Skip to main content
Advertisement
  • Loading metrics

Comparative proteomics of adult Paragonimus kellicotti excretion/secretion products released in vitro or present in the lung cyst nodule

  • Lucia S. Di Maggio ,

    Roles Conceptualization, Formal analysis, Investigation, Methodology, Validation, Visualization, Writing – original draft, Writing – review & editing

    lucia@wustl.edu

    Affiliation Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Kurt C. Curtis,

    Roles Methodology, Writing – review & editing

    Affiliation Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Petra Erdmann-Gilmore,

    Roles Investigation, Software, Writing – review & editing

    Affiliation Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Robert S. W. Sprung,

    Roles Investigation, Software, Writing – review & editing

    Affiliation Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • R. Reid Townsend,

    Roles Funding acquisition, Methodology, Resources, Writing – review & editing

    Affiliations Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America, Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Gary J. Weil,

    Roles Conceptualization, Resources, Writing – original draft, Writing – review & editing

    Affiliation Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America

  • Peter U. Fischer

    Roles Conceptualization, Funding acquisition, Methodology, Resources, Supervision, Writing – original draft, Writing – review & editing

    Affiliation Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America

Abstract

Paragonimus kellicotti is a zoonotic lung fluke infection, the agent of North American paragonimiasis, and an excellent model for other Paragonimus infections. The excretory/secretory proteins (ESP) released by parasites and presented at the parasite-host interface are frequently proposed to be useful targets for drugs and/or vaccines In vitro culture conditions may alter ESP compared to those produced in vivo. In order to investigate ESPs produced in vivo we took advantage of the fact that adult P. kellicotti reproduce in the lungs of experimentally infected gerbils in tissue cysts. We performed a mass-spectrometric analysis of adult P. kellicotti soluble somatic protein (SSPs) extracts, excreted/secreted proteins (ESPs) produced by adult worms during in vitro culture, and lung cyst fluid proteins (CFPs) from experimentally infected gerbils. We identified 2,137 P. kellicotti proteins that were present in at least two of three biological replicates and supported by at least two peptides. Among those were 1,914 proteins found in SSP, 947 in ESP and 37 in CFP. In silico analysis predicted that only 141 of the total 2,137 proteins were secreted via classical or non-classical pathways. The most abundant functional categories in SSP were storage and oxidative metabolism. The most abundant categories in ESP were proteins related to metabolism and signal transduction. The 37 parasite-related proteins in CFP belonged to 11 functional categories. The largest groups were proteins with unknown function, cytoskeletal proteins and proteasome machinery. 29 of these 37 proteins were shared among all three sample types. To our knowledge, this is the first study that compares in vitro and in vivo ESP for any Paragonimus species. This study has provided new insights into ESPs of food-borne trematodes that are produced and released in vivo. Proteins released at the host-parasite interface may help the parasite evade host immunity and may represent new targets for novel treatments or diagnostic tests for paragonimiasis.

Author summary

Neglected tropical diseases include food-borne trematode infections such as paragonimiasis that causes severe illness with important social and economic consequences. In this study, we compared excretory/secretory products (ESPs) produced by the North American lung fluke Paragonimus kellicotti that are released during in vitro culture with those that are released in vivo inside the lungs of experimentally infected animals. We identified a total of 2,137 P. kellicotti proteins. These included 1,914 proteins in soluble extracts of whole flukes, 947 proteins identified in ESPs released during in vitro culture, and 37 proteins identified in fluid recovered from lung cysts that surround adult flukes in experimentally infected gerbils. The most abundant functional protein categories varied by sample type. To our knowledge, this is the first study that has compared proteins released in vitro and in vivo by adult Paragonimus flukes. Proteins released in vivo may help the parasite evade host immunity. They may have practical value as novel drug targets or lead to improved diagnostic tests for paragonimiasis.

Introduction

Lung flukes of the genus Paragonimus include about 30 species that are mainly found in Asia, but also Africa and the Americas causing an estimated 23 million human infections [1]. Paragonimus kellicotti is the cause of North American paragonimiasis, and can lead to severe pulmonary and/or extra-pulmonary diseases in carnivorous mammals including humans. Human infections are rare, but the number of people diagnosed with this parasite species has recently been increasing [2]. Humans become infected via consumption of raw or undercooked freshwater crayfish that contain the infective metacercariae. P. kellicotti is the only species endemic to the USA and a good model for other Paragonimus species because infectious metacercariae are readily available; a small rodent model has been established and its whole genome has been sequenced [3,4].

Previous studies of Paragonimus secretomes were carried out mainly to investigate the composition and/or immunological roles of selected molecules [57]. Despite the technical progress of mass spectrometry, few proteomic studies have been conducted for any Paragonimus species. The excretory/secretory proteins (ESP) released by parasites and presented at the parasite-host interface are frequently proposed to be useful targets for drugs and/or vaccines [810]. These molecules are actively released from the parasite gut, excretory pores and surface tegument. They include proteins, carbohydrates, lipids and RNAs that are likely to be critically important for parasite survival in the mammalian host [11]. Characterization of these worm components and their functions within the host could improve our understanding of parasite-host interactions, and this could lead to the discovery of new strategies to combat these parasitic infections.

In vitro culture of adult flukes is the most common strategy to collect ESP and the implicit assumption is that these products resemble the excretome and secretome of parasites in vivo. However, verification of the worm integrity and viability during culture is a key step to avoid contamination by components derived from dying parasites. Furthermore, parasites maintained in vitro face different environmental conditions than parasites in vivo. For example the host’s immune system is not present in vitro. Therefore, the assumption that parasites release the same ESPs in vitro and in vivo is likely to be incorrect. In the gerbil model, adult P. kellicotti flukes live individually or in small groups of two to four flukes in lung cysts. Analysis of fluid from these cysts offer the unique opportunity to study ESPs in vivo [3].

The aims of this study were to study ESPs to improve understanding of parasite-host interactions and to support the discovery of novel diagnostic or therapeutic targets. We used tandem mass-spectrometry to characterize the secretome of adult P. kellicotti flukes and to compare ESPs released during in vitro culture with parasite cyst fluid proteins (CFPs) which correspond to ESPs released by adult flukes in vivo. We also compared these results to the global proteome of soluble, somatic protein (SSP) extracts of whole adult flukes.

Materials and methods

Ethics statement

Mongolian gerbils (Meriones unguiculatus) aged 5 to 8 weeks old were purchased from Charles River Laboratories (Worcester, MA, USA) and kept under specific pathogen free conditions in an animal care facility of the Department of Comparative Medicine at Washington University in St. Louis. (Missouri, USA). A protocol for the ethical use of animals in research was approved by the university’s Institutional Animal Care and Use Committee (IACUC) (protocol ID 20–0503). Procedures involving animals were performed by specially trained personnel.

Sample collection and preparation

P. kellicotti metacercariae were isolated from the heart tissue of two naturally infected species of crayfish (Orconectes luteus and Orconectes punctimanus) that were collected in Huzzah Creek (Missouri, USA) as previously described [3]. A total of four or five metacercariae, in 150μL phosphate buffered saline (PBS), were inoculated into each gerbil by intraperitoneal injection. Infected animals were sacrificed about 5 weeks post-infection, and necropsies were performed. Lungs were examined for cysts and adult flukes. Adult flukes recovered from cysts were washed three times for 5 min in warm sterile PBS and maintained in warm PBS until they emptied their gut contents. After that, parasites were incubated at 37°C, 5% CO2 in 500μL of sterile culture medium (RPMI 1640 supplemented with 30mM HEPES pH 7.2, 2% glucose and 10% penicillin/streptomycin/amphotericin mix; Sigma, St. Louis MO, USA). After 12hrs, the supernatant containing the ESP was collected under laminar flow hood. The fluid was syringe-filtered through a 0.22μm filter and store at -20°C until use.

For SSP, adult parasites (collected as described below) were washed three times for 5 min in warm and sterile PBS until they cleaned their gut content. Individual parasites were placed in a 1.5mL sterile tube containing PBS and mechanically crushed with a sterile plastic pestle under a laminar flow hood. The samples were centrifuged at 20.000X g for 10 minutes at 4°C and the supernatant with extracted proteins was syringe-filtered through a 0.22μm filter and transferred into a clean 1.5mL tube and store at -20°C until use.

Cyst fluid was collected by syringe aspiration of intact lung cysts from three different gerbils. The fluid was pooled and syringe-filtered through a 0.22μm filter and placed in a 1.5mL sterile tube and store at -20°C until use.

Protein digestion and sample preparation

Peptides were prepared as previously described using a modification of the filter-aided sample preparation method [12]. Data were acquired as three technical replicates from single biological preparations for ESP, SSP and CFP samples types. Three aliquots of 15 μg total protein from each sample were mixed with 100 μl of 100 mM Tris-HCL buffer, pH 8.5 containing 8 M urea (UA buffer) and 10 mM DTT and reduced at room temperature for an hour. The samples were transferred to the top chamber of a 10,000 MWCO cutoff filtration unit (MilliporeSigma, Burlington MA, USA) and processed to peptides as previously described [12]. The peptides were dried in a Savant DNA 120 Speedvac concentrator (Thermo Fisher Scientific, Waltham MA, USA) for 15 min. The dried peptides were dissolved in 1% (vol/vol) TFA and desalted using two micro-tips (porous graphite carbon, BIOMEKNT3CAR) (Glygen, Columbia MD, USA) on a Biomek NX Beckman robot (Beckman Coulter, Brea CA, USA), as previously described [13]. The peptides were eluted with 60 μl of 60% (vol/vol) MeCN in 0.1% (vol/vol) TFA and dried in a Savant DNA 120 Speedvac concentrator (Thermo Scientific) after adding TFA to 5% (vol/vol). The peptides were dissolved in 20 μl of 1% (vol/vol) MeCN in water. An aliquot (10%) was removed for quantification using the Pierce Quantitative Fluorometric Peptide Assay kit (Thermo Scientific). The remaining peptides were transferred to autosampler vials (Sun-Sri,), dried and stored at -80°C.

LC-MS/MS and data analysis

The peptides were separated using a nano-ELUTE chromatograph (Bruker Daltonics, Bremen, Germany) interfaced to a timsTOF Pro mass spectrometer (Bruker Daltonics) with a modified nano-electrospray source (Bruker Daltonics). The mass spectrometer was operated in Parallel accumulation-serial fragmentation (PASEF) mode [14]. The samples in 2 μL of 1% (vol/vol) FA were injected at a flow rate of 350 nL / min onto the column (75 μm i.d. × 25 cm Aurora Series) with a CSI emitter (Ionopticks, Melbourne Victoria, Australia). The column temperature was set at 50°C. The column was equilibrated using constant pressure (800 bar) with 8 column volumes of solvent A (0.1% (vol/vol) FA). Sample loading was performed at constant pressure (800 bar) in 2 μL of Solvent A.

The peptides were eluted using the following gradient of Solvent B (0.1% (vol/vol) FA/MeCN): solvent A containing 2% B was increased to 17% B over 60 min, to 25% B over 30 min, to 37% B over 10 min, to 80% B over 10 min and constant 80% B for 10 min. The MS1 and MS2 spectra were recorded from m/z 100 to 1700. Suitable precursor ions for PASEF-MS/MS were selected from TIMS-MS survey scans by a PASEF scheduling algorithm [14]. A polygon filter was applied to the m/z and ion mobility plane to select features most likely to be multiply charged peptide precursor ions. Quadrupole isolation width was set to 2 Th for m/z < 700 and 3 Th for m/z > 700. The collision energy was ramped stepwise as a function of increasing ion mobility: 52 eV for 0–19% of the ramp time; 47 eV from 19–38%; 42 eV from 38–57%; 37 eV from 57–76%; and 32 eV for the remainder. The TIMS elution voltage was calibrated linearly using the Agilent ESI-L Tuning Mix (m/z 622, 922, 1222).

Data from the mass spectrometer were converted to peak lists using DataAnalysis version 5.2 (Bruker Daltonics). The MS2 spectra with charges +2, +3 and +4 were analyzed using Mascot software version 2.5.1 (Matrix Science, London, UK) [15]. Mascot was set up to search against a custom parasite database (Paragonimus kellicotti, 12,850 entries) or host database (Meriones unguiculatus, 38,763 entries) assuming the digestion enzyme was trypsin with a maximum of 4 missed cleavages allowed [4,16]. The searches were performed with a fragment ion mass tolerance of 50 ppm and a parent ion tolerance of 25 ppm. Carbamidomethylation of cysteine was specified in Mascot as a fixed modification. Variable modifications included deamidation of asparagine and glutamine, pyro-glutamate formation from n-terminal glutamine, oxidation of methionine and acetylation of protein n-terminus.

Scaffold version 5.0.1 (Proteome Software Inc., Portland OR, USA) was used to validate MS/MS based peptide and protein identifications. Peptide identifications were accepted if they could be established at greater than 58.0% probability to achieve an FDR less than 1.0% by the Scaffold Local FDR algorithm. Protein identifications were accepted if they could be established at greater than 95.0% probability and contained at least 2 identified peptides. Protein probabilities were assigned by the Protein Prophet algorithm [17]. Proteins that contained similar peptides and could not be differentiated based on MS/MS analysis alone were grouped to satisfy the principles of parsimony. The mass spectrometry proteomics data were deposited in the ProteomeXchange Consortium (http://proteomecentral.proteomexchange.org) via the MassIVE partner repository with the dataset identifier MSV000089589.

Protein functional annotation, classification and visualization

In addition to annotation with the P. kellicotti and M. unguiculatus databases [4], proteins were functionally classified using a software developed and provided by Dr. José. M. Ribeiro [18]. The functionally annotated proteins for each dataset and sample were manually curated and plotted on a hyperlinked Excel spreadsheet (S1 and S2 Tables) (Microsoft, Redmond WA, USA). NSAF (Normalized Spectral Abundance Factor) values were used to determine the proteins relative abundance. Mean NSAF values from at least two replicates were combined and then divided by the total NSAF for the respective sample. NSAF values were recorded in an Excel spreadsheet as a percentage of the total NSAF for respective samples, the results were plotted on pie charts according to protein functional classes. Principal component analysis (PCA) and scatter plot were conducted using GraphPad Prism version 9.1.3 for Windows (GraphPad Software, San Diego CA, USA). Protein sequences were examined for the presence of signal peptides using the program SignalP 6.0 (https://services.healthtech.dtu.dk/service.php?SignalP).

Results and discussion

Proteomic profiles of P. kellicotti samples

A total of different 2,137 parasite-related proteins were identified and quantified using stringent criteria ≥ 2 peptides identified in at least 2 of the 3 replicate mass spectrometry runs from each sample (Fig 1A). ESP, SSP and CFP samples types were technically replicated in order to reduce the experimental noise of the results. Among these P. kellicotti proteins, 1,914 were detected in the SSP samples, 942 in ESP samples and 37 in CFP samples. The Venn diagram in Fig 1 shows that 29 P. kellicotti proteins were detected in all three sample types. Only 6 proteins were found exclusively in the CFP sample. These included 2 heat shock proteins, 1 histone H2A proteins and 2 proteins with unknown function (S1 Table). A principal component analysis (PCA) was applied to the protein expression data sets to show differences and similarities between sample types after functional classification of proteins (Fig 2). This analysis reduces the dimensionality of protein data sets to illustrate variation present in each data set [19]. This study is the second proteomic study of P. kellicotti. A previous mass spectrometry study to identify serodiagnostic targets by our group detected 2,555 predicted proteins in a single SSP sample [5]. While both studies used similar mass spectrometry instruments, the present study used an updated P. kellicotti database based on the recently published whole genome of P. kellicotti [4] and higher stringency analysis methods (e,g., use of NSAF values for quantitation).

thumbnail
Fig 1. Venn diagrams representing the number of proteins found in each sample type, soluble somatic proteins (SSP), excretory/secretory proteins (ESP), cyst fluid proteins (CFP).

The overlap region between the circles shows proteins present in two or more stages. In parenthesis is the total protein number for the sample. A. Comparison of P. kellicotti-derived proteins by sample type B. Comparison of M. unguiculatus (host)-derived proteins by sample type.

https://doi.org/10.1371/journal.pntd.0010679.g001

thumbnail
Fig 2. Principal component analysis.

The PCA plot represents 2,137 parasite-related proteins with biological replicates that indicated distinct proteomics profile differences between soluble somatic proteins (SSP), excretory/secretory proteins (ESP), cyst fluid proteins (CFP) samples types represented with yellow, purple and pink dots, respectively.

https://doi.org/10.1371/journal.pntd.0010679.g002

In silico analysis predicted that only 141 (6.6%) of the total 2,137 proteins were secreted via a classical or non-classical pathway. These ESPs include 17 proteases, 16 metabolism related proteins, four transporters and three protease inhibitors (S1 Table). Among these 141 proteins, 112 were found in the SSP sample type (5.9% of the proteins detected in the sample), 50 in ESP sample type (5%) and only 1 CFP sample type (2.7%) (S1 Table). Proteins, including those that lack a signal peptide, can be also secreted or excreted by the intestine, excretory pores, surface cuticle/tegument shedding or release of extracellular vesicles (EV). The presence of EV may explain the detection of proteins that are assumed to have intra-trematode functions [20]. It was shown that EV can be taken up by host cells, a process that indicates a role in the parasite’s survival [21]. It was demonstrated that EVs differ in size, cargo and function depending on the developmental stages of the trematodes [22,23].

A total of 2,053 host-related proteins were detected in the CFP sample type and the most abundant proteins were plasma proteins and constitutive cell proteins (S3 Table). This is in accordance with the cyst architecture of the parasites, which consists of dense collagenous connective tissue and various inflammatory cells such as eosinophils. From the 128 host-derived proteins detected between the SSP and ESP samples types (Figs 1B and 3 and S2 Table); 55 proteins were shared between the three sample types and 7 and 16 host proteins were found exclusively in each sample, respectively. Based on relative abundance, most of the host proteins identified in the SSP and ESP samples type were blood or cytoskeletal proteins. Interestingly we were not able to find the most abundant blood proteins in SSP or ESP as was found in the CFP sample type (S3 Table). Thus, the presence of host-derived proteins in parasite secretions may be due to a recycling system and not due to contamination during sample collection. As it was described before, parasites were washed extensively and had no microscopically visible gut content before culture. Therefore, the only way to detect host proteins in the samples is if the parasites actively secrete them. For example in F. hepatica proteins of the host’s blood were found as part of the parasite EV content and in soft ticks as part of the salivary gland proteome [20,24,25]. Host proteins have been demonstrated in the Schistosoma tegument, and they may help the parasite to evade immune recognition [26].

thumbnail
Fig 3. Functional annotation of M. unguiculatus-derived proteins found in P. kellicotti samples.

Pie chart representing the percentage of proteins found in each sample with respect to NSAF values. A. soluble somatic proteins (SSP) sample type B. excretory/secretory proteins (ESP) sample type.

https://doi.org/10.1371/journal.pntd.0010679.g003

The somatic and secreted proteome of P. kellicotti

The most abundant functional categories in the SSP sample type were storage (15%), oxidant metabolism (15%) and protease inhibitors (9%) (Fig 4A). In contrast, the most abundant categories in the ESP sample type were metabolism related proteins (oxidation, carbohydrates and lipids, 12%, 9% and 6% respectively) and signal transduction (Fig 4B). The most abundant proteins identified in the SSP sample type were two glutathione S-transferases (GSTs), fatty acid-binding protein, calcium-binding protein and actin. This list overlapped significantly with the most abundant proteins in the ESP sample type (GSTs, ferritin, fatty acid-binding protein and two unknown proteins, see S1 Table).

thumbnail
Fig 4. Functional annotation of P. kellicotti-derived proteins.

Pie chart representing the percentage of proteins found in each sample type with respect to NSAF values. A. somatic soluble proteins (SSP) sample type B. excretory/secretory proteins (ESP) sample type C. cyst fluid proteins (CFP) sample type.

https://doi.org/10.1371/journal.pntd.0010679.g004

GSTs are a versatile protein superfamily involved in cellular detoxification by either catalyzing toxin conjugation with glutathione (GSH) or passively binding to a wide range of endogenous/exogenous toxic molecules [27]. In parasites, GSTs participate in detoxifying exogenous toxins and protect the parasite from reactive oxidant damage as aldehyde products of lipid peroxidation in trematodes can be metabolized through glutathione conjugation [28]. Due to their important role in parasite-host interactions, GSTs have been targeted for pharmaceutical and vaccine purposes and have demonstrated protective effects against some parasites [29]. Fatty acid binding proteins (FABPs) are immunogenic proteins that play important roles in nutrient acquisition and parasite survival within the mammalian host. However as this family of proteins is often present in ESP, it may also have anti-inflammatory functions. For instance, a FABP secreted by F. hepatica induced human peripheral blood mononuclear cells (PBMCs) to express arginase, CHI3L1 and activated macrophages to release anti-inflammatory cytokines [30]. In addition, this FABP has been shown to downregulate production of nitric oxide and the expression of nitric oxide synthase. Several heat shock proteins (HSPs) were found in the SSP and ESP samples (17 and 8 proteins, respectively). HSPs are often detected in parasite ESPs, and they are major target of host immune responses [31]. HSPs function as proteins chaperones and they are believed to be important for stress resistance, parasite viability, infectivity and virulence [32].

Protease inhibitors play crucial roles in parasite development and survival; they are believed to counteract potentially damaging immune attack of vertebrate hosts. Many digenean trematodes rely on proteases to invade host tissues, and they also contribute to nutrition and development. We found 63 proteases in the SSP and 43 in the ESP sample with cysteine proteases as the most abundant subcategory. Several cysteine proteases were also found in P. westermani ESP, and some of them were recognized by antibodies in host serum samples [6]. Although we did not find many protease inhibitors (3%) in the P. kellicotti ESP samples, it is interesting that a cystatin was one of the most abundant proteins overall in the sample. Parasite cystatins not only participate in normal physiological processes, but they are also actively involved in host–parasite interactions including immune evasion [33]. The P. kellicotti cystatins share high sequence similarity with orthologs found in S. japonicum that are expressed in the gut and tegument of the adult parasite and inhibits the proteolytic activity of papain [34].

The study of the SSP and ESP can provide important clues to improve the serodiagnosis of parasitic diseases. For example, Sadaow and coworkers developed an immunochromatographic test for opisthorchiasis and clonorchiasis diagnosis using ESP [35]. The test used soluble ESP from adult worms cultured in vitro as the immobilized antigen. However, the preparation of ESP is time consuming and expensive, and it relies on the availability of adult parasites. In addition, ESP samples are difficult to standardize for use in serodiagnostic tests [36].

Similarities and differences between proteins in ESP and CFP

The analysis of parasite ESP released in vitro provides a window into parasite-host interactions. However, ESP may also provide false signals. The presence of intracellular proteins or apoptotic factors in ESP suggest that some proteins detected are due to nonspecific leakage of intracellular proteins into the culture medium caused by cellular damage of the parasite [37]. In vitro culture conditions are different from those in vivo in mammalian hosts. Potential differences including pH, lack of exposure to the host’s immune system, nutrients, and host-regulated parasite density may account for some differences between in vitro in vivo ESPs. We identified 37 parasite-related proteins in CFP samples that could be divided in 11 functional categories (Table 1). The largest group consist of proteins with unknown function (without and with conserved domains, 31%), cytoskeletal proteins (16%) and proteasome machinery (12%) (Figs 1 and 4C). From these 37 proteins, 29 are shared among the three samples (Fig 4 and Table 1).

thumbnail
Table 1. Proteins found in the CFP sample type.

In the first row are the accession numbers of each of the 37 parasite-derived proteins found in the CFP sample type. Presence of these proteins in the somatic soluble proteins (SSP) sample type and excretory/secretory proteins (ESP) sample type are marked with an asterisk. The GenBank accession number of the top blast hit and it similarity to the match is given in parenthesis. The proteins are ordered from the most to the least abundant in the CFP sample type.

https://doi.org/10.1371/journal.pntd.0010679.t001

Turning to parasite proteins shared by ESP and CSP, the most abundant of these proteins were peroxiredoxin-2, two cathepsins L, four Ras-related proteins and two T-complex protein 1 subunit. Peroxiredoxin are proteins able to play multiple physiological roles that include thiol-dependent peroxidase [38], chaperone holdase [39,40], regulator of H2O2, and modulator of the immune response [41,42]. Gene knock-out and RNA interference studies demonstrated that this protein plays a crucial role in survival and virulence, but there is evidences that its main physiological role may not be the same in different parasite species [4345]. Parasite survival relies on an effective defense against oxidative damage due to reactive oxygen and nitrogen species produced by the host immune system. It was also demonstrated that peroxiredoxins could activate macrophages and play a key role in promoting parasite induced Th2 type immunity while suppressing a pro-inflammatory Th1 response. In parasitic trematodes, peroxiredoxins secreted in ESP inactivate reactive oxygen species, and act as pathogen-associated molecular pattern molecules (PAMPs) where they alternatively activate macrophages, using a mechanism that is independent of antioxidant function an suppress Th1 [41,46]. Cathepsin L is a cysteine protease related to papain. Although one of the two cathepsins found in these parasite-related shared proteins was annotated as a cathepsin F, after manually curated the database they were both reclassified as cathepsin L. The alignment with several well-known cathepsins L (S1 Fig) shows that they both have the ERFNIN, GNFD and GCNGG motifs (identified with an * on S1 Fig) characteristic of the cathepsin L. This enzyme plays several essential roles in parasite survival and host-parasite interactions. They participate in nutrient acquisition and they are part of the secretome of several parasites [4749]. Cathepsin L have several functions such as nutrient acquisition by the digestion of host proteins, cleavage of the host extracellular matrix allowing parasite migration, and the impairment of the host immunity by destroying immunoglobulin and suppressing the Th1 immune response. One of the cathepsin (accession number KAF6769383.1) shares 70% sequence similarity with cathepsin Ls from P. westermani and P. pseudoheterotremus detected in adult SSP and ESP samples and is specifically expressed in the adult parasite digestive system [50]. They also demonstrate that this protein is highly sensitive and specific for serodiagnosis of human paragonimiasis, but no studies of species specificity or cross-reactivity have been performed. In Fasciola gigantica, cathepsin L was used as an antigen to develop a rapid diagnostic test for human Fasciola infection that was highly sensitive [51]. In the parasite-related shared proteins, four Ras-related proteins were found. Ras-related protein Rab-11B, Ras-related protein Rab-7a, Ras-related protein Rab-8B and Ras-related protein Rap-1b. These proteins belong to the small GTP-binding proteins family, part of the Ras superfamily, which regulate intracellular membrane trafficking of several parasites [5254]. In F. hepatica was demonstrated that Ras proteins interacts with host immune system mediators and promotes monocytes phagocytosis and inhibits cell proliferation [52]. If most Ras-related proteins have a similar function when secreted, it may help explain how endoparasites recognize host immune cells and evade them to survive.

Concluding remarks

To our knowledge, this is the first study that has compared in vitro and in vivo ESP for any Paragonimus species. Although analysis of ESPs produced in vitro can be helpful, in vitro culture does not reproduce the true biological and chemical conditions faced by the parasite inside the human host. Moreover, is well known that parasite secretomes undergo substantial changes in composition during their life cycle due to adaptation to different environments. Regarding the difference in the protein number between the ESP and CFP it is important to take into account that some proteins can be released into the cyst but diffuse from there to other host tissues. Therefore, the opportunity to analyze fluid from cysts that contained flukes allowed us to compare proteins secreted in vivo with proteins were found in culture medium after in vitro culture. Our results provide new insights into host-parasite interactions for an important food-borne parasitic trematode at the molecular level. Parasite proteins present in cyst fluid may also have practical value, because they could lead to new treatments or improved diagnostic tests for paragonimiasis.

Supporting information

S1 Table. Hyperlinked Excel spreadsheet with the functional annotation of the parasite-related-proteins and classification of the matched proteins by BLASTP searches against several databases for the three samples.

https://doi.org/10.1371/journal.pntd.0010679.s001

(XLSX)

S2 Table. Hyperlinked Excel spreadsheet with the functional annotation of the host-related-proteins and classification of the matched proteins by BLASTP searches against M. unguiculatus database for the SSP and ESP sample.

https://doi.org/10.1371/journal.pntd.0010679.s002

(XLSX)

S3 Table. Host derived protein list for the CFP.

The proteins are ordered from the most to the least abundant in the sample.

https://doi.org/10.1371/journal.pntd.0010679.s003

(XLSX)

S1 Fig. Amino acid sequences of P. kellicotti cathepsin L-like proteases (KAF6777412.1 and KAF6769383.1) aligned with known cathepsin L-like proteases retrieved from GenBank.

Conserved cathepsin L motifs (ERFNIN, GNFD and GCNGG) were marked with an *.

https://doi.org/10.1371/journal.pntd.0010679.s004

(PDF)

Acknowledgments

The authors would like to thank Dr. Yiling Mi, Alan Davis, Rose Connors and James Malone for their expert proteomics assistance and Dr. Bruce Rosa for the guidance during the proteomic analysis.

References

  1. 1. Furst T, Keiser J, Utzinger J. Global burden of human food-borne trematodiasis: a systematic review and meta-analysis. Lancet Infect Dis. 2012;12(3):210–21. WOS:000300861400026. pmid:22108757
  2. 2. Johannesen E, Nguyen V. Paragonimus kellicotti: A Lung Infection in Our Own Backyard. Case Rep Pathol. 2016;2016. Unsp 210737210.1155/2016/2107372. WOS:000378269600001. pmid:27213066
  3. 3. Fischer PU, Curtis KC, Marcos LA, Weil GJ. Molecular characterization of the North American lung fluke Paragonimus kellicotti in Missouri and its development in Mongolian gerbils. Am J Trop Med Hyg. 2011;84(6):1005–11. pmid:21633042; PubMed Central PMCID: PMC3110363.
  4. 4. Rosa BA, Choi YJ, McNulty SN, Jung H, Martin J, Agatsuma T, et al. Comparative genomics and transcriptomics of 4 Paragonimus species provide insights into lung fluke parasitism and pathogenesis. Gigascience. 2020;9(7). ARTN giaa07310.1093/gigascience/giaa073. WOS:000569171300004. pmid:32687148
  5. 5. McNulty SN, Fischer PU, Townsend RR, Curtis KC, Weil GJ, Mitreva M. Systems Biology Studies of Adult Paragonimus Lung Flukes Facilitate the Identification of Immunodominant Parasite Antigens. Plos Neglect Trop D. 2014;8(10). ARTN e324210.1371/journal.pntd.0003242. WOS:000344589000054. pmid:25329661
  6. 6. Lee EG, Na BK, Bae YA, Kim SH, Je EY, Ju JW, et al. Identification of immunodominant excretory-secretory cysteine proteases of adult Paragonimus westermani by proteome analysis. Proteomics. 2006;6(4):1290–300. WOS:000235756400023. pmid:16404718
  7. 7. Na BK, Kim SH, Lee EG, Kim TS, Bae YA, Kang I, et al. Critical roles for excretory-secretory cysteine proteases during tissue invasion of Paragonimus westermani newly excysted metacercariae. Cell Microbiol. 2006;8(6):1034–46. WOS:000237394900012. pmid:16681843
  8. 8. Hewitson JP, Grainger JR, Maizels RM. Helminth immunoregulation: The role of parasite secreted proteins in modulating host immunity. Mol Biochem Parasit. 2009;167(1):1–11. WOS:000268049600001. pmid:19406170
  9. 9. Sotillo J, Pearson MS, Becker L, Mekonnen GG, Amoah AS, van Dam G, et al. In-depth proteomic characterization of Schistosoma haematobium: Towards the development of new tools for elimination. Plos Neglect Trop D. 2019;13(5). ARTN e000736210.1371/journal.pntd.0007362. WOS:000470188100038. pmid:31091291
  10. 10. McManus DP, Dalton JP. Vaccines against the zoonotic trematodes Schistosoma japonicum, Fasciola hepatica and Fasciola gigantica. Parasitology. 2006;133:S43–S61. WOS:000244707100004. pmid:17274848
  11. 11. Lightowlers MW, Rickard MD. Excretory Secretory Products of Helminth-Parasites—Effects on Host Immune-Responses. Parasitology. 1988;96:S123–S66. WOS:A1988M840500009. pmid:3287288
  12. 12. Wisniewski JR, Zougman A, Nagaraj N, Mann M. Universal sample preparation method for proteome analysis. Nat Methods. 2009;6(5):359–62. Epub 20090419. pmid:19377485.
  13. 13. Chen ZW, Fuchs K, Sieghart W, Townsend RR, Evers AS. Deep Amino Acid Sequencing of Native Brain GABA(A) Receptors Using High-Resolution Mass Spectrometry. Mol Cell Proteomics. 2012;11(1). ARTN M111.01144510.1074/mcp.M111.011445. WOS:000299106200011. pmid:22338125
  14. 14. Meier F, Brunner AD, Koch S, Koch H, Lubeck M, Krause M, et al. Online Parallel Accumulation Serial Fragmentation (PASEF) with a Novel Trapped on Mobility Mass Spectrometer. Mol Cell Proteomics. 2018;17(12):2534–45. WOS:000452476000018. pmid:30385480
  15. 15. Perkins DN, Pappin DJC, Creasy DM, Cottrell JS. Probability-based protein identification by searching sequence databases using mass spectrometry data. Electrophoresis. 1999;20(18):3551–67. WOS:000085044900009. pmid:10612281
  16. 16. Cheng SF, Fu Y, Zhang YL, Xian WF, Wang HL, Grothe B, et al. Enhancement of de novo sequencing, assembly and annotation of the Mongolian gerbil genome with transcriptome sequencing and assembly from several different tissues. Bmc Genomics. 2019;20(1). ARTN 90310.1186/s12864-019-6276-y. WOS:000500019400002. pmid:31775624
  17. 17. Nesvizhskii AI, Keller A, Kolker E, Aebersold R. A statistical model for identifying proteins by tandem mass spectrometry. Anal Chem. 2003;75(17):4646–58. WOS:000185192300049. pmid:14632076
  18. 18. Karim S, Singh P, Ribeiro JMC. A Deep Insight into the Sialotranscriptome of the Gulf Coast Tick, Amblyomma maculatum. Plos One. 2011;6(12). ARTN e2852510.1371/journal.pone.0028525. WOS:000299113600020. pmid:22216098
  19. 19. Abdi H, Williams LJ. Principal component analysis. Wires Comput Stat. 2010;2(4):433–59. WOS:000214679600004.
  20. 20. Cwiklinski K, de la Torre-Escudero E, Trelis M, Bernal D, Dufresne PJ, Brennan GP, et al. The Extracellular Vesicles of the Helminth Pathogen, Fasciola hepatica: Biogenesis Pathways and Cargo Molecules Involved in Parasite Pathogenesis. Mol Cell Proteomics. 2015;14(12):3258–73. WOS:000365638100013. pmid:26486420
  21. 21. Marcilla A, Trelis M, Cortes A, Sotillo J, Cantalapiedra F, Minguez MT, et al. Extracellular Vesicles from Parasitic Helminths Contain Specific Excretory/Secretory Proteins and Are Internalized in Intestinal Host Cells. Plos One. 2012;7(9). ARTN e4597410.1371/journal.pone.0045974. WOS:000309554700099. pmid:23029346
  22. 22. Sanchez-Lopez CM, Trelis M, Jara L, Cantalapiedra F, Marcilla A, Bernal D. Diversity of extracellular vesicles from different developmental stages of Fasciola hepatica. Int J Parasitol. 2020;50(9):663–9. WOS:000558594000006. pmid:32531305
  23. 23. Trelis M, Sanchez-Lopez CM, Sanchez-Palencia LF, Ramirez-Toledo V, Marcilla A, Bernal D. Proteomic Analysis of Extracellular Vesicles From Fasciola hepatica Hatching Eggs and Juveniles in Culture. Front Cell Infect Mi. 2022;12. ARTN 90360210.3389/fcimb.2022.903602. WOS:000812847800001. pmid:35719328
  24. 24. Tirloni L, Reck J, Terra RMS, Martins JR, Mulenga A, Sherman NE, et al. Proteomic Analysis of Cattle Tick Rhipicephalus (Boophilus) microplus Saliva: A Comparison between Partially and Fully Engorged Females. Plos One. 2014;9(4). ARTN e9483110.1371/journal.pone.0094831. WOS:000335505000010. pmid:24762651
  25. 25. Wang H, Nuttall PA. Excretion of host immunoglobulin in tick saliva and detection of IgG-binding proteins in tick haemolymph and salivary glands. Parasitology. 1994;109 (Pt 4):525–30. pmid:7794319.
  26. 26. Wilson RA, Jones MK. Fifty years of the schistosome tegument: discoveries, controversies, and outstanding questions. Int J Parasitol. 2021;51(13–14):1213–32. WOS:000732747300011. pmid:34767805
  27. 27. Torres-Rivera A, Landa A. Glutathione transferases from parasites: A biochemical view. Acta Trop. 2008;105(2):99–112. WOS:000253323300001. pmid:17897613
  28. 28. Hong SJ, Kang SY, Chung YB, Chung MH, Oh YJ, Kang I, et al. Paragonimus westermani: A cytosolic glutathione S-transferase of a sigma-class in adult stage. Exp Parasitol. 2000;94(3):180–9. WOS:000087161700007. pmid:10831383
  29. 29. Zhan B, Perally S, Brophy PM, Xue J, Goud G, Liu S, et al. Molecular Cloning, Biochemical Characterization, and Partial Protective Immunity of the Heme-Binding Glutathione S-Transferases from the Human Hookworm Necator americanus. Infect Immun. 2010;78(4):1552–63. WOS:000275656400014. pmid:20145100
  30. 30. Figueroa-Santiago O, Espino AM. Fasciola hepatica Fatty Acid Binding Protein Induces the Alternative Activation of Human Macrophages. Infect Immun. 2014;82(12):5005–12. WOS:000346958400011. pmid:25225247
  31. 31. Cass CL, Johnson JR, Califf LL, Xu T, Hernandez HJ, Stadecker MJ, et al. Proteomic analysis of Schistosoma mansoni egg secretions. Mol Biochem Parasitol. 2007;155(2):84–93. Epub 20070619. pmid:17644200; PubMed Central PMCID: PMC2077830.
  32. 32. Perez-Morales D, Espinoza B. The role of small heat shock proteins in parasites. Cell Stress Chaperones. 2015;20(5):767–80. Epub 20150605. pmid:26045203; PubMed Central PMCID: PMC4529861.
  33. 33. Ranasinghe SL, McManus DP. Protease Inhibitors of Parasitic Flukes: Emerging Roles in Parasite Survival and Immune Defence. Trends Parasitol. 2017;33(5):400–13. WOS:000401218100010. pmid:28089171
  34. 34. He BH, Cai GB, Ni YH, Li Y, Zong HY, He L. Characterization and expression of a novel cystatin gene from Schistosoma japonicum. Mol Cell Probe. 2011;25(4):186–93. WOS:000292624500009. pmid:21601634
  35. 35. Sadaow L, Sanpool O, Rodpai R, Yamasaki H, Ittiprasert W, Mann VH, et al. Development of an Immunochromatographic Point-of-Care Test for Serodiagnosis of Opisthorchiasis and Clonorchiasis. American Journal of Tropical Medicine and Hygiene. 2019;101(5):1156–60. WOS:000497187300036. pmid:31482789
  36. 36. Phupiewkham W, Sadaow L, Sanpool O, Rodpai R, Yamasaki H, Ittiprasert W, et al. Comparative assessment of immunochromatographic test kits using somatic antigens from adult Opisthorchis viverrini and IgG and IgG4 conjugates for serodiagnosis of human opisthorchiasis. Parasitol Res. 2021;120(8):2839–46. WOS:000673447300002. pmid:34259939
  37. 37. Wang T, Ma GX, Ang CS, Korhonen PK, Koehler AV, Young ND, et al. High throughput LC-MS/MS-based proteomic analysis of excretory-secretory products from short-term in vitro culture of Haemonchus contortus. J Proteomics. 2019;204. ARTN 10337510.1016/j.jprot.2019.05.003. WOS:000475994800009. pmid:31071474
  38. 38. Gretes MC, Poole LB, Karplus PA. Peroxiredoxins in Parasites. Antioxid Redox Sign. 2012;17(4):608–33. WOS:000305470500006. pmid:22098136
  39. 39. Jang HH, Lee KO, Chi YH, Jung BG, Park SK, Park JH, et al. Two enzymes in one: Two yeast peroxiredoxins display oxidative stress-dependent switching from a peroxidase to a molecular chaperone function. Cell. 2004;117(5):625–35. WOS:000221857900009. pmid:15163410
  40. 40. Haque SJ, Majumdar T, Barik S. Redox-Assisted Protein Folding Systems in Eukaryotic Parasites. Antioxid Redox Sign. 2012;17(4):674–83. WOS:000305470500009. pmid:22122448
  41. 41. Robinson MW, Hutchinson AT, Dalton JP, Donnelly S. Peroxiredoxin: a central player in immune modulation. Parasite Immunol. 2010;32(5):305–13. WOS:000276495800001. pmid:20500659
  42. 42. Ishii T, Warabi E, Yanagawa T. Novel roles of peroxiredoxins in inflammation, cancer and innate immunity. J Clin Biochem Nutr. 2012;50(2):91–105. WOS:000302525900001. pmid:22448089
  43. 43. Angelucci F, Miele AE, Ardini M, Boumis G, Saccoccia F, Bellelli A. Typical 2-Cys peroxiredoxins in human parasites: Several physiological roles for a potential chemotherapy target. Mol Biochem Parasit. 2016;206(1–2):2–12. WOS:000379373600002. pmid:27002228
  44. 44. Mourao MD, Dinguirard N, Franco GR, Yoshino TP. Phenotypic Screen of Early-Developing Larvae of the Blood Fluke, Schistosoma mansoni, using RNA Interference. Plos Neglect Trop D. 2009;3(8). ARTN e50210.1371/journal.pntd.0000502. WOS:000269220900014. pmid:19668375
  45. 45. Sayed AA, Cook SK, Williams DL. Redox balance mechanisms in Schistosoma mansoni rely on peroxiredoxins and albumin and implicate peroxiredoxins as novel drug targets. J Biol Chem. 2006;281(25):17001–10. WOS:000238326300022. pmid:16606626
  46. 46. Donnelly S, Stack CM, O’Neill SM, Sayed AA, Williams DL, Dalton JP. Helminth 2-Cys peroxiredoxin drives Th2 responses through a mechanism involving alternatively activated macrophages. Faseb J. 2008;22(11):4022–32. WOS:000260503000030. pmid:18708590
  47. 47. Di Maggio LS, Tirloni L, Pinto AFM, Diedrich JK, Yates JR, Benavides U, et al. Across intra-mammalian stages of the liver fluke Fasciola hepatica: a proteomic study. Sci Rep-Uk. 2016;6. ARTN 3279610.1038/srep32796. WOS:000382611200001. pmid:27600774
  48. 48. Suttiprapa S, Sotillo J, Smout M, Suyapoh W, Chaiyadet S, Tripathi T, et al. Opisthorchis viverrini Proteome and Host-Parasite Interactions. Adv Parasit. 2018;102:45–72. WOS:000452100400004. pmid:30442310
  49. 49. Young ND, Campbell BE, Hall RS, Jex AR, Cantacessi C, Laha T, et al. Unlocking the Transcriptomes of Two Carcinogenic Parasites, Clonorchis sinensis and Opisthorchis viverrini. Plos Neglect Trop D. 2010;4(6). ARTN e71910.1371/journal.pntd.0000719. WOS:000279341300025. pmid:20582164
  50. 50. Yoonuan T, Nuamtanong S, Dekumyoy P, Phuphisut O, Adisakwattana P. Molecular and immunological characterization of cathepsin L-like cysteine protease of Paragonimus pseudoheterotremus. Parasitol Res. 2016;115(12):4457–70. WOS:000388191400002. pmid:27562899
  51. 51. Sadaow L, Yamasaki H, Morishima Y, Sanpool O, Rodpai R, Janwan P, et al. Effectiveness of Fasciola gigantica excretory-secretory and recombinant cathepsin L antigens for rapid diagnosis of human fascioliasis using immunochromatographic devices. Parasitol Res. 2020;119(11):3691–8. WOS:000574792100004. pmid:33009945
  52. 52. Tian AL, Lu MM, Zhang FK, Calderon-Mantilla G, Petsalaki E, Tian XW, et al. The pervasive effects of recombinant Fasciola gigantica Ras-related protein Rab10 on the functions of goat peripheral blood mononuclear cells. Parasite Vector. 2018;11. ARTN 57910.1186/s13071-018-3148-2. WOS:000449464500002. pmid:30400957
  53. 53. Langsley G, van Noort V, Carret C, Meissner M, de Villiers EP, Bishop R, et al. Comparative genomics, of the Rab protein family in Apicomplexan parasites. Microbes Infect. 2008;10(5):462–70. WOS:000256982600002. pmid:18468471
  54. 54. Kremer K, Kamin D, Rittweger E, Wilkes J, Flammer H, Mahler S, et al. An Overexpression Screen of Toxoplasma gondii Rab-GTPases Reveals Distinct Transport Routes to the Micronemes. Plos Pathog. 2013;9(3). ARTN e100321310.1371/journal.ppat.1003213. WOS:000316953800018.