2,018
25
Essay, 28 pages (7000 words)

Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host

The Importance of Sialo-Containing Glycans in Cell Biology

All living cells possess a complex and dense coating composed of glycoconjugates ( Varki, 1993 ). Such structures exist as glycolipids, glycoproteins, and proteoglycans. Among other functions, cell surface glycans mediate cell–cell recognition and control interactions between cells and other components in their local environment. Throughout evolution, glycans were designated several roles based on their physical features and structural diversity ( Varki, 2011 ). Advances in glycobiology researches have provided the means to correlate glycan structures to physiological and pathological conditions ( Lowe and Marth, 2003 ; Baum et al., 2014 ; Schmaus et al., 2014 ; Kato and Ishiwa, 2015 ; Stowell et al., 2015 ). The result is that glycoscience is emerging as a major contributor to the understanding of biology and medicine. The functional specificity of glycans is often dictated by the monosaccharides at the outermost ends of glycan chains ( Berger et al., 1982 ; Varki, 1993 ; Hart, 2013 ). In mammals, the glycan chain of glycoproteins and glycolipids often end with sialic acids (Sia), a group of sugars with distinctive chemical properties that determine their functions ( Traving and Schauer, 1998 ). Discovered about 70 years ago, Sia is a generic term referring to nine-carbon alpha (α)-keto sugars with a carboxyl group linked directly to the ring as well as its N or O substituted derivatives. There are over 50 Sia described, while the two most commonly found in mammals are N -acetyl and N -glycolyl neuraminic acid ( Varki and Schauer, 2009 ). It is estimated that a cell exhibits tens of millions Sia molecules, with the concentration in the glycocalyx approaching 100 mM ( Varki and Gagneux, 2012 ). Thanks to its bulky size and the electron density of the carboxyl group, Sia provides an important source of negative charge, which could alter the biophysical properties of cellular interactions. In eukaryotic cells, Sia residues are responsible for mediating many important interactions, including but not limited to regulation of transmembrane receptor function, altering membrane transport, control of glycoproteins (and even cell) half-life ( Schauer, 2000 ). One of the most notable functions of Sia is to regulate immune response, playing an important role in self and non-self recognition ( Traving and Schauer, 1998 ). It should not come as a surprise that several pathogenic microorganisms, such as viruses, bacteria, and protozoa have evolved strategies to exploit this feature ( Neu et al., 2011 ; Nishikawa et al., 2013 ; Chang and Nizet, 2014 ). One of the more stylish mechanisms of cell surface sialylation is exploited by the protozoal Trypanosoma cruzi , the etiological agent of Chagas disease ( Previato et al., 1985 ; Zingales et al., 1987 ; Schenkman et al., 1991 , 1992 ; Mucci et al., 2006 ; Muia et al., 2010 ).

Trypanosoma cruzi trans -Sialidase and Host Cell Invasion

Incapable of synthetizing Sia, T. cruzi has developed an elegant mechanism to take advantage of the Sia residues displayed in its mammalian host sialoglycans in order to avoid detection and elimination by immune cells ( Pereira-Chioccola and Schenkman, 1999 ; Dc-Rubin and Schenkman, 2012 ; Freire-de-Lima et al., 2015 ). Since it was shown that T. cruzi could not use the activated monosaccharide precursor CMP-Neu5Ac, as it was first considered ( Schauer et al., 1983 ), it was proposed a novel metabolic mechanism of trans -glycosylation, by which T. cruzi would incorporate Sia onto its own cell surface ( Previato et al., 1985 ). Incorporation of Sia on the surface molecules of T. cruzi parasite is catalyzed by the enzyme called trans -sialidase (Tc-TS) Previato et al., 1985 ; Schenkman et al., 1991 ). It was demonstrated in vitro and in vivo ( Zingales et al., 1987 ; Previato et al., 1990 ) the activity of Tc-TS was characterized as specific for α-2, 3 Sia, being responsible for the transfer of Sia residues from the mammal host surface sialylated molecules to the parasite mucin-like acceptors ( Schenkman et al., 1991 ; Parodi et al., 1992 ; Ruiz Rde et al., 1993 ; Previato et al., 1994 ).

In T. cruzi , the relevance of the acquisition of Sia is not completely known. However, there are proposals that such an event might help trypomastigote forms adhere and penetrate into non-phagocytic cells ( Ming et al., 1993 ; Schenkman et al., 1993 ). Sia also provides a strong negatively charged cover that protects parasites against human lytic antibodies specific for α-galactosyl residues present on the trypomastigote surface ( Pereira-Chioccola et al., 2000 ).

The transfer of a Sia residue onto an acceptor hinders the access of a second residue when two potential acceptor sites are present on the same oligosaccharide ( Previato et al., 1995 ). Tc-TS can also operate as a viral and bacterial sialidase, irreversibly transferring Sia to water in the absence of a carbohydrate acceptor ( Vandekerckhove et al., 1992 ). Analysis of the crystal structure revealed that the molecular architecture of the Tc-TS active site preserves numerous conserved features of microbial sialidases ( Buschiazzo et al., 2002 ). Furthermore, all sialidases studied so far, including Tc-TS, catalyze sialoside hydrolysis with configuration retention ( Todeschini et al., 2000 ).

There has been significant progress in understanding the importance of Tc-TS on the biology of T. cruzi . However, obstacles such as the lack of knockout strains, as well as the absence of specific Tc-TS inhibitors pose difficulties for the progress in the area. In regard to the Tc-TS inhibitors, many of them are not strong enough to serve as a scaffold to the development of drug targeting Tc-TS. In addition, most are derivatives of the substrate Sia or a transition state analog known as 2, 3-dehydro-3-deoxy- N -acetylneuraminic acid (DANA), with in vitro IC(50) values near the milimolar range and low in vivo efficacy ( Uemura et al., 1992 ; Amaya et al., 2003 ; Arioka et al., 2010 ; Dc-Rubin and Schenkman, 2012 ). Recent studies suggest that high affinity inhibitory monoclonal antibodies (mAb) for Tc-TS might provide a rational framework for novel approaches in the design of chemotherapeutic drugs ( Buschiazzo et al., 2012 ).

Over the last years, several papers described by Campetella’s group demonstrated that active Tc-TS (aTS) induces systemic effects during the acute phase of the disease ( Mucci et al., 2002 , 2005 ; Tribulatti et al., 2005 ; Risso et al., 2007 ; Ruiz Díaz et al., 2015 ), until the elicitation of broad neutralizing antibodies ( Pitcovsky et al., 2002 ). Buschiazzo et al. (2012) reported the identification and detailed characterization of the neutralizing mouse mAb 13G9, which was able to recognize and inhibit Tc-TS with high specificity. The crystal structure of the complex involving the antigen-binding fragment and the globular region of Tc-TS demonstrated that not obstructing the enzyme’s catalytic site, the antibody inhibited the movement of an assisting tyrosine (Y 119 ), which plays an important role in the trans -glycosidase mechanism ( Buschiazzo et al., 2002 ). The authors suggest that such results may bring into light new strategies for chemotherapy in Chagas disease and for disclosure of aTS function in T. cruzi pathogenesis and biology.

Besides encoding aTS, the genes of the TS family also encode its inactive analog (iTS). iTS has a His at position 342, while active members contain a Tyr at the same position ( Cremona et al., 1995 ; Egima et al., 1996 ). It has been suggested that iTS may mediate the initial contact between T. cruzi and host cells, functioning as an adhesin that contains two sugar-binding distinct sites, for Sia and β-galactose ( Todeschini et al., 2004 ). Recently, Ruiz Díaz et al. (2015) demonstrated that both aTS and iTS besides to induce a Th2-like phenotype in naive T cells, also compromise the emergence of Th1 cells. Additionally, both isoforms were associated with the parasite’s ability to reduce IL-2 production and IL-2Ra expression by T cells. The authors suggest that TS proteins are able to manipulate the TCD4 + response throughout their maturation stages to favor parasite survival and infection.

The bivalent nature of iTS could promote a glycan cross-linking believed to be essential for cellular signal transduction. Tc-TS has essentially two different domains. The N-terminal, which contains the catalytic domain of the enzyme ( Ribeirao et al., 1997 ), and the C-terminal, which is composed basically of 12 amino acids-long units repeated in tandem, termed SAPA (shed acute-phase antigen; Affranchino et al., 1989 ), which is not required for TS activity ( Campetella et al., 1994 ). Tc-TS is linked to the parasite membrane via glycosylphosphatidylinositol (GPI) anchor and shed into the bloodstream during the infection ( Schenkman et al., 1994 ). Nowadays, it is well recognized that Tc-TS is an important virulence factor involved in cell invasion and pathogenesis of Chagas disease ( Mendonça-Previato et al., 2010 ; Dc-Rubin and Schenkman, 2012 ; Miller and Roitberg, 2013 ; Freire-de-Lima et al., 2015 ). However, the studies regarding the importance of Tc-TS for cell invasion and parasite survival were paramount to understanding how T. cruzi is capable of subverting the immune system by disturbing the host cells sialoglycophenotype.

Roles of Sialic Acids on T Cell Biology

It is well documented that in T cells, different lectin families recognize distinct sialylated ligands on glycoproteins or glycolipids to regulate their functions ( Rabinovich et al., 2002a , b ; Ley and Kansas, 2004 ; Crocker et al., 2007 ; Crocker and Redelinghuys, 2008 ). Examples include siglecs, a family of Sia-binding lectins that belong to the superfamily of I-type lectins ( Crocker and Redelinghuys, 2008 ). Most siglecs bind α-2, 3 and/or α-2, 6-linked Sia on penultimate Gal residues on cell surface glycoproteins or glycolipids; however, some members bind to repeating Sia chains ( Varki and Angata, 2006 ). In the immune system, siglecs mainly function as controllers of cell signaling, although roles for some siglecs in pathogen recognition and innate immunity have been proposed ( von Gunten and Bochner, 2008 ). Erdmann et al. (2009) demonstrated that sialylated ligands on T. cruzi interact with Siglec-E (sialic acid-binding Ig-like lectin-E) on dendritic cells (DC), and such interaction suppressed the production of the proinflammatory cytokine IL-12 and subsequent T cell activation. Recently, it was proposed that sulfates from cruzipain, another important antigen expressed by T. cruzi , might play an essential role in the interaction with Siglec-E on inflammatory cells, favoring the persistence of the parasite in its mammalian hosts ( Ferrero et al., 2016 ).

Sia plays crucial roles in the regulation of host immunity, as evidenced by the sialylation changes sustained by naïve T cells during thymic selection, a tightly regulated process, essential for establishing central tolerance ( Daniels et al., 2001 ; Moody et al., 2001 ; Cao and Crocker, 2011 ; Paulson et al., 2012 ). The alterations suffered on cell surface N – and O -linked glycans, including glycoprotein sialylation, during T cell development and differentiation ( Daniels et al., 2002 ) might regulate the T cell response through a direct effect on the intrinsic properties of specific proteins, or by modulating the binding of a disparate set of cell surface proteins to a specific carbohydrate moiety. It is important to bring out that many of the changes in the degree of sialylation of carbohydrate chains observed in T cells during their development can be monitored with plant lectins ( Sharon, 1983 ; Daniels et al., 2002 ). It has been shown in vitro and in vivo that T cell activation is accompanied by loss of Sia from core 1 O -glycans (Siaα2, 3Galβ1, 3GalNAc-Ser/Thr; Galvan et al., 1998 ; Bi and Baum, 2009 ), which leads to exposure of asialo core 1 O -glycans. Such residues can be detected with the plant lectin peanut agglutinin (PNA), that recognizes Galβ1, 3GalNAc sequences on several glycoproteins including CD8, CD43, and CD45 ( Wu et al., 1996 ). It is a known fact that the loss of cell-surface Sia during T cell activation enhances TCR reactivity with antigens ( Galvan et al., 1998 ; Harrington et al., 2000 ; Bi and Baum, 2009 ). Indeed, T cell cytolytic activity may be boosted by sialidase treatment, with a simultaneous reduction in cell surface negative charge ( Sadighi Akha et al., 2006 ). In addition, de-sialylated CD8 + T cells undergo more rounds of cell division following contact with antigen ( Pappu and Shrikant, 2004 ). Recognition and disposal of pathogens rely upon the concerted actions of adaptive and innate immunological mechanisms, including recruitment of different cell types as well as production and release of cytokines, antibodies or other effector molecules ( Janeway, 2001 ). In regards to adaptive response, while the importance of CD8 + T cells is well described when it comes to the control of several viral and bacterial infections, it is seldom mentioned that eukaryotic pathogens, such as protozoans, are also subject to the action of cytotoxic lymphocytes. Chagas disease is probably the most well understood example of the role CD8 + T cells play during protozoan infections. In its vertebrate hosts, T. cruzi is perpetually changing between its intracellular amastigote forms and extracellular trypomastigote ones. There is a great range of cells that are subject to invasion by the parasite, including but not limited to fibroblasts, macrophages, myocytes, and adipocytes. During the intracellular stages, T. cruzi antigens are presented by antigen presenting cells (APCs) through by MHC class I molecules, providing many opportunities for detection by CD8 + T cells ( Garg et al., 1997 ). Several reports confirmed the importance of CD8 + T cell cytotoxicity regarding Chagas disease ( Tarleton, 2007 , 2015 ; Padilla et al., 2009 ; Dos Santos Virgilio et al., 2014 ). One of the most important was the identification of a number of epitopes expressed in proteins belonging to the TS family genes ( Martin et al., 2006 ). Until that point, there was a lack of immunodominant T cell epitopes, but now it is accepted that members of the TS gene family are a good target for CD8 + T cells. That discovery has not only led to a better understanding of how the immune system reacts to T. cruzi infection, but in a more practical venue has also fueled the research on vaccine development ( Machado et al., 2006 ; Araujo et al., 2014 ; Bontempi et al., 2015 ). Although it is universally accepted that CD8 + T cell response is an important factor in mediating host survival during T. cruzi infection, there is a substantial delay in the appearance of antigen specific CD8 + T cells following infection ( Martin et al., 2006 ; Tzelepis et al., 2006 ). This delay differs from the quick response of CD8 + T cells in other bacterial, viral, and, even protozoal infections ( Kaech et al., 2002 ), suggesting that a mechanism of immune subversion is operative.

Trypanosoma cruzi trans -Sialidase Modulates T Cell Sialoglycophenotype

As mentioned above, Sia content of T cell glycoproteins and glycolipids is regulated during both T cell development in the thymus and activation in the periphery ( Moody et al., 2001 ; Pappu and Shrikant, 2004 ). Each T cell subset expresses a specific set of glycan-modifying enzymes that regulate the pattern of sialylation on the cell surface ( Bi and Baum, 2009 ). Besides being able to sialylate mucin-like molecules expressed on the parasite surface, Tc-TS is also capable of re-sialylating host cell asialoglycoconjugates ( Mucci et al., 2006 ). Regarding T cells, such event is able to modulate both their function and half-life. Several papers published elsewhere have demonstrated that alterations in Sia residues on thymocytes and mature T cells are able to induce apoptosis ( Leguizamon et al., 1999 ; Mucci et al., 2002 , 2005 , 2006 ). Given the way TS works, it comes as a logical conclusion that the parasite’s ability to modulate the T cell sialophenotype is a powerful tool in evading detection and elimination by the host immune system. Pioneering studies on the impact of Tc-TS on T cells ( Chuenkova and Pereira, 1995 ) demonstrated that intravenous injection of minute amounts of native Tc-TS was able to increase the parasitemia and mortality of T. cruzi -infected mice. The authors suggested that such effects were entirely dependent on Tc-TS activity, since the same events did not occur in mice primed with viral or bacterial sialidases. Further in vitro experiments demonstrated that recombinant Tc-TS binds host T-lymphocytes, activating CD4 + T cells through CD43 engagement ( Todeschini et al., 2002a , b ). However, molecular evidences revealed that CD45 might be the main acceptor of Tc-TS during T. cruzi infection ( Muia et al., 2010 ). In vivo studies have highlighted the impact of Tc-TS on B and T cells sialoglycophenotypes ( Freire-de-Lima et al., 2010 ; Bermejo et al., 2013 ), reinforcing the idea that the Tc-TS may act as a virulence factor during T. cruzi infection. In B cells, the sialylation of cell surface molecules by Tc-TS elicits the production of IL-17. Interestingly, such event triggers a signaling pathway that differs from the one classically associated with IL-17, bypassing the activation of RORγt and RORα ( Bermejo et al., 2013 ). As for T cells, Freire-de-Lima et al. (2010) demonstrated the relevance of Tc-TS on the sialoglycophenotype of splenic CD8 + T cells during T. cruzi infection. T. cruzi -infected or Plasmodium berghei -infected mice were sacrificed on the eighth day post-infection (dpi), and the sialoglycophenotype of CD8 + T cells was assessed. As expected, CD8 + T cells from P. berghei -infected mice were highly positive for PNA (PNA high ). Interestingly, the glycophenotype PNA intermediate (PNA int ) exhibited by CD8 + T cells derived from T. cruzi -infected mice, becomes PNA low after intravenous administration recombinant aTS. Such event was able to inhibit the cytotoxic response mediated by antigen specific CD8 + T cells, supporting the idea that re-sialylation of asialoglycans on the surface of activated T cells might be a sophisticated evasion mechanism adopted by T. cruzi to subvert the host immune response ( Freire-de-Lima et al., 2010 ). In addition, we found that CD8 + T cells from T. cruzi -infected mice primed with recombinant iTS exhibited a glycophenotype PNA high , suggesting that iTS might have been able to compete with the native aTS by serum sialoglycoproteins, compromising the expected event of re-sialylation that takes place during the acute phase of Chagas disease ( Freire-de-Lima et al., 2010 ). Although the evidence is quite compelling, new experiments with ST3 Gal-I KO mice must be conducted in order to understand the effects triggered by Tc-Ts on CD8 + T cells during T. cruzi infection. Several immunobiological effects mediated by Tc-TS have been described elsewhere ( Mucci et al., 2006 ; Ruiz Díaz et al., 2015 ), but the chemical evidence of the enzyme’s essential role in the parasite is still lacking due to the absence of specific inhibitors of Tc-TS. As it stands Tc-TS is a promising target in drug design, an area in which further research is urgently needed.

Conclusion

Over the last years, several papers have demonstrated the importance of differential sialylation for CD8 + T cells with respect to their maturation ( Moody et al., 2001 ; Naito-Matsui et al., 2014 ), activation ( Pappu and Shrikant, 2004 ), and cytotoxic responses ( Sadighi Akha et al., 2006 ). However, few studies provided insights into the impact of surface sialylation of CD8 + T cells on infectious diseases. Infection with T. cruzi is of particular interest in this context because the parasite releases into the host plasma large amounts of proteins belonging to the TS family ( Frasch, 1994 ; Schenkman et al., 1994 ; Giorgi and de Lederkremer, 2011 ; Freire-de-Lima et al., 2012 , 2015 ). Recent studies demonstrated that cell surface asialoglycans on B and T lymphocytes might be sialylated by the action of Tc-TS ( Freire-de-Lima et al., 2010 ; Bermejo et al., 2013 ). Such events were able to disturb the host immune response and allow for the continuation of the parasite in the infected host. Since it is universally accepted that Tc-TS acts as a virulence factor during T. cruzi infection, further efforts are needed in this lively area to better understand the catalytic mechanism, as well as the functional properties of the Sia-dependent enzyme. Certainly, new impacting findings will contribute to further research on the enzyme as a therapeutic target in the fight against Chagas’ disease.

Author Contributions

Wrote the paper: LFL, LMF, VAS, KMC, AM, CGFL, JOP and LM-P. All authors read and approved the final version of the manuscript.

Funding

This work was supported by Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Coordenação de Aperfeiçoamento de Pessoal de Ensino Superior (CAPES), and Conselho Nacional de Desenvolvimento Cientifico e Tecnológico (CNPq).

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

Affranchino, J. L., Ibanez, C. F., Luquetti, A. O., Rassi, A., Reyes, M. B., Macina, R. A., et al. (1989). Identification of a Trypanosoma cruzi antigen that is shed during the acute phase of Chagas’ disease. Mol. Biochem. Parasitol. 34, 221–228. doi: 10. 1016/0166-6851(89)90050-9

||

Amaya, M. F., Buschiazzo, A., Nguyen, T., and Alzari, P. M. (2003). The high resolution structures of free and inhibitor-bound Trypanosoma rangeli sialidase and its comparison with T. cruzi trans-sialidase. J. Mol. Biol. 325, 773–784. doi: 10. 1016/S0022-2836(02)01306-2

||

Araujo, A. F., de Oliveira, G., Vasconcelos, J. F., Ersching, J., Dominguez, M. R., Vasconcelos, J. R., et al. (2014). Genetic vaccination against experimental infection with myotropic parasite strains of Trypanosoma cruzi . Mediators Inflamm. 2014: 605023. doi: 10. 1155/2014/605023

||

Arioka, S., Sakagami, M., Uematsu, R., Yamaguchi, H., Togame, H., Takemoto, H., et al. (2010). Potent inhibitor scaffold against Trypanosoma cruzi trans-sialidase. Bioorg. Med. Chem. 18, 1633–1640. doi: 10. 1016/j. bmc. 2009. 12. 062

||

Baum, L. G., Garner, O. B., Schaefer, K., and Lee, B. (2014). Microbe-host interactions are positively and negatively regulated by galectin-glycan interactions. Front. Immunol. 5: 284. doi: 10. 3389/fimmu. 2014. 00284

||

Berger, E. G., Buddecke, E., Kamerling, J. P., Kobata, A., Paulson, J. C., and Vliegenthart, J. F. (1982). Structure, biosynthesis and functions of glycoprotein glycans. Experientia 38, 1129–1162. doi: 10. 1007/BF01959725

|

Bermejo, D. A., Jackson, S. W., Gorosito-Serran, M., Acosta-Rodriguez, E. V., Amezcua-Vesely, M. C., Sather, B. D., et al. (2013). Trypanosoma cruzi trans-sialidase initiates a program independent of the transcription factors RORgammat and Ahr that leads to IL-17 production by activated B cells. Nat. Immunol. 14, 514–522. doi: 10. 1038/ni. 2569

||

Bi, S., and Baum, L. G. (2009). Sialic acids in T cell development and function. Biochim. Biophys. Acta 1790, 1599–1610. doi: 10. 1016/j. bbagen. 2009. 07. 027

||

Bontempi, I. A., Vicco, M. H., Cabrera, G., Villar, S. R., Gonzalez, F. B., Roggero, E. A., et al. (2015). Efficacy of a trans-sialidase-ISCOMATRIX subunit vaccine candidate to protect against experimental Chagas disease. Vaccine 33, 1274–1283. doi: 10. 1016/j. vaccine. 2015. 01. 044

||

Buschiazzo, A., Amaya, M. F., Cremona, M. L., Frasch, A. C., and Alzari, P. M. (2002). The crystal structure and mode of action of trans-sialidase, a key enzyme in Trypanosoma cruzi pathogenesis. Mol. Cell. 10, 757–768. doi: 10. 1016/S1097-2765(02)00680-9

||

Buschiazzo, A., Muia, R., Larrieux, N., Pitcovsky, T., Mucci, J., and Campetella, O. (2012). Trypanosoma cruzi trans-sialidase in complex with a neutralizing antibody: structure/function studies towards the rational design of inhibitors. PLoS Pathog. 8: e1002474. doi: 10. 1371/journal. ppat. 1002474

||

Campetella, O. E., Uttaro, A. D., Parodi, A. J., and Frasch, A. C. (1994). A recombinant Trypanosoma cruzi trans-sialidase lacking the amino acid repeats retains the enzymatic activity. Mol. Biochem. Parasitol. 64, 337–340. doi: 10. 1016/0166-6851(94)00036-0

||

Cao, H., and Crocker, P. R. (2011). Evolution of CD33-related siglecs: regulating host immune functions and escaping pathogen exploitation? Immunology 132, 18–26. doi: 10. 1111/j. 1365-2567. 2010. 03368. x

||

Chang, Y. C., and Nizet, V. (2014). The interplay between Siglecs and sialylated pathogens. Glycobiology 24, 818–825. doi: 10. 1093/glycob/cwu067

||

Chuenkova, M., and Pereira, M. E. (1995). Trypanosoma cruzi trans-sialidase: enhancement of virulence in a murine model of Chagas’ disease. J. Exp. Med. 181, 1693–1703. doi: 10. 1084/jem. 181. 5. 1693

||

Cremona, M. L., Sanchez, D. O., Frasch, A. C., and Campetella, O. (1995). A single tyrosine differentiates active and inactive Trypanosoma cruzi trans-sialidases. Gene 160, 123–128. doi: 10. 1016/0378-1119(95)00175-6

||

Crocker, P. R., Paulson, J. C., and Varki, A. (2007). Siglecs and their roles in the immune system. Nat. Rev. Immunol. 7, 255–266. doi: 10. 1038/nri2056

||

Crocker, P. R., and Redelinghuys, P. (2008). Siglecs as positive and negative regulators of the immune system. Biochem. Soc. Trans. 36, 1467–1471. doi: 10. 1042/BST0361467

||

Daniels, M. A., Devine, L., Miller, J. D., Moser, J. M., Lukacher, A. E., Altman, J. D., et al. (2001). CD8 binding to MHC class I molecules is influenced by T cell maturation and glycosylation. Immunity 15, 1051–1061. doi: 10. 1016/S1074-7613(01)00252-7

||

Daniels, M. A., Hogquist, K. A., and Jameson, S. C. (2002). Sweet ‘ n’ sour: the impact of differential glycosylation on T cell responses. Nat. Immunol. 3, 903–910. doi: 10. 1038/ni1002-903

||

Dc-Rubin, S. S., and Schenkman, S. (2012). T rypanosoma cruzi trans-sialidase as a multifunctional enzyme in Chagas’ disease. Cell Microbiol 14, 1522–1530. doi: 10. 1111/j. 1462-5822. 2012. 01831. x

||

Dos Santos Virgilio, F., Pontes, C., Dominguez, M. R., Ersching, J., Rodrigues, M. M., and Vasconcelos, J. R. (2014). CD8(+) T cell-mediated immunity during Trypanosoma cruzi infection: a path for vaccine development? Mediators Inflamm. 2014: 243786. doi: 10. 1155/2014/243786

|

Egima, C. M., Briones, M. R., Freitas Junior, L. H., Schenkman, R. P., Uemura, H., and Schenkman, S. (1996). Organization of trans-sialidase genes in Trypanosoma cruzi . Mol. Biochem. Parasitol. 77, 115–125. doi: 10. 1016/0166-6851(96)02582-0

||

Erdmann, H., Steeg, C., Koch-Nolte, F., Fleischer, B., and Jacobs, T. (2009). Sialylated ligands on pathogenic Trypanosoma cruzi interact with Siglec-E (sialic acid-binding Ig-like lectin-E). Cell Microbiol. 11, 1600–1611. doi: 10. 1111/j. 1462-5822. 2009. 01350. x

||

Ferrero, M. R., Heins, A. M., Soprano, L. L., Acosta, D. M., Esteva, M. I., Jacobs, T., et al. (2016). Involvement of sulfates from cruzipain, a major antigen of Trypanosoma cruzi , in the interaction with immunomodulatory molecule Siglec-E. Med. Microbiol. Immunol. 205, 21–35. doi: 10. 1007/s00430-015-0421-2

||

Frasch, A. C. (1994). Trans-sialidase, SAPA amino acid repeats and the relationship between Trypanosoma cruzi and the mammalian host. Parasitology 108(Suppl.), S37–S44. doi: 10. 1017/S0031182000075703

||

Freire-de-Lima, L., Alisson-Silva, F., Carvalho, S. T., Takiya, C. M., Rodrigues, M. M., DosReis, G. A., et al. (2010). Trypanosoma cruzi subverts host cell sialylation and may compromise antigen-specific CD8+ T cell responses. J. Biol. Chem. 285, 13388–13396. doi: 10. 1074/jbc. M109. 096305

||

Freire-de-Lima, L., Fonseca, L. M., Oeltmann, T., Mendonca-Previato, L., and Previato, J. O. (2015). The trans-sialidase, the major Trypanosoma cruzi virulence factor: three decades of studies. Glycobiology 25, 1142–1149. doi: 10. 1093/glycob/cwv057

||

Freire-de-Lima, L., Oliveira, I. A., Neves, J. L., Penha, L. L., Alisson-Silva, F., Dias, W. B., et al. (2012). Sialic acid: a sweet swing between mammalian host and Trypanosoma cruzi . Front. Immunol. 3: 356. doi: 10. 3389/fimmu. 2012. 00356

||

Galvan, M., Murali-Krishna, K., Ming, L. L., Baum, L., and Ahmed, R. (1998). Alterations in cell surface carbohydrates on T cells from virally infected mice can distinguish effector/memory CD8+ T cells from naive cells. J. Immunol. 161, 641–648.

|

Garg, N., Nunes, M. P., and Tarleton, R. L. (1997). Delivery by Trypanosoma cruzi of proteins into the MHC class I antigen processing and presentation pathway. J. Immunol. 158, 3293–3302.

|

Giorgi, M. E., and de Lederkremer, R. M. (2011). Trans-sialidase and mucins of Trypanosoma cruzi : an important interplay for the parasite. Carbohydr. Res. 346, 1389–1393. doi: 10. 1016/j. carres. 2011. 04. 006

||

Harrington, L. E., Galvan, M., Baum, L. G., Altman, J. D., and Ahmed, R. (2000). Differentiating between memory and effector CD8 T cells by altered expression of cell surface O-glycans. J. Exp. Med. 191, 1241–1246. doi: 10. 1084/jem. 191. 7. 1241

||

Hart, G. W. (2013). Thematic minireview series on glycobiology and extracellular matrices: glycan functions pervade biology at all levels. J. Biol. Chem. 288: 6903. doi: 10. 1074/jbc. R113. 453977

|

Janeway, C. A. Jr. (2001). How the immune system protects the host from infection. Microbes Infect. 3, 1167–1171. doi: 10. 1073/pnas. 131202998

||

Kaech, S. M., Hemby, S., Kersh, E., and Ahmed, R. (2002). Molecular and functional profiling of memory CD8 T cell differentiation. Cell 111, 837–851. doi: 10. 1016/S0092-8674(02)01139-X

|

Kato, K., and Ishiwa, A. (2015). The role of carbohydrates in infection strategies of enteric pathogens. Trop. Med. Health 43, 41–52. doi: 10. 2149/tmh. 2014-25

||

Leguizamon, M. S., Mocetti, E., Garcia Rivello, H., Argibay, P., and Campetella, O. (1999). Trans-sialidase from Trypanosoma cruzi induces apoptosis in cells from the immune system in vivo. J. Infect. Dis. 180, 1398–1402. doi: 10. 1086/315001

||

Ley, K., and Kansas, G. S. (2004). Selectins in T-cell recruitment to non-lymphoid tissues and sites of inflammation. Nat. Rev. Immunol. 4, 325–335. doi: 10. 1038/nri1351

||

Lowe, J. B., and Marth, J. D. (2003). A genetic approach to Mammalian glycan function. Annu. Rev. Biochem. 72, 643–691. doi: 10. 1146/annurev. biochem. 72. 121801. 161809

|

Machado, A. V., Cardoso, J. E., Claser, C., Rodrigues, M. M., Gazzinelli, R. T., and Bruna-Romero, O. (2006). Long-term protective immunity induced against Trypanosoma cruzi infection after vaccination with recombinant adenoviruses encoding amastigote surface protein-2 and trans-sialidase. Hum. Gene Ther. 17, 898–908. doi: 10. 1089/hum. 2006. 17. 898

||

Martin, D. L., Weatherly, D. B., Laucella, S. A., Cabinian, M. A., Crim, M. T., Sullivan, S., et al. (2006). CD8+ T-Cell responses to Trypanosoma cruzi are highly focused on strain-variant trans-sialidase epitopes. PLoS Pathog. 2: e77. doi: 10. 1371/journal. ppat. 0020077

||

Mendonça-Previato, L., Todeschini, A. R., de Lima, L. F., and Previato, J. O. (2010). The trans-sialidase from Trypanosoma cruzi a putative target for trypanocidal agents. Open Parasitol. J. 4, 111–115. doi: 10. 2174/1874421401004010111

|

Miller, B. R. III, and Roitberg, A. E. (2013). Trypanosoma cruzi trans-sialidase as a drug target against Chagas disease (American trypanosomiasis). Future Med. Chem. 5, 1889–1900. doi: 10. 4155/fmc. 13. 129

||

Ming, M., Chuenkova, M., Ortega-Barria, E., and Pereira, M. E. (1993). Mediation of Trypanosoma cruzi invasion by sialic acid on the host cell and trans-sialidase on the trypanosome. Mol. Biochem. Parasitol. 59, 243–252. doi: 10. 1016/0166-6851(93)90222-J

||

Moody, A. M., Chui, D., Reche, P. A., Priatel, J. J., Marth, J. D., and Reinherz, E. L. (2001). Developmentally regulated glycosylation of the CD8alphabeta coreceptor stalk modulates ligand binding. Cell 107, 501–512. doi: 10. 1016/S0092-8674(01)00577-3

||

Mucci, J., Hidalgo, A., Mocetti, E., Argibay, P. F., Leguizamon, M. S., and Campetella, O. (2002). Thymocyte depletion in Trypanosoma cruzi infection is mediated by trans-sialidase-induced apoptosis on nurse cells complex. Proc. Natl. Acad. Sci. U. S. A. 99, 3896–3901. doi: 10. 1073/pnas. 052496399

||

Mucci, J., Mocetti, E., Leguizamon, M. S., and Campetella, O. (2005). A sexual dimorphism in intrathymic sialylation survey is revealed by the trans-sialidase from Trypanosoma cruzi . J. Immunol. 174, 4545–4550. doi: 10. 4049/jimmunol. 174. 8. 4545

||

Mucci, J., Risso, M. G., Leguizamon, M. S., Frasch, A. C., and Campetella, O. (2006). The trans-sialidase from Trypanosoma cruzi triggers apoptosis by target cell sialylation. Cell Microbiol. 8, 1086–1095. doi: 10. 1111/j. 1462-5822. 2006. 00689. x

||

Muia, R. P., Yu, H., Prescher, J. A., Hellman, U., Chen, X., Bertozzi, C. R., et al. (2010). Identification of glycoproteins targeted by Trypanosoma cruzi trans-sialidase, a virulence factor that disturbs lymphocyte glycosylation. Glycobiology 20, 833–842. doi: 10. 1093/glycob/cwq037

||

Naito-Matsui, Y., Takada, S., Kano, Y., Iyoda, T., Sugai, M., Shimizu, A., et al. (2014). Functional evaluation of activation-dependent alterations in the sialoglycan composition of T cells. J. Biol. Chem. 289, 1564–1579. doi: 10. 1074/jbc. M113. 523753

||

Neu, U., Bauer, J., and Stehle, T. (2011). Viruses and sialic acids: rules of engagement. Curr. Opin. Struct. Biol. 21, 610–618. doi: 10. 1016/j. sbi. 2011. 08. 009

||

Nishikawa, Y., Ogiso, A., Kameyama, K., Nishimura, M., Xuan, X., and Ikehara, Y. (2013). alpha2-3 Sialic acid glycoconjugate loss and its effect on infection with Toxoplasma parasites. Exp. Parasitol. 135, 479–485. doi: 10. 1016/j. exppara. 2013. 08. 009

||

Padilla, A. M., Bustamante, J. M., and Tarleton, R. L. (2009). CD8+ T cells in Trypanosoma cruzi infection. Curr. Opin. Immunol. 21, 385–390. doi: 10. 1016/j. coi. 2009. 07. 006

||

Pappu, B. P., and Shrikant, P. A. (2004). Alteration of cell surface sialylation regulates antigen-induced naive CD8+ T cell responses. J. Immunol. 173, 275–284. doi: 10. 4049/jimmunol. 173. 1. 275

||

Parodi, A. J., Pollevick, G. D., Mautner, M., Buschiazzo, A., Sanchez, D. O., and Frasch, A. C. (1992). Identification of the gene(s) coding for the trans-sialidase of Trypanosoma cruzi . EMBO J. 11, 1705–1710.

|

Paulson, J. C., Macauley, M. S., and Kawasaki, N. (2012). Siglecs as sensors of self in innate and adaptive immune responses. Ann. N. Y. Acad. Sci. 1253, 37–48. doi: 10. 1111/j. 1749-6632. 2011. 06362. x

||

Pereira-Chioccola, V. L., Acosta-Serrano, A., Correia de Almeida, I., Ferguson, M. A., Souto-Padron, T., Rodrigues, M. M., et al. (2000). Mucin-like molecules form a negatively charged coat that protects Trypanosoma cruzi trypomastigotes from killing by human anti-alpha-galactosyl antibodies. J. Cell Sci. 113(Pt 7), 1299–1307.

|

Pereira-Chioccola, V. L., and Schenkman, S. (1999). Biological role of Trypanosoma cruzi trans-sialidase. Biochem. Soc. Trans. 27, 516–518. doi: 10. 1016/S0169-4758(00)01698-7

||

Pitcovsky, T. A., Buscaglia, C. A., Mucci, J., and Campetella, O. (2002). A functional network of intramolecular cross-reacting epitopes delays the elicitation of neutralizing antibodies to Trypanosoma cruzi trans-sialidase. J. Infect. Dis. 186, 397–404. doi: 10. 1086/341463

||

Previato, J., Andrade, A., Vermelho, A., Firmino, J., and Mendonça-Previato, L. (1990). Evidence for N-glycolylneuraminic acid incorporation by Trypanosoma cruzi from infected animal. Mem. Inst. Oswaldo Cruz 85, 38–39. doi: 10. 1084/jem. 175. 2. 567

|

Previato, J. O., Andrade, A. F., Pessolani, M. C., and Mendonca-Previato, L. (1985). Incorporation of sialic acid into Trypanosoma cruzi macromolecules. A proposal for a new metabolic route. Mol. Biochem. Parasitol. 16, 85–96. doi: 10. 1016/0166-6851(85)90051-9

||

Previato, J. O., Jones, C., Goncalves, L. P., Wait, R., Travassos, L. R., and Mendonca-Previato, L. (1994). O-glycosidically linked N-acetylglucosamine-bound oligosaccharides from glycoproteins of Trypanosoma cruzi . Biochem. J. 301(Pt 1), 151–159. doi: 10. 1093/glycob/11. 1. 47

||

Previato, J. O., Jones, C., Xavier, M. T., Wait, R., Travassos, L. R., Parodi, A. J., et al. (1995). Structural characterization of the major glycosylphosphatidylinositol membrane-anchored glycoprotein from epimastigote forms of Trypanosoma cruzi Y-strain. J. Biol. Chem. 270, 7241–7250. doi: 10. 1074/jbc. 270. 13. 7241

||

Rabinovich, G. A., Baum, L. G., Tinari, N., Paganelli, R., Natoli, C., Liu, F. T., et al. (2002a). Galectins and their ligands: amplifiers, silencers or tuners of the inflammatory response? Trends Immunol . 23, 313–320. doi: 10. 1016/S1471-4906(02)02232-9

|

Rabinovich, G. A., Rubinstein, N., and Toscano, M. A. (2002b). Role of galectins in inflammatory and immunomodulatory processes. Biochim. Biophys. Acta 1572, 274–284. doi: 10. 1016/S0304-4165(02)00314-8

||

Ribeirao, M., Pereira-Chioccola, V. L., Eichinger, D., Rodrigues, M. M., and Schenkman, S. (1997). Temperature differences for trans-glycosylation and hydrolysis reaction reveal an acceptor binding site in the catalytic mechanism of Trypanosoma cruzi trans-sialidase. Glycobiology 7, 1237–1246. doi: 10. 1093/glycob/7. 8. 1237

||

Risso, M. G., Pitcovsky, T. A., Caccuri, R. L., Campetella, O., and Leguizamon, M. S. (2007). Immune system pathogenesis is prevented by the neutralization of the systemic trans-sialidase from Trypanosoma cruzi during severe infections. Parasitology 134, 503–510. doi: 10. 1017/S0031182006001752

||

Ruiz Díaz, P., Mucci, J., Meira, M. A., Bogliotti, Y., Musikant, D., Leguizamon, M. S., et al. (2015). Trypanosoma cruzi trans-sialidase prevents elicitation of Th1 cell response via interleukin 10 and downregulates Th1 effector cells. Infect. Immun. 83, 2099–2108. doi: 10. 1128/IAI. 00031-15

||

Ruiz Rde, C., Rigoni, V. L., Gonzalez, J., and Yoshida, N. (1993). The 35/50 kDa surface antigen of Trypanosoma cruzi metacyclic trypomastigotes, an adhesion molecule involved in host cell invasion. Parasite Immunol. 15, 121–125. doi: 10. 1111/j. 1365-3024. 1993. tb00591. x

||

Sadighi Akha, A. A., Berger, S. B., and Miller, R. A. (2006). Enhancement of CD8 T-cell function through modifying surface glycoproteins in young and old mice. Immunology 119, 187–194. doi: 10. 1111/j. 1365-2567. 2006. 02420. x

||

Schauer, R. (2000). Achievements and challenges of sialic acid research. Glycoconj. J. 17, 485–499. doi: 10. 1023/A: 1011062223612

|

Schauer, R., Reuter, G., Muhlpfordt, H., Andrade, A. F., and Pereira, M. E. (1983). The occurrence of N-acetyl- and N-glycoloylneuraminic acid in Trypanosoma cruzi . Hoppe Seylers Z. Physiol. Chem. 364, 1053–1057. doi: 10. 1515/bchm2. 1983. 364. 2. 1053

||

Schenkman, R. P., Vandekerckhove, F., and Schenkman, S. (1993). Mammalian cell sialic acid enhances invasion by Trypanosoma cruzi . Infect. Immun. 61, 898–902.

|

Schenkman, S., Eichinger, D., Pereira, M. E., and Nussenzweig, V. (1994). Structural and functional properties of Trypanosoma trans-sialidase. Annu. Rev. Microbiol. 48, 499–523. doi: 10. 1146/annurev. mi. 48. 100194. 002435

||

Schenkman, S., Jiang, M. S., Hart, G. W., and Nussenzweig, V. (1991). A novel cell surface trans-sialidase of Trypanosoma cruzi generates a stage-specific epitope required for invasion of mammalian cells. Cell 65, 1117–1125. doi: 10. 1016/0092-8674(91)90008-M

||

Schenkman, S., Pontes de Carvalho, L., and Nussenzweig, V. (1992). Trypanosoma cruzi trans-sialidase and neuraminidase activities can be mediated by the same enzymes. J. Exp. Med. 175, 567–575. doi: 10. 1084/jem. 175. 2. 567

||

Schmaus, A., Bauer, J., and Sleeman, J. P. (2014). Sugars in the microenvironment: the sticky problem of HA turnover in tumors. Cancer Metastasis Rev. 33, 1059–1079. doi: 10. 1007/s10555-014-9532-2

||

Sharon, N. (1983). Lectin receptors as lymphocyte surface markers. Adv. Immunol. 34, 213–298. doi: 10. 1016/S0065-2776(08)60380-6

|

Stowell, S. R., Ju, T., and Cummings, R. D. (2015). Protein glycosylation in cancer. Annu. Rev. Pathol. 10, 473–510.

Tarleton, R. L. (2007). Immune system recognition of Trypanosoma cruzi . Curr. Opin. Immunol. 19, 430–434. doi: 10. 1016/j. coi. 2007. 06. 003

||

Tarleton, R. L. (2015). CD8+ T cells in Trypanosoma cruzi infection. Semin. Immunopathol. 37, 233–238. doi: 10. 1007/s00281-015-0481-9

||

Todeschini, A. R., Dias, W. B., Girard, M. F., Wieruszeski, J. M., Mendonca-Previato, L., and Previato, J. O. (2004). Enzymatically inactive trans-sialidase from Trypanosoma cruzi binds sialyl and beta-galactopyranosyl residues in a sequential ordered mechanism. J. Biol. Chem. 279, 5323–5328. doi: 10. 1074/jbc. M310663200

||

Todeschini, A. R., Girard, M. F., Wieruszeski, J. M., Nunes, M. P., DosReis, G. A., Mendonca-Previato, L., et al. (2002a). trans-Sialidase from Trypanosoma cruzi binds host T-lymphocytes in a lectin manner. J. Biol. Chem. 277, 45962–45968. doi: 10. 1074/jbc. M203185200

||

Todeschini, A. R., Mendonca-Previato, L., Previato, J. O., Varki, A., and van Halbeek, H. (2000). Trans-sialidase from Trypanosoma cruzi catalyzes sialoside hydrolysis with retention of configuration. Glycobiology 10, 213–221. doi: 10. 1093/glycob/10. 2. 213

||

Todeschini, A. R., Nunes, M. P., Pires, R. S., Lopes, M. F., Previato, J. O., Mendonca-Previato, L., et al. (2002b). Costimulation of host T lymphocytes by a trypanosomal trans-sialidase: involvement of CD43 signaling. J. Immunol. 168, 5192–5198. doi: 10. 4049/jimmunol. 168. 10. 5192

||

Traving, C., and Schauer, R. (1998). Structure, function and metabolism of sialic acids. Cell. Mol. Life. Sci. 54, 1330–1349. doi: 10. 1007/s000180050258

|

Tribulatti, M. V., Mucci, J., Van Rooijen, N., Leguizamon, M. S., and Campetella, O. (2005). The trans-sialidase from Trypanosoma cruzi induces thrombocytopenia during acute Chagas’ disease by reducing the platelet sialic acid contents. Infect. Immun. 73, 201–207. doi: 10. 1128/IAI. 73. 1. 201-207. 2005

||

Tzelepis, F., de Alencar, B. C., Penido, M. L., Gazzinelli, R. T., Persechini, P. M., and Rodrigues, M. M. (2006). Distinct kinetics of effector CD8+ cytotoxic T cells after infection with Trypanosoma cruzi in naive or vaccinated mice. Infect. Immun. 74, 2477–2481. doi: 10. 1128/IAI. 74. 4. 2477-2481. 2006

||

Uemura, H., Schenkman, S., Nussenzweig, V., and Eichinger, D. (1992). Only some members of a gene family in Trypanosoma cruzi encode proteins that express both trans-sialidase and neuraminidase activities. EMBO J. 11, 3837–3844.

|

Vandekerckhove, F., Schenkman, S., Pontes de Carvalho, L., Tomlinson, S., Kiso, M., Yoshida, M., et al. (1992). Substrate specificity of the Trypanosoma cruzi trans-sialidase. Glycobiology 2, 541–548. doi: 10. 1093/glycob/2. 6. 541

||

Varki, A. (1993). Biological roles of oligosaccharides: all of the theories are correct. Glycobiology 3, 97–130. doi: 10. 1093/glycob/3. 2. 97

|

Varki, A. (2011). Evolutionary forces shaping the Golgi glycosylation machinery: why cell surface glycans are universal to living cells. Cold Spring Harb. Perspect. Biol. 3, 1–14. doi: 10. 1101/cshperspect. a005462

||

Varki, A., and Angata, T. (2006). Siglecs–the major subfamily of I-type lectins. Glycobiology 16, 1R–27R. doi: 10. 1093/glycob/cwj008

||

Varki, A., and Gagneux, P. (2012). Multifarious roles of sialic acids in immunity. Ann. N. Y. Acad. Sci. 1253, 16–36. doi: 10. 1111/j. 1749-6632. 2012. 06517. x

||

Varki, A., and Schauer, R. (2009). “ Sialic acids,” in Essentials of Glycobiology , 2nd Edn, eds A. Varki, R. D. Cummings, J. D. Esko, H. H. Freeze, P. Stanley, C. R. Bertozzi, et al. (New York, NY: Cold Spring Harbor).

von Gunten, S., and Bochner, B. S. (2008). Basic and clinical immunology of Siglecs. Ann. N. Y. Acad. Sci. 1143, 61–82. doi: 10. 1196/annals. 1443. 011

||

Wu, W., Harley, P. H., Punt, J. A., Sharrow, S. O., and Kearse, K. P. (1996). Identification of CD8 as a peanut agglutinin (PNA) receptor molecule on immature thymocytes. J. Exp. Med. 184, 759–764. doi: 10. 1084/jem. 184. 2. 759

||

Zingales, B., Carniol, C., de Lederkremer, R. M., and Colli, W. (1987). Direct sialic acid transfer from a protein donor to glycolipids of trypomastigote forms of Trypanosoma cruzi . Mol. Biochem. Parasitol. 26, 135–144. doi: 10. 1016/0166-6851(87)90137-X

||

Thank's for Your Vote!
Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host. Page 1
Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host. Page 2
Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host. Page 3
Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host. Page 4
Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host. Page 5
Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host. Page 6
Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host. Page 7
Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host. Page 8
Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host. Page 9

This work, titled "Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host" was written and willingly shared by a fellow student. This sample can be utilized as a research and reference resource to aid in the writing of your own work. Any use of the work that does not include an appropriate citation is banned.

If you are the owner of this work and don’t want it to be published on AssignBuster, request its removal.

Request Removal
Cite this Essay

References

AssignBuster. (2022) 'Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host'. 19 January.

Reference

AssignBuster. (2022, January 19). Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host. Retrieved from https://assignbuster.com/modulation-of-cell-sialoglycophenotype-a-stylish-mechanism-adopted-by-trypanosoma-cruzi-to-ensure-its-persistence-in-the-infected-host/

References

AssignBuster. 2022. "Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host." January 19, 2022. https://assignbuster.com/modulation-of-cell-sialoglycophenotype-a-stylish-mechanism-adopted-by-trypanosoma-cruzi-to-ensure-its-persistence-in-the-infected-host/.

1. AssignBuster. "Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host." January 19, 2022. https://assignbuster.com/modulation-of-cell-sialoglycophenotype-a-stylish-mechanism-adopted-by-trypanosoma-cruzi-to-ensure-its-persistence-in-the-infected-host/.


Bibliography


AssignBuster. "Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host." January 19, 2022. https://assignbuster.com/modulation-of-cell-sialoglycophenotype-a-stylish-mechanism-adopted-by-trypanosoma-cruzi-to-ensure-its-persistence-in-the-infected-host/.

Work Cited

"Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host." AssignBuster, 19 Jan. 2022, assignbuster.com/modulation-of-cell-sialoglycophenotype-a-stylish-mechanism-adopted-by-trypanosoma-cruzi-to-ensure-its-persistence-in-the-infected-host/.

Get in Touch

Please, let us know if you have any ideas on improving Modulation of cell sialoglycophenotype: a stylish mechanism adopted by trypanosoma cruzi to ensure its persistence in the infected host, or our service. We will be happy to hear what you think: [email protected]