Triggering Ras signalling by intracellular ... - Wiley Online Library

6 downloads 118465 Views 1MB Size Report
Jul 5, 2010 - presented in the figures as a single z section after being pro- cessed using the Adobe illustrator software. Quantification of bacterial escape into ...
Cellular Microbiology (2010) 12(11), 1604–1621

doi:10.1111/j.1462-5822.2010.01494.x First published online 5 July 2010

Triggering Ras signalling by intracellular Francisella tularensis through recruitment of PKCa and bI to the SOS2/GrB2 complex is essential for bacterial proliferation in the cytosol Souhaila Al-Khodor1† and Yousef Abu Kwaik1,2* 1 Department of Microbiology and Immunology, Room 413, College of Medicine, and 2Department of Biology, University of Louisville, Louisville, KY 40202, USA. Summary Intracellular proliferation of Francisella tularensis is essential for manifestation of the fatal disease tularaemia, and is classified as a category A bioterrorism agent. The F. tularensis-containing phagosome (FCP) matures into a late endosome-like phagosome with limited fusion to lysosomes, followed by rapid bacterial escape into the cytosol. The Francisella pathogenicity island (FPI) encodes a type VI-like secretion system, and the FPIencoded IglC is essential for evasion of lysosomal fusion and phagosomal escape. Many host signalling events are likely to be modulated by F. tularensis to render the cell permissive for intracellular proliferation but they are not fully understood. Here we show that within 15 min of infection, intracellular F. tularensis ssp. novicida triggers IglC-dependent temporal activation of Ras, but attached extracellular bacteria fail to trigger Ras activation, which has never been shown for other intracellular pathogens. Intracellular F. tularensis ssp. novicida triggers activation of Ras through recruitment of PKCa and PKCbI to the SOS2/GrB2 complex. Silencing of SOS2, GrB2 and PKCa and PKCbI by RNAi has no effect on evasion of lysosomal fusion and bacterial escape into the cytosol but renders the cytosol non-permissive for replication of F. tularensis ssp. novicida. Since Ras activation promotes cell survival, we show that silencing of SOS2, GrB2 and PKCa and bI is associated with rapid early activation of caspase-3 within 8 h post infection. However, silencing of

Received 16 March, 2010; revised 25 May, 2010; accepted 28 May, 2010. *For correspondence. E-mail: [email protected]; Tel. (+1) 502 852 4117; Fax (+1) 502 852 7531. † Present address: National Institutes of Health, Bethesda, MD 20892, USA.

SOS2, GrB2 and PKCa and bI does not affect phosphorylation of Akt or Erk, indicating that activation of the PI3K/Akt and the Erk signalling cascade are independent of the F. tularensistriggered Ras activation. We conclude that intracellular F. tularensis ssp. novicida triggers temporal and early activation of Ras through the SOS2/GrB2/ PKCa/PKCbI quaternary complex. Temporal and rapid trigger of Ras signalling by intracellular F. tularensis is essential for intracellular bacterial proliferation within the cytosol, and this is associated with downregulation of early caspase-3 activation.

Introduction Francisella tularensis is a Gram-negative facultative intracellular bacterium that can cause a fatal disease designated tularaemia in humans and animals (Pechous et al., 2009; Santic et al., 2010). Being easily disseminated and highly infectious, F. tularensis has been classified as a category A select agent (Darling et al., 2002). Four closely related subspecies of F. tularensis have been identified: tularensis, holarctica, mediasiatica and novicida (Pechous et al., 2009). Two subspecies of F. tularensis cause most human illness, subspecies tularensis and subspecies holarctica (Keim et al., 2007; Champion et al., 2009). F. tularensis ssp. novicida has low virulence for humans but shares a high degree of antigenic and genetic similarities with F. tularensis ssp. tularensis and ssp. holarctica (Forsman et al., 1994) and maintains high virulence in mice (Lauriano et al., 2004; Pammit et al., 2006). In addition, F. tularensis ssp. novicida has proven to be much more amenable to genetic manipulation than the other F. tularensis subspecies (Pammit et al., 2006). Francisella tularensis enters the host cell by inducing pseudopod loops in macrophages forming the Francisella-containing phagosome (FCP) (Clemens et al., 2005; Santic et al., 2010). The FCP matures transiently to an acidified late endosome-like phagosome followed by bacterial escape into the cytosol by 30–60 min post infection (reviewed in Santic et al., 2010). Post phagosomal escape, the bacteria replicate in the cytosol and induces cmi_1494

1604..1621

© 2010 Blackwell Publishing Ltd

cellular microbiology

Ras activation by F. tularensis caspase-3-mediated apoptosis of the host cell during late stages on infection in vitro and in the mice model in vivo (Lai et al., 2001; 2004; Rajaram et al., 2009; Wickstrum et al., 2009), but F. tularensis-infected macrophages are resistant to apoptosis during early stages of infection (Santic et al., 2009). Importantly, modulation of phagosome biogenesis and escape into the cytosol by F. tularensis ssp. novicida is indistinguishable from F. tularensis ssp. tularensis and ssp. holarctica (Clemens et al., 2004; Santic et al., 2005a). While F. tularensis ssp. tularensis and holarctica have two identical copies of the FPI, F. tularensis ssp. novicida contains one single copy of the FPI (Gray et al., 2002; de Bruin et al., 2007; Santic et al., 2007). The FPI-encoded genes share homology with genes found in secretion loci of a number of bacteria, including the T6SS clusters of Vibrio cholerae and Pseudomonas aeruginosa, suggesting that the FPI encodes a T6SS-like apparatus (de Bruin et al., 2007; Barker et al., 2009). Inactivation of the FPI genes leads to decreased intramacrophage growth and decreased virulence (Gray et al., 2002; Lai et al., 2004; Tempel et al., 2006; Santic et al., 2007). The two FPIencoded proteins VgrG and IglI are translocated into the cytosol of infected macrophages (Barker et al., 2009). Both proteins are required for evasion of lysosomal fusion and bacterial escape to the cytosol (Barker et al., 2009), and the secretion of IglI is T6SS-dependent (Barker et al., 2009). The FPI-encoded iglC gene is essential for evasion of lysosomal fusion and subsequent bacterial escape into the cytosol. Recent studies have shown that bacterial escape into the cytosol of human and arthropod-derived cells is complex and requires a large portion of the bacterial genome (Asare and Abu Kwaik, 2010; Asare et al., 2010). In addition, numerous Drosophila host factors have been identified to be essential for bacterial proliferation in arthropod-derived cells and many of these factors are conserved in mammalian cells (Akimana et al., 2010). Most signalling events during infection by F. tularensis have been studied using F. tularensis ssp. novicida as a model system (Parsa et al., 2006; 2008; Rajaram et al., 2006; 2009; Butchar et al., 2008; Cremer et al., 2009a). Within the host cytosol, F. tularensis subvert host immune responses by negative regulation of TLR signalling (Telepnev et al., 2003), which results in dampening innate immunity (Butchar et al., 2008). A key element in the signalling pathways involved in transducing F. tularensisinitiated signals to cellular responses is the family of mitogen-activated protein (MAP) kinases such as p38, c-Jun N-terminal kinase (JNK) and Erk (Telepnev et al., 2005; Parsa et al., 2008). In addition to the MAP kinase pathway, the phosphoinositol 3 kinase (PI3K)/Akt pathway is also activated upon infection by F. tularensis resulting in a strong innate immune response (Parsa et al., 2006; 2008; Rajaram et al., 2006; Cremer et al., 2009b). Inhibi© 2010 Blackwell Publishing Ltd, Cellular Microbiology, 12, 1604–1621

1605

tion of PI3K/Akt results in suppression of F. tularensis ssp. novicida-induced innate immune responses (Parsa et al., 2006; Rajaram et al., 2006). The SH2 domain inositol phosphatase (SHIP) is phosphorylated upon infection by F. tularensis ssp. novicida, and negatively regulates PI3K activation resulting in dampening the innate immune response (Parsa et al., 2006; Rajaram et al., 2006). The SHIP-deficient macrophages display enhanced Akt activation upon infection by F. tularensis ssp. novicida, resulting in elevated PI3K-dependent activation and enhanced NF-kB-driven gene transcription (Parsa et al., 2006; Rajaram et al., 2006). In contrast, overexpression of the negative regulator SHIP results in decreased NF-kB activation and antagonizes the PI3K/Akt pathway (Parsa et al., 2006; Rajaram et al., 2006). In addition, a constitutive activation of Akt, or deletion of SHIP, promotes phagolysosomal fusion and limits bacterial burden in the host cytosol (Rajaram et al., 2009). Thus, signalling through the PI3K/Akt pathway by F. tularensis is required for evasion of lysosomal fusion and escape into the cytosol (Cremer et al., 2009b). It has been shown that Akt activation in macrophages occurs early during infection by F. tularensis ssp. novicida and decreases after 30 min, a pattern that is similar to the activation of Erk by F. tularensis (Rajaram et al., 2006). Activation of the Erk pathway is a critical event for immune cell activation, leading to transcriptional regulation of cytokine genes, translational regulation and other effector functions (Altman and Deckert, 1999; Ancrile et al., 2008). Activation of Erk by F. tularensis may be dependent on the GTPase-activating protein (GAP) protein Ras, which triggers activation of the Raf-1/MEK1/ERK kinase cascade (Boguski and McCormick, 1993; Kawakami et al., 2003; Byun et al., 2009). Ras is at the point of convergence for many signalling pathways, and this activity is modulated by the guanine nucleotide binding (Plyte et al., 2000). Ras is activated transiently in response to a diverse array of extracellular signals such as growth factors, cytokines, hormones and neurotransmitters that stimulate cell surface receptors such as receptor tyrosine kinases (RTKs), non-receptor tyrosine kinase-associated receptors and G protein-coupled receptors (Satoh and Kaziro, 1992; Satoh et al., 1992). The best-characterized Ras-mediated signal transduction pathway involves the activation of the epidermal growth factor receptor (Egan et al., 1993; Warner et al., 1993). Upon ligand binding, the receptor undergoes autophosphorylation, which creates binding sites of the Shc and/or Grb2 adaptor proteins (Lowenstein et al., 1992; Rotin et al., 1992). GrB2 is stably associated with the son of sevenless homologue 2 (SOS2), which is the Ras guanine nucleotide exchange factors (GEF) (Campbell et al., 1998). The recruitment of the SOS2/GrB2 complex to Ras in the plasma membrane (Plyte et al., 2000) results in a temporal activation of Ras

1606 S. Al-Khodor and Y. Abu Kwaik (Feig, 1994; Feig and Schaffhausen, 1994; Campbell et al., 1998). The recruitment of SOS2/GrB2 to Ras can occur directly or indirectly through the Shc adaptor protein (Lowenstein et al., 1992; Rotin et al., 1992). Shc, GrB2 and SOS2 provide the link between various activated cell surface receptors and Ras (Lowenstein et al., 1992; Rotin et al., 1992, Feig, 1994; Feig and Schaffhausen, 1994). The PKC-dependent Syk signalling pathway also triggers the Ras/Erk pathway (Kawakami et al., 2003; Parsa et al., 2008). Upon receptor stimulation, the serine/ threonine kinases PKCa and PKCb1 recruit the SOS2/ GrB2 complex to the vicinity of Ras, which facilitates the exchange of GTP for GDP on Ras (Kawakami et al., 2003). The active Ras-GTP stimulates four downstream effectors including Raf protein kinase, PI3K and the phospholipase-C (PLC) (Jasinski et al., 2008a). Each of these effectors regulates essential pathways of cell-cycle progression, survival and anti-apoptosis (Boguski and McCormick, 1993, Plyte et al., 2000; Kawakami et al., 2003; Honma et al., 2006; Jasinski et al., 2008a). It is not known whether F. tularensis signals Ras activation within infected cells. In this article we show that F. tularensis ssp. novicida activates Ras in human cells during early stages of infection and that this activation depends on bacterial entry and on the FPI-encoded IglC protein. Both PKCa and bI isoforms are recruited to the SOS2/GrB2 complex resulting in the activation of Ras. Silencing of SOS2, GrB2 or PKCa and bI abrogates Ras activation by F. tularensis but has no effect on evasion of lysosomal fusion and bacterial escape into the cytosol and results in non-permissiveness of the cytosol for bacterial proliferation. The SOS2, GrB2 or PKCa and bI are essential for prevention of early activation of caspase-3 in F. tularensis-infected cells.

Results F. tularensis activates Ras upon entry to the host cell It has never been examined whether F. tularensis activates Ras during infection. Therefore, we determined whether F. tularensis triggers activation of Ras in HEK293T cells and human monocytes-derived macrophages (hMDMs). The cells were infected with F. tularensis ssp. novicida strain U112 for the indicated time points, and the iglC mutant which is defective in evasion of lysosomal fusion, phagosomal escape, was used as a negative control (Fig. 1). The post-nuclear supernatant (PNS) from infected cells was incubated with the Raf1 Ras-binding domain (RBD) agarose beads and the bound Ras proteins were analysed by Western blot using anti-Ras antibody that only detects active GTP-Ras. GTPgS was used as a positive control and the untreated cells were used as a second negative control. The data showed that by

5–15 min post infection of HEK293T cells and hMDMs, wild-type (WT) F. tularensis ssp. novicida triggered rapid Ras activation while the iglC mutant and the formalinkilled (FK) WT strain failed to activate Ras, similar to non-infected cells (Fig. 1B and D). To study the kinetics of Ras activation during infection by F. tularensis ssp. novicida, we examined Ras activation at different time points of infection (Fig. 1B and D). The data showed that Ras activation was temporal and occurred within 5–15 min, but it disappeared by 2–6 h after infection (Fig. 1B and D). Importantly, the iglC mutant, and replication, did not trigger activation of Ras (Fig. 1B and D). Taken together, the data show that intracellular F. tularensis activates Ras temporally within 5–15 min of infection and the FPI-encoded IglC protein is essential for Ras activation. It is possible that attached extracellular F. tularensis triggered Ras activation through a receptor-mediated binding and signalling (Keates et al., 2001). However, it is also possible that Ras activation is triggered by the intracellular bacteria. To determine whether bacterial entry was essential to trigger Ras activation, the HEK293T cells and hMDMs were pre-treated with Cytochalasin-D (CD) prior to infection to block bacterial entry to the host cell (Goddette and Frieden, 1986; Gavrilin et al., 2006). The data clearly showed that Ras was not activated by the attached extracellular bacteria in both cells (Fig. 1C and D), indicating that bacterial entry to the host cell is essential for triggering temporal activation of Ras. Taken together, we showed that Ras activation occurs in both hMDMs and HEK293T cells infected with the WT F. tularensis ssp. novicida at 5–15 min post infection and that bacterial entry is required for the Ras activation in an IglC-dependent manner. This is the first demonstration of intracellular triggering of Ras activation by an intracellular bacterial pathogen. For the rest of this study we used HEK293T cells as a model system for infection due to the ease and efficiency of RNAi-mediated gene silencing, since, clearly, Ras was rapidly triggered by intracellular bacteria and IglC was required for this activation in both cells. In addition, we verified that evasion of lysosomal fusion by F. tularensis, bacterial escape into the cytosol, and intracellular replication within HEK293T cells is indistinguishable from hMDMs (data not shown). Mechanism of Ras activation by F. tularensis PKC has been described as a major activator of the Ras pathway (Robin et al., 2004). This activation can occur through phosphorylation of the PKC isoforms by Syk and their recruitment to the SOS2/GrB2 complex. To decipher whether the mechanism of the Ras activation pathway triggered by F. tularensis is PKC-dependent, we © 2010 Blackwell Publishing Ltd, Cellular Microbiology, 12, 1604–1621

Ras activation by F. tularensis

1607

Fig. 1. Temporal Ras activation in F. tularensis-infected cells. A. For Ras activation assay, the PNS from uninfected HEK293T cells or F. tularensis-infected cells and formalin-killed (FK) bacteria-infected cells were incubated with the Raf1 RBD (Ras-binding domain) agarose beads and the bound Ras proteins were analysed by Western blot using the Ras antibody using GTPgS as a positive control. Equal loading was verified by anti-actin antibody. B. Kinetics of Ras activation during infection by F. tularensis. PNS from uninfected HEK293T cells or F. tularensis and iglC mutant-infected cells for 15 min, 2 h and 6 h were incubated with the Raf1 RBD (Ras-binding domain) agarose beads, processed and probed as described in (A). C. Role of bacterial entry in Ras activation. HEK293T cells were pre-treated with 1 mg ml-1 Cytochalasin-D (CD) for 45 min prior to GTPgS treatment or infection for 15 min. The PNS from uninfected cells, and CD- or DMSO-treated cells were incubated with the Raf1 RBD (Ras-binding domain) agarose beads, processed and probed as described in (A). D. Kinetics of Ras activation in hMDMs: PNS from uninfected hMDMs or F. tularensis-infected cells and iglC mutant-infected cells as well as CD-treated cells infected with the WT strain were incubated with the Raf1 RBD (Ras-binding domain) agarose beads and the bound Ras proteins were analysed by Western blot using the Ras antibody, using GTPgS as a positive control. Equal loading was verified by anti-actin antibody. This experiment was performed three times and the results are representative of one experiment.

examined whether F. tularensis triggered activation of PKC and its recruitment to the SOS2/GrB2 complex. The HEK293T cells were infected with F. tularensis ssp. novicida for 15 min and Formalin-killed bacteria (FK) was used as a negative control. The PNS from both uninfected and infected cells were immunoprecipitated with the anti-SOS2, anti-GrB2, anti-PKCa or anti-PKC-b1 antibodies followed by immunoblotting. The constitutive interaction between SOS2 and GrB2 proteins was considered as a positive control. As expected, GrB2 © 2010 Blackwell Publishing Ltd, Cellular Microbiology, 12, 1604–1621

co-immunoprecipitated with SOS2 in both live WT- and FK-infected cells (Fig. 2, lanes C and D). In control experiments, PNS from uninfected cells did not show any interaction between the PKC isoforms and the SOS2/GrB2 complex (Fig. 2, lane F). The PKCa and b1 isoforms interacted with the SOS2/GrB2 complex within 15 min of infection by live F. tularensis (Fig. 2, lane A1, lane B1). However, in FK bacteria-infected cells, the PKCa and b1 isoforms did not interact with the SOS2/GrB2 complex, indicating that only viable bacteria specifically triggered

1608 S. Al-Khodor and Y. Abu Kwaik

Fig. 2. Recruitment of PKCa and bI isoforms to the SOS2/GrB2 complex during infection by F. tularensis. The PNS from WT strain or FK bacteria-infected HEK293T cells for 15 min were incubated with anti-PKC-a antibody (lanes A1 and A2), anti-PKC-bI antibody (lanes B1 and B2), anti-SOS2 antibody (lanes C1 and C2) or anti-GrB2 antibody (lanes D1 and D2). The PNS from uninfected cells (lane F) were used as a control. No antibody was added to the samples in lanes E1 and E2, as additional controls. After overnight incubation at 4°C, G protein-coupled beads were added to all samples, except the one in lane F. After serial washes, pulled-down proteins were analysed by Western blot with the respective antibodies as shown. Equal loading was verified by anti-actin antibody. This experiment was performed three times and the results are representative of one experiment.

association of both PKC isoforms with the SOS2/GrB2 bi-molecular complex (Fig. 2, lane A2, lane B2). When the blots were re-probed with the anti-Shc antibody the data showed that Shc did not bind to the SOS2/GrB2 complex in live WT- and FK bacteria-infected cells (data not shown). Taken together, intracellular F. tularensis triggers temporal Ras activation within 15 min of infection via a Shc-independent mechanism that involves interaction of both PKCa and b1 isoforms with the SOS2/GrB2 bi-molecular complex. Role of the SOS2/GrB2 complex and PKCa/b1 isoforms in F. tularensis-induced Ras activation After stimulation of a receptor, PKCa/bI recruit the SOS2/ GrB2 complex to the vicinity of Ras resulting in exchange of GDP to GTP, which activates Ras. To determine if both SOS2 and GrB2 were essential for Ras activation upon infection by F. tularensis, both factors were silenced separately by specific RNAi. Specificity of gene silencing was confirmed by Western blot (Fig. 3A). We utilized the HEK293T cells that exhibited efficient silencing. Cell viability was monitored by trypan blue staining, which

showed no effect of silencing of SOS2 or GrB2 on cell viability, similar to the untreated cells or the RNAi controltreated cells. To determine whether Ras was activated in the SOS2 and GrB2-silenced cells, the silenced and control cells were infected by F. tularensis for 15 min. The PNS were incubated with the Raf1 RBD agarose beads and the bound Ras proteins were analysed by Western blot using the anti-Ras antibody that only detects active GTP-Ras. The GTPgS treatment was performed in the untreated cells and was used as a positive control for the Ras activation assay. Active Ras was detected in the F. tularensis-infected but not uninfected cells (Fig. 3B). No difference in the levels of Ras activation was observed between untreated and RNAi control-treated cells (data not shown). By 15 min post infection, Ras was not activated by F. tularensis in the SOS2 or GrB2-silenced cells compared with the non-silenced cells (Fig. 3B). We conclude that F. tularensis triggers Ras activation through the SOS2/GrB2 bi-molecular complex and that both SOS2 and GrB2 are essential for the temporal activation of Ras by intracellular F. tularensis. To decipher the role of PKCa/bI in the activation of Ras by F. tularensis, we silenced the PKC isoforms in © 2010 Blackwell Publishing Ltd, Cellular Microbiology, 12, 1604–1621

Ras activation by F. tularensis

1609

Fig. 3. Role of PKCa/bI/SOS2/GrB2 complex in Ras activation during infection by F. tularensis. A. Western blot analysis of SOS2, GrB2, PKCa and bI gene silencing in HEK293T cells. Untreated or RNAi-treated cells were lysed at 48 h after treatment by RNAi. The PNS were analysed by SDS-PAGE and immunoblotting with specific antibodies as shown. The blots were then stripped and re-probed with the anti-actin antibody for a loading control. B. Role of SOS2/GrB2 complex and PKCa and bI isoforms in the Ras activation during infection by F. tularensis. Cells were either left untreated or treated with RNAi to silence SOS2, GrB2, PKCa and PKCbI using specific RNAi. Gene silencing was verified by Western blot. Both untreated and RNAi-treated HEK293T cells were infected with the WT strain for 15 min. The PNS were then processed for the Ras activation assay and bound proteins were analysed by Western blot using anti-Ras antibody (see legend 1 for details). GTPgS was used as positive control and was performed in the untreated cells. Equal loading was verified by anti-actin antibody. This experiment was performed three times and the results are representative of one experiment.

HEK293T cells using specific RNAi. Cell viability was monitored by trypan blue staining, which showed no effect of silencing of either PKCa or bI on cell viability, similar to untreated cells or RNAi control-treated cells. Specificity of gene silencing was confirmed by Western blot (Fig. 3A). The PKCa- or bI-silenced cells were infected by F. tularensis for 15 min and PNS were prepared as described above. The data showed that silencing of both PKC isoforms abolished the activation of Ras by F. tularensis compared with the mock-treated or RNAi control-treated cells (Fig. 3B). We conclude that in addition to the SOS2/ GrB2 bi-molecular complex, both PKCa and bI isoforms are essential for the temporal activation of Ras by intracellular F. tularensis. SOS2, GrB2, PKCa and PKCbI are required for intracellular proliferation of F. tularensis Since Ras was activated during early stages of infection by F. tularensis in an IglC-dependent mechanism and IglC is essential for intracellular proliferation, we examined whether the F. tularensis-induced Ras activation pathway is essential for bacterial replication. We examined the role of the SOS2/GrB2 complex and the PKC isoforms in intracellular replication of F. tularensis. The HEK293T © 2010 Blackwell Publishing Ltd, Cellular Microbiology, 12, 1604–1621

cells were either left untreated or treated for 48 h with the RNAi-negative control or gene-specific RNAi. Moreover, to exclude non-specific effect of silencing, we silenced calpain-9 (Cap9), as a control molecule, and showed that Cap9 silencing does not affect bacterial replication (data not shown). Gene silencing was verified by Western blot (Fig. 3A). Untreated HEK293T cells and successfully silenced cells for each of the factors were infected by F. tularensis. The infection was carried our for 1 h using multiplicity of infection (moi) of 10 followed by 1 h of gentamicin treatment and further incubation for additional 6 h and 22 h for a total of 8 h and 24 h respectively. At each time point, cells were fixed and labelled with antiF. tularensis antibodies and analysed by microscopybased single cell analysis (Fig. 4). At 2 h post infection, there was no difference in the efficiency or frequency of bacterial internalization between the non-treated or the RNAi-treated cells (one bacterium per cell) indicating that bacterial uptake was not affected upon silencing of those factors, resulting in inhibition of Ras activation (Fig. 4A). At 8 h post infection, ~80% of the untreated cells or cells transfected by the RNAi-negative control harboured 15–20 bacteria per cell (Fig. 4B). In contrast, 80–85% of the SOS2-silenced or GrB2-silenced cells did not show any bacterial proliferation at 8 h and 24 h post infection

1610 S. Al-Khodor and Y. Abu Kwaik

Fig. 4. Replication of F. tularensis within RNAi-silenced HEK293T cells. A–C. Immunofluorescence images of HEK293T cells at 2 h after infection by F. tularensis. HEK293T cells were either left untreated (Un) or treated with the RNAi-negative control or the gene-specific RNAi for 48 h before infection, then infected with the WT strain U112 or formalinkilled (FK) bacteria at moi of 10 for 1 h, followed by gentamicin treatment for 1 h to kill extracellular bacteria. The cells were further incubated for a total of 8 h and 24 h. At all time points, cells were processed for confocal microscopy as described in Experimental procedures. D. Quantitative analysis of the % of the infected cells harbouring replicating F. tularensis ssp. novicida in both untreated (UN) or RNAi-treated cells at 8 h post infection. The normal levels of replication within untreated HEK293T cells was > 15 bacteria per cell, and ~25% of the cells are infected under our experimental conditions. At least 100 cells were analysed from two different coverslips. Error bars represent standard deviation. The P-value < 0.01 is considered significant and was represented by the asterisks. This experiment was performed three times and the results are representative of one experiment.

(Fig. 4B–D). Results obtained from the RNAi-negative control-treated cells were comparable to the untreated cells confirming the specificity of silencing (Fig. 4B). These data show that the SOS2 and GrB2 factors are not required for bacterial entry but are essential for intracellular replication of F. tularensis throughout the intracellular infection. Silencing of either PKCa or PKCbI isoforms resulted in a dramatic loss of temporal activation of Ras by F. tularensis indicating an essential role of those isoforms in Ras activation (Fig. 3). To determine the role of PKCa and PKCbI isoforms in intracellular bacterial proliferation, both isoforms were silenced. The data showed that silencing of either isoforms did not affect the efficiency or frequency of bacterial internalization, and most of the infected cells harboured one bacterium at 2 h post infection. By 8–24 h post infection, silencing of either PKCa or PKCbI isoforms inhibited bacterial replication (Fig. 4B). The data showed

that by 8 h or 24 h post infection, 70–80% of the PKCaand PKCbI-silenced infected cells contained only one bacterium per cell (Fig. 4), indicating that PKCa and PKCbI are essential for permissiveness of the host cells for intracellular proliferation of F. tularensis. We conclude that components of the Ras activation pathway including the bi-molecular complex SOS2/GrB2 and both PKCa and PKCbI isoforms are dispensable for bacterial entry but are indispensable for intracellular proliferation of F. tularensis. The role of SOS2, GrB2, PKCa and PKCbI in intracellular trafficking of F. tularensis The FCP transiently matures to an acidified late endosome-like phagosome followed by bacterial escape into the cytosol by 30–60 min post infection (reviewed in Santic et al., 2010). However, the iglC mutant-containing © 2010 Blackwell Publishing Ltd, Cellular Microbiology, 12, 1604–1621

Ras activation by F. tularensis

1611

Fig. 5. Evasion of lysosomal fusion by F. tularensis is independent of the SOS2/GrB2 complex and the PKCa and bI isoforms. A. Representative confocal microscopy images of the late endosomal/lysosomal marker LAMP-2 colocalization with the phagosomes containing F. tularensis ssp. novicida. Untreated (UN) HEK293T cells or treated with the RNAi-negative control or the gene-specific RNAi for 48 h were infected with the WT strain U112 or formalin-killed (FK) bacteria using moi of 10 for 1 h followed by 1 h treatment with gentamicin to kill extracellular bacteria. The iglC mutant was used as a positive control. The white arrow points at colocalization of the iglC mutant-containing vacuole with the LAMP2 marker. B and C. Quantitative analysis of colocalization of the WT strain-containing phagosomes with LAMP2 at 30 min (B) and 2 h (C) after infection. At least 100 cells were analysed from two coverslips. A P-value < 0.01 is considered significant and is represented by the asterisks. This experiment was performed three times and the results are representative of one experiment.

phagosome fuses to the lysosomes and the mutant is defective in phagosomal escape into the cytosol (Santic et al., 2005a; 2007). The host factors involved in evasion of endocytic fusion and phagosomal escape of F. tularensis are not known. We postulated that nonpermissiveness of the SOS2-, GrB2-, PKCa- and bI-silenced cells to bacterial replication might be due to alteration of biogenesis of the FCP. To test this hypothesis, we examined intracellular trafficking within untreated or SOS2, GrB2, PKCa and bI RNAi-treated cells and used the iglC mutant as a positive control. Since the FCP transiently colocalizes with the LAMP2 late-endosomal marker for 15–30 min followed by a gradual loss of this colocalization by 1–2 h, we examined colocalization of the phagosomes with LAMP-2 and the lysosomal marker Cathepsin-D at 30 min and 2 h after infection (Fig. 5 and Fig. S1). At 30 min after infection, © 2010 Blackwell Publishing Ltd, Cellular Microbiology, 12, 1604–1621

75–80% of the iglC mutant-containing phagosomes colocalized with LAMP2 and Cathepsin-D respectively (Fig. 5B and Fig. S1A) (Santic et al., 2007). As expected, ~45–48% of the WT strain containing phagosomes in untreated cells colocalized with LAMP2 at 30 min post infection (Fig. 5B). No significant difference (Student’s t-test, P > 0.1) in LAMP2 and Cathepsin-D colocalization was observed between the WT strain-containing phagosomes in untreated or RNAi-treated cells at 30 min post infection (Fig. 5B and Fig. S1). At 2 h after infection, 65% of the WT strain-containing phagosomes did not colocalize with LAMP-2 or Cathepsin-D (Fig. 5C and Fig. S1), while ~90% of the iglC mutant-containing phagosomes showed persistent colocalization with both markers, consistent with previous observation (Fig. 5C and Fig. S1) (Santic et al., 2005a). Silencing of SOS2, GrB2 or the two PKC isoforms did not

1612 S. Al-Khodor and Y. Abu Kwaik have a significant effect on colocalization of the FCP with LAMP2 or Cathepsin-D, when compared with untreated cells or RNAi control-treated cells (Student’s t-test, P > 0.4) (Fig. 5 and Fig. S1). Taken together, neither the SOS2/GrB2 complex nor the PKCa/bI is essential for evasion of lysosomal fusion by F. tularensis. Therefore, activation of the Ras signalling by F. tularensis through recruitment of PKCa and bI to the SOS2/GrB2 complex is independent of modulation of phagosomal biogenesis and evasion of lysosomal fusion. The SOS2/GrB2 complex and PKCa/bI isoforms are dispensable for escape of F. tularensis into the cytosol Loss of the late endosomal marker LAMP-2 from the FCP by 30–60 min coincides with phagosomal escape of F. tularensis into the cytosol (Checroun et al., 2006; Santic et al., 2007; 2008; 2010; Wehrly et al., 2009). To study the role of the SOS2/GrB2 complex and PKCa/bI isoforms in bacterial escape, we utilized the fluorescence microscopy-based phagosomal integrity assay (Checroun et al., 2006; Santic et al., 2008). This assay allows differential labelling of bacteria that are cytosolic or within a compromised phagosomes versus those enclosed within intact phagosomes. After preferential permeabilization of the plasma membrane, an antibacterial antibody was loaded into the host cell cytosol to allow binding to the cytosolic bacteria, while bacteria within intact FCPs do not bind the antibody. To test whether the Ras activation cascade is involved in bacterial escape to the cytosol, we silenced the major components of the Ras activation pathway including SOS2, GrB2, PKCa/bI using gene-specific RNAi, and examined the effect on phagosomal escape. Untreated HEK293T cells and successfully silenced cells for each of the factors were infected by F. tularensis for 1 h using moi of 10, followed by 1 h of gentamicin treatment, and then processed for the fluorescence-based phagosomal integrity assay (Checroun et al., 2006; Santic et al., 2008). The iglC mutant was used as a positive control for bacteria within intact FCPs (Santic et al., 2005b). The data showed that in the untreated HEK293T cells that were infected by the WT strain, > 80% of the WT bacteria were cytosolic or within disrupted phagosomes by 2 h post infection, as expected (Fig. 6). Similarly, > 80% of the intracellular bacteria were cytosolic in the SOS2-, GrB2-, PKCa- and bI-silenced cells. Consistent with previous findings, ~80% of the iglC mutant control was within intact phagosomes (Fig. 6). These data show that the SOS2, GrB2, PKCa and PKCbII components of the Ras activation pathway play no detectable role in bacterial escape into the host cytosol. This indicates that escape of F. tularensis into the cytosol is independent of F. tularensistriggered Ras activation.

To determine whether silencing of SOS2, GrB2, PKCa and bI caused a re-entry of the bacteria into the endocytic pathway later in the infection (Checroun et al., 2006), LAMP-2 colocalization experiments were conducted at 12 and 24 h post infection. In the untreated cells and the RNAi-treated cells, F. tularensis did not re-enter LAMP-2positive vacuoles at 12 h and 24 h post infection (data not shown). Taken together, we conclude that SOS2, GrB2, PKCa and PKCbI are dispensable for evasion of lysosomal fusion and escape of F. tularensis into the cytosol, but they are indispensable for bacterial proliferation in the cytosol. Signalling of Ras through the SOS2/GrB2/PKCa/PKCbI quaternary complex protect F. tularensis-infected cells from early activation of caspase-3 It has been shown that intracellular proliferation of F. tularensis results in the activation of caspase-3mediated apoptosis during late stages of infection in tissue culture and mice organs in vivo (Rajaram et al., 2009; Wickstrum et al., 2009), but F. tularensis-infected macrophages are resistant to apoptosis during early stages of infection (Santic et al., 2009). Ras is involved in promoting cell survival through blocking activation of caspase-3 (Jasinski et al., 2008b). Therefore, we postulated that the Ras activation pathway by intracellular F. tularensis contributed to survival of the infected cells by blocking early activation of the pro-apoptotic caspase-3. To test our hypothesis, we determined the role of SOS2, GrB2, PKCa/bI in blocking early activation of caspase-3 in F. tularensis-infected cells. We silenced SOS2, GrB2, PKCa/bI in the HEK293T cells using genespecific RNAi. Untreated cells and successfully silenced cells for each of the factors were infected by F. tularensis. The infection was carried out for 1 h using moi of 10, followed by 1 h of gentamicin treatment, and incubated for additional 6 and 22 h for a total of 8 and 24 h. The cells were labelled with anti-active caspase-3 antibody and analysed by confocal microscopy. Staurosporine (STS, 1 mM), which triggers caspase-3-mediated apoptotic death in the HEK293T cells (Nakamura et al., 2007), was used as a positive control (Fig. 7B). DMSO-treated cells either uninfected or infected by F. tularensis were used as a negative control. Caspase-3 activation was not detected in mock-treated cells, but an increase of 10- to 12-fold in the number of cells exhibiting caspase-3 activation was observed among the uninfected cells after STS treatment compared with the DMSO-treated cells (Fig. 7B and C). The data showed that infection of cells silenced for SOS2, GrB2, PKCa and bI with F. tularensis resulted in early caspase-3 activation by 8 h post infection, where four- to eightfold increase in the number of © 2010 Blackwell Publishing Ltd, Cellular Microbiology, 12, 1604–1621

Ras activation by F. tularensis

1613

Fig. 6. Escape of F. tularensis into the cytosol is independent of the SOS2/GrB2 complex and the PKCa and bI isoforms. A. Representative confocal microscopy images of the WT F. tularensis within untreated (UN) or RNAi-treated cells to determine phagosomal escape using the fluorescence-based phagosomal integrity assay. This was determined by the ability of the intracellular bacteria to bind anti-F. tularensis antibody loaded into the host cell cytosol after preferential permeabilization of the plasma membrane. This is in contrast to bacteria found within intact vacuoles that are impermeable to the antibody. The iglC mutant was used as a negative control for bacteria trapped within intact phagosomes. B. Quantification of the % of disrupted phagosomes (FCP) from untreated or RNAi-treated cells. At least 100 infected cells from two different coverslips. This experiment was performed twice and the results are representative of one experiment.

cells exhibiting caspase-3 activation was detected in the RNAi-treated cells compared with the untreated or RNAi control-treated cells (Fig. 7C) (Student’s t-test, P < 0.01). Importantly, silencing of each of the four factors did not trigger early caspase-3 activation in uninfected cells (Fig. 7). A 12- to 14-fold increase in the number of cells exhibiting capsase-3 activation was also observed at 24 h post infection in the cells where expression of SOS2, GrB2, PKCa and bI was knocked down compared with untreated cells or RNAi control-treated cells (Fig. 7A and C). Therefore, early activation of caspase-3 is specifically triggered by 8 h post infection by the WT strain of F. tularensis in the cells silenced for SOS2, GrB2 or PKCa and bI (Fig. 7). We conclude that SOS2, GrB2, PKCa and bI are crucial for protecting F. tularensisinfected cells from early activation of caspase-3 within F. tularensis-infected cells. © 2010 Blackwell Publishing Ltd, Cellular Microbiology, 12, 1604–1621

SOS2, GrB2, PKCa and bI are not required for activation of Akt or Erk by F. tularensis It has been shown that F. tularensis induces Akt and Erk phosphorylation that peaks during the first few minutes of infection and Akt phosphorylation occurs downstream of the PI3K pathway (Rajaram et al., 2006). It is also possible the Erk phosphorylation occurs downstream of Ras activation by F. tularensis. Moreover, class I PI3K contain RBDs and can also be activated through association with active Ras. To determine if the Ras activation pathway interacted with the PI3K pathway or Erk upon infection by F. tularensis, we analysed Akt and Erk phosphorylation upon silencing the major players of the Ras activation pathway including SOS2, GrB2 and both PKC isoforms. Untreated and RNAi-treated cells were infected with F. tularensis and verification of the specific gene silencing was

1614 S. Al-Khodor and Y. Abu Kwaik

Fig. 7. Early activation of caspase-3 in the SOS2-, GrB2-, PKCa- and bI-silenced cells upon infection by F. tularensis. A. Representative confocal microscopy images of the caspase-3 activation after infection with the F. tularensis WT strain. HEK293T cells were either left untreated of treated with the SOS2, GrB2, PKCa and bI respective RNAi. After verifying specific gene silencing, cells were infected with the WT strain U112 using moi of 10 for 1 h followed by 1 h treatment with gentamicin to kill extracellular bacteria. Cells were further incubated for a total of 24 h. Caspase-3 activation was detected by anti-active caspase-3 antibody at 24 h post-infection. B. Controls for caspase-3 activation: Staurosporine (STS)-treated cells were used as a positive control and DMSO-treated cells were used as a negative control. Caspase-3 activation was detected by anti-active caspase-3 antibody at 24 h post infection, compared with Staurosporine (STS)-treated cells were used as a positive control and DMSO-treated cells used as a negative control. C. Quantitative analysis of caspase-3 activation at 8 and 24 h post infection. At least 100 cells were analysed from two different coverslips. The levels of caspase-3 activation are presented as ratio of number of cells with active caspase-3 in RNAi-treated cells that were infected by F. tularensis, versus uninfected cells treated with the same RNAi. This experiment was performed twice and the results are representative of one experiment.

accomplished by Western blot. The cells were lysed 5–30 min after infection and protein lysates were analysed by Western blotting with the anti-pAkt or anti-pErk antibody (Fig. 8 and data not shown). To ensure equal protein loading, the membranes were re-probed with the anti-Akt and Anti-Erk antibody. The results showed that infection of untreated cells by F. tularensis induced robust phosphorylation of Akt and Erk (Fig. 8 and data not shown). Silencing of SOS2, GrB2, PKCa and bI did not have any detectable effect on the phosphorylation of Akt or Erk by 5–30 min after infection (Fig. 8 and data not shown). This indicates the activation of the PI3K/Akt pathway and Erk activation are independent of Ras activation by F. tularensis. Independence of F. tularensismediated Ras activation from F. tularensis-mediated activation of the PI3k/Akt pathway is consistent with the

Fig. 8. Triggering the PI3K and the Ras activation pathways by F. tularensis is independent. Untreated or RNAi-treated HEK293T cells were infected with the WT strain F. tularensis ssp. novicida for 5 min and compared with uninfected cells. Phosphorylation of Akt was detected using anti-pAkt antibody. The same membrane was re-probed with anti-Akt antibody to verify equal loading. © 2010 Blackwell Publishing Ltd, Cellular Microbiology, 12, 1604–1621

Ras activation by F. tularensis role of the PI3K/Akt pathway in evasion of lysosomal fusion and phagosomal escape of F. tularensis (Rajaram et al., 2009), while Ras signalling by F. tularensis through SOS2, GrB2, PKCa and bI is independent of evasion of lysosomal fusion and phagosomal escape. Discussion Early phases of the interaction of F. tularensis with the host and modulating the signalling cascades are thought to be key events in subsequent intracellular proliferation (Butchar et al., 2008; Santic et al., 2010). However, the signalling steps that are triggered during this crucial stage of initiating the infection are largely unknown. Many studies have demonstrated the role of the PI3K/Akt/SHIP/ MIR-155 in signalling during infection by F. tularensis (Parsa et al., 2006; 2008; Rajaram et al., 2006; 2009). The receptor-mediated stimulation of Ras and subsequent activation of its downstream effector molecules is critical in the biological response to numerous signals (Meadows et al., 2001), but Ras activation has never been shown to be triggered intracellularly by any intracellular bacterial pathogen. Whether Ras is activated by intracellular F. tularensis has not been reported yet. In this article we describe for the first time the mechanism of Ras activation by intracellular F. tularensis ssp. novicida and its indispensable role in intracellular bacterial proliferation, which has never been shown for any intracellular pathogen. We show a SOS2/GrB2/PKCa- and PKCbI-dependent signalling pathway is temporally triggered within 15 min of infection by F. tularensis, leading to the activation of Ras in F. tularensis-infected cells. Fifteen minutes after infection, Ras activation peaks then declines, indicating that the temporal Ras activation occurs rapidly during early stages of F. tularensis–host cell interaction. Activation of Ras within gastric cells has been shown to be triggered by Helicobacter pylori, which binds the EGF receptor leading to the activation of Ras (Keates et al., 2001). Moreover, Internalin B (InlB), a surface protein of Listeria monocytogenes binds the c-Met receptor leading to Ras activation, which is essential for uptake of L. monocytogenes, and for activation of the PI3K/Akt signalling pathway (Mansell et al., 2001). In contrast, we show that entry of F. tularensis into the host cell is essential to trigger Ras activation and that bacterial attachment to the host cell membrane is not sufficient to trigger Ras activation. Moreover, Ras signalling by F. tularensis is independent of the PI3k/Akt signalling cascade, which is triggered by attached extracellular F. tularensis (Jasinski et al., 2008b). Importantly, entry of F. tularensis ssp. novicida into the host cell is independent of the SOS2, GrB2, or PKCa and bI isoforms factors and Ras activation. We conclude that the temporal activation of Ras by F. tularensis ssp. novicida is not © 2010 Blackwell Publishing Ltd, Cellular Microbiology, 12, 1604–1621

1615

surface receptor-mediated; but is triggered by intracellular bacteria and is essential for intracellular bacterial proliferation, which is novel. A F. tularensis-secreted effector is likely responsible of the activation of Ras. This may be mediated by interacting with some components of the Ras activation pathway such as SOS2 or GrB2. The FPI encodes a type T6SSlike apparatus that plays an essential role in evasion of lysosomal fusion and bacterial escape into the cytosol (Broms et al., 2009; Santic et al., 2010). The IglC protein is encoded by the FPI and in addition to its role in evasion of lysosomal fusion and escape into the cytosol, we show that the IglC protein plays an essential role in triggering Ras activation. Bacterial effectors of various pathogens can interfere with the Ras signalling pathways through distinct mechanisms (Chen et al., 1999; Mimuro et al., 2002). For example, the H. pylori CagA secreted effector interacts with GrB2, which results in the activation of the Ras signalling cascade (Mimuro et al., 2002). Similarly, Salmonella typhimurium activates JNK, a downstream effector of Ras signalling, which is mediated by the effector protein SopE (Chen et al., 1999). Our data clearly show that activation of Ras by F. tularensis is IglCdependent. However, it is still to be determined whether F. tularensis translocates IglC that interacts directly or indirectly with a component of the Ras activation pathway. This mechanism of Ras activation occurs at the early stages of infection of both primary macrophages and the HEK293T cell line. Activation of Ras does not occur if bacterial entry to the cell is blocked, and occurs earlier (~5 min) than bacterial escape to the cytosol (30–60 min) (Santic et al., 2010). These observations exclude the possibility that bacterial escape into the cytosol is required is for Ras activation. The small GTP-binding proteins of the Ras superfamily are master controllers of cell physiology, and their functions range from cell cycle progression, cell morphology, actin rearrangement, endocytosis to intracellular trafficking of vesicles and organelles (Jasinski et al., 2008a,b). Moreover, the Ras activation pathway is involved in promoting survival and suppressing cell death (Jasinski et al., 2008a,b). Cell morphology and actin rearrangement are not affected upon knocking down the Ras activation pathway, which excludes both processes from being involved in Ras signalling by F. tularensis. We have tested two possibilities to unravel the role of Ras activation by F. tularensis in bacterial proliferation within the cytosol. First, our data confirm that the SOS2/GrB2/PKCa/PKCbI quaternary complex does not play roles in evasion of lysosomal fusion or in escape of F. tularensis into the host cytosol. Second, since Ras is essential for cell survival and anti-apoptosis (Jasinski et al., 2008a), we show that an early activation of caspase-3 is exhibited in infected cells in which expression of SOS2, GrB2, PKCa or PKCbI has

1616 S. Al-Khodor and Y. Abu Kwaik been knocked down. This indicates that temporal and early triggering of the Ras activation pathway by F. tularensis plays a role in promoting cell survival during early stages of infection through prevention of caspase-3 activation, which is the executioner of apoptosis (Lai et al., 2001). We conclude that the temporal early activation of Ras by F. tularensis ssp. novicida is essential for intracellular proliferation of F. tularensis ssp. novicida and this is mediated, at least in part, by inhibition of early activation of caspase-3 in the infected cell. The time difference between early Ras activation and its effect in protecting cells from early caspase-3 activation may be due to a time-dependent event that may be triggered upon Ras activation. This may involve activation of other cellular pathways required to block caspase-3 activation, such as triggering expression of anti-apoptotic genes (Santic et al., 2009). The PI3K/Akt signalling pathway is rapidly activated upon infection by F. tularensis, leading to phosphorylation of Akt, which is downregulated by SHIP (Parsa et al., 2006; 2008; Butchar et al., 2007; 2008; Cremer et al., 2009b; 2010; Rajaram et al., 2009). Infection by L. monocytogenes triggers the PI3K/Akt signalling pathway via the surface protein InlB leading to Ras activation, which is required for bacterial entry (Mansell et al., 2001). However, based on four lines of evidence, our data exclude the role of Ras activation in triggering the PI3K/Akt pathway by F. tularensis and show that the role of the Ras activation pathway in bacterial replication is independent of the PI3K/Akt pathway. First, triggering the PI3k/Akt by F. tularensis is mediated by attached extracellular bacteria, while Ras signalling is not triggered by attached extracellular bacteria (Mansell et al., 2001). Second, constitutive expression of Akt or deletion of SHIP promotes phagolysosomal fusion and prevents bacterial escape into the cytosol (Mansell et al., 2001). In contrast, triggering the Ras signalling pathway by F. tularensis does not play any detectable role in evasion of lysosomal fusion or bacterial escape to the cytosol. Third, the role of SHIP in the response of infected cells to F. tularensis is independent of Erk and Ras (Rajaram et al., 2009). Fourth, silencing SOS2, GrB2, PKCa and PKCbI by RNAi blocks activation of Ras by F. tularensis and inhibits intracellular bacterial proliferation, but has no effect on phosphorylation of Akt or Erk. Therefore, triggering the PI3K/Akt signalling pathway or Erk is independent of Ras activation by F. tularensis. In conclusion, our data clearly demonstrate that upon entry into the host cell, intracellular F. tularensis triggers recruitment of the PKCa and PKCbI isoforms to the SOS2/GrB2 complex resulting in activation of the Ras pathway (Fig. 9), which is a novel strategy of triggering Ras signalling by an intracellular pathogen. The SOS2/ GrB2/PKCa/PKCbI quaternary complex is essential for activation of the Ras pathway by F. tularensis, which is essential for bacterial proliferation within the cytosol asso-

ciated with blocking early activation of the pro-apoptotic caspase-3.

Experimental procedures Bacterial strains and cells The F. tularensis ssp. novicida strain U112 and its isogenic mutant iglC have been described previously (Lauriano et al., 2003). The bacterial strain was grown for 2 days on tryptic soy agar (TSA) supplemented with 0.1% cysteine and 10 mg ml-1 kanamycin was supplemented for the mutant. Isolation and preparation of the hMDMs from peripheral blood of volunteers was carried out as previously described (Al-Khodor et al., 2008). The HEK293T cells were maintained as we described previously (Price et al., 2009).

Silencing of SOS2, GrB2 and PCK isoforms in HEK293T cells One day before transfection, 3 ml of 5 ¥ 105 cells per ml of HEK293T were seeded in each well of the six-well plates and 1 ml of the same cell density was seeded on glass coverslip in 24-well plates. In both cases, the wells were treated with 0.2 mg ml-1 poly-L lysine for 30 min at room temperature to increase cell attachment. The SOS2, GrB2, PKCa, bI isoforms RNAi was purchased from Santa Cruz (Santa Cruz, CA). A scrambled RNAi was used as our negative control for silencing. At the time of the transfection, the cells were washed once with the transfection media and processed as recommended by the manufacturers. After 6 h of incubation at 37°C in a CO2 incubator, the growth medium containing 2¥ FBS was added and cells were incubated for additional 18 h. The media were then aspirated and new media containing 1¥ FBS were added to the cells and incubated for additional 24 h. Cells in the six-well plates were than washed in 1 ¥ PBS and lysed using the mammalian protein extraction reagent M-Per (Pierce-Thermo Scientific, Rockford, IL) supplemented with one tablet of the complete mini protease inhibitors EDTA-free cocktail (Roche, Indianapolis, IN). The PNS were boiled using 1¥ reducing buffer (Thermo Scientific, Waltham, MA), separated by SDS-PAGE and transferred onto PVDF membranes (Millipore, Billerica, MA). Western blotting was performed using 1:200 dilution of rabbit polyclonal anti-PKC-a, anti-PKC-bI, anti-SOS2 and anti-GrB2 antisera (Santa Cruz, Santa Cruz, CA), rabbit polyclonal anti-Erk1/2 antiserum (Abacam, Cambridge, MA) that detects total Erk, anti-P42/44 that detected phosphorylated Erk (Cell Signaling, Danvers, MA). Polyclonal rabbit anti-actin antiserum was used at 1:100 dilution (Sigma, St Louis, MO).

Ras activation assay One week before infection hMDMs from volunteers’ blood were seeded in an ultra low attachment plate, then cells were detached after 4 days and 3 ml of 5 ¥ 105 cells per well hMDMs were seeded in six-well plates. One day before infection, 3 ml of 5 ¥ 105 HEK293T cells ml-1 were seeded in six-well plates pre-treated with 0.2 mg ml-1 poly-L lysine for 30 min at room temperature in order to increase cell attachment. © 2010 Blackwell Publishing Ltd, Cellular Microbiology, 12, 1604–1621

Ras activation by F. tularensis

5 -1

PKC βI SOS2

m

Ras-GTP

PKCα GrB2

30-60 min

PI3K

Caspase-3 activation

5-15 min

Akt

in

PKCGrB2 α

5

SHIP

Ras-GDP

PKC βI SOS2

F. tularensis

1617

escape

Apoptosis proliferation

Fig. 9. A model for the Ras activation pathway by intracellular F. tularensis. Within few minutes after bacterial entry into the host cell and biogenesis of the phagosome, F. tularensis ssp. novicida triggers activation of Ras. PKCa and bI isoforms in the cytosol are recruited to the plasma membrane where they bind to the SOS2/GrB2 complex which in turn activates Ras to the GTP-bound form. Activation of Ras through SOS2/GrB2/PKCa-bI is essential for bacterial proliferation. The Ras activation pathway by F. tularensis ssp. novicida inhibits early activation of caspase-3 in the infected cells. The Ras activation pathway is independent of evasion of lysosomal fusion and bacterial escape to the cytosol. The major PI3K/Akt and the Ras activation signalling pathways are independently triggered by F. tularensis ssp. novicida.

Both untreated and RNAi-treated cells were infected with the F. tularensis ssp. novicida strain U112 or its iglC isogenic mutant for the indicated time points using moi of 10. When the infection was performed longer than 1 h, 50 mg ml-1 gentamicin was added to the cells for 1 h in order to kill extracellular bacteria. Where indicated, cells were pre-treated with 1 mg ml-1 Cytochalasin-D or 0.1% DMSO for 45 min at 37°C prior to infection. For Formalinkilled bacteria, the bacteria were killed by exposure to 3.7% formalin for 30 min at room temperature prior to infection. After the indicated times of infection, the medium was carefully removed and cells were rinsed once with ice-cold PBS. Both uninfected and infected cells, mock-treated cells or cells treated with RNAi were washed with ice-cold 1¥ PBS then lysed with 1¥ lysis/assay buffer (Ras activation assay from Cell Biolabs, San Diego, CA). To detect Ras activation, the agarose beads coated with Raf1 Ras-binding domain (RBD) were added to the PNS incubated at 4°C for 1 h with gentle mixing. Beads were then washed and after boiling the beads, the proteins were eluted and subjected to Western blot probed with the anti-Ras mouse monoclonal antibody (1:1000) (Cell Biolabs). The GTPgS was used as a positive control.

washed in 1 ¥ PBS and lysed using the mammalian protein extraction reagent M-Per (Pierce-Thermo Scientific, Rockford, IL). Cell lysates were centrifuged for 10 min at 13 000 g and the supernatant was collected and treated with one tablet of the complete mini EDTA-free protease inhibitor cocktail (Roche, Indianapolis, IN). Approximately, 1 ml of the PNS (250 mg proteins) of both WT and FK-infected cells were incubated overnight at 4°C with 20 ml (equivalent to 4 mg) of rabbit anti-PKC-a, rabbit anti-PKC-bI, rabbit anti-SOS2 or rabbit anti-GrB2 antisera (Santa Cruz Biotechnology, Santa Cruz, CA). The PNS from uninfected cells incubated without antibodies were used as negative control. A total of 100 ml of immobilized G protein resin slurry (PierceThermo Scientific, Rockford, IL) was added to the PNS and incubated with gentle end-to-end mixing for additional 2 h at room temperature. Five hundred microlitres of Tris buffer saline was added to the mixture and the beads were recovered and washed three times followed by centrifugation at 2500 g for 2–3 min. After the final wash, the complexes were processed for Western blot. The immunoprecipitated proteins were examined by immunoblotting with the rabbit polyclonal anti-PKC-a, anti-PKC-bI, anti-SOS2, anti-GrB2 and anti-Shc antisera (Santa Cruz) and re-probed with the mouse monoclonal anti-actin antibody.

Co-immunoprecipitation A total of 3 ¥ 106 HEK293T cells in 3 ml were seeded in six-well plates and infected by the WT strain U112 or the formalin-killed (FK) F. tularensis for 15 min using moi of 10. Cells were then © 2010 Blackwell Publishing Ltd, Cellular Microbiology, 12, 1604–1621

Confocal laser scanning microscopy After confirming specific gene silencing by Western blot, cell viability was determined by trypan blue exclusion. Untreated

1618 S. Al-Khodor and Y. Abu Kwaik 5 ¥ 105 HEK293T cells and successfully silenced cells seeded on coverslips in 24-well plates, cells were infected with F. tularensis ssp. novicida for 1 h (unless otherwise indicated) using moi of 10. After infection, the cells were washed three times with warm culture medium, and 50 mg ml-1 gentamicin was added for additional 1 h to kill extracellular bacteria. After three washes with DMEM, infected cells were incubated for the indicated time points after infection. The infected cells were fixed and permeabilized with ice-cold methanol for 5 min, then blocked in 3% BSA for 1 h. To label bacteria, goat anti-F. tularensis antibody (1:4000) was used followed by the anti-goat secondary antibody conjugated to Alexa fluor-488 (Molecular Probes Invitrogen, Carlsbad, CA). The anti-LAMP-2 (H4B4) 1:2000 mouse monoclonal antibody (developed by J.T. August and J.E.K. Hildreth) was obtained from the Developmental Studies Hybridoma Bank (University of Iowa, IA), mAbs anti-Cathepsin-D (1:200) was obtained from BD transduction (San Jose, CA). Anti-mouse secondary antibodies conjugated to Alexa fluor-555 were obtained from Molecular Probes. Colocalization of the FCP with LAMP-2 and Cathepsin-D was performed as we described previously (Santic et al., 2005a; 2008). To detect activation of caspase-3 in the infected cells, anti-active caspase-3 rabbit polyclonal antiserum (1:200) (Cell Signaling, Danvers, MA) was used, followed by the donkey anti-rabbit secondary antibody conjugated to Alexa fluor-555 (Molecular Probes Invitrogen, Carlsbad, CA). As a negative control, uninfected HEK293T cells were mock-treated with 0.1% DMSO. As a positive control for caspase-3 activation, HEK293T cells were pre-treated with 1 mM Staurosporine (Sigma) for 18 h, as previously described (Nakamura et al., 2007). After fixation, cells were labelled and processed for confocal microscopy as we described previously (Santic et al., 2005b). The images were captured using the Fluoview FV-1000 confocal microscope and presented in the figures as a single z section after being processed using the Adobe illustrator software.

Quantification of bacterial escape into the cytosol To quantify escape of F. tularensis from its phagosome into the host cytosol, we performed the fluorescence-based integrity assay, as described previously (Checroun et al., 2006; Santic et al., 2008). Briefly, untreated or RNAi-treated HEK293T cells in 24-well plate were infected with the WT strain or the iglC mutant. At 2 h after infection, cells were washed once with KHM buffer (110 mM potassium acetate/20 mM Hepes/2 mM MgCl2, pH 7.3), and their plasma membranes were selectively permeabilized and processed as described previously (Checroun et al., 2006; Santic et al., 2008). The images were captured using the Fluoview FV-1000 confocal microscope, and presented in the figures as a single z section after being processed using the Adobe illustrator software.

Detection of Akt phosphorylation To study phosphorylation of Akt during infection by F. tularensis in the RNAi-silenced cells, untreated or RNAi-treated HEK293T cells were infected with an moi of 100 for 5 min. Uninfected and infected cells were lysed in M-Per buffer treated with the Complete EDTA-free protease inhibitor cocktail. PNS were processed by Western blot using rabbit polyclonal anti-pAkt antiserum

(1:1000) (Cell Signaling, Danvers, MA). To ensure equal loading, the membrane was re-probed with the rabbit polyclonal anti-Akt antibody (1:200) (Santa Cruz, Santa Cruz, CA).

Data and statistical analyses All experiments have been performed three times, the standard deviation was calculated, and statistically significant difference is indicated by asterisk in the bar graphs in the figures. Statistical analyses were performed using the two-tail Student’s t-test and GraphPad Prism-5 software.

Acknowledgements Y.A.K. is supported by Public Health Service Awards R01AI43965 and R01AI069321 from NIAID and by the commonwealth of Kentucky Research Challenge Trust Fund.

References Akimana, C., Al-khodor, S., and Abu Kwaik, Y. (2010) Host factors required for modulation of phagosome biogenesis and proliferation of Francisella tularensis within the cytosol. PLoS ONE 5: e11025. Al-Khodor, S., Price, C.T., Habyarimana, F., Kalia, A., and Abu Kwaik, Y. (2008) A Dot/Icm-translocated ankyrin protein of Legionella pneumophila is required for intracellular proliferation within human macrophages and protozoa. Mol Microbiol 70: 908–923. Altman, A., and Deckert, M. (1999) The function of small GTPases in signaling by immune recognition and other leukocyte receptors. Adv Immunol 72: 1–101. Ancrile, B.B., O’Hayer, K.M., and Counter, C.M. (2008) Oncogenic ras-induced expression of cytokines: a new target of anti-cancer therapeutics. Mol Interv 8: 22–27. Asare, R., and Abu Kwaik, Y. (2010) Molecular complexity orchestrates modulation of phagosome biogenesis and escape to the cytosol of macrophages by Francisella tularensis. Environ Microbiol (in press). doi: 10.1111/j.14622920.2010.02229.x Asare, R., Akimana, C., Jones, S., and Abu Kwaik, Y. (2010) Molecular bases of proliferation of Francisella tularensis in arthropod vectors. Environ Microbiol (in press). doi: 10.1111/j.1462-2920.2010.02230.x Barker, J.R., Chong, A., Wehrly, T.D., Yu, J.J., Rodriguez, S.A., Liu, J., et al. (2009) The Francisella tularensis pathogenicity island encodes a secretion system that is required for phagosome escape and virulence. Mol Microbiol 74: 1459–1470. Boguski, M.S., and McCormick, F. (1993) Proteins regulating Ras and its relatives. Nature 366: 643–654. Broms, J.E., Lavander, M., and Sjostedt, A. (2009) A conserved alpha-helix essential for a type VI secretion-like system of Francisella tularensis. J Bacteriol 191: 2431– 2446. de Bruin, O.M., Ludu, J.S., and Nano, F.E. (2007) The Francisella pathogenicity island protein IglA localizes to the bacterial cytoplasm and is needed for intracellular growth. BMC Microbiol 7: 1. © 2010 Blackwell Publishing Ltd, Cellular Microbiology, 12, 1604–1621

Ras activation by F. tularensis Butchar, J.P., Rajaram, M.V., Ganesan, L.P., Parsa, K.V., Clay, C.D., Schlesinger, L.S., and Tridandapani, S. (2007) Francisella tularensis induces IL-23 production in human monocytes. J Immunol 178: 4445–4454. Butchar, J.P., Cremer, T.J., Clay, C.D., Gavrilin, M.A., Wewers, M.D., Marsh, C.B., et al. (2008) Microarray analysis of human monocytes infected with Francisella tularensis identifies new targets of host response subversion. PLoS ONE 3: e2924. Byun, J.Y., Kim, M.J., Yoon, C.H., Cha, H., Yoon, G., and Lee, S.J. (2009) Oncogenic Ras signals through activation of both phosphoinositide 3-kinase and rac1 to induce c-Jun NH2-terminal kinase-mediated, caspase-independent cell death. Mol Cancer Res 7: 1534–1542. Campbell, S.L., Khosravi-Far, R., Rossman, K.L., Clark, G.J., and Der, C.J. (1998) Increasing complexity of Ras signaling. Oncogene 17: 1395–1413. Champion, M.D., Zeng, Q., Nix, E.B., Nano, F.E., Keim, P., Kodira, C.D., et al. (2009) Comparative genomic characterization of Francisella tularensis strains belonging to low and high virulence subspecies. PLoS Pathog 5: e1000459. Checroun, C., Wehrly, T.D., Fischer, E.R., Hayes, S.F., and Celli, J. (2006) Autophagy-mediated reentry of Francisella tularensis into the endocytic compartment after cytoplasmic replication. Proc Natl Acad Sci USA 103: 14578–14583. Chen, L.M., Bagrodia, S., Cerione, R.A., and Galan, J.E. (1999) Requirement of p21-activated kinase (PAK) for Salmonella typhimurium-induced nuclear responses. J Exp Med 189: 1479–1488. Clemens, D.L., Lee, B.Y., and Horwitz, M.A. (2004) Virulent and avirulent strains of Francisella tularensis prevent acidification and maturation of their phagosomes and escape into the cytoplasm in human macrophages. Infect Immun 72: 3204–3217. Clemens, D.L., Lee, B.Y., and Horwitz, M.A. (2005) Francisella tularensis enters macrophages via a novel process involving pseudopod loops. Infect Immun 73: 5892–5902. Cremer, T.J., Ravneberg, D.H., Clay, C.D., Piper-Hunter, M.G., Marsh, C.B., Elton, T.S., et al. (2009a) MiR-155 induction by F. novicida but not the virulent F. tularensis results in SHIP down-regulation and enhanced proinflammatory cytokine response. PLoS ONE 4: e8508. Cremer, T.J., Amer, A., Tridandapani, S., and Butchar, J.P. (2009b) Francisella tularensis regulates autophagy-related host cell signaling pathways. Autophagy 5: 125–128. Cremer, T.J., Ravneberg, D.H., Clay, C.D., Piper-Hunter, M.G., Marsh, C.B., Elton, T.S., et al. (2010) MiR-155 induction by F. novicida but not the virulent F. tularensis results in SHIP down-regulation and enhanced pro-inflammatory cytokine response. PLoS ONE (in press). Darling, R.G., Catlett, C.L., Huebner, K.D., and Jarrett, D.G. (2002) Threats in bioterrorism. I: CDC category A agents. Emerg Med Clin North Am 20: 273–309. Egan, S.E., Giddings, B.W., Brooks, M.W., Buday, L., Sizeland, A.M., and Weinberg, R.A. (1993) Association of Sos Ras exchange protein with Grb2 is implicated in tyrosine kinase signal transduction and transformation. Nature 363: 45–51. Feig, L.A. (1994) Guanine-nucleotide exchange factors: a family of positive regulators of Ras and related GTPases. Curr Opin Cell Biol 6: 204–211. © 2010 Blackwell Publishing Ltd, Cellular Microbiology, 12, 1604–1621

1619

Feig, L.A., and Schaffhausen, B. (1994) Signal transduction. The hunt for Ras targets. Nature 370: 508–509. Forsman, M., Sandström, G., and Sjöstedt, A. (1994) Analysis of 16S ribosomal DNA sequences of Francisella strains and utilization for determination of the phylogeny of the genus and for identification of strains by PCR. Int J Syst Bacteriol 44: 38–46. Gavrilin, M.A., Bouakl, I.J., Knatz, N.L., Duncan, M.D., Hall, M.W., Gunn, J.S., and Wewers, M.D. (2006) Internalization and phagosome escape required for Francisella to induce human monocyte IL-1beta processing and release. Proc Natl Acad Sci USA 103: 141–146. Goddette, D.W., and Frieden, C. (1986) Actin polymerization. The mechanism of action of cytochalasin D. J Biol Chem 261: 15974–15980. Gray, C.G., Cowley, S.C., Cheung, K.K., and Nano, F.E. (2002) The identification of five genetic loci of Francisella novicida associated with intracellular growth. FEMS Microbiol Lett 215: 53–56. Honma, M., Higuchi, O., Shirakata, M., Yasuda, T., Shibuya, H., Iemura, S., et al. (2006) Dok-3 sequesters Grb2 and inhibits the Ras-Erk pathway downstream of proteintyrosine kinases. Genes Cells 11: 143–151. Jasinski, P., Zwolak, P., Terai, K., and Dudek, A.Z. (2008a) Novel Ras pathway inhibitor induces apoptosis and growth inhibition of K-ras-mutated cancer cells in vitro and in vivo. Transl Res 152: 203–212. Jasinski, P., Welsh, B., Galvez, J., Land, D., Zwolak, P., Ghandi, L., et al. (2008b) A novel quinoline, MT477: suppresses cell signaling through Ras molecular pathway, inhibits PKC activity, and demonstrates in vivo anti-tumor activity against human carcinoma cell lines. Invest New Drugs 26: 223–232. Kawakami, Y., Kitaura, J., Yao, L., McHenry, R.W., Kawakami, Y., Newton, A.C., et al. (2003) A Ras activation pathway dependent on Syk phosphorylation of protein kinase C. Proc Natl Acad Sci USA 100: 9470–9475. Keates, S., Sougioultzis, S., Keates, A.C., Zhao, D., Peek, R.M., Jr, Shaw, L.M., and Kelly, C.P. (2001) cag+ Helicobacter pylori induce transactivation of the epidermal growth factor receptor in AGS gastric epithelial cells. J Biol Chem 276: 48127–48134. Keim, P., Johansson, A., and Wagner, D.M. (2007) Molecular epidemiology, evolution, and ecology of Francisella. Ann N Y Acad Sci 1105: 30–66. Lai, X.H., Golovliov, I., and Sjostedt, A. (2001) Francisella tularensis induces cytopathogenicity and apoptosis in murine macrophages via a mechanism that requires intracellular bacterial multiplication. Infect Immun 69: 4691– 4694. Lai, X.H., Golovliov, I., and Sjostedt, A. (2004) Expression of IglC is necessary for intracellular growth and induction of apoptosis in murine macrophages by Francisella tularensis. Microb Pathog 37: 225–230. Lauriano, C.M., Barker, J.R., Nano, F.E., Arulanandam, B.P., and Klose, K.E. (2003) Allelic exchange in Francisella tularensis using PCR products. FEMS Microbiol Lett 229: 195– 202. Lauriano, C.M., Barker, J.R., Yoon, S.S., Nano, F.E., Arulanandam, B.P., Hassett, D.J., and Klose, K.E. (2004) MglA regulates transcription of virulence factors necessary for

1620 S. Al-Khodor and Y. Abu Kwaik Francisella tularensis intraamoebae and intramacrophage survival. Proc Natl Acad Sci USA 101: 4246–4249. Lowenstein, E.J., Daly, R.J., Batzer, A.G., Li, W., Margolis, B., Lammers, R., et al. (1992) The SH2 and SH3 domaincontaining protein GRB2 links receptor tyrosine kinases to ras signaling. Cell 70: 431–442. Mansell, A., Khelef, N., Cossart, P., and O’Neill, L.A. (2001) Internalin B activates nuclear factor-kappa B via Ras, phosphoinositide 3-kinase, and Akt. J Biol Chem 276: 43597–43603. Meadows, K.N., Bryant, P., and Pumiglia, K. (2001) Vascular endothelial growth factor induction of the angiogenic phenotype requires Ras activation. J Biol Chem 276: 49289– 49298. Mimuro, H., Suzuki, T., Tanaka, J., Asahi, M., Haas, R., and Sasakawa, C. (2002) Grb2 is a key mediator of Helicobacter pylori CagA protein activities. Mol Cell 10: 745–755. Nakamura, R.E., Hunter, D.D., Yi, H., Brunken, W.J., and Hackam, A.S. (2007) Identification of two novel activities of the Wnt signaling regulator Dickkopf 3 and characterization of its expression in the mouse retina. BMC Cell Biol 8: 52. Pammit, M.A., Raulie, E.K., Lauriano, C.M., Klose, K.E., and Arulanandam, B.P. (2006) Intranasal vaccination with a defined attenuated Francisella novicida strain induces gamma interferon-dependent antibody-mediated protection against tularemia. Infect Immun 74: 2063–2071. Parsa, K.V., Ganesan, L.P., Rajaram, M.V., Gavrilin, M.A., Balagopal, A., Mohapatra, N.P., et al. (2006) Macrophage pro-inflammatory response to Francisella novicida infection is regulated by SHIP. PLoS Pathog 2: e71. Parsa, K.V., Butchar, J.P., Rajaram, M.V., Cremer, T.J., and Tridandapani, S. (2008) The tyrosine kinase Syk promotes phagocytosis of Francisella through the activation of Erk. Mol Immunol 45: 3012–3021. Pechous, R.D., McCarthy, T.R., and Zahrt, T.C. (2009) Working toward the future: insights into Francisella tularensis pathogenesis and vaccine development. Microbiol Mol Biol Rev 73: 684–711. Plyte, S., Majolini, M.B., Pacini, S., Scarpini, F., Bianchini, C., Lanfrancone, L., et al. (2000) Constitutive activation of the Ras/MAP kinase pathway and enhanced TCR signaling by targeting the Shc adaptor to membrane rafts. Oncogene 19: 1529–1537. Price, C.T., Al-Khodor, S., Al-Quadan, T., Santic, M., Habyarimana, F., Kalia, A., and Kwaik, Y.A. (2009) Molecular mimicry by an F-box effector of Legionella pneumophila hijacks a conserved polyubiquitination machinery within macrophages and protozoa. PLoS Pathog 5: e1000704. Rajaram, M.V., Ganesan, L.P., Parsa, K.V., Butchar, J.P., Gunn, J.S., and Tridandapani, S. (2006) Akt/Protein kinase B modulates macrophage inflammatory response to Francisella infection and confers a survival advantage in mice. J Immunol 177: 6317–6324. Rajaram, M.V., Butchar, J.P., Parsa, K.V., Cremer, T.J., Amer, A., Schlesinger, L.S., and Tridandapani, S. (2009) Akt and SHIP modulate Francisella escape from the phagosome and induction of the Fas-mediated death pathway. PLoS ONE 4: e7919. Robin, P., Boulven, I., Bole-Feysot, C., Tanfin, Z., and Leiber, D. (2004) Contribution of PKC-dependent and -independent processes in temporal ERK regulation by

ET-1, PDGF, and EGF in rat myometrial cells. Am J Physiol Cell Physiol 286: C798–C806. Rotin, D., Honegger, A.M., Margolis, B.L., Ullrich, A., and Schlessinger, J. (1992) Presence of SH2 domains of phospholipase C gamma 1 enhances substrate phosphorylation by increasing the affinity toward the epidermal growth factor receptor. J Biol Chem 267: 9678–9683. Santic, M., Molmeret, M., and Abu Kwaik, Y. (2005a) Modulation of biogenesis of the Francisella tularensis subsp. novicida-containing phagosome in quiescent human macrophages and its maturation into a phagolysosome upon activation by IFN-gamma. Cell Microbiol 7: 957–967. Santic, M., Molmeret, M., Klose, K.E., Jones, S., and Abu Kwaik, Y. (2005b) The Francisella tularensis pathogenicity island protein IglC and its regulator MglA are essential for modulating phagosome biogenesis and subsequent bacterial escape into the cytoplasm. Cell Microbiol 7: 969– 979. Santic, M., Molmeret, M., Barker, J.R., Klose, K.E., Dekanic, A., Doric, M., and Abu Kwaik, Y. (2007) A Francisella tularensis pathogenicity island protein essential for bacterial proliferation within the host cell cytosol. Cell Microbiol 9: 2391–2403. Santic, M., Asare, R., Skrobonja, I., Jones, S., and Abu Kwaik, Y. (2008) Acquisition of the vacuolar ATPase proton pump and phagosome acidification are essential for escape of Francisella tularensis into the macrophage cytosol. Infect Immun 76: 2671–2677. Santic, M., Pavokovic, G., Jones, S., Asare, R., and Kwaik, Y.A. (2009) Regulation of apoptosis and anti-apoptosis signalling by Francisella tularensis. Microbes Infect 12: 126– 134. Santic, M., Al-Khodor, S., and Abu Kwaik, Y. (2010) Cell biology and molecular ecology of Francisella tularensis. Cell Microbiol 12: 129–139. Satoh, T., and Kaziro, Y. (1992) Ras in signal transduction. Semin Cancer Biol 3: 169–177. Satoh, T., Nakafuku, M., and Kaziro, Y. (1992) Function of Ras as a molecular switch in signal transduction. J Biol Chem 267: 24149–24152. Telepnev, M., Golovliov, I., Grundstrom, T., Tarnvik, A., and Sjostedt, A. (2003) Francisella tularensis inhibits Toll-like receptor-mediated activation of intracellular signalling and secretion of TNF-alpha and IL-1 from murine macrophages. Cell Microbiol 5: 41–51. Telepnev, M., Golovliov, I., and Sjostedt, A. (2005) Francisella tularensis LVS initially activates but subsequently down-regulates intracellular signaling and cytokine secretion in mouse monocytic and human peripheral blood mononuclear cells. Microb Pathog 38: 239–247. Tempel, R., Lai, X.H., Crosa, L., Kozlowicz, B., and Heffron, F. (2006) Attenuated Francisella novicida transposon mutants protect mice against wild-type challenge. Infect Immun 74: 5095–5105. Warner, L.C., Hack, N., Egan, S.E., Goldberg, H.J., Weinberg, R.A., and Skorecki, K.L. (1993) RAS is required for epidermal growth factor-stimulated arachidonic acid release in rat-1 fibroblasts. Oncogene 8: 3249–3255. Wehrly, T.D., Chong, A., Virtaneva, K., Sturdevant, D.E., Child, R., Edwards, J.A., et al. (2009) Intracellular biology and virulence determinants of Francisella tularensis © 2010 Blackwell Publishing Ltd, Cellular Microbiology, 12, 1604–1621

Ras activation by F. tularensis revealed by transcriptional profiling inside macrophages. Cell Microbiol 11: 1128–1150. Wickstrum, J.R., Bokhari, S.M., Fischer, J.L., Pinson, D.M., Yeh, H.W., Horvat, R.T., and Parmely, M.J. (2009) Francisella tularensis induces extensive caspase-3 activation and apoptotic cell death in the tissues of infected mice. Infect Immun 77: 4827–4836.

Supporting information Additional Supporting Information may be found in the online version of this article: Fig. S1. Colocalization of Cathepsin-D with the FCP. A. Representative confocal microscopy images of colocalization of the lysosomal marker Cathepsin-D (red) with the

© 2010 Blackwell Publishing Ltd, Cellular Microbiology, 12, 1604–1621

1621

phagosomes containing F. tularensis ssp. novicida (green). Untreated (Un) HEK293T cells or treated with the RNAi-negative control or the gene-specific RNAi for 48 h were infected with the WT strain U112 or formalin-killed (FK) bacteria using moi of 10 for 1 h followed by 1 h treatment with gentamicin to kill extracellular bacteria. The iglC mutant was used as a positive control. B. Quantitative analysis of colocalization of the WT straincontaining phagosomes with Cathepsin-D at 30 min and 2 h after infection. At least 100 cells were analysed from different coverslips. This experiment was performed three times and the results are representative of one experiment. Please note: Wiley-Blackwell are not responsible for the content or functionality of any supporting materials supplied by the authors. Any queries (other than missing material) should be directed to the corresponding author for the article.