#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Exoproteome profiling of Trypanosoma cruzi during amastigogenesis early stages


Authors: Samuel C. Mandacaru aff001;  Rayner M. L. Queiroz aff001;  Marcos R. Alborghetti aff001;  Lucas S. de Oliveira aff001;  Consuelo M. R. de Lima aff001;  Izabela M. D. Bastos aff003;  Jaime M. Santana aff003;  Peter Roepstorff aff002;  Carlos André O. Ricart aff001;  Sébastien Charneau aff001
Authors place of work: Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil aff001;  Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark aff002;  Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil aff003
Published in the journal: PLoS ONE 14(11)
Category: Research Article
doi: https://doi.org/10.1371/journal.pone.0225386

Summary

Chagas disease is caused by the protozoan Trypanosoma cruzi, affecting around 8 million people worldwide. After host cell invasion, the infective trypomastigote form remains 2–4 hours inside acidic phagolysosomes to differentiate into replicative amastigote form. In vitro acidic-pH-induced axenic amastigogenesis was used here to study this step of the parasite life cycle. After three hours of trypomastigote incubation in amastigogenesis promoting acidic medium (pH 5.0) or control physiological pH (7.4) medium samples were subjected to three rounds of centrifugation followed by ultrafiltration of the supernatants. The resulting exoproteome samples were trypsin digested and analysed by nano flow liquid chromatography coupled to tandem mass spectrometry. Computational protein identification searches yielded 271 and 483 protein groups in the exoproteome at pH 7.4 and pH 5.0, respectively, with 180 common proteins between both conditions. The total amount and diversity of proteins released by parasites almost doubled upon acidic incubation compared to control. Overall, 76.5% of proteins were predicted to be secreted by classical or non-classical pathways and 35.1% of these proteins have predicted transmembrane domains. Classical secretory pathway analysis showed an increased number of mucins and mucin-associated surface proteins after acidic incubation. However, the number of released trans-sialidases and surface GP63 peptidases was higher at pH 7.4. Trans-sialidases and mucins are anchored to the membrane and exhibit an enzyme-substrate relationship. In general, mucins are glycoproteins with immunomodulatory functions in Chagas disease, present mainly in the epimastigote and trypomastigote surfaces and could be enzymatically cleaved and released in the phagolysosome during amastigogenesis. Moreover, evidence for flagella discard during amastigogenesis are addressed. This study provides the first comparative analysis of the exoproteome during amastigogenesis, and the presented data evidence the dynamism of its profile in response to acidic pH-induced differentiation.

Keywords:

Membrane proteins – Parasitic diseases – Mucin – Cytoskeletal proteins – Motor proteins – Host cells – Trypanosoma cruzi – Trypomastigotes

Introduction

Trypanosoma cruzi is the etiologic agent of Chagas disease in Central and South America, with more than 10,000 deaths annually worldwide [1]. An increasing number of cases are being reported in non-endemic regions including the United States and Europe due to intense migration of individuals from endemic areas of Latin America [26]. In humans, T. cruzi infection usually develops from an acute phase characterized by high parasitemia and a robust immune response, into a clinically variable chronic phase. In the absence of treatment during the chronic phase, parasite proliferation is highly contained by a humoral and cellular immune response. However the infection remains persistent, particularly in the myocardium and smooth muscle, which may lead to the development of cardiac and digestive complications [7,8]. The treatment of Chagas disease is currently based around chemotherapy, since no effective vaccine is available. Only two drugs are available: nifurtimox and benznidazole. Benznidazole is considered the first-line treatment in most countries due to its effectiveness for the treatment of acute, congenital, reactive and early chronic infections. In many cases, this drug is able to reduce disease progression, but the high toxicity has the potential to cause serious side effects, leading to interruption of patient treatment. Additionally, the low efficacy of the current drugs is low in adult patients with the chronic disease [9,10].

In order to carry out its lifecycle, infective T. cruzi trypomastigotes invade nucleated mammalian host cells and differentiate intracellularly into replicative amastigote forms (amastigogenesis). After internalization, trypomastigotes remain 2–4 h inside acidic phagolysosomes [11], escaping into the host cell cytoplasm to complete its differentiation. To facilitate the escape process, the parasite recruits trans-sialidase proteins and release pore-forming molecules called Tc-TOX [12,13]. It has also been recently reported that T. cruzi infection blocks the expression of host cell immunoproteasome subunits, proteasome activator protein PA28b, TAP1 and MHC class I molecule by an unknown posttranscriptional control [14]. This indicates that trypomastigotes release proteins and/or other molecules in response to processes caused by or following the differentiation induction.

The secreted/excreted protein repertoire (here generically referred as exoproteome) plays important roles in homeostasis, immune response, development, proteolysis, adhesion, cell proliferation, cell differentiation, morphogenesis and cellular communication [15]. Furthermore, secreted/excreted proteins account for approximately 10% of the proteins encoded by a genome [1518]. Trypomastigotes are able to release membranous vesicles filled with virulent factors such as trans-sialidases [19,20]. It is these extracellular vesicles that have been shown to be involved in the pathogenesis of Chagas disease by increasing heart parasitism and inflammation [20].

Classically secreted proteins can be identified by the presence of an N-terminal cleavable signal peptide (SP) that is typically 15–30 amino acids long. Furthermore, a class of secretory proteins, known as leaderless proteins, is exported from the cell without signal sequences through non-classical secretion pathways. For example cell surface shedding and inclusion into exosomes and other secretory vesicles [21], or even release from the plasma membrane through the enzymatic cleavage of their lipid anchor [22].

The acidic milieu is a key step in triggering amastigogenesis and parasite molecular response during this process and has been studied through high-throughput quantitative proteomic and phosphoproteomic approaches [23]. Queiroz and colleagues, analysing intracellular proteins 2 hours after induction, reported the overexpression of several proteins predicted to be secreted, indicating an increase in vesicular traffic. This observation leads us to hypothesize a change in parasite exoproteome repertoire after acidic-pH induction. To address this hypothesis, we evaluate the exoproteome changes of T. cruzi trypomastigote upon the first three hours of acidic-pH-induced axenic amastigogenesis compared to the exoproteome of trypomastigote incubated at physiological pH for the same period.

Materials and methods

Trypomastigote cell culture

Trypomastigotes, Y strain [24], were maintained in monolayers of HeLa cells grown in DMEM supplemented with 5% fetal bovine serum, at pH 7.4, according to [25,26]. The parasites of the outbreak from the 4th to 5th day after infection were carefully collected from the supernatant, consisting of over 98% trypomastigotes [26].

Exoproteome samples

Trypomastigote cells were washed 3 times with DMEM, pH 7.4, without serum, by centrifugation at 2,500 × g for 10 min. Then, 1.0 × 109 washed parasites were resuspended in 5 mL DMEM without serum at pH 7.4 or pH 5.0, (2.0 × 108 cells/mL final concentration) and incubated in a 25 cm2 culture flasks at 37°C for 3 h with gently shaking every 20 min. After incubation, the parasites motility was microscopically monitored and the samples were collected only if ~ 95% of the cells remained mobile [27]. For pH 7.4 we obtained samples in duplicate and for pH 5.0 in triplicate. In order to remove cells following incubation, the medium was centrifuged for 5 min at room temperature in 3 rounds to ensure complete removal of cells and avoid mechanical cell lysis: firstly, at 2,000 × g, then at 4,000 × g and the last at 6,000 × g, with the supernatants transferred to new tubes after each centrifugation. After cell removal, the supernatants containing the exoproteomes were concentrated and buffer exchanged to 20 mM triethylammonium bicarbonate using AmiconTM filter units with 3 kDa cut-off membrane (Millipore, Billerica, MA, USA), dried and stored at -20°C.

Sample preparation for LC-MS/MS

The exoproteome samples were resuspended in 20 mM triethylammonium bicarbonate, reduced with 20 mM dithiothreitol at 56°C for 45 min, alkylated with 40 mM iodoacetamide in the dark at room temperature for 60 min and digested overnight at 37°C with 1 μg modified trypsin (Promega, Madison, USA). After digestion, the sample was acidified to 0.1% trifluoracetic acid (TFA), final concentration, and desalted with homemade microcolumns of Poros Oligo R3 resin (PerSeptive Biosystems, Framingham, USA) packed (1 cm long) in p200 tips (adapted from [28]). Prior to lyophilization, a Biochrom 30 amino acid analyzer (Biochrom, Cambridge, U.K.) was employed to determine peptide concentration according to the manufacturers protocol [29].

LC–MS/MS and data analysis

Samples were analysed by an EASY-nano LC system (Proxeon Biosystems, Odense, Denmark) coupled online to an LTQ-Orbitrap Velos mass spectrometer (Thermo Scientific, Waltham, USA). The exoproteomes at physiological pH and acidic pH were analysed in duplicate and in triplicate, respectively. Two μg of peptides from each fraction were loaded onto an 18 cm fused silica emitter (75 μm inner diameter) manually packed with reverse phase capillary column ReproSil-Pur C18-AQ 3 μm resin (Dr. Maisch GmbH, Germany) and eluted using a gradient from 100% phase A (0.1% formic acid) to 35% phase B (0.1% formic acid, 95% acetonitrile) for 210 min for each sample, 35% to 100% phase B for 5 min and 100% phase B for 8 min in (a total of 223 min at 250 nL/min) [29]. After each run, the column was washed with 90% phase B and re-equilibrated with phase A. Mass spectra were acquired in positive ion mode applying data-dependent automatic survey MS scan and tandem mass spectra (MS/MS) acquisition. Each MS scan in the orbitrap (mass range of m/z of 400–1800 and resolution 60,000) was followed by MS/MS of the seven most intense ions in the LTQ. Fragmentation in the LTQ was performed by HCD and selected sequenced ions were dynamically excluded for 30 s. Raw data were viewed in Xcalibur v.2.1 (Thermo Scientific, Waltham, USA). Data processing was performed using Proteome Discoverer v.1.3 (Thermo Scientific, Waltham, USA). Raw files were generated, and these were searched using Proteome Discoverer with SequestHT algorithm against Trypanosoma cruzi database containing the proteins of the parasite reference proteome database downloaded from UniProt (early 2017). Contaminant proteins (several types of human keratins, BSA and porcine trypsin) were also added to the database and all contaminant proteins identified were manually removed from the result lists. The searches were performed with the following parameters: MS accuracy 10 ppm, MS/MS accuracy 0.5 Da, trypsin digestion with up to 2 missed cleavage allowed, fixed carbamidomethyl modification of cysteine and variable modification of oxidized methionine. The number of proteins, protein groups and number of peptides were filtered for a false discovery rate (FDR) less than 1%; peptides with rank 1 and proteins with at least 3 peptides using Proteome Discoverer. ProteinCenter software (Thermo Scientific, Waltham, USA) was used to generate FASTA formatted files of groups of proteins of interest, GO annotation and statistical analysis between conditions (Fisher's exact test). Improved annotation of the identified proteins was acquired using Blast2GO software (http://www.blast2go.com/b2ghome) using default parameters. SignalP v.4.1 (http://www.cbs.dtu.dk/services/SignalP/) and SecretomeP v.2.0 (http://www.cbs.dtu.dk/services/SecretomeP/) were used to predict proteins secreted by classical and non-classical pathways, respectively. The parameters, eukaryotes/mammal, gram positive and gram negative were set to predict the secretion pathways. The TMHMM algorithm (http://www.cbs.dtu.dk/services/TMHMM/) was used to predict the number of transmembrane helixes in the protein sequences.

Results and discussion

In order to identify proteins secreted by T. cruzi during amastigogenesis, we performed a qualitative exoproteome analysis of trypomastigote in two different conditions. Thus, samples from parasites incubated for 3 hours at pH 7.4 (control) or at pH 5.0 (amastigogenesis) were investigated by shotgun/bottom-up proteomics (Fig 1). The computational analysis of LC-MS/MS data identified 271 and 483 protein groups at pH 7.4 and pH 5.0 respectively, with 180 common protein groups being present in both conditions (Fig 2A; S1 Table).

Experimental setup.
Fig. 1. Experimental setup.
Tissue culture-derived trypomastigotes were harvested and washed before incubation in DMEM without FBS at pH 7.4 or pH 5.0 for 3 h. After incubation, the parasites were removed by three cycles of centrifugation and the proteins presented in the supernatant were TCA/acetone precipitated. Following protein digestion, peptides were subjected to nanoLC-MS/MS analysis.
Comparison of <i>T</i>. <i>cruzi</i> trypomastigote exoproteome at pH 7.4 and pH 5.0.
Fig. 2. Comparison of T. cruzi trypomastigote exoproteome at pH 7.4 and pH 5.0.
Venn diagram of protein groups identified in each condition (A). Protein categorization by GO annotation for cell compartment (B), molecular function (C) and biological activity (D). Y-axis represents the number of proteins present in each GO term.

The acidic pH-induced differentiation causes drastic metabolic and morphologic changes in T. cruzi trypomastigotes [23], preparing the parasite for a replicative stage. As demonstrated by Engel et al. (1985), the amastigote pre-replicative lag period spans from 18.2 to 34.2 hours, depending on the cloned stock analyzed. In 1995, Tomlinson et al. showed that, 2 hours after pH induction, almost no trypomastigotes were observed in culture and 4h after pH induction, more than 90% of trypomastigotes were transformed into amastigotes. Focusing on the transformation phase of this parasite, the exoproteome was analyzed at a time point of 3 hours after pH-induced differentiation. After this time, the exoproteome reflects changes in the trypomastigote transformation to amastigote and not the amastigote exoproteome "per se". In fact, amastigogenesis dynamics after pH lowering was also evaluated by Hernández-Osorio [30] and they found that, after 3 hours, intermediate forms were predominant (over 80%). Therefore, the exoproteome analysed here can be related to early trypomastigote morphological changes, microenvironment modulation to the next replicative phase and host cell metabolism modulation to parasite survival and replication. In 2013, Caradonna et al. [31], demonstrated that several host metabolism and cytoskeleton modulations which support parasite intracellular growth. Energy production, nucleotide metabolism, pteridine metabolism and fat acid oxidation were shown as interconnected pathways between host and parasite, regulated by host Akt signaling. The capability of adaptation would be especially relevant in the context of a natural dynamic infection in the mammalian host as observed during amastigogenesis. Plasticity within the same population could reflect the ability to change the environment in order to control growth rates. Some specific microenvironments can be a critical issue underlying tissue tropism and persistent infection [31]. An example of this can be observed during T. cruzi infection in adipose tissue and muscle. The fatty acid rich environment and high metabolism of energy production of these cells, provide a space against immune system [31]. The total amount and diversity of proteins released by the parasites almost doubled upon acidic induction within the three hours of incubation (Fig 2A). While hundreds of proteins were identified in both conditions, a significant number of identified proteins were specific for each one (Fig 2A). While 33.6% (91/271) of proteins were specific of pH 7.4 exoproteome, 62.7% (303/483) were specific of pH 5.0 condition (S1 Table). It suggests that there is an increase in protein diversity in exoproteome during amastigogenesis.

Overall, at pH 5.0 all GO categories increased except for proteasome complex and mitochondrion (cell compartment), cellular homeostasis (biological activity) and catalytic activity and antioxidant activity (molecular function) (Fig 2B–2D). Fisher's exact test between conditions also showed proteins with catalytic activity and hydrolase activity (a subcategory of catalytic activity) under-represented at pH 5.0 (Table 1). Fig 2B highlights the common membrane components in exoproteome, particularly at pH 5.0—indicating potential parasite surface remodeling. Metabolic process, regulation of biological process, response to stimulus and transport (Fig 2C) are the most represented term of the biological activity category. In terms of molecular function, the high number of proteins with catalytic activity corroborate the myriad of metabolic processes in the exoproteome (Fig 2D). Altogether, it is likely that the parasite renews its metabolism during the first hours of amastigogenesis.

Tab. 1. Under-represented GO terms in parasites incubated at pH 5.0 compared to pH 7.4.
Under-represented GO terms in parasites incubated at pH 5.0 compared to pH 7.4.

Secretome analysis of T. cruzi epimastigotes and metacyclic trypomastigotes reported a considerable amount of microvesicles and exosomes [32]. The parasite has different strategies to mediate intercellular communication [33,34], and these vesicles can be used to interact directly with host cells by transferring several small molecules such as proteins, mRNAs, microRNAs and small molecules [33]. Vesicles can transport proteins in soluble form, associated or as integral components of membranes. Transmembrane domains were predicted in 27% of proteins (155/574), with 33 proteins exclusively detected at pH 7.4, 70 at pH 5.0 and 52 in both conditions. These proteins presented up to four predicted transmembrane helices (Fig 3A). Based on this analysis, our results indicate that cell-derived trypomastigotes in both pH conditions could also release vesicles. Moreover, the increased proportion of transmembrane proteins in the exoproteome at pH 5.0, compared to pH 7.4 condition, indicates that transition of trypomastigotes to the early stage of amastigotes may trigger other types of secretion/excretion besides vesicles. However, further experiments to confirm this hypothesis are necessary. All 155 proteins with predicted transmembrane domains were also predicted to be secreted (Fig 3B), suggesting that these proteins could be present in vesicles or being secreted/excreted.

TMHMM analysis.
Fig. 3. TMHMM analysis.
Increased number of proteins predicted to possess transmembrane domain in pH 5.0 related to pH 7.4 (A) and all proteins with predicted transmembrane domains are also predicted to be secreted/excreted (B).

In silico screening of excreted/secreted proteins based on genomic information cannot be considered self-sustaining evidence for its secretion as the prediction accuracy is highly dependent on the tool performance and quality of the genomic annotations. Furthermore, predicted secretory proteins may not be expressed in the particular cell/condition under examination or have a retention signal that prevents their secretion [15]. Integrating bioinformatic analysis to predict secreted proteins with proteomic data reinforce the excreted/secreted status of a particular protein and provides validation for the subproteome enrichment and its quality. Herein, in silico prediction of secretion through classical (SignalP) and non-classical pathways (SecretomeP) of trypomastigote exoproteomes at pH 7.4 and pH 5.0 showed remarkably that 76.5% (439/574) of all detected proteins are predicted to be secreted in both algorithms (Fig 4A). Almost twice the number of proteins predicted to be secreted was identified in the acidic condition (199 proteins at pH 7.4 versus 370 proteins at pH 5.0, Fig 4A). This corroborates the hypothesis raised from previously published findings on over expression of several proteins (e.g. some Rab proteins and ADP-ribosylation factors), within 2 hours of acidic induction, indicating an increase in vesicular traffic [23]. In addition, most proteins were predicted to be released through non-classical pathways at pH 5.0 (131 versus 109). While a modest difference, it is in accordance with other findings for members of the kinetoplastidae order [3537]. Despite computational prediction presents some issues (e.g. none of the algorithms were designed specifically for Trypanosomatids), a very high percentage of predicted proteins together with the GO annotation profiles are concordant synergic and ratify our sampling and results.

Secretory pathway prediction.
Fig. 4. Secretory pathway prediction.
Total number of predicted proteins secreted and by classical (SignalP) and non-classical pathways (SecretomeP) (A). Blast2GO annotation of T. cruzi trypomastigote exoproteome at pH 7.4 and pH 5.0 and comparison of most abundant biological activity (B) and molecular function GO terms (C) of annotated secrete/excreted proteins.

Kinetoplastids database was explored with Blast2GO software in order to characterize the total of proteins predicted to be secreted/excreted. The biological processes: translation, response to stress, protein folding, biosynthetic process, generation of precursor metabolites and energy displayed higher number of proteins at pH 5.0 (Fig 4B). DNA binding, ion binding, structural constituent of ribosome and unfolded protein binding were molecular functions predominantly represented at pH 5.0 and hydrolase activity acting on glycosyl bonds and peptidase activity were under represented in this condition (Fig 4C). As expected, the most assigned term was hydrolase activity on glycosyl bonds due to the extent of trans-sialidases family members.

The most abundant proteins identified on classical secretory pathway analysis are shown on Fig 5. Trans-sialidases (TS) are enzymes that transfer sialic acid to mucins and both macromolecules are anchored to the plasma membrane by glycosylphoshatidylinositol [38,39]. This enzymatic reaction promotes protection to the parasite against the host immune system and promotes cell invasion. TS function is also important for parasitic escape from parasitophorous vacuole. The number of trans-sialidases identified in acidic conditions was lower than at pH 7.4 (35 and 44, respectively) (Fig 5). Furthermore, specific trans-sialidases were identified in each condition, as previously reported [40]. Specific trans-sialidases, at pH 5.0, could be involved in parasitophorous escape and, at pH 7.4, could be involved in immune system escape and cell invasion (Fig 5).

Representative proteins from classical secretory pathway in <i>T</i>. <i>cruzi</i>.
Fig. 5. Representative proteins from classical secretory pathway in T. cruzi.
Most abundant proteins released through classical pathway within T. cruzi trypomastigote exoproteome at pH 7.4 and pH 5.0.

Mucins repertory changes along parasite cell cycle [22] and, as expected, striking identification differences between both conditions for mucin and mucin-like proteins were observed. These families jumped from 2 representatives in the exoproteome at pH 7.4 to 38 different proteins at pH 5.0. Interestingly, a transcriptomic analysis showed a decrease in mRNA for mucins during amastigogenesis and an increase of transcripts for membrane-bound/secreted phospholipase A1 [41] and for surface-localized phosphatidylinositol-phospholipase C (PI-PLC) [42]. This increase in phospholipase transcripts may explain the increase of mucins in this secretome analysis [43]. Similarly, a study performed an analysis of glycoconjugate mucin secretion in cultured rat conjunctival goblet cells, and observed an increase of mucin secretion directly related to the phospholipase C and phospholipase A2 dependent-Ca2+ performance under physiological conditions [43]. This mechanism may be employed as the same manner in T. cruzi parasites, considering that many cellular processes are conserved among eukaryotes. This data provides evidence that the parasite remodels the cell coat releasing surface proteins during amastigogenesis.

Mucin-associated surface proteins (MASP) exhibited the same pattern of mucins with a considerable increase in acidic pH, from 13 proteins identified specifically at pH 7.4 to 58 specifically at pH 5.0 and 56 in both conditions. The T. cruzi genome has approximately 1,300 clustered genes coding for MASPs and these proteins can be shed by circulating and infective parasites [44,45]. Using anti-MASP antibodies, Bartholomeu et al., (2009), detected MASP in the supernatant of PI-PLC treated trypomastigotes [44]. In this scenario, increased MASP levels after acidification condition could be explained by increasing action of phospholipases, in the same fashion for mucins, even with downregulation of MASP transcripts during early amastigote development [46]. In addition, few members of these families (trans-sialidases, mucins and MASP) were also predicted to be secreted through non-classical pathway.

In addition, some heat shock proteins (HSPs) and HSP-associated proteins, such as chaperonins, co-chaperones, prefoldins, calreticulin and cyclophilins or peptidyl prolyl isomerases were found in both exoproteome conditions. In the acidic environment, 13 HSPs and HSP-associated proteins were detected, and some of them playing important roles in the macrophage activity. For example, Hsp10 inhibits classical LPS-induced activation of macrophages due to pro-inflammatory cytokine synthesis [47,48]. T. cruzi parasites also infect macrophages, and require an ideal environment to replicate, nonetheless, this mechanism needs to be experimentally demonstrated in this organism. Moreover (change), Hsp10 is considered a circulating anti-inflammatory factor that possibly acts to contain macrophage activation [49].

Conversely, in both conditions, cyclophilins or peptidyl prolyl isomerases, Hsp70 and clusterin are involved in the classically activation of macrophages [50]. In Toxoplasma gondii, cyclophilin-18 induced IL-12 production by dendritic cells and triggered cell signalling through CCR5. This mechanism may provide a strong protective response to the parasite allowing its transmission, avoiding host’s intermediates [51]. Interestingly, Hsp70 of Mycobacterium tuberculosis inhibits the infection of CD4+ T cells by HIV-1, blocking the CCR5 co-receptor [52] in other words, despite its role as a chaperone in the activation of macrophages [50] and protein folding, this protein may help the parasite in its survival and proliferation, inhibiting invasion of competitive microorganisms. Further, clusterin is known as a secreted extracellular chaperone capable to bind unfolded proteins, which could promote receptor-mediated endocytosis and intracellular lysosomal degradation [53]. Likewise, prefoldin, only found at pH 5.0, seems to act as a co-chaperone mediating chaperone-substrate interactions [54]. These observations seem to be feasible to occur in vivo, considering the high acidic environment of phagolysosome and the whole arsenal of T. cruzi HSP and HSP-associated proteins.

Since proteases are involved in crucial steps of the biological life cycle of T. cruzi including aspects of host-parasite interaction, these enzymes are subjects of special attention [5557]. The most studied zinc-dependent metalloproteases, also termed as GP63 family in trypanosomatids, are described as major surface glycoproteins with acid protease activity and virulence factors [58,59]. GP63 genes are present at high-copy-number [60] and encode proteins involved in parasite-host interaction. After 3 hours of incubation, the number of specific identifications at pH 5.0 and pH 7.4 was 2 and 7, respectively and 2 were present in both conditions. GP63, MASP, mucin-like proteins are surface membrane proteins that compose extracellular vesicles of trypomastigote [61] and of the early stages of amastigote differentiation accordingly to our data. Besides its ability to interact with the host extracellular matrix, GP63 is also able to inhibit NK cellular function. In trypanosomatids, this promotes resistance to antimicrobial peptides, intracellular amastigote survival in macrophages and degradation of cytosolic proteins of host cells. Altogether it demonstrates the versatility of GP63 in parasite survival in conditions of stress [6264].

Calpain-like proteins is another family of proteins with a large number of different genes present in trypanosomatid genomes [65]. Calpains are found as microtubule-interacting proteins in T. cruzi and T. brucei [56]. Two calpain-like cysteine peptidases were only released at pH 7.4 as well as at pH 5.0, respectively, and 2 more in both conditions. One calpain peptidase and the calpain-like CAP5.5 (cytoskeleton-associated protein 5.5) were recently described as immunoreactive proteins recognized by serum immunoglobulin from chagasic patients with early cardiomyopathy [66]. Moreover, CAP5.5 was shown to be secreted/excreted by metacyclic trypomastigotes [32]. According to our data, calpain cysteine peptidases and CAP5.5 are released (associated or not to vesicles) during the trypomastigote differentiation into amastigote.

Several proteins related to ubiquitin signaling were identified from the exoproteome at pH 5.0 and pH 7.4 (S2 Table). None of them were classified as secreted through classical pathway. For the parasites, ubiquination and ubiquitin-proteasome pathway are crucial in key steps in host colonization (proliferation and cell differentiation) [67] and for the host cells to modify immunoregulatory functions [68]. Bacteria and viruses secrete ubiquitin signaling related proteins into host cell. In T. cruzi, a protein related to ubiquitin signaling was shown to be secreted into the host cell and to localize in the nucleus [69]. Our data suggests that such proteins may function in the host cell nuclei orchestrating host regulatory elements towards parasite survival inside the host cell.

Intraflagellar vesicle transport occurs via microtubules driven by motors such as proteins belonging to the kinesin and the dynein family [70]. SNF-7 is a protein related to cargo transport through cytoskeleton and vesicle coating, which was secreted at pH 5.0 conditions only. Secretion profiles of transport and vesicle structures may indicate a dynamic parasite behaviour including an actively remodelling intracellular expression pattern related to the exportation of molecules during early stages of amastigogenesis. It fits with the idea that after cell invasion, trypomastigote disassemble and discard their flagella into host cell cytoplasm where it is degraded. This process releases flagellar proteins that enter the MHC-I processing pathway and presentation to CD8+ T cells as demonstrated for paraflagellar rod protein PAR4. Additionally, it was demonstrated that TcPAR4 immunization in mice enhanced resistance to T. cruzi [71]. Our results corroborate previous observations that TcPAR4 is released after parasite cell invasion. Paraflagellar rod component Par4 putative (Q4CUM0) was found exclusively in the parasite exoproteome at pH 5.0 (Table 2). This work provides 22 flagellar proteins exclusively identified at pH 5.0 and 12 proteins shared at pH 5.0 and pH 7.4. Among these proteins, kinesins and dyneins (or associated proteins) were found only at pH 5.0 or shared in both conditions. Strikingly, no flagellar proteins were identified as being released exclusively at pH 7.4. As demonstrated to TcPAR4, these proteins can be candidates for vaccines or good protein targets for new chemotherapy strategies.

Tab. 2. Flagellar and flagellar-associated proteins in the T. cruzi exoproteome.
Flagellar and flagellar-associated proteins in the <i>T</i>. <i>cruzi</i> exoproteome.

Conclusion

Acidic-pH-induced axenic amastigogenesis creates a lysosome-like environment, mimicking conditions when the parasite enters into the host cell. Exploiting this model, exoproteome analyses of early stages of amastigogenesis allowed the identification of several exclusive proteins at pH 5.0 related to cell communication, response to stimulus, regulation of biological process and cell division. In this scenario, exclusive proteins identified at pH 5.0 have the potential to modulate host cellular metabolism, allowing parasite survival, differentiation and proliferation. T. cruzi exoproteome changes during its life stage may provide advantages to parasites over host. Regarding trypomastigotes maintained in pH 7.4 or pH 5.0 for 3 hours, this is the first study to investigate the T. cruzi exoproteome change during the amastigogenesis. Our data provide evidence and direction for further studies to explore exoproteome changes during the first hours of amastigogenesis; highlighting the increase in number and diversity of proteins in acidic condition. This corroborates previous studies on the increase in vesicular trafficking during amastigogenesis. Furthermore, this work provides a list of vesicular and flagellar proteins released after acidic induction that could be explored as potential candidates to multitarget vaccines.

Supporting information

S1 Table [xlsx]
Proteins identified exclusively at pH7.4, at pH 5.0 and in both conditions.

S2 Table [pdf]
proteins related to ubiquitin signaling and ubiquitin-proteasome pathway from exoproteome at pH 5.0 and pH 7.4.


Zdroje

1. WHO (2010) First WHO report on neglected tropical diseases.

2. Albajar-Vinas P, Jannin J (2011) The hidden Chagas disease burden in Europe. Euro Surveill 16.

3. Rassi A Jr., Rassi A, Marin-Neto JA (2010) Chagas disease. Lancet 375: 1388–1402. doi: 10.1016/S0140-6736(10)60061-X 20399979

4. Roca C, Pinazo MJ, Lopez-Chejade P, Bayo J, Posada E, López-Solana J, et al. (2011) Chagas disease among the Latin American adult population attending in a primary care center in Barcelona, Spain. PLoS Negl Trop Dis 5: e1135. doi: 10.1371/journal.pntd.0001135 21572511

5. Coura JR, Vinas PA (2010) Chagas disease: a new worldwide challenge. Nature 465: S6–7. doi: 10.1038/nature09221 20571554

6. Bern C, Montgomery SP (2009) An estimate of the burden of Chagas disease in the United States. Clin Infect Dis 49: e52–54. doi: 10.1086/605091 19640226

7. Basso B (2013) Modulation of immune response in experimental Chagas disease. World J Exp Med 3: 1–10. doi: 10.5493/wjem.v3.i1.1 24520540

8. Cardillo F, de Pinho RT, Antas PR, Mengel J (2015) Immunity and immune modulation in Trypanosoma cruzi infection. Pathog Dis 73: ftv082. doi: 10.1093/femspd/ftv082 26438729

9. Hasslocher-Moreno AM, do Brasil PE, de Sousa AS, Xavier SS, Chambela MC, Sperandio da Silva GM (2012) Safety of benznidazole use in the treatment of chronic Chagas' disease. J Antimicrob Chemother 67: 1261–1266. doi: 10.1093/jac/dks027 22331592

10. Bern C (2011) Antitrypanosomal therapy for chronic Chagas' disease. N Engl J Med 364: 2527–2534. doi: 10.1056/NEJMct1014204 21714649

11. Burleigh BA, Andrews NW (1995) The mechanisms of Trypanosoma cruzi invasion of mammalian cells. Annu Rev Microbiol 49: 175–200. doi: 10.1146/annurev.mi.49.100195.001135 8561458

12. Rubin-de-Celis SS, Uemura H, Yoshida N, Schenkman S (2006) Expression of trypomastigote trans-sialidase in metacyclic forms of Trypanosoma cruzi increases parasite escape from its parasitophorous vacuole. Cell Microbiol 8: 1888–1898. doi: 10.1111/j.1462-5822.2006.00755.x 16824037

13. Andrews NW (1993) Living dangerously: how Trypanosoma cruzi uses lysosomes to get inside host cells, and then escapes into the cytoplasm. Biol Res 26: 65–67. 7670547

14. Camargo R, Faria LO, Kloss A, Favali CB, Kuckelkorn U, Kloetzel PM, et al. (2014) Trypanosoma cruzi infection down-modulates the immunoproteasome biosynthesis and the MHC class I cell surface expression in HeLa cells. PLoS One 9: e95977. doi: 10.1371/journal.pone.0095977 24752321

15. Caccia D, Dugo M, Callari M, Bongarzone I (2013) Bioinformatics tools for secretome analysis. Biochim Biophys Acta 1834: 2442–2453. doi: 10.1016/j.bbapap.2013.01.039 23395702

16. Clamp M, Fry B, Kamal M, Xie X, Cuff J, Lin MF, et al. (2007) Distinguishing protein-coding and noncoding genes in the human genome. Proc Natl Acad Sci U S A 104: 19428–19433. doi: 10.1073/pnas.0709013104 18040051

17. Skalnikova H, Motlik J, Gadher SJ, Kovarova H (2011) Mapping of the secretome of primary isolates of mammalian cells, stem cells and derived cell lines. Proteomics 11: 691–708. doi: 10.1002/pmic.201000402 21241017

18. Pavlou MP, Diamandis EP (2010) The cancer cell secretome: a good source for discovering biomarkers? J Proteomics 73: 1896–1906. doi: 10.1016/j.jprot.2010.04.003 20394844

19. Affranchino JL, Ibanez CF, Luquetti AO, Rassi A, Reyes MB, Macina RA, et al. (1989) Identification of a Trypanosoma cruzi antigen that is shed during the acute phase of Chagas' disease. Mol Biochem Parasitol 34: 221–228. doi: 10.1016/0166-6851(89)90050-9 2499788

20. Buschiazzo A, Muia R, Larrieux N, Pitcovsky T, Mucci J, Campetella O (2012) Trypanosoma cruzi trans-sialidase in complex with a neutralizing antibody: structure/function studies towards the rational design of inhibitors. PLoS Pathog 8: e1002474. doi: 10.1371/journal.ppat.1002474 22241998

21. Raimondo F, Morosi L, Chinello C, Magni F, Pitto M (2011) Advances in membranous vesicle and exosome proteomics improving biological understanding and biomarker discovery. Proteomics 11: 709–720. doi: 10.1002/pmic.201000422 21241021

22. Buscaglia CA, Campo VA, Frasch AC, Di Noia JM (2006) Trypanosoma cruzi surface mucins: host-dependent coat diversity. Nat Rev Microbiol 4: 229–236. doi: 10.1038/nrmicro1351 16489349

23. Queiroz RM, Charneau S, Mandacaru SC, Schwammle V, Lima BD, Roepstorff P, et al. (2014) Quantitative proteomic and phosphoproteomic analysis of Trypanosoma cruzi amastigogenesis. Mol Cell Proteomics 13: 3457–3472. doi: 10.1074/mcp.M114.040329 25225356

24. Ld Silva (1953) Sobre uma cepa de Trypanosoma cruzi altamente virulenta para o camundongo branco. Fol Clin Biol 20: 191–207.

25. Andrews NW, Colli W (1982) Adhesion and interiorization of Trypanosoma cruzi in mammalian cells. J Protozool 29: 264–269. doi: 10.1111/j.1550-7408.1982.tb04024.x 7047731

26. Queiroz RM, Ricart CA, Machado MO, Bastos IM, de Santana JM, de Sousa MV, et al. (2016) Insight into the Exoproteome of the Tissue-Derived Trypomastigote form of Trypanosoma cruzi. Front Chem 4: 42. doi: 10.3389/fchem.2016.00042 27872839

27. Tomlinson S, Vandekerckhove F, Frevert U, Nussenzweig V (1995) The induction of Trypanosoma cruzi trypomastigote to amastigote transformation by low pH. Parasitology 110 (Pt 5): 547–554.

28. Gobom J, Nordhoff E, Mirgorodskaya E, Ekman R, Roepstorff P (1999) Sample purification and preparation technique based on nano-scale reversed-phase columns for the sensitive analysis of complex peptide mixtures by matrix-assisted laser desorption/ionization mass spectrometry. J Mass Spectrom 34: 105–116. doi: 10.1002/(SICI)1096-9888(199902)34:2<105::AID-JMS768>3.0.CO;2-4 10093212

29. Queiroz RM, Charneau S, Bastos IM, Santana JM, Sousa MV, Roepstorff P, et al. (2014) Cell surface proteome analysis of human-hosted Trypanosoma cruzi life stages. J Proteome Res 13: 3530–3541. doi: 10.1021/pr401120y 24978697

30. Hernandez-Osorio LA, Marquez-Duenas C, Florencio-Martinez LE, Ballesteros-Rodea G, Martinez-Calvillo S, Manning-Cela RG (2010) Improved method for in vitro secondary amastigogenesis of Trypanosoma cruzi: morphometrical and molecular analysis of intermediate developmental forms. J Biomed Biotechnol 2010: 283842. doi: 10.1155/2010/283842 20037731

31. Caradonna KL, Engel JC, Jacobi D, Lee CH, Burleigh BA (2013) Host metabolism regulates intracellular growth of Trypanosoma cruzi. Cell Host Microbe 13: 108–117. doi: 10.1016/j.chom.2012.11.011 23332160

32. Bayer-Santos E, Aguilar-Bonavides C, Rodrigues SP, Cordero EM, Marques AF, Varela-Ramirez A, et al. (2012) Proteomic analysis of Trypanosoma cruzi secretome: characterization of two populations of extracellular vesicles and soluble proteins. J Proteome Res.

33. Freire-de-Lima L, da Fonseca LM, da Silva VA, da Costa KM, Morrot A, Freire-de-Lima CG, et al. (2016) Modulation of Cell Sialoglycophenotype: A Stylish Mechanism Adopted by Trypanosoma cruzi to Ensure Its Persistence in the Infected Host. Front Microbiol 7: 698. doi: 10.3389/fmicb.2016.00698 27242722

34. Cestari I, Ansa-Addo E, Deolindo P, Inal JM, Ramirez MI (2012) Trypanosoma cruzi immune evasion mediated by host cell-derived microvesicles. J Immunol 188: 1942–1952. doi: 10.4049/jimmunol.1102053 22262654

35. Cuervo P, De Jesus JB, Saboia-Vahia L, Mendonca-Lima L, Domont GB, Cupolillo E (2009) Proteomic characterization of the released/secreted proteins of Leishmania (Viannia) braziliensis promastigotes. J Proteomics 73: 79–92. doi: 10.1016/j.jprot.2009.08.006 19703603

36. Grebaut P, Chuchana P, Brizard JP, Demettre E, Seveno M, Bossard G, et al. (2009) Identification of total and differentially expressed excreted-secreted proteins from Trypanosoma congolense strains exhibiting different virulence and pathogenicity. Int J Parasitol 39: 1137–1150. doi: 10.1016/j.ijpara.2009.02.018 19285981

37. Geiger A, Hirtz C, Becue T, Bellard E, Centeno D, Gargani D, et al. (2010) Exocytosis and protein secretion in Trypanosoma. BMC Microbiol 10: 20. doi: 10.1186/1471-2180-10-20 20102621

38. Dc-Rubin SS, Schenkman S (2012) Trypanosoma cruzi trans-sialidase as a multifunctional enzyme in Chagas' disease. Cell Microbiol 14: 1522–1530. doi: 10.1111/j.1462-5822.2012.01831.x 22747789

39. Freire-de-Lima L, Oliveira IA, Neves JL, Penha LL, Alisson-Silva F, Dias WB, et al. (2012) Sialic acid: a sweet swing between mammalian host and Trypanosoma cruzi. Front Immunol 3: 356. doi: 10.3389/fimmu.2012.00356 23230438

40. Atwood JA, 3rd, Weatherly DB, Minning TA, Bundy B, Cavola C, Opperdoes FR, et al. (2005) The Trypanosoma cruzi proteome. Science 309: 473–476. doi: 10.1126/science.1110289 16020736

41. Belaunzaran ML, Wilkowsky SE, Lammel EM, Gimenez G, Bott E, Barbieri MA, et al. (2013) Phospholipase A1: a novel virulence factor in Trypanosoma cruzi. Mol Biochem Parasitol 187: 77–86. doi: 10.1016/j.molbiopara.2012.12.004 23275096

42. de Paulo Martins V, Okura M, Maric D, Engman DM, Vieira M, Docampo R, et al. (2010) Acylation-dependent export of Trypanosoma cruzi phosphoinositide-specific phospholipase C to the outer surface of amastigotes. J Biol Chem 285: 30906–30917. doi: 10.1074/jbc.M110.142190 20647312

43. Lippestad M, Hodges RR, Utheim TP, Serhan CN, Dartt DA (2018) Signaling pathways activated by resolvin E1 to stimulate mucin secretion and increase intracellular Ca(2+) in cultured rat conjunctival goblet cells. Exp Eye Res 173: 64–72. doi: 10.1016/j.exer.2018.04.015 29702100

44. Bartholomeu DC, Cerqueira GC, Leao AC, daRocha WD, Pais FS, Macedo C, et al. (2009) Genomic organization and expression profile of the mucin-associated surface protein (masp) family of the human pathogen Trypanosoma cruzi. Nucleic Acids Res 37: 3407–3417. doi: 10.1093/nar/gkp172 19336417

45. dos Santos SL, Freitas LM, Lobo FP, Rodrigues-Luiz GF, Mendes TA, Oliveira AC, et al. (2012) The MASP family of Trypanosoma cruzi: changes in gene expression and antigenic profile during the acute phase of experimental infection. PLoS Negl Trop Dis 6: e1779. doi: 10.1371/journal.pntd.0001779 22905275

46. Li Y, Shah-Simpson S, Okrah K, Belew AT, Choi J, Caradonna KL, et al. (2016) Transcriptome Remodeling in Trypanosoma cruzi and Human Cells during Intracellular Infection. PLoS Pathog 12: e1005511. doi: 10.1371/journal.ppat.1005511 27046031

47. Vanags D, Williams B, Johnson B, Hall S, Nash P, Taylor A, et al. (2006) Therapeutic efficacy and safety of chaperonin 10 in patients with rheumatoid arthritis: a double-blind randomised trial. Lancet 368: 855–863. doi: 10.1016/S0140-6736(06)69210-6 16950363

48. Johnson BJ, Le TT, Dobbin CA, Banovic T, Howard CB, Flores Fde M, et al. (2005) Heat shock protein 10 inhibits lipopolysaccharide-induced inflammatory mediator production. J Biol Chem 280: 4037–4047. doi: 10.1074/jbc.M411569200 15546885

49. Shamaei-Tousi A, D'Aiuto F, Nibali L, Steptoe A, Coates AR, Parkar M, et al. (2007) Differential regulation of circulating levels of molecular chaperones in patients undergoing treatment for periodontal disease. PloS one 2: e1198. doi: 10.1371/journal.pone.0001198 18030332

50. Henderson B, Henderson S (2009) Unfolding the relationship between secreted molecular chaperones and macrophage activation states. Cell Stress and Chaperones 14: 329–341. doi: 10.1007/s12192-008-0087-4 18958583

51. Aliberti J, Valenzuela JG, Carruthers VB, Hieny S, Andersen J, Charest H, et al. (2003) Molecular mimicry of a CCR5 binding-domain in the microbial activation of dendritic cells. Nature immunology 4: 485. doi: 10.1038/ni915 12665855

52. Babaahmady K, Oehlmann W, Singh M, Lehner T (2007) Inhibition of human immunodeficiency virus type 1 infection of human CD4+ T cells by microbial HSP70 and the peptide epitope 407–426. J Virol 81: 3354–3360. doi: 10.1128/JVI.02320-06 17251296

53. Wilson MR, Easterbrook-Smith SB (2000) Clusterin is a secreted mammalian chaperone. Trends in biochemical sciences 25: 95–98. doi: 10.1016/s0968-0004(99)01534-0 10694874

54. Vainberg IE, Lewis SA, Rommelaere H, Ampe C, Vandekerckhove J, Klein HL, et al. (1998) Prefoldin, a chaperone that delivers unfolded proteins to cytosolic chaperonin. Cell 93: 863–873. doi: 10.1016/s0092-8674(00)81446-4 9630229

55. Alvarez VE, Niemirowicz GT, Cazzulo JJ (2012) The peptidases of Trypanosoma cruzi: digestive enzymes, virulence factors, and mediators of autophagy and programmed cell death. Biochim Biophys Acta 1824: 195–206. doi: 10.1016/j.bbapap.2011.05.011 21621652

56. Branquinha MH, Marinho FA, Sangenito LS, Oliveira SS, Goncalves KC, Ennes-Vidal V, et al. (2013) Calpains: potential targets for alternative chemotherapeutic intervention against human pathogenic trypanosomatids. Curr Med Chem 20: 3174–3185. doi: 10.2174/0929867311320250010 23899207

57. Bastos IM, Motta FN, Grellier P, Santana JM (2013) Parasite prolyl oligopeptidases and the challenge of designing chemotherapeuticals for Chagas disease, leishmaniasis and African trypanosomiasis. Curr Med Chem 20: 3103–3115. doi: 10.2174/0929867311320250006 23514419

58. Kulkarni MM, Olson CL, Engman DM, McGwire BS (2009) Trypanosoma cruzi GP63 proteins undergo stage-specific differential posttranslational modification and are important for host cell infection. Infect Immun 77: 2193–2200. doi: 10.1128/IAI.01542-08 19273559

59. Pech-Canul AC, Monteon V, Solis-Oviedo RL (2017) A Brief View of the Surface Membrane Proteins from Trypanosoma cruzi. J Parasitol Res 2017: 3751403. doi: 10.1155/2017/3751403 28656101

60. Ma L, Chen K, Meng Q, Liu Q, Tang P, Hu S, et al. (2011) An evolutionary analysis of trypanosomatid GP63 proteases. Parasitol Res 109: 1075–1084. doi: 10.1007/s00436-011-2348-x 21503641

61. Bernabo G, Levy G, Ziliani M, Caeiro LD, Sanchez DO, Tekiel V (2013) TcTASV-C, a protein family in Trypanosoma cruzi that is predominantly trypomastigote-stage specific and secreted to the medium. PLoS One 8: e71192. doi: 10.1371/journal.pone.0071192 23923058

62. Yao C (2010) Major surface protease of trypanosomatids: one size fits all? Infection and immunity 78: 22–31. doi: 10.1128/IAI.00776-09 19858295

63. Olivier M, Atayde VD, Isnard A, Hassani K, Shio MT (2012) Leishmania virulence factors: focus on the metalloprotease GP63. Microbes and infection 14: 1377–1389. doi: 10.1016/j.micinf.2012.05.014 22683718

64. d’Avila-Levy CM, Altoé EC, Uehara LA, Branquinha MH, Santos AL (2014) GP63 function in the interaction of trypanosomatids with the invertebrate host: facts and prospects. Proteins and Proteomics of Leishmania and Trypanosoma: Springer. pp. 253–270.

65. Ersfeld K, Barraclough H, Gull K (2005) Evolutionary relationships and protein domain architecture in an expanded calpain superfamily in kinetoplastid parasites. J Mol Evol 61: 742–757. doi: 10.1007/s00239-004-0272-8 16315106

66. Caminha MA, de Lorena VMB, de Oliveira Junior W, Perales J, Carvalho PC, Lima DB, et al. (2019) Trypanosoma cruzi immunoproteome: Calpain-like CAP5.5 differentially detected throughout distinct stages of human Chagas disease cardiomyopathy. J Proteomics 194: 179–190. doi: 10.1016/j.jprot.2018.11.019 30503829

67. Munoz C, San Francisco J, Gutierrez B, Gonzalez J (2015) Role of the Ubiquitin-Proteasome Systems in the Biology and Virulence of Protozoan Parasites. Biomed Res Int 2015: 141526. doi: 10.1155/2015/141526 26090380

68. Hu H, Sun SC (2016) Ubiquitin signaling in immune responses. Cell Res 26: 457–483. doi: 10.1038/cr.2016.40 27012466

69. Hashimoto M, Murata E, Aoki T (2010) Secretory protein with RING finger domain (SPRING) specific to Trypanosoma cruzi is directed, as a ubiquitin ligase related protein, to the nucleus of host cells. Cell Microbiol 12: 19–30. doi: 10.1111/j.1462-5822.2009.01375.x 19702650

70. Wren KN, Craft JM, Tritschler D, Schauer A, Patel DK, Smith EF, et al. (2013) A differential cargo-loading model of ciliary length regulation by IFT. Curr Biol 23: 2463–2471. doi: 10.1016/j.cub.2013.10.044 24316207

71. Kurup SP, Tarleton RL (2014) The Trypanosoma cruzi flagellum is discarded via asymmetric cell division following invasion and provides early targets for protective CD8(+) T cells. Cell Host Microbe 16: 439–449. doi: 10.1016/j.chom.2014.09.003 25299330


Článok vyšiel v časopise

PLOS One


2019 Číslo 11
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#