cytoplasm metabolism, Cytoplasm microbiology, DNA-Binding Proteins metabolism, Humans, Membrane Proteins metabolism, Mutation, Shigella flexneri genetics, Transferases (Other Substituted Phosphate Groups) metabolism, Virulence, Bacterial Outer Membrane Proteins metabolism, Cardiolipins biosynthesis, Shigella flexneri metabolism, Shigella flexneri pathogenicity
- Abstract
Cardiolipin, an anionic phospholipid that resides at the poles of the inner and outer membranes, is synthesized primarily by the putative cardiolipin synthase ClsA in Shigella flexneri An S. flexneri clsA mutant had no cardiolipin detected within its membrane, grew normally in vitro , and invaded cultured epithelial cells, but it failed to form plaques in epithelial cell monolayers, indicating that cardiolipin is required for virulence. The clsA mutant was initially motile within the host cell cytoplasm but formed filaments and lost motility during replication and failed to spread efficiently to neighboring cells. Mutation of pbgA , which encodes the transporter for cardiolipin from the inner membrane to the outer membrane, also resulted in loss of plaque formation. The S. flexneri pbgA mutant had normal levels of cardiolipin in the inner membrane, but no cardiolipin was detected in the outer membrane. The pbgA mutant invaded and replicated normally within cultured epithelial cells but failed to localize the actin polymerization protein IcsA properly on the bacterial surface and was unable to spread to neighboring cells. The clsA mutant, but not the pbgA mutant, had increased phosphatidylglycerol in the outer membrane. This appeared to compensate partially for the loss of cardiolipin in the outer membrane, allowing some IcsA localization in the outer membrane of the clsA mutant. We propose a dual function for cardiolipin in S. flexneri pathogenesis. In the inner membrane, cardiolipin is essential for proper cell division during intracellular growth. In the outer membrane, cardiolipin facilitates proper presentation of IcsA on the bacterial surface. IMPORTANCE The human pathogen Shigella flexneri causes bacterial dysentery by invading colonic epithelial cells, rapidly multiplying within their cytoplasm, and then spreading intercellularly to neighboring cells. Worldwide, Shigella spp. infect hundreds of millions of people annually, with fatality rates up to 15%. Antibiotic treatment of Shigella infections is compromised by increasing antibiotic resistance, and there is no approved vaccine to prevent future infections. This has created a growing need to understand Shigella pathogenesis and identify new targets for antimicrobial therapeutics. Here we show a previously unknown role of phospholipids in S. flexneri pathogenesis. We demonstrate that cardiolipin is required in the outer membrane for proper surface localization of IcsA and in the inner membrane for cell division during growth in the host cell cytoplasm., (Copyright © 2017 Rossi et al.)
- Published
- 2017
- Full Text
- View/download PDF
25. The Metalloprotease Mpl Supports Listeria monocytogenes Dissemination through Resolution of Membrane Protrusions into Vacuoles.
- Author
-
Alvarez DE and Agaisse H
- Subjects
- Amino Acid Sequence, Bacterial Proteins immunology, Binding Sites, Cell Membrane immunology, Cell Membrane microbiology, Cell Membrane ultrastructure, Cytoplasm immunology, Cytoplasm microbiology, Cytoplasm ultrastructure, Gene Deletion, HeLa Cells, Humans, Listeria monocytogenes growth & development, Listeria monocytogenes immunology, Membrane Proteins immunology, Metalloendopeptidases immunology, Operon, Protein Binding, Type C Phospholipases immunology, Vacuoles immunology, Vacuoles ultrastructure, Bacterial Proteins genetics, Gene Expression Regulation, Bacterial, Host-Pathogen Interactions, Listeria monocytogenes genetics, Membrane Proteins genetics, Metalloendopeptidases genetics, Type C Phospholipases genetics, Vacuoles microbiology
- Abstract
Listeria monocytogenes is an intracellular pathogen that disseminates within the intestinal epithelium through acquisition of actin-based motility and formation of plasma membrane protrusions that project into adjacent cells. The resolution of membrane protrusions into vacuoles from which the pathogen escapes results in bacterial spread from cell to cell. This dissemination process relies on the mlp-actA-plcB operon, which encodes ActA, a bacterial nucleation-promoting factor that mediates actin-based motility, and PlcB, a phospholipase that mediates vacuole escape. Here we investigated the role of the metalloprotease Mpl in the dissemination process. In agreement with previous findings showing that Mpl is required for PlcB activation, infection of epithelial cells with the ΔplcB or Δmpl strains resulted in the formation of small infection foci. As expected, the ΔplcB strain displayed a strong defect in vacuole escape. However, the Δmpl strain showed an unexpected defect in the resolution of protrusions into vacuoles, in addition to the expected but mild defect in vacuole escape. The Δmpl strain displayed increased levels of ActA on the bacterial surface in protrusions. We mapped an Mpl-dependent processing site in ActA between amino acid residues 207 to 238. Similar to the Δmpl strain, the ΔactA207-238 strain displayed increased levels of ActA on the bacterial surface in protrusions. Although the ΔactA207-238 strain displayed wild-type actin-based motility, it formed small infection foci and failed to resolve protrusions into vacuoles. We propose that, in addition to its role in PlcB processing and vacuole escape, the metalloprotease Mpl is required for ActA processing and protrusion resolution., (Copyright © 2016, American Society for Microbiology. All Rights Reserved.)
- Published
- 2016
- Full Text
- View/download PDF
26. Molecular and Cellular Mechanisms of Shigella flexneri Dissemination.
- Author
-
Agaisse H
- Subjects
- Actin-Related Protein 2-3 Complex metabolism, Antigens, Bacterial metabolism, Bacterial Proteins biosynthesis, Bacterial Proteins metabolism, Biological Transport, Cell Surface Extensions, DNA-Binding Proteins biosynthesis, Dysentery, Bacillary pathology, Epithelial Cells microbiology, Humans, Intestinal Mucosa microbiology, Transcription Factors biosynthesis, Virulence Factors metabolism, Wiskott-Aldrich Syndrome Protein, Neuronal metabolism, Actins metabolism, Dysentery, Bacillary microbiology, Intestinal Mucosa pathology, Shigella flexneri pathogenicity, Type III Secretion Systems metabolism, Vacuoles microbiology
- Abstract
The intracellular pathogen Shigella flexneri is the causative agent of bacillary dysentery in humans. The disease is characterized by bacterial invasion of intestinal cells, dissemination within the colonic epithelium through direct spread from cell to cell, and massive inflammation of the intestinal mucosa. Here, we review the mechanisms supporting S. flexneri dissemination. The dissemination process primarily relies on actin assembly at the bacterial pole, which propels the pathogen throughout the cytosol of primary infected cells. Polar actin assembly is supported by polar expression of the bacterial autotransporter family member IcsA, which recruits the N-WASP/ARP2/3 actin assembly machinery. As motile bacteria encounter cell-cell contacts, they form plasma membrane protrusions that project into adjacent cells. In addition to the ARP2/3-dependent actin assembly machinery, protrusion formation relies on formins and myosins. The resolution of protrusions into vacuoles occurs through the collapse of the protrusion neck, leading to the formation of an intermediate membrane-bound compartment termed vacuole-like protrusions (VLPs). VLP formation requires tyrosine kinase and phosphoinositide signaling in protrusions, which relies on the integrity of the bacterial type 3 secretion system (T3SS). The T3SS is also required for escaping double membrane vacuoles through the activity of the T3SS translocases IpaB and IpaC, and the effector proteins VirA and IcsB. Numerous factors supporting envelope biogenesis contribute to IcsA exposure and maintenance at the bacterial pole, including LPS synthesis, membrane proteases, and periplasmic chaperones. Although less characterized, the assembly and function of the T3SS in the context of bacterial dissemination also relies on factors supporting envelope biogenesis. Finally, the dissemination process requires the adaptation of the pathogen to various cellular compartments through transcriptional and post-transcriptional mechanisms.
- Published
- 2016
- Full Text
- View/download PDF
27. Integration of linear and dendritic actin nucleation in Nck-induced actin comets.
- Author
-
Borinskaya S, Velle KB, Campellone KG, Talman A, Alvarez D, Agaisse H, Wu YI, Loew LM, and Mayer BJ
- Subjects
- Animals, Dendrites metabolism, Fetal Proteins metabolism, Formins, HeLa Cells, Humans, Mice, Microfilament Proteins metabolism, NIH 3T3 Cells, Nuclear Proteins metabolism, Structure-Activity Relationship, Wiskott-Aldrich Syndrome Protein, Neuronal metabolism, src Homology Domains, Actin Cytoskeleton metabolism, Actins metabolism, Adaptor Proteins, Signal Transducing metabolism, Oncogene Proteins metabolism
- Abstract
The Nck adaptor protein recruits cytosolic effectors such as N-WASP that induce localized actin polymerization. Experimental aggregation of Nck SH3 domains at the membrane induces actin comet tails--dynamic, elongated filamentous actin structures similar to those that drive the movement of microbial pathogens such as vaccinia virus. Here we show that experimental manipulation of the balance between unbranched/branched nucleation altered the morphology and dynamics of Nck-induced actin comets. Inhibition of linear, formin-based nucleation with the small-molecule inhibitor SMIFH2 or overexpression of the formin FH1 domain resulted in formation of predominantly circular-shaped actin structures with low mobility (actin blobs). These results indicate that formin-based linear actin polymerization is critical for the formation and maintenance of Nck-dependent actin comet tails. Consistent with this, aggregation of an exclusively branched nucleation-promoting factor (the VCA domain of N-WASP), with density and turnover similar to those of N-WASP in Nck comets, did not reconstitute dynamic, elongated actin comets. Furthermore, enhancement of branched Arp2/3-mediated nucleation by N-WASP overexpression caused loss of the typical actin comet tail shape induced by Nck aggregation. Thus the ratio of linear to dendritic nucleation activity may serve to distinguish the properties of actin structures induced by various viral and bacterial pathogens., (© 2016 Borinskaya et al. This article is distributed by The American Society for Cell Biology under license from the author(s). Two months after publication it is available to the public under an Attribution–Noncommercial–Share Alike 3.0 Unported Creative Commons License (http://creativecommons.org/licenses/by-nc-sa/3.0).)
- Published
- 2016
- Full Text
- View/download PDF
28. Bacterial spread from cell to cell: beyond actin-based motility.
- Author
-
Kuehl CJ, Dragoi AM, Talman A, and Agaisse H
- Subjects
- Animals, Cell Surface Extensions physiology, Cell Surface Extensions ultrastructure, Host-Pathogen Interactions, Humans, Listeria monocytogenes pathogenicity, Macrophages microbiology, Movement, Shigella flexneri pathogenicity, Vacuoles microbiology, Actins metabolism, Bacterial Proteins metabolism, Cytosol microbiology, Listeria monocytogenes physiology, Shigella flexneri physiology
- Abstract
Several intracellular pathogens display the ability to propagate within host tissues by displaying actin-based motility in the cytosol of infected cells. As motile bacteria reach cell-cell contacts they form plasma membrane protrusions that project into adjacent cells and resolve into vacuoles from which the pathogen escapes, thereby achieving spread from cell to cell. Seminal studies have defined the bacterial and cellular factors that support actin-based motility. By contrast, the mechanisms supporting the formation of protrusions and their resolution into vacuoles have remained elusive. Here, we review recent advances in the field showing that Listeria monocytogenes and Shigella flexneri have evolved pathogen-specific mechanisms of bacterial spread from cell to cell., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
29. Shigella flexneri serotype 3a: the rise of a superbug.
- Author
-
Agaisse H
- Subjects
- Female, Humans, Male, Azithromycin therapeutic use, Drug Resistance, Bacterial drug effects, Dysentery, Bacillary drug therapy, Dysentery, Bacillary epidemiology, Sexually Transmitted Diseases drug therapy, Sexually Transmitted Diseases epidemiology
- Published
- 2015
- Full Text
- View/download PDF
30. STIM1 Is a Novel Component of ER-Chlamydia trachomatis Inclusion Membrane Contact Sites.
- Author
-
Agaisse H and Derré I
- Subjects
- Calcium metabolism, DNA-Binding Proteins metabolism, HeLa Cells, Humans, Inclusion Bodies metabolism, Membrane Glycoproteins metabolism, Protein Serine-Threonine Kinases metabolism, Stromal Interaction Molecule 1, Bacterial Proteins metabolism, Chlamydia trachomatis physiology, Endoplasmic Reticulum metabolism, Membrane Proteins metabolism, Neoplasm Proteins metabolism
- Abstract
Productive developmental cycle of the obligate intracellular bacterial pathogen Chlamydia trachomatis depends on the interaction of the replicative vacuole, named the inclusion, with cellular organelles. We have recently reported the formation of ER-Inclusion membrane contact sites (MCSs), where the endoplasmic reticulum (ER) is in apposition to the inclusion membrane. These platforms contain the C. trachomatis inclusion membrane protein IncD, the mammalian ceramide transfer protein CERT and the ER resident proteins VAPA/B and were proposed to play a role in the non-vesicular trafficking of lipids to the inclusion. Here, we identify STIM1 as a novel component of ER-Inclusion MCSs. STIM1, an ER calcium (Ca2+) sensor that relocate to ER-Plasma Membrane (PM) MCSs upon Ca2+ store depletion, associated with C. trachomatis inclusion. STIM1, but not the general ER markers Rtn3C and Sec61ß, was enriched at the inclusion membrane. Ultra-structural studies demonstrated that STIM1 localized to ER-Inclusion MCSs. Time-course experiments showed that STIM1, CERT and VAPB co-localized throughout the developmental cycle. By contrast, Orai1, the PM Ca2+ channel that interacts with STIM1 at ER-PM MCSs, did not associate with C. trachomatis inclusion. Upon ER Ca2+ store depletion, a pool of STIM1 relocated to ER-PM MCSs, while the existing ER-Inclusion MCSs remained enriched in STIM1. Finally, we have identified the CAD domain, which mediates STIM1-Orai1 interaction, as the minimal domain required for STIM1 enrichment at ER-Inclusion MCSs. Altogether this study identifies STIM1 as a novel component of ER-C. trachomatis inclusion MCSs. We discuss the potential role(s) of STIM1 during the infection process.
- Published
- 2015
- Full Text
- View/download PDF
31. The class II phosphatidylinositol 3-phosphate kinase PIK3C2A promotes Shigella flexneri dissemination through formation of vacuole-like protrusions.
- Author
-
Dragoi AM and Agaisse H
- Subjects
- Bacterial Secretion Systems physiology, Cell Line, Tumor, Cell Membrane metabolism, Cell Surface Extensions microbiology, Dysentery, Bacillary pathology, Dysentery, Bacillary transmission, HT29 Cells, Host-Pathogen Interactions, Humans, Intestinal Mucosa microbiology, Listeria monocytogenes pathogenicity, Listeriosis pathology, Phosphatidylinositol 3-Kinases biosynthesis, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol Phosphates biosynthesis, RNA Interference, RNA, Small Interfering, Vacuoles metabolism, Cell Surface Extensions metabolism, Listeriosis transmission, Phosphatidylinositol 3-Kinases metabolism, Shigella flexneri pathogenicity, Vacuoles microbiology
- Abstract
Intracellular pathogens such as Shigella flexneri and Listeria monocytogenes achieve dissemination in the intestinal epithelium by displaying actin-based motility in the cytosol of infected cells. As they reach the cell periphery, motile bacteria form plasma membrane protrusions that resolve into vacuoles in adjacent cells, through a poorly understood mechanism. Here, we report on the role of the class II phosphatidylinositol 3-phosphate kinase PIK3C2A in S. flexneri dissemination. Time-lapse microscopy revealed that PIK3C2A was required for the resolution of protrusions into vacuoles through the formation of an intermediate membrane-bound compartment that we refer to as a vacuole-like protrusion (VLP). Genetic rescue of PIK3C2A depletion with RNA interference (RNAi)-resistant cDNA constructs demonstrated that VLP formation required the activity of PIK3C2A in primary infected cells. PIK3C2A expression was required for production of phosphatidylinositol 3-phosphate [PtdIns(3)P] at the plasma membrane surrounding protrusions. PtdIns(3)P production was not observed in the protrusions formed by L. monocytogenes, whose dissemination did not rely on PIK3C2A. PIK3C2A-mediated PtdIns(3)P production in S. flexneri protrusions was regulated by host cell tyrosine kinase signaling and relied on the integrity of the S. flexneri type 3 secretion system (T3SS). We suggest a model of S. flexneri dissemination in which the formation of VLPs is mediated by the PIK3C2A-dependent production of the signaling lipid PtdIns(3)P in the protrusion membrane, which relies on the T3SS-dependent activation of tyrosine kinase signaling in protrusions., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
32. A role for the small GTPase Rac1 in vaccinia actin-based motility.
- Author
-
Alvarez DE and Agaisse H
- Subjects
- Actin-Related Protein 2-3 Complex metabolism, Actins metabolism, Animals, Humans, Vaccinia virology, Vaccinia virus metabolism, Vaccinia virus pathogenicity, Viral Proteins metabolism, Wiskott-Aldrich Syndrome Protein, Neuronal metabolism, Vaccinia metabolism, Vaccinia virus physiology, Virus Release, rac1 GTP-Binding Protein metabolism
- Abstract
Vaccinia virus dissemination relies on the recruitment of the nucleation promoting factor N-WASP underneath cell-associated extracellular virus (CEVs) and subsequent recruitment and activation of the ARP2/3 complex, a major actin nucleator of the host cell. We have recently discovered that, in addition to the N-WASP/ARP2/3 pathway, vaccinia actin-based motility also relies on the small GTPase Rac1 and its downstream effector the formin-type actin nucleator FHOD1. Here we discuss the potential signaling mechanisms supporting the integration of the N-WASP/ARP2/3 and Rac1/FHOD1 pathways. We suggest the existence of a receptor tyrosine kinase family member that would integrate the Src-dependent activation of the N-WASP/ARP2/3 pathway and the GTP exchange factor-dependent activation of the Rac1/FHOD1 pathway.
- Published
- 2015
- Full Text
- View/download PDF
33. The Shigella flexneri type 3 secretion system is required for tyrosine kinase-dependent protrusion resolution, and vacuole escape during bacterial dissemination.
- Author
-
Kuehl CJ, Dragoi AM, and Agaisse H
- Subjects
- Bacterial Proteins genetics, DNA Primers genetics, Dysentery, Bacillary metabolism, Fluorescent Antibody Technique, HT29 Cells, Humans, Phosphorylation, Protein-Tyrosine Kinases metabolism, Bacterial Proteins metabolism, Cell Surface Extensions physiology, Dysentery, Bacillary physiopathology, Shigella flexneri metabolism, Type III Secretion Systems metabolism, Vacuoles metabolism
- Abstract
Shigella flexneri is a human pathogen that triggers its own entry into intestinal cells and escapes primary vacuoles to gain access to the cytosolic compartment. As cytosolic and motile bacteria encounter the cell cortex, they spread from cell to cell through formation of membrane protrusions that resolve into secondary vacuoles in adjacent cells. Here, we examined the roles of the Type 3 Secretion System (T3SS) in S. flexneri dissemination in HT-29 intestinal cells infected with the serotype 2a strain 2457T. We generated a 2457T strain defective in the expression of MxiG, a central component of the T3SS needle apparatus. As expected, the ΔmxiG strain was severely affected in its ability to invade HT-29 cells, and expression of mxiG under the control of an arabinose inducible expression system (ΔmxiG/pmxiG) restored full infectivity. In this experimental system, removal of the inducer after the invasion steps (ΔmxiG/pmxiG (Ara withdrawal)) led to normal actin-based motility in the cytosol of HT-29 cells. However, the time spent in protrusions until vacuole formation was significantly increased. Moreover, the number of formed protrusions that failed to resolve into vacuoles was also increased. Accordingly, the ΔmxiG/pmxiG (Ara withdrawal) strain failed to trigger tyrosine phosphorylation in membrane protrusions, a signaling event that is required for the resolution of protrusions into vacuoles. Finally, the ΔmxiG/pmxiG (Ara withdrawal) strain failed to escape from the formed secondary vacuoles, as previously reported in non-intestinal cells. Thus, the T3SS system displays multiple roles in S. flexneri dissemination in intestinal cells, including the tyrosine kinase signaling-dependent resolution of membrane protrusions into secondary vacuoles, and the escape from the formed secondary vacuoles.
- Published
- 2014
- Full Text
- View/download PDF
34. The serine/threonine kinase STK11 promotes Shigella flexneri dissemination through establishment of cell-cell contacts competent for tyrosine kinase signaling.
- Author
-
Dragoi AM and Agaisse H
- Subjects
- AMP-Activated Protein Kinase Kinases, Actins, Gene Silencing, HT29 Cells, HeLa Cells, Humans, Protein Serine-Threonine Kinases genetics, Protein-Tyrosine Kinases genetics, Signal Transduction, Vacuoles, Epithelial Cells metabolism, Epithelial Cells microbiology, Protein Serine-Threonine Kinases metabolism, Protein-Tyrosine Kinases metabolism, Shigella flexneri physiology
- Abstract
Shigella flexneri is an intracellular pathogen that disseminates in the intestinal epithelium by displaying actin-based motility. We found that although S. flexneri displayed comparable actin-based motilities in the cytosols of HeLa229 and HT-29 epithelial cell lines, the overall dissemination process was much more efficient in HT-29 cells. Time-lapse microscopy demonstrated that as motile bacteria reached the cell cortex in HT-29 cells, they formed membrane protrusions that resolved into vacuoles, from which the bacteria escaped and gained access to the cytosol of adjacent cells. In HeLa229 cells, S. flexneri also formed membrane protrusions that extended into adjacent cells, but the protrusions rarely resolved into vacuoles. Instead, the formed protrusions collapsed and retracted, bringing the bacteria back to the cytosol of the primary infected cells. Silencing the serine/threonine kinase STK11 (also known as LKB1) in HT-29 cells decreased the efficiency of protrusion resolution into vacuoles. Conversely, expressing STK11 in HeLa229 cells, which lack the STK11 locus, dramatically increased the efficiency of protrusion resolution into vacuoles. S. flexneri dissemination in HT-29 cells led to the local phosphorylation of tyrosine residues in protrusions, a signaling event that was not observed in HeLa229 cells but was restored in STK11-expressing HeLa229 cells. Treatment of HT-29 cells with the tyrosine kinase inhibitor imatinib abrogated tyrosine kinase signaling in protrusions, which correlated with a severe decrease in the efficiency of protrusion resolution into vacuoles. We suggest that the formation of STK11-dependent lateral cell-cell contacts competent for tyrosine kinase signaling promotes S. flexneri dissemination in epithelial cells., (Copyright © 2014, American Society for Microbiology. All Rights Reserved.)
- Published
- 2014
- Full Text
- View/download PDF
35. Antivirulence properties of an antifreeze protein.
- Author
-
Heisig M, Abraham NM, Liu L, Neelakanta G, Mattessich S, Sultana H, Shang Z, Ansari JM, Killiam C, Walker W, Cooley L, Flavell RA, Agaisse H, and Fikrig E
- Subjects
- Animals, Antifreeze Proteins genetics, Biofilms drug effects, Disease Resistance genetics, Drosophila genetics, Drosophila microbiology, Insect Proteins genetics, Ixodes chemistry, Methicillin-Resistant Staphylococcus aureus pathogenicity, Mice, Recombinant Proteins genetics, Recombinant Proteins pharmacology, Staphylococcal Infections genetics, Staphylococcal Infections immunology, Anti-Bacterial Agents pharmacology, Antifreeze Proteins pharmacology, Insect Proteins pharmacology, Methicillin-Resistant Staphylococcus aureus drug effects
- Abstract
As microbial drug-resistance increases, there is a critical need for new classes of compounds to combat infectious diseases. The Ixodes scapularis tick antifreeze glycoprotein, IAFGP, functions as an antivirulence agent against diverse bacteria, including methicillin-resistant Staphylococcus aureus. Recombinant IAFGP and a peptide, P1, derived from this protein bind to microbes and alter biofilm formation. Transgenic iafgp-expressing flies and mice challenged with bacteria, as well as wild-type animals administered P1, were resistant to infection, septic shock, or biofilm development on implanted catheter tubing. These data show that an antifreeze protein facilitates host control of bacterial infections and suggest therapeutic strategies for countering pathogens.
- Published
- 2014
- Full Text
- View/download PDF
36. Novel strategies to enforce an epithelial phenotype in mesenchymal cells.
- Author
-
Dragoi AM, Swiss R, Gao B, and Agaisse H
- Subjects
- Apoptosis Regulatory Proteins genetics, Apoptosis Regulatory Proteins metabolism, Cadherins genetics, Cadherins metabolism, Calcium-Binding Proteins genetics, Calcium-Binding Proteins metabolism, Cell Line, Tumor, Cluster Analysis, Epithelial Cells, Gene Expression Regulation, Neoplastic, HeLa Cells, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Humans, Mesenchymal Stem Cells metabolism, Neoplasm Metastasis, Neoplasms genetics, Neoplasms metabolism, Neoplasms pathology, RNA Interference, RNA Processing, Post-Transcriptional, Reproducibility of Results, Transcription Factors genetics, Transcription Factors metabolism, Transcription, Genetic, Transcriptome, Zinc Finger E-box-Binding Homeobox 1, Epithelial-Mesenchymal Transition genetics, Phenotype
- Abstract
E-cadherin downregulation in cancer cells is associated with epithelial-to-mesenchymal transition (EMT) and metastatic prowess, but the underlying mechanisms are incompletely characterized. In this study, we probed E-cadherin expression at the plasma membrane as a functional assay to identify genes involved in E-cadherin downregulation. The assay was based on the E-cadherin-dependent invasion properties of the intracellular pathogen Listeria monocytogenes. On the basis of a functional readout, automated microscopy and computer-assisted image analysis were used to screen siRNAs targeting 7,000 human genes. The validity of the screen was supported by its definition of several known regulators of E-cadherin expression, including ZEB1, HDAC1, and MMP14. We identified three new regulators (FLASH, CASP7, and PCGF1), the silencing of which was sufficient to restore high levels of E-cadherin transcription. In addition, we identified two new regulators (FBXL5 and CAV2), the silencing of which was sufficient to increase E-cadherin expression at a posttranscriptional level. FLASH silencing regulated the expression of E-cadherin and other ZEB1-dependent genes, through posttranscriptional regulation of ZEB1, but it also regulated the expression of numerous ZEB1-independent genes with functions predicted to contribute to a restoration of the epithelial phenotype. Finally, we also report the identification of siRNA duplexes that potently restored the epithelial phenotype by mimicking the activity of known and putative microRNAs. Our findings suggest new ways to enforce epithelial phenotypes as a general strategy to treat cancer by blocking invasive and metastatic phenotypes associated with EMT., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
37. Expression of the effector protein IncD in Chlamydia trachomatis mediates recruitment of the lipid transfer protein CERT and the endoplasmic reticulum-resident protein VAPB to the inclusion membrane.
- Author
-
Agaisse H and Derré I
- Subjects
- Bacterial Proteins genetics, Bacterial Proteins metabolism, Carrier Proteins genetics, Chlamydia trachomatis genetics, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, HeLa Cells, Humans, Membrane Glycoproteins genetics, Membrane Glycoproteins metabolism, Membrane Proteins genetics, Oligopeptides, Carrier Proteins metabolism, Chlamydia trachomatis metabolism, Endoplasmic Reticulum metabolism, Gene Expression Regulation, Bacterial physiology, Membrane Proteins metabolism, Protein Transport physiology
- Abstract
Chlamydia trachomatis is an obligate intracellular human pathogen responsible for ocular and genital infections. To establish its membrane-bound intracellular niche, the inclusion, C. trachomatis relies on a set of effector proteins that are injected into the host cells or inserted into the inclusion membrane. We previously proposed that insertion of the C. trachomatis effector protein IncD into the inclusion membrane contributes to the recruitment of the lipid transfer protein CERT to the inclusion. Due to the genetically intractable status of C. trachomatis at that time, this model of IncD-CERT interaction was inferred from ectopic expression of IncD and CERT in the host cell. In the present study, we investigated the impact of conditionally expressing a FLAG-tagged version of IncD in C. trachomatis. This genetic approach allowed us to establish that IncD-3×FLAG localized to the inclusion membrane and caused a massive recruitment of the lipid transfer protein CERT that relied on the PH domain of CERT. In addition, we showed that the massive IncD-dependent association of CERT with the inclusion led to an increased recruitment of the endoplasmic reticulum (ER)-resident protein VAPB, and we determined that, at the inclusion, CERT-VAPB interaction relied on the FFAT domain of CERT. Altogether, the data presented here show that expression of the C. trachomatis effector protein IncD mediates the recruitment of the lipid transfer protein CERT and the ER-resident protein VAPB to the inclusion.
- Published
- 2014
- Full Text
- View/download PDF
38. Actin network disassembly powers dissemination of Listeria monocytogenes.
- Author
-
Talman AM, Chong R, Chia J, Svitkina T, and Agaisse H
- Subjects
- Actin Cytoskeleton metabolism, Actin Cytoskeleton microbiology, Actins antagonists & inhibitors, Actins metabolism, Cell Membrane metabolism, Cell Membrane microbiology, Cofilin 1 genetics, Cofilin 1 metabolism, Cytosol metabolism, Cytosol microbiology, Gene Expression Regulation, Glia Maturation Factor genetics, Glia Maturation Factor metabolism, HeLa Cells, Host-Pathogen Interactions, Humans, Microfilament Proteins genetics, Microfilament Proteins metabolism, Microscopy, Confocal, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Signal Transduction, Vacuoles metabolism, Vacuoles microbiology, Actin Cytoskeleton genetics, Actins genetics, Listeria monocytogenes physiology
- Abstract
Several bacterial pathogens hijack the actin assembly machinery and display intracellular motility in the cytosol of infected cells. At the cell cortex, intracellular motility leads to bacterial dissemination through formation of plasma membrane protrusions that resolve into vacuoles in adjacent cells. Here, we uncover a crucial role for actin network disassembly in dissemination of Listeria monocytogenes. We found that defects in the disassembly machinery decreased the rate of actin tail turnover but did not affect the velocity of the bacteria in the cytosol. By contrast, defects in the disassembly machinery had a dramatic impact on bacterial dissemination. Our results suggest a model of L. monocytogenes dissemination in which the disassembly machinery, through local recycling of the actin network in protrusions, fuels continuous actin assembly at the bacterial pole and concurrently exhausts cytoskeleton components from the network distal to the bacterium, which enables membrane apposition and resolution of protrusions into vacuoles.
- Published
- 2014
- Full Text
- View/download PDF
39. A role for the small GTPase Rac1 in vaccinia actin-based motility.
- Author
-
Alvarez DE and Agaisse H
- Subjects
- Animals, Humans, Actin-Related Protein 2-3 Complex metabolism, Actins metabolism, Cell Movement physiology, Fetal Proteins metabolism, Nuclear Proteins metabolism, Vaccinia virus physiology, Wiskott-Aldrich Syndrome Protein, Neuronal metabolism, rac1 GTP-Binding Protein metabolism
- Abstract
Vaccinia virus dissemination relies on the recruitment of the nucleation promoting factor N-WASP underneath cell-associated extracellular virus (CEVs) and subsequent recruitment and activation of the ARP2/3 complex, a major actin nucleator of the host cell. We have recently discovered that, in addition to the N-WASP/ARP2/3 pathway, vaccinia actin-based motility also relies on the small GTPase Rac1 and its downstream effector the formin-type actin nucleator FHOD1. Here we discuss the potential signaling mechanisms supporting the integration of the N-WASP/ARP2/3 and Rac1/FHOD1 pathways. We suggest the existence of a receptor tyrosine kinase family member that would integrate the Src-dependent activation of the N-WASP/ARP2/3 pathway and the GTP exchange factor-dependent activation of the Rac1/FHOD1 pathway.
- Published
- 2014
- Full Text
- View/download PDF
40. Tyrosine kinases, drugs, and Shigella flexneri dissemination.
- Author
-
Dragoi AM and Agaisse H
- Subjects
- Humans, Dysentery, Bacillary metabolism, Intestinal Mucosa metabolism, Intestinal Mucosa microbiology, Protein-Tyrosine Kinases metabolism, Shigella flexneri metabolism
- Abstract
Shigella flexneri is an enteropathogenic bacterium responsible for approximately 100 million cases of severe dysentery each year. S. flexneri colonization of the human colonic epithelium is supported by direct spread from cell to cell, which relies on actin-based motility. We have recently uncovered that, in intestinal epithelial cells, S. flexneri actin-based motility is regulated by the Bruton's tyrosine kinase (Btk). Consequently, treatment with Ibrutinib, a specific Btk inhibitor currently used in the treatment of B-cell malignancies, effectively impaired S. flexneri spread from cell to cell. Thus, therapeutic intervention capitalizing on drugs interfering with host factors supporting the infection process may represent an effective alternative to treatments with antimicrobial compounds.
- Published
- 2014
- Full Text
- View/download PDF
41. The formin FHOD1 and the small GTPase Rac1 promote vaccinia virus actin-based motility.
- Author
-
Alvarez DE and Agaisse H
- Subjects
- Animals, Fetal Proteins antagonists & inhibitors, Fetal Proteins genetics, Fluorescent Antibody Technique, Formins, HeLa Cells, Humans, Image Processing, Computer-Assisted, Immunoblotting, Mice, Nuclear Proteins antagonists & inhibitors, Nuclear Proteins genetics, Protein Binding, RNA, Small Interfering genetics, Vaccinia metabolism, Vaccinia virology, Actin-Related Protein 2-3 Complex metabolism, Actins metabolism, Cell Movement physiology, Fetal Proteins metabolism, Nuclear Proteins metabolism, Vaccinia virus physiology, Wiskott-Aldrich Syndrome Protein, Neuronal metabolism, rac1 GTP-Binding Protein metabolism
- Abstract
Vaccinia virus dissemination relies on the N-WASP-ARP2/3 pathway, which mediates actin tail formation underneath cell-associated extracellular viruses (CEVs). Here, we uncover a previously unappreciated role for the formin FHOD1 and the small GTPase Rac1 in vaccinia actin tail formation. FHOD1 depletion decreased the number of CEVs forming actin tails and impaired the elongation rate of the formed actin tails. Recruitment of FHOD1 to actin tails relied on its GTPase binding domain in addition to its FH2 domain. In agreement with previous studies showing that FHOD1 is activated by the small GTPase Rac1, Rac1 was enriched and activated at the membrane surrounding actin tails. Rac1 depletion or expression of dominant-negative Rac1 phenocopied the effects of FHOD1 depletion and impaired the recruitment of FHOD1 to actin tails. FHOD1 overexpression rescued the actin tail formation defects observed in cells overexpressing dominant-negative Rac1. Altogether, our results indicate that, to display robust actin-based motility, vaccinia virus integrates the activity of the N-WASP-ARP2/3 and Rac1-FHOD1 pathways.
- Published
- 2013
- Full Text
- View/download PDF
42. Bruton's tyrosine kinase regulates Shigella flexneri dissemination in HT-29 intestinal cells.
- Author
-
Dragoi AM, Talman AM, and Agaisse H
- Subjects
- Actins metabolism, Agammaglobulinaemia Tyrosine Kinase, Cell Line, Tumor, Cytosol metabolism, Cytosol microbiology, Dysentery, Bacillary enzymology, Dysentery, Bacillary microbiology, HT29 Cells, Humans, Intestinal Mucosa enzymology, Phosphorylation, Shigella flexneri pathogenicity, Wiskott-Aldrich Syndrome Protein, Neuronal metabolism, Dysentery, Bacillary metabolism, Intestinal Mucosa metabolism, Intestinal Mucosa microbiology, Protein-Tyrosine Kinases metabolism, Shigella flexneri metabolism
- Abstract
Shigella flexneri is a Gram-negative intracellular pathogen that infects the intestinal epithelium and utilizes actin-based motility to spread from cell to cell. S. flexneri actin-based motility has been characterized in various cell lines, but studies in intestinal cells are limited. Here we characterized S. flexneri actin-based motility in HT-29 intestinal cells. In agreement with studies conducted in various cell lines, we showed that S. flexneri relies on neural Wiskott-Aldrich Syndrome protein (N-WASP) in HT-29 cells. We tested the potential role of various tyrosine kinases involved in N-WASP activation and uncovered a previously unappreciated role for Bruton's tyrosine kinase (Btk) in actin tail formation in intestinal cells. We showed that Btk depletion led to a decrease in N-WASP phosphorylation which affected N-WASP recruitment to the bacterial surface, decreased the number of bacteria displaying actin-based motility, and ultimately affected the efficiency of spread from cell to cell. Finally, we showed that the levels of N-WASP phosphorylation and Btk expression were increased in response to infection, which suggests that S. flexneri infection not only triggers the production of proinflammatory factors as previously described but also manipulates cellular processes required for dissemination in intestinal cells.
- Published
- 2013
- Full Text
- View/download PDF
43. Host pathways important for Coxiella burnetii infection revealed by genome-wide RNA interference screening.
- Author
-
McDonough JA, Newton HJ, Klum S, Swiss R, Agaisse H, and Roy CR
- Subjects
- Coxiella burnetii growth & development, Epithelial Cells microbiology, Genetic Testing methods, Genome, Human, HeLa Cells, Humans, Microscopy, Fluorescence, RNA Interference, Vacuoles microbiology, Coxiella burnetii pathogenicity, Host-Pathogen Interactions, Q Fever pathology
- Abstract
Unlabelled: Coxiella burnetii is an intracellular pathogen that replicates within a lysosome-like vacuole. A Dot/Icm type IVB secretion system is used by C. burnetii to translocate effector proteins into the host cytosol that likely modulate host factor function. To identify host determinants required for C. burnetii intracellular growth, a genome-wide screen was performed using gene silencing by small interfering RNA (siRNA). Replication of C. burnetii was measured by immunofluorescence microscopy in siRNA-transfected HeLa cells. Newly identified host factors included components of the retromer complex, which mediates cargo cycling between the endocytic pathway and the Golgi apparatus. Reducing the levels of the retromer cargo-adapter VPS26-VPS29-VPS35 complex or retromer-associated sorting nexins abrogated C. burnetii replication. Several genes, when silenced, resulted in enlarged vacuoles or an increased number of vacuoles within C. burnetii-infected cells. Silencing of the STX17 gene encoding syntaxin-17 resulted in a striking defect in homotypic fusion of vacuoles containing C. burnetii, suggesting a role for syntaxin-17 in regulating this process. Lastly, silencing host genes needed for C. burnetii replication correlated with defects in the translocation of Dot/Icm effectors, whereas, silencing of genes that affected vacuole morphology, but did not impact replication, did not affect Dot/Icm translocation. These data demonstrate that C. burnetii vacuole maturation is important for creating a niche that permits Dot/Icm function. Thus, genome-wide screening has revealed host determinants involved in sequential events that occur during C. burnetii infection as defined by bacterial uptake, vacuole transport and acidification, activation of the Dot/Icm system, homotypic fusion of vacuoles, and intracellular replication., Importance: Q fever in humans is caused by the bacterium Coxiella burnetii. Infection with C. burnetii is marked by its unique ability to replicate within a large vacuolar compartment inside cells that resembles the harsh, acidic environment of a lysosome. Central to its pathogenesis is the delivery of bacterial effector proteins into the host cell cytosol by a Dot/Icm type IVB secretion system. These proteins can interact with and manipulate host factors, thereby leading to creation and maintenance of the vacuole that the bacteria grow within. Using high-throughput genome-wide screening in human cells, we identified host factors important for several facets of C. burnetii infection, including vacuole transport and membrane fusion events that promote vacuole expansion. In addition, we show that maturation of the C. burnetii vacuole is necessary for creating an environment permissive for the Dot/Icm delivery of bacterial effector proteins into the host cytosol.
- Published
- 2013
- Full Text
- View/download PDF
44. The UPD3 cytokine couples environmental challenge and intestinal stem cell division through modulation of JAK/STAT signaling in the stem cell microenvironment.
- Author
-
Zhou F, Rasmussen A, Lee S, and Agaisse H
- Subjects
- Animals, Basement Membrane metabolism, Cell Communication, Cytokines metabolism, Drosophila melanogaster metabolism, Drosophila melanogaster microbiology, Enterocytes cytology, Enterocytes metabolism, Enterocytes microbiology, Environment, Enzyme Activation, ErbB Receptors metabolism, Female, Intestines microbiology, Janus Kinases metabolism, Muscles metabolism, Pectobacterium carotovorum physiology, Receptors, Invertebrate Peptide metabolism, STAT Transcription Factors metabolism, Stem Cells metabolism, Viscera metabolism, Cell Division, Drosophila Proteins metabolism, Drosophila melanogaster cytology, Intestines cytology, Signal Transduction, Stem Cell Niche, Stem Cells cytology
- Abstract
In Drosophila, the replacement of spent enterocytes (ECs) relies on division of intestinal stem cells (ISCs) and differentiation of their progeny, the enteroblasts (EBs). Recent studies have revealed a role for JAK/STAT signaling in the modulation of the rate of ISC division in response to environmental challenge. Here, we demonstrate the critical role of the UPD3 cytokine in the JAK/STAT-dependent response to enteric infection. We show that upd3 expression is activated in ECs and in EBs that massively differentiate in response to challenge. We show that the UPD3 cytokine, which is secreted basally and accumulates at the basement membrane, is required for stimulation of JAK/STAT signaling in EBs and visceral muscles (VMs). We further show that stimulation of ISC division requires active JAK/STAT signaling in EBs and VMs, but apparently not in ISCs. Our results suggest that EBs and VMs modulate the rate of the EGFR-dependent ISC division through upd3-dependent production of the EGF ligands Spitz and Vein, respectively. This study therefore supports the notion that the production of the UPD3 cytokine in stem cell progeny (ECs and EBs) stimulates intestinal stem cell division through modulation of JAK/STAT signaling in the stem cell microenvironment (EBs and VMs)., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
45. A C. trachomatis cloning vector and the generation of C. trachomatis strains expressing fluorescent proteins under the control of a C. trachomatis promoter.
- Author
-
Agaisse H and Derré I
- Subjects
- Cell Line, Chlamydia trachomatis growth & development, DNA Replication, Gene Order, Humans, Luminescent Proteins metabolism, Chlamydia trachomatis genetics, Gene Expression Regulation, Bacterial, Genetic Vectors genetics, Luminescent Proteins genetics, Operon
- Abstract
Here we describe a versatile cloning vector for conducting genetic experiments in C. trachomatis. We successfully expressed various fluorescent proteins (i.e. GFP, mCherry and CFP) from C. trachomatis regulatory elements (i.e. the promoter and terminator of the incDEFG operon) and showed that the transformed strains produced wild type amounts of infectious particles and recapitulated major features of the C. trachomatis developmental cycle. C. trachomatis strains expressing fluorescent proteins are valuable tools for studying the C. trachomatis developmental cycle. For instance, we show the feasibility of investigating the dynamics of inclusion fusion and interaction with host proteins and organelles by time-lapse video microscopy.
- Published
- 2013
- Full Text
- View/download PDF
46. Casein kinase 2 regulates vaccinia virus actin tail formation.
- Author
-
Alvarez DE and Agaisse H
- Subjects
- Actins genetics, Casein Kinase II genetics, Cell Line, Humans, Phosphorylation, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Vaccinia enzymology, Vaccinia virology, Vaccinia virus genetics, Actins metabolism, Casein Kinase II metabolism, Vaccinia metabolism, Vaccinia virus physiology
- Abstract
Casein kinase 2 (CK2) is a pleiotropic serine/threonine kinase that regulates numerous cellular processes and is essential to the infectious cycle of several viruses. Here we investigated the potential role of CK2 in vaccinia virus (VACV) infection. We used the CK2 inhibitor TBB and found that CK2 inactivation impaired VACV dissemination and actin tail formation. We used RNAi and confirmed that CK2 depletion impaired VACV actin tail formation. Furthermore, we designed a recombinant virus that allowed us to specifically detect cell-associated enveloped viruses (CEVs) at the plasma membrane and demonstrated that CK2 inactivation does not affect CEV formation. Finally, we showed that CK2 depletion impaired the recruitment of Src to CEVs. We discuss the possibility that CK2 may stimulate the A36-dependent recruitment of Src through A36 phosphorylation., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
47. The lipid transfer protein CERT interacts with the Chlamydia inclusion protein IncD and participates to ER-Chlamydia inclusion membrane contact sites.
- Author
-
Derré I, Swiss R, and Agaisse H
- Subjects
- Binding Sites, Endoplasmic Reticulum microbiology, Host-Pathogen Interactions, Humans, Intracellular Membranes metabolism, Intracellular Membranes microbiology, Protein Binding, Protein Transport, Vesicular Transport Proteins, Bacterial Proteins metabolism, Carrier Proteins metabolism, Chlamydia chemistry, Endoplasmic Reticulum metabolism, Inclusion Bodies metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
Bacterial pathogens that reside in membrane bound compartment manipulate the host cell machinery to establish and maintain their intracellular niche. The hijacking of inter-organelle vesicular trafficking through the targeting of small GTPases or SNARE proteins has been well established. Here, we show that intracellular pathogens also establish direct membrane contact sites with organelles and exploit non-vesicular transport machinery. We identified the ER-to-Golgi ceramide transfer protein CERT as a host cell factor specifically recruited to the inclusion, a membrane-bound compartment harboring the obligate intracellular pathogen Chlamydia trachomatis. We further showed that CERT recruitment to the inclusion correlated with the recruitment of VAPA/B-positive tubules in close proximity of the inclusion membrane, suggesting that ER-Inclusion membrane contact sites are formed upon C. trachomatis infection. Moreover, we identified the C. trachomatis effector protein IncD as a specific binding partner for CERT. Finally we showed that depletion of either CERT or the VAP proteins impaired bacterial development. We propose that the presence of IncD, CERT, VAPA/B, and potentially additional host and/or bacterial factors, at points of contact between the ER and the inclusion membrane provides a specialized metabolic and/or signaling microenvironment favorable to bacterial development.
- Published
- 2011
- Full Text
- View/download PDF
48. RNAi screen reveals host cell kinases specifically involved in Listeria monocytogenes spread from cell to cell.
- Author
-
Chong R, Squires R, Swiss R, and Agaisse H
- Subjects
- Actins metabolism, Cell Surface Extensions metabolism, Cytosol metabolism, HeLa Cells, Humans, Image Processing, Computer-Assisted, Microscopy, Fluorescence, Movement, Reproducibility of Results, Rickettsia conorii cytology, Genetic Testing, Host-Pathogen Interactions genetics, Listeria monocytogenes cytology, Phosphotransferases metabolism, RNA Interference
- Abstract
Intracellular bacterial pathogens, such as Listeria monocytogenes and Rickettsia conorii display actin-based motility in the cytosol of infected cells and spread from cell to cell through the formation of membrane protrusions at the cell cortex. Whereas the mechanisms supporting cytosolic actin-based motility are fairly well understood, it is unclear whether specific host factors may be required for supporting the formation and resolution of membrane protrusions. To address this gap in knowledge, we have developed high-throughput fluorescence microscopy and computer-assisted image analysis procedures to quantify pathogen spread in human epithelial cells. We used the approach to screen a siRNA library covering the human kinome and identified 7 candidate kinases whose depletion led to severe spreading defects in cells infected with L. monocytogenes. We conducted systematic validation procedures with redundant silencing reagents and confirmed the involvement of the serine/threonine kinases, CSNK1A1 and CSNK2B. We conducted secondary assays showing that, in contrast with the situation observed in CSNK2B-depleted cells, L. monocytogenes formed wild-type cytosolic tails and displayed wild-type actin-based motility in the cytosol of CSNK1A1-depleted cells. Furthermore, we developed a protrusion formation assay and showed that the spreading defect observed in CSNK1A1-depleted cells correlated with the formation of protrusion that did not resolve into double-membrane vacuoles. Moreover, we developed sending and receiving cell-specific RNAi procedures and showed that CSNK1A was required in the sending cells, but was dispensable in the receiving cells, for protrusion resolution. Finally, we showed that the observed defects were specific to Listeria monocytogenes, as Rickettsia conorii displayed wild-type cell-to-cell spread in CSNK1A1- and CSNK2B-depleted cells. We conclude that, in addition to the specific host factors supporting cytosolic actin-based motility, such as CSNK2B, Listeria monocytogenes requires specific host factors, such as CSNK1A1 in order to form productive membrane protrusions and spread from cell to cell.
- Published
- 2011
- Full Text
- View/download PDF
49. Regulatory mimicry in Listeria monocytogenes actin-based motility.
- Author
-
Chong R, Swiss R, Briones G, Stone KL, Gulcicek EE, and Agaisse H
- Subjects
- Amino Acid Sequence, Animals, Bacterial Proteins chemistry, Bacterial Proteins genetics, Casein Kinase II metabolism, Female, HeLa Cells, Humans, Listeria monocytogenes chemistry, Listeria monocytogenes genetics, Listeriosis enzymology, Listeriosis microbiology, Membrane Proteins chemistry, Membrane Proteins genetics, Mice, Mice, Inbred BALB C, Molecular Sequence Data, Protein Binding, Sequence Alignment, Actin-Related Protein 2-3 Complex metabolism, Bacterial Proteins metabolism, Listeria monocytogenes physiology, Listeriosis metabolism, Membrane Proteins metabolism
- Abstract
The actin-based motility of the intracellular pathogen Listeria monocytogenes relies on ActA, a bacterial factor with a structural domain allowing it to mimic the actin nucleation-promoting activity of host cell proteins of the WASP/WAVE family. Here, we used an RNAi-based genetic approach in combination with computer-assisted image analysis to investigate the role of host factors in L. monocytogenes cell-to-cell spread. We showed that the host cell serine/threonine kinase CK2 is required for efficient actin tail formation by L. monocytogenes. Furthermore, CK2-mediated phosphorylation of ActA regulated its affinity for the actin-nucleating ARP2/3 complex, as is the case for CK2-mediated phosphorylation of WASP and WAVE. Thus, ActA not only displays structural mimicry of WASP/WAVE family members, but also regulatory mimicry, having precisely co-opted the host machinery regulating these proteins. Comparisons based on ActA amino acid sequence suggest that unrelated pathogens that display actin-based motility may have evolved a similar strategy of regulatory mimicry.
- Published
- 2009
- Full Text
- View/download PDF
50. RNA interference screen for human genes associated with West Nile virus infection.
- Author
-
Krishnan MN, Ng A, Sukumaran B, Gilfoy FD, Uchil PD, Sultana H, Brass AL, Adametz R, Tsui M, Qian F, Montgomery RR, Lev S, Mason PW, Koski RA, Elledge SJ, Xavier RJ, Agaisse H, and Fikrig E
- Subjects
- Computational Biology, Dengue Virus physiology, Endoplasmic Reticulum metabolism, Gene Expression Profiling, Genome, Human, HIV, HeLa Cells, Humans, Immunity genetics, Monocarboxylic Acid Transporters deficiency, Monocarboxylic Acid Transporters genetics, Monocarboxylic Acid Transporters metabolism, Muscle Proteins deficiency, Muscle Proteins genetics, Muscle Proteins metabolism, Protein Binding, Ubiquitin-Protein Ligases deficiency, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Ubiquitination genetics, Vesiculovirus, Virus Replication, RNA Interference, West Nile Fever genetics, West Nile Fever virology, West Nile virus physiology
- Abstract
West Nile virus (WNV), and related flaviviruses such as tick-borne encephalitis, Japanese encephalitis, yellow fever and dengue viruses, constitute a significant global human health problem. However, our understanding of the molecular interaction of such flaviviruses with mammalian host cells is limited. WNV encodes only 10 proteins, implying that it may use many cellular proteins for infection. WNV enters the cytoplasm through pH-dependent endocytosis, undergoes cycles of translation and replication, assembles progeny virions in association with endoplasmic reticulum, and exits along the secretory pathway. RNA interference (RNAi) presents a powerful forward genetics approach to dissect virus-host cell interactions. Here we report the identification of 305 host proteins that affect WNV infection, using a human-genome-wide RNAi screen. Functional clustering of the genes revealed a complex dependence of this virus on host cell physiology, requiring a wide variety of molecules and cellular pathways for successful infection. We further demonstrate a requirement for the ubiquitin ligase CBLL1 in WNV internalization, a post-entry role for the endoplasmic-reticulum-associated degradation pathway in viral infection, and the monocarboxylic acid transporter MCT4 as a viral replication resistance factor. By extending this study to dengue virus, we show that flaviviruses have both overlapping and unique interaction strategies with host cells. This study provides a comprehensive molecular portrait of WNV-human cell interactions that forms a model for understanding single plus-stranded RNA virus infection, and reveals potential antiviral targets.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.