Skip to main content
Advertisement
  • Loading metrics

Cysteine proteases in protozoan parasites

  • Jair L. Siqueira-Neto ,

    jairlage@ucsd.edu

    Affiliation Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America

  • Anjan Debnath,

    Affiliation Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America

  • Laura-Isobel McCall,

    Current address: Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, United States of America

    Affiliation Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America

  • Jean A. Bernatchez,

    Affiliation Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America

  • Momar Ndao,

    Affiliations National Reference Centre for Parasitology, The Research Institute of the McGill University Health Center, Montreal, Canada, Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada

  • Sharon L. Reed,

    Affiliation Departments of Pathology and Medicine, University of California San Diego School of Medicine, La Jolla, California, United States of America

  • Philip J. Rosenthal

    Affiliation Department of Medicine, University of California, San Francisco, San Francisco, California, United States of America

Abstract

Cysteine proteases (CPs) play key roles in the pathogenesis of protozoan parasites, including cell/tissue penetration, hydrolysis of host or parasite proteins, autophagy, and evasion or modulation of the host immune response, making them attractive chemotherapeutic and vaccine targets. This review highlights current knowledge on clan CA cysteine proteases, the best-characterized group of cysteine proteases, from 7 protozoan organisms causing human diseases with significant impact: Entamoeba histolytica, Leishmania species (sp.), Trypanosoma brucei, T. cruzi, Cryptosporidium sp., Plasmodium sp., and Toxoplasma gondii. Clan CA proteases from three organisms (T. brucei, T. cruzi, and Plasmodium sp.) are well characterized as druggable targets based on in vitro and in vivo models. A number of candidate inhibitors are under development. CPs from these organisms and from other protozoan parasites should be further characterized to improve our understanding of their biological functions and identify novel targets for chemotherapy.

Introduction

Proteases are enzymes that catalyze the hydrolysis of peptide bonds and are important in a number of biological activities, including digestion of peptides, activation of other enzymes, modulation of the immune system, participation in the cell cycle, and differentiation and autophagy. There are at least 6 classes of proteases classified according to the nucleophilic group responsible for the first step in the proteolysis: serine, cysteine, metallo, aspartate, glutamate, and threonine proteases. Cysteine proteases (CPs) are categorized into 72 families, but not all are represented in protozoan parasites [1]. The most abundant and well characterized CPs in these organisms are the clan CA papain-family enzymes, named after an abundant protease present in papaya fruit. We selected 7 protozoan organisms with medical relevance for review of the current knowledge regarding their clan CA CPs. Table 1 summarizes well-characterized clan CA CPs from these organisms, and Table 2 lists some well-studied inhibitors.

Despite the essentiality of CPs for parasite survival, there are, as yet, no approved drugs targeting this group of enzymes. The most advanced drug in development against a parasite CP is the vinyl sulfone inhibitor K11777, with in vivo efficacy in an animal model of Chagas disease [2, 3]; K11777 is currently moving forward to clinical trials.

Entamoeba histolytica

E. histolytica causes amebiasis and is responsible for about 70,000 deaths per year [4, 5]. The parasite infects the intestinal tract and can cause diarrhea, colitis, and peritonitis. Extraintestinal amebiasis can also occur, commonly leading to amoebic liver abscesses. E. histolytica CPs are encoded by approximately 50 genes and are responsible for epithelial barrier penetration and degradation of host extracellular matrix components. Only four proteases, EhCP1, EhCP2, EhCP5, and EhCP7, are highly expressed by E. histolytica in culture and have identified cellular localizations. EhCP1 is present in intracellular vesicles, EhCP2 localizes to the internal and external cell membrane, and EhCP5 is on the cell surface. Invasive E. histolytica clinical isolates have 10×–1,000× more CP activity in culture than noninvasive E. dispar isolates, suggesting a role for CPs in parasite virulence and invasion [6].

Secreted E. histolytica CPs cleave the colonic mucus layer and overcome mucosal host defenses in the gut [7]. Villin and tight junction proteolysis are also mediated by these CPs [8]. EhCP1, EhCP2, and EhCP5 have been shown to degrade extracellular matrix components, such as fibronectin, laminin, and collagen [9]. Transcriptional studies identified increased expression of EhCP1 and EhCP2 when E. histolytica interacted with human collagen and mucin [10]. Those results corroborate the key role of these CPs in parasite invasion of host tissues.

E. histolytica CPs are also hypothesized to interfere with both innate and adaptive host immune responses. They degrade human IgA, the most abundant immunoglobulin defense at the mucosal surface [11]. Extracellular CPs disrupt the heavy chain of IgG, and this is considered to be a key strategy for E. histolytica survival during tissue invasion [12]. E. histolytica CPs activate the alternative pathway of the host complement system and generate hemolytically active C3b, but extracellular CPs circumvent host immunity by degrading and inactivating anaphylatoxins C3a and C5a [13]. E. histolytica CPs modulate cell-mediated immunity as well. Extracellular CPs degrade pro–IL-1β that is released by damaged intestinal epithelial cells to form the active mature proinflammatory cytokine IL-1β [14]. On the other hand, E. histolytica CPs cleave both pro–and mature IL-18 [15], and the degradation of IL-18 may contribute to either parasite or host survival. A recent study demonstrated the function of EhCP1, EhCP4, and EhCP5 in the intercellular junction of E. histolytica and macrophages to modulate the host cell cytoskeleton and trigger subsequent IL-1β secretion, detailing the cascade of molecular events triggered by the initial interaction of E. histolytica with macrophages [16]. That study suggests new roles for EhCP1 and EhCP4 in inducing a proinflammatory response by host macrophages upon cellular contact. EhCPs may also contribute to parasite encystation–excystation [17] and in nutrient acquisition through erythrophagocystosis [18], both essential to parasite infectivity and survival.

The vinyl sulfone inhibitors K11777 and WRR483 were tested against recombinant EhCP1, and both demonstrated potent inhibitory activity, significantly reducing parasite invasion of human colonic xenografts [19]. Further studies should be carried out to develop an antiamebiasis drug targeting CPs.

Leishmania sp.

Leishmaniasis is a spectrum of diseases, including skin ulcers; degradation of nose and palate mucosal tissue; and liver, spleen, and bone marrow infiltration. This latter visceral form of the disease is the most lethal, killing over 60,000 people per year [20]. The factors determining clinical manifestations are not fully understood; these are strongly influenced by parasite strain or species and by immunological factors [21]. Leishmania express a broad range of CPs, the best characterized of which are CPA, CPB, and CPC (CPs A, B, and C), all part of clan CA, family C1.

CPA and CPB are cathepsin L-like and likely show some functional redundancy [22], while CPC is cathepsin B-like. Cathepsin B-like and cathepsin L-like enzymes have key structural differences associated with divergent substrate binding and proteolytic mechanisms. In cathepsin L, binding of the peptide substrates spans the entire channel between the two domains of the protein, leading to endopeptidolytic activity. In contrast, cathepsin B has an additional occlusion loop that restricts substrate binding under low pH, leading to carboxypeptidase activity; this loop is displaced at high pH, at which point cathepsin B displays endopeptidase activity [23, 24].

CPB genes are arranged in tandem arrays [25], with 19 copies in L. mexicana, 8 in L. major [26], and 5 in L. chagasi [27], while CPA and CPC are single copy [28, 29]. CP gene expression is stage regulated; most are expressed at higher levels in the mammalian amastigote stage than in the insect promastigote stage [25, 29]. However, CPB1 and CPB2 are expressed at higher levels in metacyclic promastigotes [30] and CPC in procyclic promastigotes [28]. Consistent with elevated CP expression in the amastigote stage, Leishmania CPs play key roles in the interactions between Leishmania and its mammalian host. CPA-knockdown or knockout L. chagasi and L. infantum show decreased in vitro [31] and in vivo infectivity [32]. Likewise, CPB-knockout L. mexicana displayed impaired macrophage infectivity and delayed lesion progression [33]. Multiple CPB copies are required for significant restoration of virulence [34]. CPB modulates host responses to L. mexicana by down-regulating protective Th1 immune responses, and in particular IFN-γ production, via degradation of the transcription factor NF-κB and subsequent inhibition of IL-12 production by infected macrophages [35]. L. mexicana CPs also cleave JNK and ERK MAP kinases. Both kinases negatively regulate IL-12 production, and therefore the protozoan cysteine protease CPB can alter host macrophage signaling by increasing IL-12 transcription [35]. L. amazonensis likewise inhibits antigen presentation via CP-mediated degradation of MHC class II molecules [36]. CPB also modulates levels of parasite proteins, including gp63 [37], a major virulence factor in Leishmania. Episomal expression of gp63 in CPB-deficient parasites was sufficient to restore virulence to wild-type levels [37]. Several of the effects attributed to CPB via knockout studies may therefore occur via modulation of other parasite proteins rather than through direct cleavage by CPB. CPC, in contrast, is involved in Leishmania cell death [38] and secretome remodeling [39]; knockouts were attenuated in macrophage infection in vitro but were nevertheless able to cause lesions in vivo, suggesting compensatory mechanisms [40].

Other less-studied clan CA members in Leishmania include Atg4.1 and Atg4.2 (clan CA, family C54) and otubain-like enzymes (clan CA, family C65). Atg4.2 knockout was associated with impaired metacyclogenesis, altered autophagosome accumulation (especially under nutritional stress), increased amastigote length, and impaired amastigote proliferation; in contrast, Atg4.1 knockout only showed minor effects on promastigote infectivity [41]. A novel otubain-like CP is a deubiquitinating enzyme recently characterized from L. infantum that may also have proinflammatory activity by stimulating lipid droplet biogenesis and inducing IL-6 and TNF-α secretion [42].

Early work with vinyl sulfone and dihydrazide compounds showed successful in vivo treatment of cutaneous leishmaniasis by CPC inhibition [43]. Since then, several other CPC inhibitors have shown in vivo efficacy to treat cutaneous (palladacycle DPPE 1.2 [44]; organotellurane RT-01 [45]) and visceral (organotellurane RF07 [46]) leishmaniasis. In vitro assays identified multiple compound classes active against CPB (e.g., semicarbazones, thiosemicarbazones, triazine nitriles [47], and benzophenones [48]), but these have yet to be tested in vivo.

Trypanosoma brucei

Human African trypanosomiasis (sleeping sickness), caused by various subspecies of T. brucei, is found in sub-Saharan Africa. The flagellated parasite invades the blood–brain barrier, causing fatal damage in the central nervous system, with approximately 7,000 deaths per year [20]. Similar to Leishmania CPs, the best characterized T. brucei CPs are cathepsin-like enzymes, cathepsin L (TbCatL, also known as brucipain and rhodesain in different T. brucei subspecies) and cathepsin B (TbCatB) [49].

TbCatL and TbCatB are both involved in virulence. TbCatL promotes parasite crossing of the blood–brain barrier via activation of host G-protein–coupled receptors such as PAR2 (protease-activated receptor 2) and subsequent induction of host calcium-signaling pathways [50, 51]. TbCatL also protects T. brucei from lysis by host serum [52, 53] and from opsonization via degradation of internalized variant surface-glycoprotein–bound antibodies [54]. As for TbCatL [55], TbCatB expression is greatest in the bloodstream stage [56]; it mediates the degradation of endocytosed host transferrin in the parasite endosomal/lysosomal compartment as part of T. brucei iron acquisition pathways [57, 58]. TbCatB may also be involved in cytokinesis [56]. A number of studies using RNA interference (RNAi) [58] or small-molecule inhibitors in vitro [59] and in animal infection models [60, 61] have validated these proteases as drug targets. K11777 in particular targets TbCatL: it showed synergy when combined with eflornithine and might be considered for development as a new therapy for African trypanosomiasis [62]. Other small molecules have been successfully tested against TbCatL, including dipeptide nitriles [63] and triazine nitriles [64], but their efficacy in vivo is yet to be determined. A recent review highlights a thorough list of candidate T. brucei and T. cruzi CP inhibitors [65].

Trypanosoma cruzi

Chagas disease is endemic in the Americas and is the main cause of heart failure in Latin America, leading to more than 12,000 deaths every year [66]. It is caused by the parasite T. cruzi, which encodes genes from four clans of CPs: CA, CD, CE, and CF. Clan CA includes the most abundant protease of this parasite: cruzain (cruzipain), a papain-like cathepsin L-like member of family C1.

Cruzain is present in epimastigotes and bloodstream trypomastigotes and is identified as a major antigen in infected humans [67], and for that reason, it has been considered as a vaccine candidate [68]. It plays important roles in differentiation [69], metabolism [70], evasion of the immune response, and invasion of host cells [71]. In trypomastigotes (infective stage), cruzain is localized in the flagellar pocket, and in intracellular amastigotes, the enzyme is on the cell surface. Recombinant cruzain was expressed in bacteria and demonstrated protease activity after autocatalytic activation [72]. The crystal structure of cruzain complexed with the inhibitor Z-Phe-Ala-fluoromethyl ketone was resolved, confirming structural similarities with papain.

Genes coding for a 30-kDa cathepsin B-like CP, two other cathepsins, and other homologues of calpains (family C2) are also present [74]. Other members of clan CA include autophagin-like Atg4 protease (family of C54), responsible for processing Atg8 for the formation of the autophagosomes involved in the parasitic autophagy process essential for metacyclogenesis and virulence [75, 76]. Atg4 has recently been proposed as a potential new target for chemotherapy [76].

The idea of developing CP inhibitors as Chagas disease chemotherapy originated in observations of in vitro antiparasitic effects associated with cruzain inhibition. Because of multiple copies of the cruzain gene, gene knockout could not be achieved to confirm the essentiality of the enzyme, even though “chemical validation” with protease inhibitors suggested that it is essential. The first proof of concept in an animal model showed that mice could be rescued from a lethal T. cruzi infection with the vinyl sulfone inhibitor K11777 dosed for 20 days [3]. Surviving mice had negative hemoculture, indicating parasitological cure. The same inhibitor was later tested in a dog model of Chagas disease, and it prevented cardiomyopathy after 7 days of treatment [2].

A number of other cruzain inhibitors are under study. Computational screening of the ZINC database identified inhibitors with nanomolar potency against cruzain [77]. Some isatins also have the thiosemicarbazone functionality, a well-known cruzain inhibitor group [78]. However, isatin compounds without a thiosemicarbazone also demonstrated inhibitory activity against cruzain [79]. These compounds have a peptide sequence recognized by the catalytic site of cruzain as well as the epoxy electrophile reminiscent of the E-64 inhibitor. Odanacatib, a reversible inhibitor of human cathepsin K with an amino nitrile warhead, was in Phase III trials to treat osteoporosis, but its development was discontinued due to risk of stroke (see the associated chapter, “Cysteine proteases as digestive enzymes in parasitic helminths,” for information on odanacatib efficacy against hookworm infection). The nitrile warhead forms a reversible (albeit with a slow off rate) thioimidate with the active site of human cathepsin K [80]. Inspired by this approach, reversible inhibitors have been developed against cruzain with 100× selectivity compared to human cathepsins L, B, S, and F. Further optimization of these compounds led to the discovery of Cz007 and Cz008, with IC50s against cultured parasites in the nanomolar range and antiparasitic efficacy in a mouse model of Chagas disease [81]. More recently, the vinyl sulfone WRR-669 was demonstrated to be a noncovalent inhibitor of cruzain. These results, showing both in vitro and in vivo efficacy against T. cruzi, support cruzain as a valid and druggable target for Chagas disease. A review published in 2015 highlights inhibitors tested against cruzain [83].

Cryptosporidium parvum

Cryptosporidiosis, caused by Cryptosporidium sp., is a concern worldwide. In a large study in sub-Saharan Africa and southern Asia, this protozoan was the second most common cause of moderate-to-severe diarrhea in infants and the third most common cause in toddlers, accounting for an estimated 2.9 and 4.7 million cases annually in children <2 years old in these regions, respectively [84]. Acquired immune deficiency syndrome (AIDS) patients, people under immunosuppressive treatments, patients with inheritable immunodeficiency, diabetic people, infants, and old or malnourished people are the most susceptible to severe cryptosporidiosis [85]. Outbreaks have been described among caregivers and students of veterinary hospitals after contact with calves infected with C. parvum, the zoonotic species [86]. A major outbreak was reported in Milwaukee in 1993, in which 403,000 people were infected and the cost of outbreak-associated illness was US$96.2 million [87, 88].

Genes encoding 20 clan CA cathepsin L-like proteases have been reported in Cryptosporidium parvum. Five genes coding for cryptopains—cathepsin L-like proteases, a representative of clan CA—were identified in the C. parvum genome [89], but only cryptopain-1 has been analyzed biochemically [90]. Cryptopain-1 is actively transcribed and expressed in sporozoites, the infectious stage of the parasite [90]. At physiologically achievable concentrations, K11777 arrested the growth of C. parvum in human intestinal cell lines [91]. In C57BL/6 interferon-γ receptor knockout mice, which are highly susceptible to C. parvum infection (mimicking cryptosporidiosis in AIDS patients), K11777 administered either orally or intraperitoneally rescued mice from a lethal C. parvum infection [91].This potent anticryptosporidial activity is hypothesized to be the result of K11777 inhibiting the active site of cryptopain-1 [91].

Recently, otubain protease (OTU), a CP that participates in the ubiquitin pathway, has also been identified [92]. The biochemical properties of the otubain-like CP of C. parvum (CpOTU) were characterized, and the enzyme may have an essential function during the oocyst stage of the parasite, when its expression reached maximum levels [93]. The protein contains an unusual C-terminal extension (217 amino acids) compared to other OTUs previously identified in human, mouse, and Drosophila, and deletion of the extension resulted in complete loss of enzyme activity.

Plasmodium sp.

Malaria is by far the deadliest parasitic disease of humans, with 446,000 or 631,000 [94, 95] deaths estimated in 2015 using different modeling approaches. Plasmodium falciparum and P. vivax are the most common species infecting humans, with P. falciparum responsible for nearly all deaths. The parasites express multiple CPs, some of which are the subjects of recent reviews [9698]. For P. falciparum, the genome sequence predicts 33 CP-like proteins, although a number of these are probably not active enzymes. The best characterized are 4 falcipains and 3 dipeptidyl peptidases, all clan CA proteases [96].

The functions of plasmodial CPs have been characterized using selective CP inhibitors and in some cases by gene knockout. Erythrocytic malaria parasites import erythrocyte cytosol and degrade hemoglobin in an acidic food vacuole as a source of amino acids [99] in a cooperative process involving enzymes of multiple catalytic classes, including CPs, aspartic proteases, and metalloproteases. Incubating parasites with broadly active CP inhibitors causes the food vacuole to swell and fill with undegraded hemoglobin, suggesting CP essentiality in hemoglobin processing. CPs also contribute indirectly to hemoglobin hydrolysis via the processing of plasmepsins to active enzymes [100]. The CPs with clear roles in hemoglobin hydrolysis are falcipain-2, falcipain-3, and dipeptidyl aminopeptidase 1. Knockout of falcipain-2 caused the food vacuoles of P. falciparum trophozoites to fill with undegraded hemoglobin, but this abnormality resolved later in the life cycle, presumably due to expression of falcipain-3 [101]. In contrast, knockout of falcipain-3 was not tolerated, suggesting that this protease is essential [102]. Some studies have also suggested roles for CPs in erythrocyte rupture at the completion of the erythrocytic cycle or in merozoite invasion of erythrocytes [103]. CP inhibitors blocked the rupture of erythrocytes by mature schizonts. Mediators of this process are predicted to be members of the SERA family, including the pseudo-CP SERA5 [104], the functional CP SERA6 [105], and dipeptidyl aminopeptidase 3 [103, 106], although recent reports refute activity of dipeptidyl aminopeptidase 3 in erythrocyte rupture [98, 107]. Recent advances have demonstrated a proteolytic cascade responsible for egress of merozoites from host erythrocytes; this cascade includes cleavage of the actin-binding domain of the erythrocyte cytoskeletal protein β-spectrin by SERA6 to mediate erythrocyte rupture, the final step required for merozoite egress [108]. Some studies have also suggested that CPs participate in erythrocyte invasion by asexual parasites, notably falcipain-1 [109] and dipeptidyl aminopeptidase 3 [98]. However, studies with protease inhibitors have generally not supported this conclusion; knockout of falcipain-1 did not block P. falciparum development [110, 111], and antibodies against the endogenous P. falciparum CP inhibitor falstatin blocked invasion, suggesting that inhibiting falcipain-like CP activity facilitates invasion [112]. Considering another family of clan CA CP, a nucleolar calpain-like P. falciparum CP appears to be required for the development of erythrocytic parasites [113, 114]. Also, a P. falciparum otubain-like CP was recently shown to localize to the apicoplast organelle and to be required for normal apicoplast and parasite development via inhibition of the predicted role of P. falciparum Atg8 in protein import to the apicoplast [115].

CPs appear to play additional roles in nonerythrocytic plasmodial stages. Considering liver stages, an unidentified plasmodial CP cleaves the circumsporozoite protein, which coats the sporozoites injected by mosquitoes, to enable invasion of hepatocytes [116]. In the murine parasite P. berghei, the orthologue of falcipain-1 appears to be critical for invasion of erythrocytes by hepatocyte-derived merozoites [117]. Considering mosquito stages, CP inhibitors and the knockout of falcipain-1 decreased oocyst production in mosquitoes [110, 118]. Also, dipeptidyl aminopeptidase 2, which is expressed in gametocytes, may contribute to gamete egress [119].

Our understanding of the roles of CPs in the plasmodial life cycle suggests numerous potential drug targets. Falcipain-2 and falcipain-3 have low pH optima, consistent with activity in the acidic food vacuole, and both enzymes were localized to the food vacuole [120]. Falcipain-2 is more active against peptidyl substrates, uniquely able to activate and undergo autohydrolysis at neutral pH, and more stable at neutral pH. Considering specificity for peptide substrates and inhibitors, important differences were seen between falcipain-2, falcipain-3, and homologs from the rodent parasites P. berghei and P. vinckei; differences in specificity between falcipain-2 and falcipain-3 were less pronounced [121]. Both enzymes are synthesized as membrane-bound proforms that are processed, probably by autohydrolysis, to soluble mature forms. A related enzyme, falcipain-2′, is nearly identical in sequence and biochemical features to falcipain-2, but its role is uncertain, as in contrast to the case with falcipain-2 and falcipain-3, knockout of falcipain-2′ had no clear phenotype [122]. The falcipains have some unique features for papain-family proteases, including unusually long N-terminal domains and insertions in the catalytic domain. Identified functions of these domains include trafficking of falcipain-2 to the food vacuole by upstream portions of the prodomain; enzyme inhibition by downstream portions of the prodomain; mediation of enzyme folding by short peptides immediately upstream of the catalytic domain [123]; and mediation of binding to the native substrate, hemoglobin, by a small insertion near the C-terminus of the catalytic domain.

Multiple studies have demonstrated that CP inhibitors have potent antimalarial effects [124]. With these inhibitors, a block in P. falciparum development is accompanied by a specific block in hemoglobin hydrolysis, marked by the appearance of swollen, hemoglobin-filled food vacuoles, and antiparasitic effects correlated with the degree of inhibition of falcipain-2 and falcipain-3. Drug discovery directed against falcipains is facilitated by the available structures of falcipain-2 and falcipain-3 complexed with small-molecule and protein inhibitors [125].

Peptidyl falcipain inhibitors with nanomolar antimalarial activity have included fluoromethyl ketones, vinyl sulfones, and aldehydes; in some cases, in vivo activity against murine malaria has also been demonstrated [126]. Promising nonpeptidyl falcipain inhibitors have included a series of nitriles that was extensively studied with many promising features, including excellent in vitro and in vivo potency [127]; this project was halted because of tissue binding that might predict idiosyncratic toxicity, but the evaluation of nitrile inhibitors is ongoing. Another interesting approach is the optimization of natural products, including analogues of gallinamide A, a compound from cyanobacteria with nanomolar antimalarial activity [128], and sugarcane cystatin [129].

Concerning the potential for resistance, parasites were selected for resistance to a vinyl sulfone falcipain inhibitor, but the selection was slow and the mechanism of resistance complex, without mutations in target enzymes, suggesting that resistance to falcipain inhibitors may develop slowly, especially with combination therapy [130]. Considering combinations, the activity of artemisinins, the mainstay of modern treatment for falciparum malaria, requires falcipain activity [131]; thus, falcipain inhibitors should probably not be combined with artemisinins. In contrast, inhibitors of CPs and aspartic proteases showed synergistic antimalarial effects, consistent with a complementary role for these two classes of enzymes and suggesting the potential for synergistic combination antimalarial therapy [132].

Toxoplasma gondii

Toxoplasma gondii is a foodborne pathogen with seroprevalence ranging from 10%–30% in North America and northern Europe to more than 80% in areas of Latin America and Africa [133]. Most infected individuals remain asymptomatic despite lifelong infection. In contrast, congenital transmission or infection of immunocompromised patients with AIDS or organ transplants can lead to fatal, disseminated disease [133]. T. gondii CPs have been shown to be important for invasion, digestion of host proteins for nutrition [134, 137], and autophagy for cyst survival [138]. The Toxoplasma genome project revealed that the redundancy of CP genes is lower in T. gondii than in most other studied parasites. For example, E. histolytica has more than 50 genes encoding CPs with similar structure and specificity [139]. In contrast, T. gondii has genes encoding only one cathepsin B (TgCPB), one cathepsin L (TgCPL), and three cathepsin Cs (TgCPC1, 2, and 3), potentially making them more amenable drug targets. Active recombinant proteases have been expressed for all the T. gondii CPs, simplifying structure-based drug design.

TgCPB and TgCPL have been linked to host cell invasion. Targeting TgCPB with a peptidyl cathepsin B inhibitor or antisense RNA inhibited host cell invasion and in vitro growth and blocked infection in a chick embryo model of toxoplasmosis [142]. TgCPL acts as a maturase for TgCPB [143] and key adhesins, the microneme proteins MIC2-associated protein (M2AP) and MIC3. TgCPL knockouts were attenuated in virulence in acute infection in mice [134]. Both TgCPB and TgCPL have been localized in the vacuolar compartment, a lysosome-like organelle, where TgCPL has been shown to digest host cytosolic proteins. Most recently, TgCPL has been shown to be important for cyst survival in latent infection [138]. In TgCPL knockout strains or cysts incubated with the vinyl sulfone inhibitor LHVS, autophagy of autophagosomes in the vacuolar compartment was inhibited, resulting in abnormal cyst morphology and decreased survival.

The most developed peptidyl inhibitors target TgCPB and/or TgCPL. The crystal structure of TgCPL with morpholinurea-leucyl-homophenyl-vinyl sulfone has been determined, and the inhibitor can block host cell invasion [136] and cyst viability in vitro [138]. K11777 inhibits purified recombinant TgCPB and TgCPL in the nanomolar range and blocks host cell invasion, parasite replication, and viability in a chick embryo egg model [144]. Unfortunately, neither vinyl sulfone inhibitor is likely to cross the blood–brain barrier to prevent latent infection, so further optimization will be required.

The T. gondii cathepsin Cs are exopeptidases that are also potential drug targets. TgCPC1 was the most highly expressed cathepsin mRNA in tachyzoites; TgCPC3 was only identified in oocysts [137]. Both TgCPC1 and TgCPC2 localize to the dense granules and are secreted into the parasitophorous vacuole, where they degrade peptides. Both TgCPC1 and TgCPC2 were inhibited by Gly-Phe-dimethylketone, reducing parasite intracellular growth and proliferation. The same phenotype was not seen with a TgCPC1 knockout, as TgCPC2 expression was up-regulated, suggesting the importance of inhibiting both enzymes [137].

Autophagy is a key process in all eukaryotic cells to remove and recycle misfolded proteins and damaged organelles. Autophagy is likely to be important in T. gondii tachyzoites to survive extracellular stress and for bradyzoites during latent infection [138]. Although only a limited number of autophagy proteins (Atg) are encoded in the T. gondii genome, and there are no classic lysosomes in this organism, autophagosome-like bodies are formed [138, 145]. TgCPL appears to play an important role in the degradation of autophagosomes, as knockout or inhibition of TgCPL results in undigested proteins and organelles in the vacuolar compartment, limiting chronic infection in mice.

The clan CA cysteine proteinases of T. gondii have been identified as potential vaccine candidates. DNA vaccines containing TgCPB or TgCPL gene sequences individually or together produced both humoral and cellular immune responses in BALB/c mice. Following immunization, survival was prolonged following intraperitoneal challenge with tachyzoites, with the most significant effect from the combined TgCPB/TgCPL vaccine [146]. Similar results were seen with a TgCPC1 DNA vaccine [147].

Conclusion

From the 7 protozoan parasites causing human disease that are of interest in this review, only 2 genera (Trypanosoma and Plasmodium) have at least one well-characterized clan CA CP: cruzain in T. cruzi, TbCatL in T. brucei, and falcipain-2 and falcipain-3 in the malaria parasite. These enzymes were validated as drug targets using a variety of inhibitor chemistries.

One common theme is the observation that distinct pathogens employ related CPs to perform similar functions. For example, the process of invading a tissue in the case of extracellular parasites [10, 19, 51, 148] and invading a host cell in the case of intracellular parasites [71, 135, 136, 140] is highly dependent on clan CA proteases. It is also interesting to note that parasites have evolved different mechanisms to utilize CPs to modulate the immune system of the host. EhCP can directly degrade IgA, IgG, and IL-18 [1115]. By contrast, CPB in Leishmania sp. modulates host responses by down-regulating protective Th1 immune responses, and in particular IFN-γ production, via degradation of the transcription factor NF-κB and the subsequent inhibition of IL-12 production by infected macrophages [26, 35, 149].

Apart from the more well-studied Trypanosoma and Plasmodium sp., it is important to continue investigations regarding the therapeutic potential of other protozoan CPs. In support of this, essentiality has been suggested or demonstrated for the CPs in many of the species discussed here. Furthermore, the potential for the emergence of other protease targets is great, considering that less than 10% of putative CPs found in the respective genomes have been so far characterized.

Key learning points

  • Gaps in our knowledge: there are many genome copies of CPs per protozoan organism. Less than 10 enzymes have been well characterized per parasite and not more than 2 enzymes per pathogen have had their structures resolved by X-ray crystallography.
  • Importance: the major functions of CPs shared by these protozoan parasites are host invasion and tissue penetration, virulence and evasion/modulation of the host immune system.
  • Potential targets for chemotherapy: the most well studied CPs of protozoan parasites are the cathepsin L-like enzymes from T. brucei and T. cruzi, and falcipain-2 and falcipain-3 from Plasmodium sp. They are validated targets in vitro and in vivo for the development of novel therapies.

Top four papers

  1. McKerrow JH. Development of cysteine protease inhibitors as chemotherapy for parasitic diseases: insights on safety, target validation, and mechanism of action. Int J Parasitol. 1999;29(6):833–7. PubMed PMID: 10480720. [162]
  2. Sijwali PS, Rosenthal PJ. Gene disruption confirms a critical role for the cysteine protease falcipain-2 in hemoglobin hydrolysis by Plasmodium falciparum. Proc Natl Acad Sci U S A. 2004;101(13):4384–9. PubMed PMID: 15070727.
  3. Sajid M, McKerrow JH. Cysteine proteases of parasitic organisms. Mol Biochem Parasitol. 2002;120(1):1–21. PubMed PMID: 11849701.
  4. McKerrow JH, Caffrey C, Kelly B, Loke P, Sajid M. Proteases in parasitic diseases. Annu Rev Pathol. 2006;1:497–536. doi: 10.1146/annurev.pathol.1.110304.100151. PubMed PMID: 18039124 [163]

References

  1. 1. Sajid M, McKerrow JH. Cysteine proteases of parasitic organisms. Mol Biochem Parasitol. 2002;120(1):1–21. pmid:11849701.
  2. 2. Barr SC, Warner KL, Kornreic BG, Piscitelli J, Wolfe A, Benet L, et al. A cysteine protease inhibitor protects dogs from cardiac damage during infection by Trypanosoma cruzi. Antimicrob Agents Chemother. 2005;49(12):5160–1. pmid:16304193; PubMed Central PMCID: PMCPMC1315979.
  3. 3. Engel JC, Doyle PS, Hsieh I, McKerrow JH. Cysteine protease inhibitors cure an experimental Trypanosoma cruzi infection. J Exp Med. 1998;188(4):725–34. pmid:9705954; PubMed Central PMCID: PMCPMC2213346.
  4. 4. Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380(9859):2095–128. pmid:23245604.
  5. 5. Nakajima H. The world health report 1998—Life in the 21st century: A vision for all. World Health Organization, 1998.
  6. 6. Reed SL, Keene WE, McKerrow JH. Thiol proteinase expression and pathogenicity of Entamoeba histolytica. J Clin Microbiol. 1989;27(12):2772–7. pmid:2556432; PubMed Central PMCID: PMCPMC267124.
  7. 7. Moncada D, Keller K, Chadee K. Entamoeba histolytica cysteine proteinases disrupt the polymeric structure of colonic mucin and alter its protective function. Infect Immun. 2003;71(2):838–44. pmid:12540564; PubMed Central PMCID: PMCPMC145371.
  8. 8. Lauwaet T, Oliveira MJ, Callewaert B, De Bruyne G, Saelens X, Ankri S, et al. Proteolysis of enteric cell villin by Entamoeba histolytica cysteine proteinases. J Biol Chem. 2003;278(25):22650–6. pmid:12690119.
  9. 9. Espinosa-Cantellano M, Martinez-Palomo A. Pathogenesis of intestinal amebiasis: from molecules to disease. Clin Microbiol Rev. 2000;13(2):318–31. pmid:10756002; PubMed Central PMCID: PMCPMC100155.
  10. 10. Debnath A, Tashker JS, Sajid M, McKerrow JH. Transcriptional and secretory responses of Entamoeba histolytica to mucins, epithelial cells and bacteria. Int J Parasitol. 2007;37(8–9):897–906. pmid:17362964.
  11. 11. Garcia-Nieto RM, Rico-Mata R, Arias-Negrete S, Avila EE. Degradation of human secretory IgA1 and IgA2 by Entamoeba histolytica surface-associated proteolytic activity. Parasitol Int. 2008;57(4):417–23. pmid:18571975.
  12. 12. Tran VQ, Herdman DS, Torian BE, Reed SL. The neutral cysteine proteinase of Entamoeba histolytica degrades IgG and prevents its binding. J Infect Dis. 1998;177(2):508–11. pmid:9466550.
  13. 13. Reed SL, Ember JA, Herdman DS, DiScipio RG, Hugli TE, Gigli I. The extracellular neutral cysteine proteinase of Entamoeba histolytica degrades anaphylatoxins C3a and C5a. J Immunol. 1995;155(1):266–74. pmid:7602103.
  14. 14. Zhang Z, Wang L, Seydel KB, Li E, Ankri S, Mirelman D, et al. Entamoeba histolytica cysteine proteinases with interleukin-1 beta converting enzyme (ICE) activity cause intestinal inflammation and tissue damage in amoebiasis. Mol Microbiol. 2000;37(3):542–8. pmid:10931347.
  15. 15. Que X, Kim SH, Sajid M, Eckmann L, Dinarello CA, McKerrow JH, et al. A surface amebic cysteine proteinase inactivates interleukin-18. Infect Immun. 2003;71(3):1274–80. pmid:12595442; PubMed Central PMCID: PMCPMC148873.
  16. 16. St-Pierre J, Moreau F, Cornick S, Quach J, Begum S, Aracely Fernandez L, et al. The macrophage cytoskeleton acts as a contact sensor upon interaction with Entamoeba histolytica to trigger IL-1beta secretion. PLoS Pathog. 2017;13(8):e1006592. pmid:28837696; PubMed Central PMCID: PMCPMC5587335.
  17. 17. Ebert F, Bachmann A, Nakada-Tsukui K, Hennings I, Drescher B, Nozaki T, et al. An Entamoeba cysteine peptidase specifically expressed during encystation. Parasitol Int. 2008;57(4):521–4. pmid:18723116.
  18. 18. Irmer H, Tillack M, Biller L, Handal G, Leippe M, Roeder T, et al. Major cysteine peptidases of Entamoeba histolytica are required for aggregation and digestion of erythrocytes but are dispensable for phagocytosis and cytopathogenicity. Mol Microbiol. 2009;72(3):658–67. pmid:19426210.
  19. 19. Melendez-Lopez SG, Herdman S, Hirata K, Choi MH, Choe Y, Craik C, et al. Use of recombinant Entamoeba histolytica cysteine proteinase 1 to identify a potent inhibitor of amebic invasion in a human colonic model. Eukaryot Cell. 2007;6(7):1130–6. Epub 2007/05/22. pmid:17513563; PubMed Central PMCID: PMCPmc1951106.
  20. 20. Mortality GBD, Causes of Death C. Global, regional, and national age-sex specific all-cause and cause-specific mortality for 240 causes of death, 1990–2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet. 2015;385(9963):117–71. pmid:25530442; PubMed Central PMCID: PMCPMC4340604.
  21. 21. McCall LI, Zhang WW, Matlashewski G. Determinants for the development of visceral leishmaniasis disease. PLoS Pathog. 2013;9(1):e1003053. pmid:23300451; PubMed Central PMCID: PMCPMC3536654.
  22. 22. Alexander J, Coombs GH, Mottram JC. Leishmania mexicana cysteine proteinase-deficient mutants have attenuated virulence for mice and potentiate a Th1 response. J Immunol. 1998;161(12):6794–801. Epub 1998/12/23. pmid:9862710.
  23. 23. Novinec M, Lenarcic B. Papain-like peptidases: structure, function, and evolution. Biomol Concepts. 2013;4(3):287–308. pmid:25436581.
  24. 24. Turk V, Stoka V, Vasiljeva O, Renko M, Sun T, Turk B, et al. Cysteine cathepsins: from structure, function and regulation to new frontiers. Biochim Biophys Acta. 2012;1824(1):68–88. pmid:22024571.
  25. 25. Souza AE, Waugh S, Coombs GH, Mottram JC. Characterization of a multi-copy gene for a major stage-specific cysteine proteinase of Leishmania mexicana. FEBS Lett. 1992;311(2):124–7. pmid:1397299.
  26. 26. Mottram JC, Coombs GH, Alexander J. Cysteine peptidases as virulence factors of Leishmania. Curr Opin Microbiol. 2004;7(4):375–81. ISI:000223458200010. pmid:15358255
  27. 27. Mundodi V, Somanna A, Farrell PJ, Gedamu L. Genomic organization and functional expression of differentially regulated cysteine protease genes of Leishmania donovani complex. Gene. 2002;282(1–2):257–65. pmid:11814698.
  28. 28. Bart G, Coombs GH, Mottram JC. Isolation of lmcpc, a gene encoding a Leishmania mexicana cathepsin-B-like cysteine proteinase. Mol Biochem Parasitol. 1995;73(1–2):271–4. pmid:8577339.
  29. 29. Mottram JC, Robertson CD, Coombs GH, Barry JD. A developmentally regulated cysteine proteinase gene of Leishmania mexicana. Mol Microbiol. 1992;6(14):1925–32. pmid:1508041.
  30. 30. Mottram JC, Frame MJ, Brooks DR, Tetley L, Hutchison JE, Souza AE, et al. The multiple cpb cysteine proteinase genes of Leishmania mexicana encode isoenzymes that differ in their stage regulation and substrate preferences. J Biol Chem. 1997;272(22):14285–93. WOS:A1997XB49200050. pmid:9162063
  31. 31. Mundodi V, Kucknoor AS, Gedamu L. Role of Leishmania (Leishmania) chagasi amastigote cysteine protease in intracellular parasite survival: studies by gene disruption and antisense mRNA inhibition. BMC Mol Biol. 2005;6:3. PMC549197. pmid:15691375
  32. 32. Denise H, Poot J, Jiménez M, Ambit A, Herrmann DC, Vermeulen AN, et al. Studies on the CPA cysteine peptidase in the Leishmania infantum genome strain JPCM5. BMC Mol Biol. 2006;7:42. PMC1657026. pmid:17101050
  33. 33. Mottram JC, Souza AE, Hutchison JE, Carter R, Frame MJ, Coombs GH. Evidence from disruption of the lmcpb gene array of Leishmania mexicana that cysteine proteinases are virulence factors. P Natl Acad Sci USA. 1996;93(12):6008–13. WOS:A1996UQ45500062.
  34. 34. Denise H, McNeil K, Brooks DR, Alexander J, Coombs GH, Mottram JC. Expression of multiple CPB genes encoding cysteine proteases is required for Leishmania mexicana virulence in vivo. Infect Immun. 2003;71(6):3190–5. WOS:000183116300026. pmid:12761098
  35. 35. Cameron P, McGachy A, Anderson M, Paul A, Coombs GH, Mottram JC, et al. Inhibition of lipopolysaccharide-induced macrophage IL-12 production by Leishmania mexicana amastigotes: the role of cysteine peptidases and the NF-kappaB signaling pathway. J Immunol. 2004;173(5):3297–304. pmid:15322192.
  36. 36. Leao SD, Lang T, Prina E, Hellio R, Antoine JC. Intracellular Leishmania-Amazonensis Amastigotes Internalize and Degrade Mhc Class-Ii Molecules of Their Host-Cells. J Cell Sci. 1995;108:3219–31. WOS:A1995RZ34900008. pmid:7593283
  37. 37. Casgrain PA, Martel C, McMaster WR, Mottram JC, Olivier M, Descoteaux A. Cysteine Peptidase B Regulates Leishmania mexicana Virulence through the Modulation of GP63 Expression. PLoS Pathog. 2016;12(5):e1005658. pmid:27191844; PubMed Central PMCID: PMCPMC4871588.
  38. 38. El-Fadili AK, Zangger H, Desponds C, Gonzalez IJ, Zalila H, Schaff C, et al. Cathepsin B-like and cell death in the unicellular human pathogen Leishmania. Cell Death Dis. 2010;1:e71. Epub 2011/03/03. pmid:21364675; PubMed Central PMCID: PMCPmc3032344.
  39. 39. Gerbaba TK, Gedamu L. Cathepsin B gene disruption induced Leishmania donovani proteome remodeling implies cathepsin B role in secretome regulation. PLoS ONE. 2013;8(11):e79951. pmid:24244582; PubMed Central PMCID: PMCPMC3828211.
  40. 40. Bart G, Frame MJ, Carter R, Coombs GH, Mottram JC. Cathepsin B-like cysteine proteinase-deficient mutants of Leishmania mexicana. Mol Biochem Parasitol. 1997;88(1–2):53–61. Epub 1997/09/01. pmid:9274867.
  41. 41. Williams RA, Mottram JC, Coombs GH. Distinct roles in autophagy and importance in infectivity of the two ATG4 cysteine peptidases of Leishmania major. J Biol Chem. 2013;288(5):3678–90. pmid:23166325; PubMed Central PMCID: PMCPMC3561585.
  42. 42. Azevedo CS, Guido BC, Pereira JL, Nolasco DO, Correa R, Magalhaes KG, et al. Revealing a Novel Otubain-Like Enzyme from Leishmania infantum with Deubiquitinating Activity toward K48-Linked Substrate. Front Chem. 2017;5:13. pmid:28386537; PubMed Central PMCID: PMCPMC5362604.
  43. 43. Selzer PM, Pingel S, Hsieh I, Ugele B, Chan VJ, Engel JC, et al. Cysteine protease inhibitors as chemotherapy: lessons from a parasite target. Proc Natl Acad Sci U S A. 1999;96(20):11015–22. pmid:10500116; PubMed Central PMCID: PMCPMC34234.
  44. 44. Paladi Cde S, Pimentel IA, Katz S, Cunha RL, Judice WA, Caires AC, et al. In vitro and in vivo activity of a palladacycle complex on Leishmania (Leishmania) amazonensis. PLoS Negl Trop Dis. 2012;6(5):e1626. pmid:22616018; PubMed Central PMCID: PMCPMC3352823.
  45. 45. Lima CB, Arrais-Silva WW, Cunha RL, Giorgio S. A novel organotellurium compound (RT-01) as a new antileishmanial agent. Korean J Parasitol. 2009;47(3):213–8. pmid:19724693; PubMed Central PMCID: PMCPMC2735685.
  46. 46. Salerno Pimentel IA, Paladi Cde S, Katz S, de Souza Judice WA, Cunha RL, Barbieri CL. In vitro and in vivo activity of an organic tellurium compound on Leishmania (Leishmania) chagasi. PLoS ONE. 2012;7(11):e48780. pmid:23144968; PubMed Central PMCID: PMCPMC3492430.
  47. 47. Schroder J, Noack S, Marhofer RJ, Mottram JC, Coombs GH, Selzer PM. Identification of semicarbazones, thiosemicarbazones and triazine nitriles as inhibitors of Leishmania mexicana cysteine protease CPB. PLoS ONE. 2013;8(10):e77460. pmid:24146999; PubMed Central PMCID: PMCPMC3797739.
  48. 48. de Almeida L, Alves KF, Maciel-Rezende CM, Jesus Lde O, Pires FR, Junior CV, et al. Benzophenone derivatives as cysteine protease inhibitors and biological activity against Leishmania(L.) amazonensis amastigotes. Biomed Pharmacother. 2015;75:93–9. pmid:26463637.
  49. 49. Caffrey CR, Lima AP, Steverding D. Cysteine peptidases of kinetoplastid parasites. Adv Exp Med Biol. 2011;712:84–99. pmid:21660660.
  50. 50. Grab DJ, Garcia-Garcia JC, Nikolskaia OV, Kim YV, Brown A, Pardo CA, et al. Protease Activated Receptor Signaling Is Required for African Trypanosome Traversal of Human Brain Microvascular Endothelial Cells. PloS Negl Trop Dis. 2009;3(7):e479. ARTN e479 WOS:000268452200010. pmid:19621073
  51. 51. Nikolskaia OV, de ALAP, Kim YV, Lonsdale-Eccles JD, Fukuma T, Scharfstein J, et al. Blood-brain barrier traversal by African trypanosomes requires calcium signaling induced by parasite cysteine protease. J Clin Invest. 2006;116(10):2739–47. pmid:16998589; PubMed Central PMCID: PMCPMC1570376.
  52. 52. Alsford S, Currier RB, Guerra-Assuncao JA, Clark TG, Horn D. Cathepsin-L Can Resist Lysis by Human Serum in Trypanosoma brucei brucei. PLoS Pathog. 2014;10(5):e1004130. ARTN e1004130 WOS:000337732300038. pmid:24830321
  53. 53. Uzureau P, Uzureau S, Lecordier L, Fontaine F, Tebabi P, Homble F, et al. Mechanism of Trypanosoma brucei gambiense resistance to human serum. Nature. 2013;501(7467):430–4. pmid:23965626.
  54. 54. Santos CC, Coombs GH, Lima AP, Mottram JC. Role of the Trypanosoma brucei natural cysteine peptidase inhibitor ICP in differentiation and virulence. Mol Microbiol. 2007;66(4):991–1002. pmid:17944830; PubMed Central PMCID: PMCPMC2680270.
  55. 55. Caffrey CR, Hansell E, Lucas KD, Brinen LS, Alvarez Hernandez A, Cheng J, et al. Active site mapping, biochemical properties and subcellular localization of rhodesain, the major cysteine protease of Trypanosoma brucei rhodesiense. Mol Biochem Parasitol. 2001;118(1):61–73. pmid:11704274.
  56. 56. Mackey ZB, O'Brien , Greenbaum DC, Blank RB, McKerrow JH. A cathepsin B-like protease is required for host protein degradation in Trypanosoma brucei. J Biol Chem. 2004;279(46):48426–33. pmid:15326171.
  57. 57. O'Brien TC, Mackey ZB, Fetter RD, Choe Y, O'Donoghue AJ, Zhou M, et al. A Parasite Cysteine Protease Is Key to Host Protein Degradation and Iron Acquisition. J Biol Chem. 2008;283(43):28934–43. ISI:000260179900019. pmid:18701454
  58. 58. O'Brien TC, Mackey ZB, Fetter RD, Choe Y, O'Donoghue AJ, Zhou M, et al. A parasite cysteine protease is key to host protein degradation and iron acquisition. J Biol Chem. 2008;283(43):28934–43. pmid:18701454; PubMed Central PMCID: PMCPMC2570886.
  59. 59. Steverding D, Sexton DW, Wang X, Gehrke SS, Wagner GK, Caffrey CR. Trypanosoma brucei: chemical evidence that cathepsin L is essential for survival and a relevant drug target. Int J Parasitol. 2012;42(5):481–8. pmid:22549023.
  60. 60. Caffrey CR, Scory S, Steverding D. Cysteine proteinases of trypanosome parasites: novel targets for chemotherapy. Curr Drug Targets. 2000;1(2):155–62. pmid:11465068.
  61. 61. Scory S, Caffrey CR, Stierhof YD, Ruppel A, Steverding D. Trypanosoma brucei: killing of bloodstream forms in vitro and in vivo by the cysteine proteinase inhibitor Z-phe-ala-CHN2. Exp Parasitol. 1999;91(4):327–33. pmid:10092476.
  62. 62. Steverding D. Evaluation of trypanocidal activity of combinations of anti-sleeping sickness drugs with cysteine protease inhibitors. Exp Parasitol. 2015;151–152:28–33. pmid:25662707.
  63. 63. Schirmeister T, Schmitz J, Jung S, Schmenger T, Krauth-Siegel RL, Gutschow M. Evaluation of dipeptide nitriles as inhibitors of rhodesain, a major cysteine protease of Trypanosoma brucei. Bioorg Med Chem Lett. 2017;27(1):45–50. pmid:27890381.
  64. 64. Ehmke V, Winkler E, Banner DW, Haap W, Schweizer WB, Rottmann M, et al. Optimization of triazine nitriles as rhodesain inhibitors: structure-activity relationships, bioisosteric imidazopyridine nitriles, and X-ray crystal structure analysis with human cathepsin L. ChemMedChem. 2013;8(6):967–75. pmid:23658062.
  65. 65. Ferreira LG, Andricopulo AD. Targeting cysteine proteases in trypanosomatid disease drug discovery. Pharmacol Ther. 2017;180:49–61. pmid:28579388.
  66. 66. Perez-Molina JA, Molina I. Chagas disease. Lancet. 2017;391(10115):82–94. pmid:28673423.
  67. 67. Scharfstein J, Schechter M, Senna M, Peralta JM, Mendonca-Previato L, Miles MA. Trypanosoma cruzi: characterization and isolation of a 57/51,000 m.w. surface glycoprotein (GP57/51) expressed by epimastigotes and bloodstream trypomastigotes. J Immunol. 1986;137(4):1336–41. pmid:3090146.
  68. 68. Duschak VG, Couto AS. Cruzipain, the major cysteine protease of Trypanosoma cruzi: a sulfated glycoprotein antigen as relevant candidate for vaccine development and drug target. A review. Curr Med Chem. 2009;16(24):3174–202. pmid:19689291.
  69. 69. Franke de Cazzulo BM, Martinez J, North MJ, Coombs GH, Cazzulo JJ. Effects of proteinase inhibitors on the growth and differentiation of Trypanosoma cruzi. FEMS Microbiol Lett. 1994;124(1):81–6. pmid:8001773.
  70. 70. Cazzulo JJ, Stoka V, Turk V. Cruzipain, the major cysteine proteinase from the protozoan parasite Trypanosoma cruzi. Biol Chem. 1997;378(1):1–10. pmid:9049059.
  71. 71. Meirelles MN, Juliano L, Carmona E, Silva SG, Costa EM, Murta AC, et al. Inhibitors of the major cysteinyl proteinase (GP57/51) impair host cell invasion and arrest the intracellular development of Trypanosoma cruzi in vitro. Mol Biochem Parasitol. 1992;52(2):175–84. pmid:1620157.
  72. 72. Eakin AE, Mills AA, Harth G, McKerrow JH, Craik CS. The sequence, organization, and expression of the major cysteine protease (cruzain) from Trypanosoma cruzi. J Biol Chem. 1992;267(11):7411–20. pmid:1559982.
  73. 73. McGrath ME, Eakin AE, Engel JC, McKerrow JH, Craik CS, Fletterick RJ. The crystal structure of cruzain: a therapeutic target for Chagas' disease. J Mol Biol. 1995;247(2):251–9. pmid:7707373.
  74. 74. Yong V, Schmitz V, Vannier-Santos MA, de Lima AP, Lalmanach G, Juliano L, et al. Altered expression of cruzipain and a cathepsin B-like target in a Trypanosoma cruzi cell line displaying resistance to synthetic inhibitors of cysteine-proteinases. Mol Biochem Parasitol. 2000;109(1):47–59. pmid:10924756.
  75. 75. Kosec G, Alvarez V, Cazzulo JJ. Cysteine proteinases of Trypanosoma cruzi: from digestive enzymes to programmed cell death mediators. Biocell. 2006;30(3):479–90. pmid:17375468.
  76. 76. Vanrell MC, Losinno AD, Cueto JA, Balcazar D, Fraccaroli LV, Carrillo C, et al. The regulation of autophagy differentially affects Trypanosoma cruzi metacyclogenesis. PLoS Negl Trop Dis. 2017;11(11):e0006049. pmid:29091711.
  77. 77. Ferreira RS, Bryant C, Ang KK, McKerrow JH, Shoichet BK, Renslo AR. Divergent modes of enzyme inhibition in a homologous structure-activity series. J Med Chem. 2009;52(16):5005–8. pmid:19637873; PubMed Central PMCID: PMCPMC3760508.
  78. 78. Fonseca NC, da Cruz LF, da Silva Villela F, do Nascimento Pereira GA, de Siqueira-Neto JL, Kellar D, et al. Synthesis of a sugar-based thiosemicarbazone series and structure-activity relationship versus the parasite cysteine proteases rhodesain, cruzain, and Schistosoma mansoni cathepsin B1. Antimicrob Agents Chemother. 2015;59(5):2666–77. pmid:25712353; PubMed Central PMCID: PMCPMC4394791.
  79. 79. Chiyanzu I, Hansell E, Gut J, Rosenthal PJ, McKerrow JH, Chibale K. Synthesis and evaluation of isatins and thiosemicarbazone derivatives against cruzain, falcipain-2 and rhodesain. Bioorg Med Chem Lett. 2003;13(20):3527–30. Epub 2003/09/25. S0960894X0300756X [pii]. pmid:14505663.
  80. 80. Stoch SA, Zajic S, Stone JA, Miller DL, van Bortel L, Lasseter KC, et al. Odanacatib, a selective cathepsin K inhibitor to treat osteoporosis: safety, tolerability, pharmacokinetics and pharmacodynamics—results from single oral dose studies in healthy volunteers. Br J Clin Pharmacol. 2013;75(5):1240–54. pmid:23013236; PubMed Central PMCID: PMCPMC3635595.
  81. 81. Ndao M, Beaulieu C, Black WC, Isabel E, Vasquez-Camargo F, Nath-Chowdhury M, et al. Reversible cysteine protease inhibitors show promise for a Chagas disease cure. Antimicrob Agents Chemother. 2014;58(2):1167–78. pmid:24323474; PubMed Central PMCID: PMCPMC3910870.
  82. 82. Jones BD, Tochowicz A, Tang Y, Cameron MD, McCall LI, Hirata K, et al. Synthesis and Evaluation of Oxyguanidine Analogues of the Cysteine Protease Inhibitor WRR-483 against Cruzain. ACS Med Chem Lett. 2016;7(1):77–82. pmid:26819670; PubMed Central PMCID: PMCPMC4716606.
  83. 83. Martinez-Mayorga K, Byler KG, Ramirez-Hernandez AI, Terrazas-Alvares DE. Cruzain inhibitors: efforts made, current leads and a structural outlook of new hits. Drug Discov Today. 2015;20(7):890–8. pmid:25697479.
  84. 84. Sow SO, Muhsen K, Nasrin D, Blackwelder WC, Wu Y, Farag TH, et al. The Burden of Cryptosporidium Diarrheal Disease among Children < 24 Months of Age in Moderate/High Mortality Regions of Sub-Saharan Africa and South Asia, Utilizing Data from the Global Enteric Multicenter Study (GEMS). PLoS Negl Trop Dis. 2016;10(5):e0004729. pmid:27219054; PubMed Central PMCID: PMCPMC4878811.
  85. 85. Armson A, Thompson RC, Reynoldson JA. A review of chemotherapeutic approaches to the treatment of cryptosporidiosis. Expert Rev Anti Infect Ther. 2003;1(2):297–305. pmid:15482125.
  86. 86. Benschop J, Booker CM, Shadbolt T, Weston JF. A Retrospective Cohort Study of an Outbreak of Cryptosporidiosis among Veterinary Students. Vet Sci. 2017;4(2). pmid:29056688.
  87. 87. Mac Kenzie WR, Hoxie NJ, Proctor ME, Gradus MS, Blair KA, Peterson DE, et al. A massive outbreak in Milwaukee of cryptosporidium infection transmitted through the public water supply. N Engl J Med. 1994;331(3):161–7. pmid:7818640.
  88. 88. Corso PS, Kramer MH, Blair KA, Addiss DG, Davis JP, Haddix AC. Cost of illness in the 1993 waterborne Cryptosporidium outbreak, Milwaukee, Wisconsin. Emerg Infect Dis. 2003;9(4):426–31. pmid:12702221; PubMed Central PMCID: PMCPMC2957981.
  89. 89. Abrahamsen MS, Templeton TJ, Enomoto S, Abrahante JE, Zhu G, Lancto CA, et al. Complete genome sequence of the apicomplexan, Cryptosporidium parvum. Science. 2004;304(5669):441–5. pmid:15044751.
  90. 90. Na BK, Kang JM, Cheun HI, Cho SH, Moon SU, Kim TS, et al. Cryptopain-1, a cysteine protease of Cryptosporidium parvum, does not require the pro-domain for folding. Parasitology. 2009;136(2):149–57. pmid:19091155.
  91. 91. Ndao M, Nath-Chowdhury M, Sajid M, Marcus V, Mashiyama ST, Sakanari J, et al. A cysteine protease inhibitor rescues mice from a lethal Cryptosporidium parvum infection. Antimicrob Agents Chemother. 2013;57(12):6063–73. pmid:24060869; PubMed Central PMCID: PMCPMC3837922.
  92. 92. Nijman SM, Luna-Vargas MP, Velds A, Brummelkamp TR, Dirac AM, Sixma TK, et al. A genomic and functional inventory of deubiquitinating enzymes. Cell. 2005;123(5):773–86. pmid:16325574.
  93. 93. Ju HL, Kang JM, Noh HS, Kim DR, Hong Y, Sohn WM, et al. Characterization of a novel otubain-like cysteine protease of Cryptosporidium parvum. Parasitol Int. 2014;63(4):580–3. pmid:24709083.
  94. 94. Gething PW, Casey DC, Weiss DJ, Bisanzio D, Bhatt S, Cameron E, et al. Mapping Plasmodium falciparum Mortality in Africa between 1990 and 2015. N Engl J Med. 2016;375(25):2435–45. pmid:27723434; PubMed Central PMCID: PMCPMC5484406.
  95. 95. Ghebreyesus TA. World malaria report 2017: Foreword. World Health Organization, 2017. Accessed 15 Jun 2018. Available from: http://apps.who.int/iris/bitstream/handle/10665/259492/9789241565523-eng.pdf?sequence=1.
  96. 96. Rosenthal PJ. Falcipains and other cysteine proteases of malaria parasites. Adv Exp Med Biol. 2011;712:30–48. pmid:21660657.
  97. 97. Rosenthal PJ. Falcipains. In: Rawlings ND, Salvesen GS, editors. Handbook of Proteolytic Enzymes. Oxford: Academic Press; 2013. p. 1907–12.
  98. 98. Deu E. Proteases as antimalarial targets: strategies for genetic, chemical, and therapeutic validation. FEBS J. 2017;284(16):2604–28. pmid:28599096; PubMed Central PMCID: PMCPMC5575534.
  99. 99. Francis SE, Sullivan DJ Jr., Goldberg DE. Hemoglobin metabolism in the malaria parasite Plasmodium falciparum. Annu Rev Microbiol. 1997;51:97–123. pmid:9343345.
  100. 100. Drew ME, Banerjee R, Uffman EW, Gilbertson S, Rosenthal PJ, Goldberg DE. Plasmodium food vacuole plasmepsins are activated by falcipains. J Biol Chem. 2008;283(19):12870–6. Epub 2008/03/01. M708949200 [pii] pmid:18308731; PubMed Central PMCID: PMC2442342.
  101. 101. Sijwali PS, Rosenthal PJ. Gene disruption confirms a critical role for the cysteine protease falcipain-2 in hemoglobin hydrolysis by Plasmodium falciparum. Proc Natl Acad Sci U S A. 2004;101(13):4384–9. pmid:15070727.
  102. 102. Sijwali PS, Koo J, Singh N, Rosenthal PJ. Gene disruptions demonstrate independent roles for the four falcipain cysteine proteases of Plasmodium falciparum. Mol Biochem Parasitol. 2006;150(1):96–106. pmid:16890302.
  103. 103. Blackman MJ. Malarial proteases and host cell egress: an 'emerging' cascade. Cell Microbiol. 2008;10(10):1925–34. pmid:18503638; PubMed Central PMCID: PMCPMC2610400.
  104. 104. Collins CR, Hackett F, Atid J, Tan MSY, Blackman MJ. The Plasmodium falciparum pseudoprotease SERA5 regulates the kinetics and efficiency of malaria parasite egress from host erythrocytes. PLoS Pathog. 2017;13(7):e1006453. pmid:28683142; PubMed Central PMCID: PMCPMC5500368.
  105. 105. Ruecker A, Shea M, Hackett F, Suarez C, Hirst EM, Milutinovic K, et al. Proteolytic activation of the essential parasitophorous vacuole cysteine protease SERA6 accompanies malaria parasite egress from its host erythrocyte. J Biol Chem. 2012;287(45):37949–63. pmid:22984267; PubMed Central PMCID: PMCPMC3488066.
  106. 106. Arastu-Kapur S, Ponder EL, Fonovic UP, Yeoh S, Yuan F, Fonovic M, et al. Identification of proteases that regulate erythrocyte rupture by the malaria parasite Plasmodium falciparum. Nat Chem Biol. 2008;4(3):203–13. pmid:18246061.
  107. 107. Ghosh S, Chisholm SA, Dans M, Lakkavaram A, Kennedy K, Ralph SA, et al. The cysteine protease dipeptidyl aminopeptidase 3 does not contribute to egress of Plasmodium falciparum from host red blood cells. PLoS ONE. 2018;13(3):e0193538. pmid:29509772; PubMed Central PMCID: PMCPMC5839547.
  108. 108. Thomas JA, Tan MSY, Bisson C, Borg A, Umrekar TR, Hackett F, et al. A protease cascade regulates release of the human malaria parasite Plasmodium falciparum from host red blood cells. Nat Microbiol. 2018;3(4):447–55. pmid:29459732.
  109. 109. Greenbaum DC, Baruch A, Grainger M, Bozdech Z, Medzihradszky KF, Engel J, et al. A role for the protease falcipain 1 in host cell invasion by the human malaria parasite. Science. 2002;298(5600):2002–6. pmid:12471262.
  110. 110. Eksi S, Czesny B, Greenbaum DC, Bogyo M, Williamson KC. Targeted disruption of Plasmodium falciparum cysteine protease, falcipain 1, reduces oocyst production, not erythrocytic stage growth. Mol Microbiol. 2004;53(1):243–50. pmid:15225318.
  111. 111. Sijwali PS, Kato K, Seydel KB, Gut J, Lehman J, Klemba M, et al. Plasmodium falciparum cysteine protease falcipain-1 is not essential in erythrocytic stage malaria parasites. Proc Natl Acad Sci U S A. 2004;101(23):8721–6. pmid:15166288; PubMed Central PMCID: PMCPMC423262.
  112. 112. Pandey KC, Singh N, Arastu-Kapur S, Bogyo M, Rosenthal PJ. Falstatin, a cysteine protease inhibitor of Plasmodium falciparum, facilitates erythrocyte invasion. PLoS Pathog. 2006;2(11):e117. pmid:17083274; PubMed Central PMCID: PMCPMC1630708.
  113. 113. Russo I, Oksman A, Goldberg DE. Fatty acid acylation regulates trafficking of the unusual Plasmodium falciparum calpain to the nucleolus. Mol Microbiol. 2009;72(1):229–45. pmid:19239622; PubMed Central PMCID: PMCPMC2746569.
  114. 114. Russo I, Oksman A, Vaupel B, Goldberg DE. A calpain unique to alveolates is essential in Plasmodium falciparum and its knockdown reveals an involvement in pre-S-phase development. Proc Natl Acad Sci U S A. 2009;106(5):1554–9. pmid:19164769; PubMed Central PMCID: PMCPMC2629787.
  115. 115. Datta G, Hossain ME, Asad M, Rathore S, Mohmmed A. Plasmodium falciparum OTU-like cysteine protease (PfOTU) is essential for apicoplast homeostasis and associates with noncanonical role of Atg8. Cell Microbiol. 2017;19(9):e12748. pmid:28423214.
  116. 116. Coppi A, Pinzon-Ortiz C, Hutter C, Sinnis P. The Plasmodium circumsporozoite protein is proteolytically processed during cell invasion. J Exp Med. 2005;201(1):27–33. pmid:15630135; PubMed Central PMCID: PMCPMC1995445.
  117. 117. Hopp CS, Bennett BL, Mishra S, Lehmann C, Hanson KK, Lin JW, et al. Deletion of the rodent malaria ortholog for falcipain-1 highlights differences between hepatic and blood stage merozoites. PLoS Pathog. 2017;13(9):e1006586. pmid:28922424; PubMed Central PMCID: PMCPMC5602738.
  118. 118. Eksi S, Czesny B, van Gemert GJ, Sauerwein RW, Eling W, Williamson KC. Inhibition of Plasmodium falciparum oocyst production by membrane-permeant cysteine protease inhibitor E64d. Antimicrob Agents Chemother. 2007;51(3):1064–70. pmid:17178799; PubMed Central PMCID: PMCPMC1803139.
  119. 119. Suarez-Cortes P, Sharma V, Bertuccini L, Costa G, Bannerman NL, Sannella AR, et al. Comparative Proteomics and Functional Analysis Reveal a Role of Plasmodium falciparum Osmiophilic Bodies in Malaria Parasite Transmission. Mol Cell Proteomics. 2016;15(10):3243–55. pmid:27432909; PubMed Central PMCID: PMCPMC5054347.
  120. 120. Dahl EL, Rosenthal PJ. Biosynthesis, localization, and processing of falcipain cysteine proteases of Plasmodium falciparum. Mol Biochem Parasitol. 2005;139(2):205–12. Epub 2005/01/25. S0166-6851(04)00313-5 [pii] pmid:15664655.
  121. 121. Shenai BR, Sijwali PS, Singh A, Rosenthal PJ. Characterization of native and recombinant falcipain-2, a principal trophozoite cysteine protease and essential hemoglobinase of Plasmodium falciparum. J Biol Chem. 2000;275(37):29000–10. pmid:10887194.
  122. 122. Singh N, Sijwali PS, Pandey KC, Rosenthal PJ. Plasmodium falciparum: biochemical characterization of the cysteine protease falcipain-2'. Exp Parasitol. 2006;112(3):187–92. Epub 2005/12/13. S0014-4894(05)00280-8 [pii] pmid:16337629.
  123. 123. Sijwali PS, Shenai BR, Rosenthal PJ. Folding of the Plasmodium falciparum cysteine protease falcipain-2 is mediated by a chaperone-like peptide and not the prodomain. J Biol Chem. 2002;277(17):14910–5. Epub 2002/02/06. [pii]. pmid:11827964.
  124. 124. Marco M, Coteron JM. Falcipain inhibition as a promising antimalarial target. Curr Top Med Chem. 2012;12(5):408–44. pmid:22242849.
  125. 125. Kerr ID, Lee JH, Pandey KC, Harrison A, Sajid M, Rosenthal PJ, et al. Structures of falcipain-2 and falcipain-3 bound to small molecule inhibitors: implications for substrate specificity. J Med Chem. 2009;52(3):852–7. Epub 2009/01/09. [pii]. pmid:19128015; PubMed Central PMCID: PMC2651692.
  126. 126. Olson JE, Lee GK, Semenov A, Rosenthal PJ. Antimalarial effects in mice of orally administered peptidyl cysteine protease inhibitors. Bioorg Med Chem. 1999;7(4):633–8. pmid:10353642.
  127. 127. Coteron JM, Catterick D, Castro J, Chaparro MJ, Diaz B, Fernandez E, et al. Falcipain inhibitors: optimization studies of the 2-pyrimidinecarbonitrile lead series. J Med Chem. 2010;53(16):6129–52. pmid:20672841.
  128. 128. Conroy T, Guo JT, Elias N, Cergol KM, Gut J, Legac J, et al. Synthesis of gallinamide A analogues as potent falcipain inhibitors and antimalarials. J Med Chem. 2014;57(24):10557–63. pmid:25412465.
  129. 129. Melo PMS, El Chamy Maluf S, Azevedo MF, Paschoalin T, Budu A, Bagnaresi P, et al. Inhibition of Plasmodium falciparum cysteine proteases by the sugarcane cystatin CaneCPI-4. Parasitol Int. 2018;67(2):233–6. pmid:29288140.
  130. 130. Singh A, Rosenthal PJ. Selection of cysteine protease inhibitor-resistant malaria parasites is accompanied by amplification of falcipain genes and alteration in inhibitor transport. J Biol Chem. 2004;279(34):35236–41. pmid:15192087.
  131. 131. Klonis N, Crespo-Ortiz MP, Bottova I, Abu-Bakar N, Kenny S, Rosenthal PJ, et al. Artemisinin activity against Plasmodium falciparum requires hemoglobin uptake and digestion. Proc Natl Acad Sci U S A. 2011;108(28):11405–10. pmid:21709259; PubMed Central PMCID: PMC3136263.
  132. 132. Semenov A, Olson JE, Rosenthal PJ. Antimalarial synergy of cysteine and aspartic protease inhibitors. Antimicrob Agents Chemother. 1998;42(9):2254–8. pmid:9736544.
  133. 133. Robert-Gangneux F, Darde ML. Epidemiology of and diagnostic strategies for toxoplasmosis. Clin Microbiol Rev. 2012;25(2):264–96. pmid:22491772; PubMed Central PMCID: PMCPMC3346298.
  134. 134. Dou Z, McGovern OL, Di Cristina M, Carruthers VB. Toxoplasma gondii ingests and digests host cytosolic proteins. MBio. 2014;5(4):e01188–14. pmid:25028423; PubMed Central PMCID: PMCPMC4161261.
  135. 135. Larson ET, Parussini F, Huynh MH, Giebel JD, Kelley AM, Zhang L, et al. Toxoplasma gondii cathepsin L is the primary target of the invasion-inhibitory compound morpholinurea-leucyl-homophenyl-vinyl sulfone phenyl. J Biol Chem. 2009;284(39):26839–50. pmid:19596863; PubMed Central PMCID: PMCPMC2785372.
  136. 136. Teo CF, Zhou XW, Bogyo M, Carruthers VB. Cysteine protease inhibitors block Toxoplasma gondii microneme secretion and cell invasion. Antimicrob Agents Chemother. 2007;51(2):679–88. pmid:17145790; PubMed Central PMCID: PMCPMC1797762.
  137. 137. Que X, Engel JC, Ferguson D, Wunderlich A, Tomavo S, Reed SL. Cathepsin Cs are key for the intracellular survival of the protozoan parasite, Toxoplasma gondii. J Biol Chem. 2007;282(7):4994–5003. pmid:17164247.
  138. 138. Di Cristina M, Dou Z, Lunghi M, Kannan G, Huynh MH, McGovern OL, et al. Toxoplasma depends on lysosomal consumption of autophagosomes for persistent infection. Nat Microbiol. 2017;2:17096. pmid:28628099; PubMed Central PMCID: PMCPMC5527684.
  139. 139. Tillack M, Biller L, Irmer H, Freitas M, Gomes MA, Tannich E, et al. The Entamoeba histolytica genome: primary structure and expression of proteolytic enzymes. BMC Genomics. 2007;8:170. pmid:17567921; PubMed Central PMCID: PMCPMC1913524.
  140. 140. Que X, Ngo H, Lawton J, Gray M, Liu Q, Engel J, et al. The cathepsin B of Toxoplasma gondii, toxopain-1, is critical for parasite invasion and rhoptry protein processing. J Biol Chem. 2002;277(28):25791–7. pmid:12000756.
  141. 141. Huang R, Que X, Hirata K, Brinen LS, Lee JH, Hansell E, et al. The cathepsin L of Toxoplasma gondii (TgCPL) and its endogenous macromolecular inhibitor, toxostatin. Mol Biochem Parasitol. 2009;164(1):86–94. pmid:19111576; PubMed Central PMCID: PMCPMC2663568.
  142. 142. Que X, Wunderlich A, Joiner KA, Reed SL. Toxopain-1 is critical for infection in a novel chicken embryo model of congenital toxoplasmosis. Infect Immun. 2004;72(5):2915–21. pmid:15102804; PubMed Central PMCID: PMCPMC387868.
  143. 143. Dou Z, Coppens I, Carruthers VB. Non-canonical maturation of two papain-family proteases in Toxoplasma gondii. J Biol Chem. 2013;288(5):3523–34. pmid:23250753; PubMed Central PMCID: PMCPMC3561571.
  144. 144. Chaparro JD, Cheng T, Tran UP, Andrade RM, Brenner SBT, Hwang G, et al. Two key cathepsins, TgCPB and TgCPL, are targeted by the vinyl sulfone inhibitor K11777 in in vitro and in vivo models of toxoplasmosis. PLoS ONE. 2018;13(3):e0193982. pmid:29565998; PubMed Central PMCID: PMCPMC5863946.
  145. 145. Nguyen HM, El Hajj H, El Hajj R, Tawil N, Berry L, Lebrun M, et al. Toxoplasma gondii autophagy-related protein ATG9 is crucial for the survival of parasites in their host. Cell Microbiol. 2017;19(6):e12712. pmid:27992947.
  146. 146. Zhao G, Zhou A, Lv G, Meng M, Sun M, Bai Y, et al. Toxoplasma gondii cathepsin proteases are undeveloped prominent vaccine antigens against toxoplasmosis. BMC Infect Dis. 2013;13:207. pmid:23651838; PubMed Central PMCID: PMCPMC3659040.
  147. 147. Han Y, Zhou A, Lu G, Zhao G, Sha W, Wang L, et al. DNA Vaccines Encoding Toxoplasma gondii Cathepsin C 1 Induce Protection against Toxoplasmosis in Mice. Korean J Parasitol. 2017;55(5):505–12. pmid:29103265; PubMed Central PMCID: PMCPMC5678475.
  148. 148. Grab DJ, Garcia-Garcia JC, Nikolskaia OV, Kim YV, Brown A, Pardo CA, et al. Protease activated receptor signaling is required for African trypanosome traversal of human brain microvascular endothelial cells. PLoS Negl Trop Dis. 2009;3(7):e479. pmid:19621073; PubMed Central PMCID: PMCPMC2707606.
  149. 149. Mahmoudzadeh-Niknam H, McKerrow JH. Leishmania tropica: cysteine proteases are essential for growth and pathogenicity. Exp Parasitol. 2004;106(3–4):158–63. Epub 2004/06/03. pmid:15172223.
  150. 150. Besteiro S, Williams RA, Morrison LS, Coombs GH, Mottram JC. Endosome sorting and autophagy are essential for differentiation and virulence of Leishmania major. J Biol Chem. 2006;281(16):11384–96. pmid:16497676.
  151. 151. De Souza Leao S, Lang T, Prina E, Hellio R, Antoine JC. Intracellular Leishmania amazonensis amastigotes internalize and degrade MHC class II molecules of their host cells. J Cell Sci. 1995;108(Pt 10):3219–31. pmid:7593283.
  152. 152. Abdulla MH, O'Brien T, Mackey ZB, Sajid M, Grab DJ, McKerrow JH. RNA interference of Trypanosoma brucei cathepsin B and L affects disease progression in a mouse model. PLoS Negl Trop Dis. 2008;2(9):e298. Epub 2008/09/30. pmid:18820745; PubMed Central PMCID: PMC2553486.
  153. 153. Garcia MP, Nobrega OT, Teixeira AR, Sousa MV, Santana JM. Characterisation of a Trypanosoma cruzi acidic 30 kDa cysteine protease. Mol Biochem Parasitol. 1998;91(2):263–72. pmid:9566519.
  154. 154. Tolbert MK, Brand MD, Gould EN. In vitro effects of cysteine protease inhibitors on Trichomonas foetus-induced cytopathic changes in porcine intestinal epithelial cells. Am J Vet Res. 2016;77(8):890–7. pmid:27463553.
  155. 155. Cobo ER, Reed SL, Corbeil LB. Effect of vinyl sulfone inhibitors of cysteine proteinases on Tritrichomonas foetus infection. Int J Antimicrob Agents. 2012;39(3):259–62. pmid:22104282; PubMed Central PMCID: PMCPMC3279618.
  156. 156. Chen YT, Brinen LS, Kerr ID, Hansell E, Doyle PS, McKerrow JH, et al. In vitro and in vivo studies of the trypanocidal properties of WRR-483 against Trypanosoma cruzi. PLoS Negl Trop Dis. 2010;4(9):e825. pmid:20856868; PubMed Central PMCID: PMCPMC2939063.
  157. 157. Stolze SC, Deu E, Kaschani F, Li N, Florea BI, Richau KH, et al. The antimalarial natural product symplostatin 4 is a nanomolar inhibitor of the food vacuole falcipains. Chem Biol. 2012;19(12):1546–55. pmid:23261598; PubMed Central PMCID: PMCPMC3601557.
  158. 158. Brak K, Kerr ID, Barrett KT, Fuchi N, Debnath M, Ang K, et al. Nonpeptidic tetrafluorophenoxymethyl ketone cruzain inhibitors as promising new leads for Chagas disease chemotherapy. J Med Chem. 2010;53(4):1763–73. pmid:20088534; PubMed Central PMCID: PMCPMC2838180.
  159. 159. Gauthier JY, Chauret N, Cromlish W, Desmarais S, Duong LT, Falgueyret JP, et al. The discovery of odanacatib (MK-0822), a selective inhibitor of cathepsin K. Bioorg Med Chem Lett. 2008;18(3):923–8. pmid:18226527.
  160. 160. Duong le T, Leung AT, Langdahl B. Cathepsin K Inhibition: A New Mechanism for the Treatment of Osteoporosis. Calcif Tissue Int. 2016;98(4):381–97. pmid:26335104.
  161. 161. Brumatti G, Ma C, Lalaoui N, Nguyen NY, Navarro M, Tanzer MC, et al. The caspase-8 inhibitor emricasan combines with the SMAC mimetic birinapant to induce necroptosis and treat acute myeloid leukemia. Sci Transl Med. 2016;8(339):339ra69. pmid:27194727.
  162. 162. McKerrow JH. Development of cysteine protease inhibitors as chemotherapy for parasitic diseases: insights on safety, target validation, and mechanism of action. Int J Parasitol. 1999;29(6):833–7. pmid:10480720.
  163. 163. McKerrow JH, Caffrey C, Kelly B, Loke P, Sajid M. Proteases in parasitic diseases. Annu Rev Pathol. 2006;1:497–536. pmid:18039124.