Skip to main content
Advertisement
  • Loading metrics

Host Cell Rap1b mediates cAMP-dependent invasion by Trypanosoma cruzi

  • Gabriel Ferri,

    Roles Formal analysis, Methodology, Validation, Writing – original draft

    Affiliations CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina, Laboratorio de Biología Molecular de Trypanosomas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos, Ciudad de Buenos Aires, Argentina

  • Daniel Musikant,

    Roles Formal analysis, Investigation

    Affiliations CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina, Laboratorio de Biología Molecular de Trypanosomas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos, Ciudad de Buenos Aires, Argentina

  • Martin M. Edreira

    Roles Conceptualization, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Supervision, Writing – original draft, Writing – review & editing

    mme2@pitt.edu

    Affiliations CONICET-Universidad de Buenos Aires, IQUIBICEN, Ciudad de Buenos Aires, Argentina, Laboratorio de Biología Molecular de Trypanosomas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos, Ciudad de Buenos Aires, Argentina

Abstract

Trypanosoma cruzi cAMP-mediated invasion has long been described, however, the detailed mechanism of action of the pathway activated by this cyclic nucleotide still remains unknown. We have recently demonstrated a crucial role for Epac in the cAMP-mediated invasion of the host cell. In this work, we gathered evidence indicating that the cAMP/Epac pathway is activated in different cells lines. In accordance, data collected from pull-down experiments designed to identify only the active form of Rap1b (Rap1b-GTP), and infection assays using cells transfected with a constitutively active mutant of Rap1b (Rap1b-G12V), strongly suggest the participation of Rap1b as mediator of the pathway. In addition to the activation of this small GTPase, fluorescence microscopy allowed us to demonstrate the relocalization of Rap1b to the entry site of the parasite. Moreover, phospho-mimetic and non-phosphorylable mutants of Rap1b were used to demonstrate a PKA-dependent antagonistic effect on the pathway, by phosphorylation of Rap1b, and potentially of Epac. Finally, Western Blot analysis was used to determine the involvement of the MEK/ERK signalling downstream of cAMP/Epac/Rap1b-mediated invasion.

Author summary

As an obligate intracellular parasite, T. cruzi needs to gain access to the host cell cytosol, where replication and differentiation takes place. The process of invasion is mediated by the activation of different signaling pathways in the host. Activation of cAMP signaling has been implicated in invasion, with high intracellular levels of cAMP associated with an increase in invasion. In this regard, we have recently shown that Epac (Exchange protein activated by cAMP), and not PKA, mediates the effect of the cyclic nucleotide on parasite internalization. In this work, in order to shed some light into the detailed mechanism of invasion mediated by cAMP/Epac, we have explored the activation of downstream effectors of this pathway during host cell infection. Our results established that Rap1b, a known effector of Epac, and MEK/ERK, are downstream effectors of the pathway. In addition, we found a PKA-dependent inhibition of infection, presumably by phosphorylation of Rap1b, and potentially of Epac.

Introduction

As an obligate intracellular parasite, Trypanosoma cruzi replicates in the cytoplasm of infected mammalian host cells. The ability to infect host cells depends on the stage of the parasite, the strain and the DTU (Discrete Typing Units), as the result of the expression of a differential repertoire of surface/secreted molecules, that activates different signalling pathways in the host cell. In general, the attachment of trypomastigotes activates several hosts signalling pathways, including the elevation of intracellular cAMP levels in the host cell [1]. It has been shown that invasion involves the recruitment and fusion of lysosomes to the entry site [2], and that cAMP potentiates the Ca2+-dependent exocytosis of lysosomes and lysosome-mediated cell invasion [3]. Although a transient increase of Ca2+ and the recruitment of lysosomes are common features in the invasion of metacyclic trypomastigotes (MTs) and tissue culture-derived trypomastigotes (TCTs) [4,5], the signalling pathways that these parasites promote in the host cell are different. Among them, the activation of cAMP-mediated signalling by TCTs is a poorly studied process. It has been previously demonstrated that the pharmacologic intervention of the cAMP pathway was able to modulate parasite invasion [4,6,7]. To determine the specific role of cAMP main effectors, PKA and Epac, in T. cruzi invasion, we used a set of pharmacological tools to selectively activate or inhibit these proteins. Whereas differential activation of PKA had no effect on invasion, a significant increase in invasion was observed in cells treated with a cAMP analogue that selectively and exclusively activates Epac [7]. Accordingly, the inhibition of Epac by ESI-09 [8] showed a significant decrease in invasion. Unexpectedly, specific inhibition of PKA also showed a positive effect on invasion, suggesting a PKA/Epac crosstalk during the process of invasion [7]. In this regard, it has been described that both proteins can be recruited to the same microdomain through the association with radixin [9,10], an ERM structural protein that attaches the plasma membrane to the cortical actin cytoskeleton [11]. Respectively, confocal studies have shown that ERM proteins are associated with the invasion site of extracellular amastigotes (EAs), where colocalize with F-actin [12]. Moreover, a link between radixin and the cAMP/Epac-dependent pathway during TCT invasion was confirmed by blocking host cell invasion with a permeable version of 15-mer sequence (stearate-KPRACSYDLLLEHQRP-amide peptide) corresponding to the minimal Epac1 ERM binding domain. This peptide displaces the Epac protein from its association with radixin and delocalized it from the microdomain. Under these conditions, the percentage of invasion is similar to that obtained when the Epac protein is inhibited by ESI-09 [7]. Taken together, these results clearly established a crucial role for Epac in the cAMP-mediated invasion of the host cell.

However, downstream effectors involved in this pathway are still unknown. Epac1 has been involved in PI3K/Akt and MEK/ERK pathways [13,14], and members of these pathways, including Rap1, were localized at late endosomes/lysosomes [15]. In cardiomyocytes, the cAMP/Epac/Rap1 pathway modulates the excitation-contraction mechanism by stimulating Ca2+ release through ryanodine receptors (RyR) [16]. In smooth muscle cells, Rap1 inhibits RhoA activity and promotes Ca2+ desensitization and smooth muscle relaxation [17]. Rap1 activation also induces muscle hyperpolarization by decreasing Ca2+ influx by inducing the opening of Ca2+-sensitive K+ channels, generating a boost in vasodilation [18]. In addition, Rap1 was shown to modulate mitogen-activated kinases (MAPKs), like extracellular signal-regulated kinase (ERK1/2), inducing the stimulation or inhibition of these kinases depending on the cell type. More recently, the role of Rap1 in ERK phosphorylation and activation in smooth muscle was demonstrated [19]. Ral-GDS, an effector of Rap1, promotes cardiomyocyte autophagy [20], and the downstream effector of Ral-GDS, RalB, binds specific subunits of the exocytosis machinery and mediates activation of autophagosome assembly [21].

Smooth muscle and heart are the most important target organs for T. cruzi infection and persistence during the chronic phase of Chagas disease. Taking into account that Epac has a critical role in cAMP-mediated invasion and the regulation of various cAMP-dependent functions in smooth muscle and heart, possibly modulating the intracellular concentration of Ca2+ through the activation of Rap1 and the participation of ERK1/2 [18,22,23], deciphering the detailed functioning of the cAMP/Epac pathway would provide a deeper insight into the host cell invasion mechanisms mediated by this cyclic nucleotide. In this work, we investigated the involvement of two known effectors, Rap1b and ERK, as potential mediators in the cAMP/Epac-dependent invasion by T. cruzi and the role of PKA-dependent Rap1b phosphorylation.

Materials and methods

Cells and parasites

NRK (ATCC CRL-6509), VERO (ATCC CCL-81) and HELA (ATCC CCL-2) cell lines were cultured in DMEM medium supplemented with Glutamax (Gibco), 10% (v/v) FBS (Natocor), 100 U/ml penicillin and 0.1 mg/ml streptomycin (Sigma), and maintained at 37°C in a 5% CO2 atmosphere. The HL-1 cell line [24] was cultured in a gelatin/fibronectin matrix (5 μg fibronectin / 0.02% gelatin (m/v)-Sigma) and Claycomb culture medium supplemented with Glutamax(Gibco), 10% (v/v) FBS, 100 U/ml penicillin, 0.1 mg/ml streptomycin and 0.1 mM norepinephrine (Sigma). Tissue culture-derived trypomastigotes forms (TCT) of T. cruzi Y strain were routinely maintained in VERO cells cultured in DMEM supplemented with 4% FBS and penicillin/streptomycin. Trypomastigotes were obtained from supernatants of infected VERO cells by centrifugation. First, the supernatant conditioned medium was centrifuged at low speed (500 g) to remove intact cells and cell debris Then, the supernatant obtained was centrifugated at 3,000 g for 15 min. and the pellet with the parasites was washed in PBS three times.

Invasion assay

Cells were grown on glass cover slides in a 24 multi-well plate with DMEM 10% FBS for 24 hours at 2x104 cells/well density at 37°C, 5% CO2 and incubated with: 37,5 μM of the Epac1 inhibitor ESI-09 (Sigma); 300 μM of 8-Br-cAMP (Biolog); 50 μM of the MEK1/2 inhbitor PD98059; or 0,1% DMSO as a control condition. Cells were then washed and infected with trypomastigotes of the Y strain (moi 100:1) for 2 hours. Parasite were removed and cells incubated for 48 hs. Cells were fixed, stained with DAPI and infection level determined by fluorescence microscopy. Percentage of infection (calculated as the (#Infected cells/total counted cells)*100) and amastigotes/100 cells were calculated counting 3,000 cells, expressed as mean ± SD of three or more independent experiments and performed in triplicate. Infection of non-treated cells was considered as basal infection.

Host cell transfection

A transient transfection protocol with polyethyleneimine (PEI) was used [25]. Briefly, cells were grown at about 60% confluence and incubated at 37°C in a 5% CO2, 95% humidified air environment. Next day, cells were transfected with pCGN empty vector (EMPTY), pCGN-HA-Rap1b (HA-Rap1) or HA-Rap1b mutants (G12V, S179A, S179D, or combinations) (kindly provided by Dr D. Altschuler, University of Pittsburgh, USA) using a ratio of 4:1 PEI:DNA mix in OptiMEM medium (Gibco). The mixture was kept for 30 min. at room temperature and then added to the cells and incubated at 37 C and 5% CO2. After 24h, cells were washed with PBS and complete medium (DMEM or Claycomb 10% FBS) was added. The transfected cells were used at 24h post-transfection.

Trypomastigote release assay

HL-1 cells were seeded on a 24-well plate at a concentration of 7000 cells/mL in Claycomb medium supplemented with 10% FBS. After 24 hours, cells were infected and treated as described above. 72 hours later, medium was replaced with fresh prepared treatments until trypomastigotes were observed under microscope at six days post infection (pi). Supernatants were transferred to a new plate, washed to avoid mammalian cell contamination and a solution of resazurin sodium salt was added as a fluorogenic oxidation-reduction indicator (final concentration 0.1 mM). After 3 hours of incubation, fluorescence was measured with a FLUOstar OPTIMA (BMG LABTECH) microplate reader at 590 nm (excitation: 570 nm). Baseline corrected values of fluorescence were normalized to the negative control. Results are expressed as mean ± SD of three or more independent experiments and performed in triplicate.

GST Pull-down

Detection of active Rap1 (GTP-bound) was performed through pull-down assays using a recombinant GST-RBD protein (GST fusion to the Rap1b-binding domain of the RalGDS protein, which only recognizes active Rap). A total of 1 mL bacteria lysates containing GST or GST-RBD were mixed by rotation with 40 μl 50% GSH-Sepharose at 4°C for 1 h. The beads were centrifuged at 800 g for 2 min. at 4°C and washed with lysis buffer. Lysates from HA-Rap1 transfected cells pre-treated for 2h with 8Br-cAMP, infected with trypomastigotes of the Y strain (Tp Y) or mock infected (Ctrl) were incubated with RBD-glutathione-agarose resin for 1h at 4°C. Resin was washed and eluted with cracking buffer for WB analysis.

Western Blot

After electrophoresis, the gel was equilibrated in 25 mM Trizma base, 192 mM L-1 glycine and 20% v/v methanol pH 8.3. Then, proteins were transferred to previously hydrated with methanol PVDF membranes (Amersham Hybond, GE Healthcare) in a vertical tank (Mini-PROTEAN Tetra Cell, Bio-Rad). After transfer, membranes were blocked with 20 mM L−1 Tris-HCl, 500 mM NaCl, 0.05% Tween and 5% non-fat milk, pH 7.5, incubated with anti-GST (Genscript), anti-p44/42 MAPK (ERK1/2, Cell Signaling), anti-phospho-p44/42 MAPK (pERK1/2, Cell Signaling) or anti-GAPDH (Santa Cruz Biotechnology) antibodies. After incubation, membrane was washed and incubated with rabbit horseradish peroxidase (HRP)-IgGs antibody (Santa Cruz Biotechnology), washed again and then revealed using 0.88 mg/ml luminol, 0.066 mg/ml p-coumaric acid, 6 mM H2O2; 100 mM Tris-HCl, pH 8.8 solution. Chemiluminescence was recorded with the C-DiGit scanner (LI-COR), and bands intensity were quantified with ImageJ and ImageLab 6.1 (Bio-Rad) software.

ERK phosphorylation

Cells were treated with or without PD98059 for 2h and incubated with trypomastigotes of the Y strain for 30 min., treated with 750 μM H2O2 for 5 min. or mock infected. Then, cells were lysed and cracking buffer added for WB analysis.

Indirect immunofluorescence assay

Cells were adhered to glass previously treated with 40 μg/ml of poly-D-lysine (Sigma), fixed with PBS-PFA 4% (Sigma), washed with PBS and incubated with NH4Cl for 15 min. Then, were permeabilized with 0.2% Triton-x100 and incubated with anti-RAP1 antibody (Genscript) at 4°C. After 16 h, washed with PBS, incubated with mouse anti-IgG (H+L) anti-conjugated to Alexa Fluor594 antibody (Jackson InmunoResearch), and nuclei stained with DAPI. Finally, glasses were mounted on slides with FluorSave mounting solution (Merk Millipore). Preparations were analysed in a Nikon Eclipse E600 fluorescence microscope.

Results

cAMP/Epac activation as a ubiquitous mechanism of invasion in T. cruzi

The crucial role of Epac during invasion by T. cruzi was recently described in NRK cells [7]. In order to assess the ubiquity of the cAMP/Epac pathway, other cell lines were used in invasion assays. Similar to what happened in NRK cells [4,7], high levels of cAMP induced by a non-hydrolysable permeable analogue of cAMP, 8-Br-cAMP (8-Bromoadenosine 3′,5′-cyclic monophosphate) (Biolog), positively modulated invasion in both HELA and HL-1 cells (Fig 1). Consistent with this result, specific pharmacological inhibition of Epac by ESI-09 (Sigma), resulted in a significant decrease in invasion in both cell lines (Fig 1).

thumbnail
Fig 1. cAMP/Epac pathway is required for T. cruzi invasion in different cell lines.

Pre-treated NRK, HELA or HL-1 cells (30 min at 300 μM of 8-Br-cAMP or 37.5 μM of ESI-09) were infected with trypomastigotes from T. cruzi Y strain (100:1 parasite to cell ratio for 2 h). 48 hs post-infection cells were fixed, stained with DAPI and percentage of infection determined by fluorescence microscopy. Infection of untreated cells was considered as basal infection. Results are expressed as mean ± SD (n ≥ 3). **** p<0.0001, *** p <0.001, ** p <0.01; ANOVA and Dunnett’s post-test.

https://doi.org/10.1371/journal.pntd.0011191.g001

Rap1b as a mediator of the cAMP/Epac-dependent invasion

The participation of Rap1b as mediator of the cAMP/Epac-dependent invasion was evaluated by pull-down experiments using agarose bound GST-RalGDS Rap-binding domain (GST-RBD), designed to pull down only the active form of Rap1b (Rap1b-GTP). Briefly, cells transfected with HA-Rap1b were incubated in the presence of DMSO (vehicle), 8-Br-cAMP, ESI-09 or trypomastigotes of Y strain of T. cruzi for 2 h. Cells were lysed and lysates used in pull-down experiments (Figs 2 and S1 and S2). As shown in Fig 2, higher levels of activated GTP-bound Rap1 were detected in lysates from cells incubated with 8-Br-cAMP and trypomastigotes, while inhibition of Epac by ESI-09 reduced Rap1b activation as expected (S2 Fig), supporting the involvement of Rap1b in cAMP-mediated invasion.

thumbnail
Fig 2. Rap1b pull-down assays.

A) HA-Rap1 transfected NRK, HELA or HL-1 cells were incubated for 2 h with 8-Br-cAMP, infected with trypomastigotes from T. cruzi Y strain or mock infected (DMSO). Then, cells were lysed and pull-down assay with glutathione-agarose resin performed for 1h at 4°C. Resin was washed and eluted with cracking buffer for WB analysis. B) Bands were quantified and normalized against the input using ImageJ cell software. Results are expressed as mean ± SD (n≥3). * p<0.05, ** p <0.005, One-way ANOVA–Dunnett´s multiple comparison test.

https://doi.org/10.1371/journal.pntd.0011191.g002

In accordance with these results, when HELA and HL-1 cells transfected with a constitutively active mutant of Rap1b, Rap1b-G12V, in which a single point substitution, glycine-to-valine at codon 12 of Rap1b, a significant increase in infection was observed when compared with the control (Fig 3A–3D, respectively). Noteworthy, when the complete invasion-differentiation-release cycle was evaluated in HL-1 cells overexpressing Rap1b-G12V (Fig 3E), trypomastigotes released into the medium showed similar results than the results obtained for percentage of infected cells and amastigotes/100cells (Fig 3C and 3D), suggesting the cAMP/Epac/Rap1b pathway would play a role in the early steps of the establishment of the infection, as previously hypothesized [7]. In addition to these results suggesting that Rap1b-GTP is required as a mediator of the cAMP/Epac1 pathway during the invasion by T. cruzi, fluorescence microscopy in HL-1 (Figs 4 and S3) and Hela cells (S4 Fig), revealed the relocalization of Rap1b, reflected as an increase in the fluorescence intensity of Rap1b, to the site of entry of T. cruzi, supporting the hypothesis that Rap1b needs to be activated and properly localized in the entry site.

thumbnail
Fig 3. Invasion assays.

Rap1 transfected Hela and HL-1 cells were infected with trypomastigotes from T. cruzi Y strain (100:1 parasite to cell ratio for 2 h). 48 hs post-infection cells were fixed, stained with DAPI and percentage of invasion determined by fluorescence microscopy. Infection of untreated cells was considered as basal infection. Percentage of infection (calculated as the (#Infected cells/total counted cells)*100) and amastigotes/100 cells were calculated counting 3,000 cells, results are expressed as mean ± SD (n ≥ 3), ** p <0.01, * p <0.1; ANOVA and Dunnett’s post-test. In the case of the trypomastigote release assay, HL-1 cells were infected and treated as described above. 72 hours later, medium was replaced with fresh prepared treatments until trypomastigotes were observed under microscope at six days post infection (pi). Supernatants were transferred to a new plate and quantification of trypomastigotes was performed with resazurin method. Results are expressed as mean ± SD (n ≥ 3). **** p<0.0001, t student test.

https://doi.org/10.1371/journal.pntd.0011191.g003

thumbnail
Fig 4. Parasite-Rap1 colocalization.

For immunofluorescence, HL-1 cells were infected for 5 to 15 min with (Tp Y) trypomastigotes from T. cruzi Y strain (20:1 parasite to cell ratio) or mock infected (Control) and then fixed and incubated with primary antibody against Rap1 protein and a secondary antibody conjugated to Alexa594. Photos were taken with a fluorescence microscope.

https://doi.org/10.1371/journal.pntd.0011191.g004

Role of PKA-dependent Rap1b phosphorylation

While the specific activation of PKA had no effect on invasion, an increase in internalized parasites was observed as a result of PKA inhibition [7]. Therefore, under physiological conditions, PKA-mediated phosphorylation would negatively regulate the cAMP/Epac pathway of invasion. The inhibition of the Epac-mediated invasion pathway could be achieved, at least, at two different levels: through direct phosphorylation of Epac or at the level of Rap1, an Epac effector and a known target for PKA-mediated phosphorylation [26]. Since trypomastigotes released (Fig 3E), showed similar results than the percentage of infected cells and amastigotes/100cells (Fig 3C and 3D), we used this more reliable technique, in comparison to cell counting in the microscope, to evaluate the role of Rap1b phosphorylation. HL-1 cells overexpressing phospho-mimetic (S179D) or phospho-deficient (S179A) Rap1b mutants were infected and trypomastigotes released at day 6 pi were counted using resazurin method [27]. As shown in Fig 5, cells transfected with the phospho-mimetic Rap1b-S179D mutant presented a decrease in the number of released trypomastigotes, with respect to control cells or cells overexpressing the non-phosphorylable mutant Rap1b-S179A, supporting a PKA-dependent antagonistic effect on the pathway, as previously described in NRK cells [7]. Interestingly, the effect of phosphorylation could be reverted by transfecting cells with the double mutant G12V/S179D, a constitutive active phospho-mimetic Rap1, opening the possibility of a two-level regulation of PKA on the Epac/Rap1 pathway.

thumbnail
Fig 5. PKA-dependent phosphorylation effect on parasite release.

Transfected HL-1 cells were infected and treated as described above. 72 hours later, medium was replaced with fresh prepared treatments until trypomastigotes were observed under microscope at six days post infection (pi). Supernatants were transferred to a new plate and quantification of trypomastigotes was performed with resazurin method. Infection of cells transfected with pCGN empty vector was considered as basal infection. Results are expressed as mean ± SD (n ≥ 3), *** p <0.001, ** p <0.01; ANOVA and Dunnett’s post-test.

https://doi.org/10.1371/journal.pntd.0011191.g005

MEK/ERK as a downstream effector of cAMP/Epac-mediated invasion of T. cruzi

In order to elucidate the involvement of MEK/ERK in the cAMP-dependent invasion, activation of ERK1/2 was analysed by Western Blot. An increase in ERK1/2 phosphorylation in both NRK (Fig 6A) and HL-1 (S5 Fig) cells was observed during the host cell infection. Interestingly, parasite-induced ERK phosphorylation was diminished by the addition of ESI-09 (S6 Fig). To determine whether the activation of ERK1/2 modulates the infection levels, cells pre-treated with the MEK1/2 kinase inhibitor PD98059 were infected with the parasite. In accordance, the inhibition of ERK phosphorylation produced a significant decrease in infection (Fig 6B). MEK/ERK could be independently activated or a downstream effector of Epac/Rap1. The fact that the inhibition of MEK or Epac induced a similar decrease in invasion, and no additive or synergic effects were observed when both proteins were simultaneously inhibited (Fig 6C), suggests that MEK/ERK is a downstream effector of cAMP/Epac1/Rap1b-mediated invasion.

thumbnail
Fig 6. ERK phosphorylation.

A) NRK cells were incubated for 2 h with trypomastigotes from T. cruzi Y strain (Tp Y), treated with 750 μM H2O2 for 5 min (positive control) or mock infected (Control). Then, cells were lysed and cracking buffer added for WB analysis. Representative WB performed on NRK cells is shown. B) and C) Invasion assay. Pretreated NRK cells were infected with trypomastigotes from T. cruzi Y strain (100:1 parasite to cell ratio) for 2 h, washed and incubated with fresh 10% FBS DMEM medium for 48 hs. Then, cells were fixed, stained with DAPI and percentage of infection determined by fluorescence microscopy. Infection of untreated cells was considered as basal infection. Results are expressed as mean ± SD (n ≥ 3) *** p <0.001, ** p <0.01; ANOVA and Dunnett’s post-test.

https://doi.org/10.1371/journal.pntd.0011191.g006

Discussion

Infection of cell cultures shown to be a useful model to study host-cell interaction and invasion [28], however, it has to be considered that T. cruzi invasion showed to be a complex process just taking into account the different stages of the parasite with the ability to infect host cells. In addition, despite of being able to infect any nucleated cell in vitro, it has been shown in vivo that T. cruzi exhibits a certain cellular tropism [29] and that the signalling pathways activated in the host cell differ according to the stage of the parasite [30]. This complexity is even higher when considering different DTUs, strains and the repertoire of surface/secreted molecules that activates different signalling pathways in the host cell [1]. Additionally, differences in cell recognition and invasion, when performing assays in different cell lines, should be also consider [31]. With all these contemplations, and when it is clear that the possibility of replicating human biology in a dish is quite limited, in vitro models are an essential tool to dissect the phenomenon of parasite-host cell interaction and invasion.

In this context, it has been reported that the activation of cAMP-mediated signalling pathways triggers Ca2+-dependent lysosomal exocytosis and promotes host cell invasion by T. cruzi [4]. The Ca2+ release from intracellular compartments, such as the endoplasmic reticulum, is associated with an increase in intracellular levels of cAMP. In mammalian cells, cAMP downstream effectors, PKA and Epac, are involved in Ca2+-activated exocytosis events [32]. Furthermore, members of these pathways, including Rap1, have been localized to late endosomes/lysosomes [15], and Epac-mediated activation of Rap1 has been identified in regulated exocytosis in human sperm [33], insulin secretion [34], and pancreatic amylase release [35]. It was previously shown that Epac1-mediated signalling represents the main mechanism for cAMP-mediated invasion by T. cruzi [7]. In addition, ERM proteins, which are essential for the function and architecture of the cell cortex by linking the plasma membrane to the underlying actin cytoskeleton [11], have been associated with the invasion of EAs [12]. Moreover, in confocal studies, it was shown that ERM proteins are recruited at the entry site of the parasites where they colocalize with F-actin, while its depletion inhibits HELA cells invasion [12]. Remarkably, one of its members, radixin, was identified as a scaffold unit for cAMP effectors in the spatial regulation of Epac1/Rap1-mediated signalling [9,10]. In this regard, we have previously revealed a link between Epac1 and radixin in the cAMP-mediated invasion of TCTs, by blocking the invasion of NRK cells with a permeable peptide of 15 amino acids that binds to the minimal ERM-binding domain of Epac [7]. In order to elucidate the role of cAMP downstream effectors involved in T. cruzi invasion, we evaluated the activation of the cAMP/Epac pathway by TCTs of Y strain in NRK, HELA and HL-1 cell lines. NRK cells are normal fibroblasts from rat kidney, originally used in the establishment of cAMP as a modulator of invasion events [4] and to demonstrate the participation of Epac1 as the main effector of this modulation [7]. On the other hand, HELA cells are epithelial human cervix cells that have been widely used in invasion assays [3638] and HL-1 cells, previously used in invasion assays, as well [39], are cardiomyocytes from mouse heart, one of the most important target organs in the infection and persistence of T. cruzi. Our data showed that the activation of the cAMP/Epac pathway by TCTs occurs regardless of the origin (rat, mouse, human) or the cell type (kidney, cervix, heart) that the parasite is invading. In addition, we investigated the role of Rap1b during the cAMP/Epac1-mediated invasion. Rap1b, a GTPase of the Ras family, is known to integrate Epac- and/or PKA-dependent events to achieve an efficient cAMP signal transduction [18,40,41]. Pull-down assays were used to detect higher levels of activated GTP-bound Rap1 in lysates from infected cells. Likewise, as shown in Fig 3, cells transfected with the constitutively active form of Rap1b (G12V) were more susceptible to infection, compared to the control. However, it is important to note that due to the fact that cells are constitutively overexpressing Rap1b (G12V), and cells were incubated for 48 hs after invasion in order to achieve sensitive in the parasite count, an effect of the overexpression of this small GTPase on parasite replication could not be excluded, and needs to be further explored. In addition to Rap1b activation, and in accordance to our hypothesis, data obtained from fluorescence microscopy assays evidenced the recruitment of Rap1b to the parasite entry site. Interestingly, when studying PKA participation using a specific inhibitors of this kinase, it was observed that the invasion levels of TCTs increased compared to the control [7], suggesting a PKA-dependent antagonist effect. This effect could be mediated by PKA phosphorylation of the effectors of the cAMP pathway, such as Epac and Rap1b. PKA-dependent phosphorylation at S179 of Rap1b has long been established [42]. Results presented in this work support the antagonistic effect of PKA through, at least, Rap1b phosphorylation, since trypomastigote release was affected in cells transfected with phospho-mimetic Rap1b-S179D, with respect to control cells and cells overexpressing Rap1b-S179A, the non-phosphorylable version of Rap1b. In line with these observations, it has been shown that Rap1b phosphorylation destabilizes the association of this protein with the plasma membrane and promotes Rap1b inactivation [43,44]. Nevertheless, as mentioned above, the experiments were carried out in transfected cells, and an effect of the overexpressed proteins on parasite replication could not be ruled out.

Overall, our data suggest that activation and relocalization of Rap1b are required as a mediator of the cAMP/Epac1 pathway during the TCT infection. In this scenario, the fact that PKA negative regulation on infection was abrogated in the presence of the constitutively active G12V mutation, suggests that Rap1b is required in the phosphorylated and inactive form to completely abolish the cAMP/Epac/Rap1b pathway of infection.

It has been described that the MEK/ERK pathway can be activated or inhibited by cAMP [45]. Furthermore, the activation of this pathway participates in the invasion of T. cruzi by way of the interaction of the host cell with parasite surface molecules, such as TS [46], Tc85 [47] or TSSAII [48]. Also, Rap1 is associated with the phosphorylation and activation of ERK1/2 in smooth muscle [19]. Accordingly, our data revealed that TCTs induce ERK1/2 phosphorylation in mammalian cells and ERK1/2 activation modulates the invasion of these parasites as a downstream effector of Epac/Rap1-mediated invasion.

Although the transient increase in cytosolic Ca2+ concentration and lysosome recruitment that occur during invasion are characteristics shared between MTs and TCTs [4,5], the signalling pathways triggered by both forms of parasites in the host cell are different. In TCT invasion, ERK1/2 activation is a distinctive feature that is mediated by Ca2+-dependent lysosomal exocytosis through the regulation of F-actin and the activation of the focal adhesion kinase (FAK) [49]. During MT invasion, in contrast, PKC promotes Ca2+ release from inositol 3-phosphate (IP3)-sensitive compartments through the binding of the surface glycoprotein gp82 to LAMP-2 receptors [30,50]. On the contrary, the activity of PKC is not required for the invasion of TCTs in NRK cells, since treatment with PKC inhibitors did not affect the response to Ca2+ or the reorganization of F-actin, and has no effect on parasite internalization [51]. The divergence between the signalling pathways triggered by MTs and TCTs might be associated with the fact that the internalization of TCT is initiated by an invagination of the plasma membrane [52], in a lysosomal exocytosis-dependent process induced by a membrane injury and the following activation of the PMR mechanism [53]. These mechanisms lead to changes that take place through the inhibition of the Rho/Rho signaling pathway by PKA [54,55]. The fact that RhoA promotes actin polymerization but has a negative effect on EAs internalization during HELA cell invasion [56] and that Rap1b inhibits RhoA/ROCK activity in the muscle smooth tissue [57], suggest the hypothesis that the cAMP/Epac1/Rap1b signalling pathway could be activated in the first steps of the invasion by T. cruzi, promoting Ca2+-dependent lysosomal exocytosis and the reorganization of the cytoskeleton. Once the parasite is inside the cell, a PKA-mediated inhibition of Epac/Rap1b might be necessary for the parasite retention. In accordance, our results showed that Rap1b seems to be associated with the plasma membrane at the parasite entry site where it could be required during the internalization process and PKA had an antagonistic effect, probably through the phosphorylation of the S179 of Rap1b.

In this work, we have gathered evidence strongly suggesting that the cAMP/Epac/Rap1b/ERK pathway is activated during the early steps of host cell infection and that it would be negatively regulated by PKA, possibly through the phosphorylation of Epac and/or Rap1b. Importantly, a detailed characterization of effectors involved in T. cruzi invasion would provide an attractive set of new therapeutic targets for the repositioning or the development of new antiparasitic drugs, since there is a large variety of therapies that target cAMP-mediated signalling [58].

Supporting information

S1 Fig. Densitometry analysis.

Chemiluminescence was recorded with the C-DiGit scanner (LI-COR), and bands were quantified and normalized against the input using ImageJ and ImageLab 6.1 (Bio-Rad) software. The normalization was performed following the “Western Blot Normalization Using Image Lab Software” guide. Results are expressed as mean ± SD (n≥3). * p<0.05, ** p <0.005, One-way ANOVA–Dunnett´s multiple comparison test.

https://doi.org/10.1371/journal.pntd.0011191.s001

(PDF)

S2 Fig. Rap1b pull-down assays.

A) HA-Rap1 transfected HELA cells were incubated for 2 h 37.5uM ESI-09 or 0.1% DMSO. Then, cells were lysed and pull-down assay with glutathione-agarose resin performed for 1 h at 4°C. Resin was washed and eluted with cracking buffer for WB analysis. B) Bands were quantified and normalized against the input using ImageJ cell software. Results are expressed as mean ± SD (n≥3). ** p<0.01, t student test.

https://doi.org/10.1371/journal.pntd.0011191.s002

(TIFF)

S3 Fig. Immunofluorescence of HL-1 cells.

A) Cells were infected for 5 to 15 min with (Tp Y) trypomastigotes from T. cruzi Y strain or mock infected (Control) and then fixed and incubated with primary antibody against Rap1 protein and a secondary antibody conjugated to Alexa594. B) Line profiles obtained from the line shown in the Rap1 quadrant. Photos were taken with a fluorescence microscope. Only a representative image is shown. Scale bar: 10 μm.

https://doi.org/10.1371/journal.pntd.0011191.s003

(PDF)

S4 Fig.

A) HeLa cells were incubated with trypomastigotes and the percentage of infected cells with Rap1 positive signal at the site of parasite entry were quantified at 5 and 20 minutes. Quantification was performed counting at least 30 infected cells of each time point in 3 independent experiments. * p < 0.01 (t test). B) Immunofluorescence of HL-1 cells were infected for 20 min with trypomastigotes from T. cruzi Y strain (Tp Y) (20:1 parasite to cell ratio) or mock infected (Control) and then fixed and incubated with primary antibody against Rap1 protein and a secondary antibody conjugated to Alexa594. Photos were taken with a fluorescence microscope.

https://doi.org/10.1371/journal.pntd.0011191.s004

(TIF)

S5 Fig. Raw data ERK phosphorylation.

Upper) NRK cells were incubated for 2 h with trypomastigotes from T. cruzi Y strain (Tp Y), treated with 750 μM H2O2 for 5 min (positive control) or mock infected (Control). Then, cells were lysed and cracking buffer added for WB analysis. Lower) HL-1 cells were incubated for 2 h with trypomastigotes from T. cruzi Y strain (Tp Y) or mock infected (Control). Then, cells were lysed and cracking buffer added for WB analysis.

https://doi.org/10.1371/journal.pntd.0011191.s005

(PDF)

S6 Fig.

A) HeLa cells were pretreated with H2O2 (750 mM, 5 min) or ESI-09 (37.5mM, 30 min), then cells were washed and incubated with trypomastigotes for 2 hours. Cells were washed and lyzed. WB with ERK, P-ERK or GAPDH antibodies were performed. B) Erk and P-Erk band density were normalized to GAPDH, then P-Erk expression was relativized to total Erk. Band density were quantified using ImageJ. * p < 0.01 (t test).

https://doi.org/10.1371/journal.pntd.0011191.s006

(TIFF)

References

  1. 1. Ferri G, Edreira MM. All Roads Lead to Cytosol: Trypanosoma cruzi Multi-Strategic Approach to Invasion. Front Cell Infect Microbiol [Internet]. 2021;11:89. Available from: https://www.frontiersin.org/article/10.3389/fcimb.2021.634793
  2. 2. Andrews NW. Lysosome recruitment during host cell invasion by Trypanosoma cruzi. Trends Cell Biol. 1995/03/01. Dept of Cell Biology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA.; 1995;5:133–7. pmid:14732170
  3. 3. Rodríguez A, Samoff E, Rioult MG, Chung A, Andrews NW. Host cell invasion by trypanosomes requires lysosomes and microtubule/kinesin-mediated transport. J Cell Biol. 1996;134:349–62. pmid:8707821
  4. 4. Rodriguez A, Martinez I, Chung A, Berlot CH, Andrews NW. cAMP regulates Ca2+-dependent exocytosis of lysosomes and lysosome-mediated cell invasion by trypanosomes. J Biol Chem. 1999/06/08. Departments of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, 06520, USA.; 1999;274:16754–9. pmid:10358016
  5. 5. Martins RM, Alves RM, Macedo S, Yoshida N. Starvation and rapamycin differentially regulate host cell lysosome exocytosis and invasion by Trypanosoma cruzi metacyclic forms. Cell Microbiol. 2011;13:943–54. pmid:21501360
  6. 6. Fernandes AB, Neira I, Ferreira AT, Mortara RA. Cell invasion by Trypanosoma cruzi amastigotes of distinct infectivities: studies on signaling pathways. Parasitol Res. 2006/06/23. Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Sao Paulo-UNIFESP, Escola Paulista de Medicina, Rua Botucatu, 862 6th Floor, 04023–062, Sao Paulo, Brazil.; 2006;100:59–68. pmid:16791632
  7. 7. Musikant D, Ferri G, Durante IM, Buscaglia CA, Altschuler DL, Edreira MM. Host Epac1 is required for cAMP-mediated invasion by Trypanosoma cruzi. Mol Biochem Parasitol [Internet]. Elsevier B.V.; 2017;211:67–70. Available from: pmid:27984073
  8. 8. Enserink JM, Christensen AE, De Rooij J, Van Triest M, Schwede F, Genieser HG, et al. A novel Epac-specific cAMP analogue demonstrates independent regulation of Rap1 and ERK. Nat Cell Biol. 2002;4:1–6.
  9. 9. Gloerich M, Ponsioen B, Vliem MJ, Zhang Z, Zhao J, Kooistra MR, et al. Spatial Regulation of Cyclic AMP-Epac1 Signaling in Cell Adhesion by ERM Proteins. Mol Cell Biol. 2010;30:5421–31. pmid:20855527
  10. 10. Hochbaum D, Barila G, Ribeiro-Neto F, Altschuler DL. Radixin Assembles cAMP Effectors Epac and PKA into a Functional cAMP Compartment. J Biol Chem [Internet]. 2010/11/05. Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.; 2011;286:859–66. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0021925820542749
  11. 11. McClatchey AI. ERM proteins at a glance. J Cell Sci. 2014;127:3199–204. pmid:24951115
  12. 12. Ferreira ÉR, Bonfim-Melo A, Cordero EM, Mortara RA. ERM proteins play distinct roles in cell invasion by extracellular amastigotes of Trypanosoma cruzi. Front Microbiol. 2017;8:1–11.
  13. 13. Gündüz D, Troidl C, Tanislav C, Rohrbach S, Hamm C, Aslam M. Role of PI3K/Akt and MEK/ERK Signalling in cAMP/Epac-Mediated Endothelial Barrier Stabilisation. Front Physiol. 2019;10:1–13.
  14. 14. Baviera AM, Zanon NM, Navegantes LCC, Kettelhut IC. Involvement of cAMP/Epac/PI3K-dependent pathway in the antiproteolytic effect of epinephrine on rat skeletal muscle. Mol Cell Endocrinol. 2010;315:104–12. pmid:19804812
  15. 15. Pizon V, Desjardins M, Bucci C, Parton RG, Zerial M. Association of Rap1a and Rap1b proteins with late endocytic/phagocytic compartments and Rap2a with the Golgi complex. J Cell Sci. 1994/06/01. INSERM U-248, Faculte de Medecine Lariboisiere-Saint Louis, Paris, France.; 1994;107:1661–70. pmid:7962206
  16. 16. Oestreich EA, Malik S, Goonasekera SA, Blaxall BC, Kelley GG, Dirksen RT, et al. Epac and phospholipase Cε regulate Ca2+ release in the heart by activation of protein kinase Cε and calcium-calmodulin kinase II. J Biol Chem. 2009;284:1514–22.
  17. 17. Zieba BJ, Artamonov M V, Jin L, Momotani K, Ho R, Franke AS, et al. The cAMP-responsive Rap1 Guanine Nucleotide Exchange Factor, Epac, Induces Smooth Muscle Relaxation by Down-regulation of RhoA Activity. J Biol Chem [Internet]. Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22908, USA.; 2011;286:16681–92. Available from: http://europepmc.org/abstract/MED/21454546
  18. 18. Kosuru R, Chrzanowska M. Integration of rap1 and calcium signaling. Int J Mol Sci. 2020;21:1–21. pmid:32120817
  19. 19. Li Q, Teng Y, Wang J, Yu M, Li Y, Zheng H. Rap1 promotes proliferation and migration of vascular smooth muscle cell via the ERK pathway. Pathol—Res Pract [Internet]. Elsevier; 2018;214:1045–50. Available from: pmid:29789158
  20. 20. Rifki OF, Bodemann BO, Battiprolu PK, White MA, Hill JA. RalGDS-dependent cardiomyocyte autophagy is required for load-induced ventricular hypertrophy. J Mol Cell Cardiol. Department of Internal Medicine, Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.; 2013;59:128–38. pmid:23473774
  21. 21. Bodemann BO, Orvedahl A, Cheng T, Ram RR, Ou Y-H, Formstecher E, et al. RalB and the Exocyst Mediate the Cellular Starvation Response by Direct Activation of Autophagosome Assembly. Cell [Internet]. 2011;144:253–67. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0092867410014364 pmid:21241894
  22. 22. Ruiz-Hurtado G, Morel E, Domínguez-Rodríguez A, Llach A, Lezoualc’h F, Benitah J-P, et al. Epac in cardiac calcium signaling. J Mol Cell Cardiol [Internet]. Elsevier Ltd; 2013;58:162–71. Available from: pmid:23220153
  23. 23. Lezoualc’H F, Fazal L, Laudette M, Conte C. Cyclic AMP sensor EPAC proteins and their role in cardiovascular function and disease. Circ Res. 2016;118:881–97. pmid:26941424
  24. 24. Claycomb WC, Lanson NAJ, Stallworth BS, Egeland DB, Delcarpio JB, Bahinski A, et al. HL-1 cells: a cardiac muscle cell line that contracts and retains phenotypic characteristics of the adult cardiomyocyte. Proc Natl Acad Sci U S A. 1998;95:2979–84. pmid:9501201
  25. 25. Longo PA, Kavran JM, Kim MS, Leahy DJ. Transient mammalian cell transfection with polyethylenimine (PEI). Methods Enzymol. Academic Press Inc.; 2013. p. 227–40. pmid:24011049
  26. 26. Edreira MM, Li S, Hochbaum D, Wong S, Gorfe AA, Ribeiro-Neto F, et al. Phosphorylation-induced Conformational Changes in Rap1b. J Biol Chem [Internet]. 2009;284:27480–6. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0021925820384180
  27. 27. Rolón M, Vega C, Escario JA, Gómez-Barrio A. Development of resazurin microtiter assay for drug sensibility testing of Trypanosoma cruzi epimastigotes. Parasitol Res [Internet]. 2006;99:103–7. Available from: http://link.springer.com/10.1007/s00436-006-0126-y pmid:16506080
  28. 28. Breyner NM, Hecht M, Nitz N, Rose E, Carvalho JL. In vitro models for investigation of the host-parasite interface—possible applications in acute Chagas disease. Acta Trop [Internet]. 2020;202:105262. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0001706X19310824 pmid:31706861
  29. 29. Santi-Rocca J, Fernandez-Cortes F, Chillón-Marinas C, González-Rubio ML, Martin D, Gironès N, et al. A multi-parametric analysis of Trypanosoma cruzi infection: Common pathophysiologic patterns beyond extreme heterogeneity of host responses. Sci Rep. 2017;7:1–12.
  30. 30. Maeda FY, Cortez C, Yoshida N. Cell signaling during Trypanosoma cruzi invasion. Front Immunol. Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de Sao Paulo Sao Paulo, Sao Paulo, Brazil.; 2012;3:361. pmid:23230440
  31. 31. Duran-Rehbein GA, Vargas-Zambrano JC, Cuéllar A, Puerta CJ, Gonzalez JM. Mammalian cellular culture models of Trypanosoma cruzi infection: a review of the published literature. Parasite [Internet]. 2014;21:38. Available from: http://www.parasite-journal.org/10.1051/parasite/2014040 pmid:25083732
  32. 32. Seino S, Shibasaki T. PKA-dependent and PKA-independent pathways for cAMP-regulated exocytosis. Physiol Rev. 2005/09/27. Division of Cellular and Molecular Medicine, Kobe University Graduate School of Medicine, Kobe 650–0017, Japan. seino@med.kobe-u.ac.jp; 2005;85:1303–42. pmid:16183914
  33. 33. Miro-Moran A, Jardin I, Ortega-Ferrusola C, Salido GM, Peña FJ, Tapia JA, et al. Identification and function of exchange proteins activated directly by cyclic AMP (Epac) in mammalian spermatozoa. PLoS One. 2012;7:e37713. pmid:22662198
  34. 34. Tengholm A, Gylfe E. cAMP signalling in insulin and glucagon secretion. Diabetes, Obes Metab. 2017;19:42–53. pmid:28466587
  35. 35. Sabbatini ME, Chen X, Ernst SA, Williams JA. Rap1 activation plays a regulatory role in pancreatic amylase secretion. J Biol Chem. 2008;283:23884–94. pmid:18577515
  36. 36. Clemente TM, Cortez C, Novaes A da S, Yoshida N. Surface Molecules Released by Trypanosoma cruzi Metacyclic Forms Downregulate Host Cell Invasion. PLoS Negl Trop Dis. 2016;10:1–18.
  37. 37. Ferreira BL, Ferreira ÉR, Bonfim-Melo A, Mortara RA, Bahia D. Trypanosoma cruzi extracellular amastigotes selectively trigger the PI3K/Akt and Erk pathways during HeLa cell invasion. Microbes Infect. 2019;21:485–9. pmid:31247328
  38. 38. Rodrigues JPF, Souza Onofre T, Barbosa BC, Ferreira ÉR, Bonfim-Melo A, Yoshida N. Host cell protein LAMP-2 is the receptor for Trypanosoma cruzi surface molecule gp82 that mediates invasion. Cell Microbiol. 2019;21:1–11. pmid:30609224
  39. 39. Benatar AF, García GA, Bua J, Cerliani JP, Postan M, Tasso LM, et al. Galectin-1 Prevents Infection and Damage Induced by Trypanosoma cruzi on Cardiac Cells. PLoS Negl Trop Dis. 2015;9:1–23. pmid:26451839
  40. 40. Hochbaum D, Hong K, Barila G, Ribeiro-Neto F, Altschuler DL. Epac, in synergy with cAMP-dependent protein kinase (PKA), is required for cAMP-mediated mitogenesis. J Biol Chem. 2008;283:4464–8. pmid:18063584
  41. 41. Jaśkiewicz A, Pajką B, Orzechowski A. The many faces of rap1 GTPase. Int J Mol Sci. 2018;19:1–20. pmid:30241315
  42. 42. Altschuler D, Lapetina EG. Mutational analysis of the cAMP-dependent protein kinase-mediated phosphorylation site of Rap1b. J Biol Chem [Internet]. © 1993 ASBMB. Currently published by Elsevier Inc; originally published by American Society for Biochemistry and Molecular Biology.; 1993;268:7527–31. Available from: https://doi.org/10.1016/S0021-9258(18)53207-5 pmid:8463283
  43. 43. Ntantie E, Gonyo P, Lorimer EL, Hauser AD, Schuld N, McAllister D, et al. An Adenosine-Mediated Signaling Pathway Suppresses Prenylation of the GTPase Rap1B and Promotes Cell Scattering. Sci Signal [Internet]. 2013;6. Available from: https://www.science.org/doi/10.1126/scisignal.2003374 pmid:23716716
  44. 44. Takahashi M, Dillon TJ, Liu C, Kariya Y, Wang Z, Stork PJS. Protein kinase a-dependent phosphorylation of Rap1 regulates its membrane localization and cell migration. J Biol Chem. 2013;288:27712–23. pmid:23946483
  45. 45. Stork PJ., Schmitt JM. Crosstalk between cAMP and MAP kinase signaling in the regulation of cell proliferation. Trends Cell Biol [Internet]. 2002;12:258–66. Available from: pmid:12074885
  46. 46. Chuenkova M V., Pereira MA. The T. cruzi trans-sialidase induces PC12 cell differentiation via MAPK/ERK pathway. Neuroreport. 2001;12:3715–8. pmid:11726780
  47. 47. Magdesian MH, Tonelli RR, Fessel MR, Silveira MS, Schumacher RI, Linden R, et al. A conserved domain of the gp85/trans-sialidase family activates host cell extracellular signal-regulated kinase and facilitates Trypanosoma cruzi infection. Exp Cell Res. United States; 2007;313:210–8. pmid:17101128
  48. 48. Cánepa GE, Degese MS, Budu A, Garcia CRS, Buscaglia CA. Involvement of TSSA (trypomastigote small surface antigen) in Trypanosoma cruzi invasion of mammalian cells 1. Biochem J. 2012;444:211–8.
  49. 49. Onofre TS, Rodrigues JPF, Yoshida N. Depletion of host cell focal adhesion kinase increases the susceptibility to invasion by trypanosoma cruzi metacyclic forms. Front Cell Infect Microbiol. 2019;9:1–10.
  50. 50. Onofre TS, Rodrigues JPF, Shio MT, Macedo S, Juliano MA, Yoshida N. Interaction of Trypanosoma cruzi Gp82 With Host Cell LAMP2 Induces Protein Kinase C Activation and Promotes Invasion. Front Cell Infect Microbiol. 2021;11:1–16. pmid:33777840
  51. 51. Rodriguez A, Rioult MG, Ora A, Andrews NW. A trypanosome-soluble factor induces IP3 formation, intracellular Ca2+ mobilization and microfilament rearrangement in host cells. J Cell Biol. 1995;129:1263–73. pmid:7775573
  52. 52. Woolsey AM, Sunwoo L, Petersen CA, Brachmann SM, Cantley LC, Burleigh BA. Novel Pl 3-kinase-dependent mechanisms of trypanosome invasion and vacuole maturation. J Cell Sci. 2003;116:3611–22.
  53. 53. Fernandes M, Cortez M, Flannery A, Tam C, Mortara R, Andrews N. Trypanosoma cruzi subverts the sphingomyelinase-mediated plasma membrane repair pathway for cell invasion. J Exp Med. 2011;208:909–21. pmid:21536739
  54. 54. Woolsey AM, Burleigh BA. Host cell actin polymerization is required for cellular retention of Trypanosoma cruzi and early association with endosomal/lysosomal compartments. Cell Microbiol [Internet]. 2004;6:829–38. Available from: https://onlinelibrary.wiley.com/doi/10.1111/j.1462-5822.2004.00405.x pmid:15272864
  55. 55. Mott A, Lenormand G, Costales J, Fredberg JJ, Burleigh BA. Modulation of host cell mechanics by Trypanosoma cruzi. J Cell Physiol. 2009;218:315–22. pmid:18853412
  56. 56. Bonfim-Melo A, Ferreira ÉR, Mortara RA. Rac1/WAVE2 and Cdc42/N-WASP participation in actin-dependent host cell invasion by extracellular amastigotes of Trypanosoma cruzi. Front Microbiol. 2018;9:1–14.
  57. 57. Lakshmikanthan S, Zieba BJ, Ge Z-D, Momotani K, Zheng X, Lund H, et al. Rap1b in Smooth Muscle and Endothelium Is Required for Maintenance of Vascular Tone and Normal Blood Pressure. Arterioscler Thromb Vasc Biol [Internet]. 2014;34:1486–94. Available from: http://atvb.ahajournals.org/cgi/doi/10.1161/ATVBAHA.114.303678 pmid:24790136
  58. 58. Parnell E, Palmer TM, Yarwood SJ. The future of EPAC-targeted therapies: agonism versus antagonism. Trends Pharmacol Sci [Internet]. Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK. School of Pharmacy, University of Bradford, Bradford BD7 1DP, UK. Institute of Molecular, Cell, and Systems B; 2015;36:203–14. Available from: https://linkinghub.elsevier.com/retrieve/pii/S016561471500022X pmid:25744542