Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1729 word(s) 1729 2021-09-02 08:51:19 |
2 update references and layout Meta information modification 1729 2021-09-03 06:32:02 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Henriques Pons, A. Trypanosoma cruzi Infection; liver involvement. Encyclopedia. Available online: https://encyclopedia.pub/entry/13856 (accessed on 23 April 2024).
Henriques Pons A. Trypanosoma cruzi Infection; liver involvement. Encyclopedia. Available at: https://encyclopedia.pub/entry/13856. Accessed April 23, 2024.
Henriques Pons, Andrea. "Trypanosoma cruzi Infection; liver involvement" Encyclopedia, https://encyclopedia.pub/entry/13856 (accessed April 23, 2024).
Henriques Pons, A. (2021, September 02). Trypanosoma cruzi Infection; liver involvement. In Encyclopedia. https://encyclopedia.pub/entry/13856
Henriques Pons, Andrea. "Trypanosoma cruzi Infection; liver involvement." Encyclopedia. Web. 02 September, 2021.
Trypanosoma cruzi Infection; liver involvement
Edit

Currently, in Chagas disease, hepatomegaly is cited in most papers published which either study acutely infected patients or experimental models, and we know that the Trypanosoma cruzi can infect multiple cell types in the liver, especially Kupffer cells and dendritic cells. Moreover, liver damage is more pronounced in cases of oral infection, which is mainly found in the Amazon region. However, the importance of liver involvement, including the hepatic immune response, in disease progression does not receive much attention.

infection Chagas disease liver hepatic immune response Trypanosoma cruzi cellular immunology

1. Introduction

Hepatocytes compose approximately 60% of the hepatic cells and about 90% of the liver volume [1], and their primary function is formation and excretion of bile; lipid synthesis and plasma lipoprotein secretion; control of cholesterol metabolism; regulation of carbohydrate homeostasis; formation of urea, serum albumin, coagulation factors, enzymes, and other molecules; and metabolism or detoxification of drugs and other exogenous substances [2].

Although previous authors suggested that hepatic stellate cells (HSC) are professional intrahepatic APCs that elicit many T cell responses, in contrast to other liver cells that lead primarily to tolerance, additional results should be considered. For example, it was also demonstrated that HSCs are central in the liver’s regulatory response, as they can inhibit T cell responses via B7-H1-mediated apoptosis [3]. Moreover, it was shown that HSCs alone do not present antigens to naive CD4 + T lymphocytes but, in the presence of dendritic cells and TGF-β, preferentially induce FoxP3 + Treg cells [4].

Besides the main hepatic dendritic cells (HDCs) observed, an extraordinary and minor population of DC is also found in the liver, possibly an intermediate population with cytotoxic properties that can efficiently lead to T lymphocyte activation. These NK1.1 + cytotoxic HDCs, also found in other tissues, are inflammatory cells that can produce high levels of IFN-γ via autocrine IL-12 stimulation, leading to the development and activation of cytotoxic T lymphocytes [5][6][7][8].

Reversing or breaking hepatic immune tolerance in persistent infections or cancer is of central importance. This can be demonstrated by the reversion of liver T cell tolerance and viral persistence in the case of hepaitis B virus (HBV) infection after the blockade of inhibitory receptors, such as PD-1 [9][10]. Additionally, in tumors such as hepatocellular carcinoma, PD-1 blockade has good results in inducing a protective immune response [11][12][13].

2. The Participation of the Liver in T. cruzi Infection, a More Recent Perspective

Today we know that the liver plays an essential role in the infection, and its importance in parasite clearance and destruction of blood (trypomastigote) forms, for example, is well documented. Experimental murine infections showed that specific antibodies against the parasite plus phagocytic cells are required for extracellular trypomastigote clearance. IgG-coated parasites are phagocytosed by resident mononuclear cells, especially in the liver but also in the lungs and spleen [14]. Moreover, pro-inflammatory cytokines, mainly IFN-γ, potentiate the trypanocidal activity of the phagocytic cells [15], and intact Fc portions of IgGs are required [16]. Sardinha and cols. published in 2010 that the liver is the primary site of parasite accumulation just one hour after intravenous injection of T. cruzi trypomastigote forms in chronically infected mice [17]. At this time point, viable parasites and parasite remnants were observed scattered in the liver parenchyma, which considerably diminished after 48 hours, and no intracellular parasites were observed in the liver seven days after the challenge [17]. Moreover, transmission electron microscopy showed platelet thrombi occluding small vessels in the lung, liver, and spleen, and phagocytosed parasites in different stages of destruction were found within macrophages, neutrophils, and eosinophils. Therefore, it seems that not a particular cell population, but different cells, act in concert to destroy the parasites in the liver [14].
The liver is the main synthesis site for the complement system’s components, and it has long been evaluated if this lytic pathway could play a role in removing blood parasites. Although trypomastigote clearance is dependent on C3 [18], it is primarily independent of the lytic terminal pathway. A more detailed analysis of the complement system’s importance in parasite clearance showed that C1q, C3, mannan-binding lectin, and ficolin molecules bind to trypomastigote forms. Moreover, C3b and C4b deposition assays revealed that T. cruzi activates mainly the lectin and alternative complement pathways in non-immune human serum [19]. Experiments using C5-deficient mice showed no difference in parasite clearance compared with wild-type mice [16].
It is long known that blood trypomastigote forms express several complement system inhibitors, such as a decay-accelerating factor expressed by T. cruzi (T-DAF) [20], complement C2 receptor inhibitor trispanning [21], complement regulatory protein, and others [22]. Nevertheless, some parasite strains seem to be susceptible to the complement system [19].
It is also known that the infection subverts the host lipid metabolism in multiple ways [23], mainly affecting the low-density lipoprotein- (LDL) and high-density lipoprotein-dependent (HDL) pathways and their receptors. LDL is generated from liver-derived very-low-density lipoprotein (VLDL) and is a potent inhibitor of T. cruzi trans-sialidase, an enzyme expressed mainly by epimastigote and trypomastigote forms of T. cruzi that transfers sialic acid from the environment to the parasite surface [24]. Moreover, the addition of LDL and HDL to cells in culture enhances infection by T. cruzi trypomastigote forms in a dose-dependent manner [25]. The LDL receptor (LDLr) is one of the molecules used by the parasite during cell invasion, and in vitro infection in the presence of an LDLr blocker resulted in a 42% reduction of intracellular infection [26]. The LDLr is also involved in the trafficking of lysosomes to the cytoplasmic parasitophorous vacuole, and the disruption of the LDLr pathway affects the intracellular parasite load.
Today we know some of the lipid metabolic pathways that led to the first observations of hepatic steatosis by Dr. Chagas. For example, it was recently demonstrated that T. cruzi interaction with LDLr leads to the accumulation of LDL cholesterol in host tissue in acute and chronic chagasic patients [27]. Moreover, murine experimental infection revealed a significant increase in the absolute amount of triacylglycerides, cholesterol, and cholesterol esters in liver microsomal membranes [28]. Additionally, T. cruzi experimental infection was considered a potent risk factor for non-alcoholic steatohepatitis, associated with strong oxidative stress and metabolic disorders [29].
Regarding the hepatic immune response, we have just recently started to understand the integration and possible interdependency between hepatic and peripheral immunity after infection. As primarily observed at the beginning of the last century, the T. cruzi infection leads to inflammatory mononuclear cell infiltration in the liver parenchyma. Today we know some of the main cell types that compose the inflammatory foci and their inflammatory mediators. Briefly, as this topic will be discussed in more detail in the next section, the infection induces an increase in Mac1+, activated CD8+ and CD4+ T lymphocytes expressing CD25, CD69, and/or CD122, natural killer (NK), and NKT cells [17] in the liver. Moreover, one of the significant roles of NK cells and CD4+ T lymphocytes in liver protection and infection control is interferon-gamma production (IFN-γ) [30]. In addition, we observed that experimental murine infection leads to increased hepatic regulatory T (Treg) cell numbers, higher expression of programmed death ligand 1 (PD-L1 or B7-H1) in the liver stroma, increased blood activity of ALT and AST transaminases, and other alterations [31].

3. The Immune Response in the Liver

The liver is the second largest organ in the human body. It performs many essential functions, including metabolic regulation, digestion, production of bile, detoxification (conjugations with sulfate, glucuronic acid, glutathione, acetate, and glycine), and biotransformation of drugs and toxins (oxidations-reductions and hydrolysis) [32][33][34]. Approximately 80% of the blood supply that reaches the liver comes through the hepatic portal vein, consisting of blood that is low in oxygen and rich in nutrients and molecules of the intestinal microbiota. This anatomical characteristic determines that the liver typically meets very high levels of bacterial components that, in the periphery, would be recognized as danger signals and potent pro-inflammatory stimuli. The liver must then be able to individually discriminate pathogenic damage-associated molecular patterns (DAMP) [35] and especially pathogen-associated molecular patterns (PAMP) [36] from harmless DAMPs and PAMPs. This means that the liver must retain its tolerogenic bias and selectively recognize proper danger signals for pro-inflammatory response against infections or tumors, for example. These fascinating properties are just beginning to be elucidated.
Many anatomical, immunological, and environmental aspects play a central role in balancing tolerance versus immune responses in the liver. For example, the organ is highly vascularized, and these hepatic microvessels are known as hepatic sinusoids. The epithelial cells that line the sinusoids are fenestrated, allowing the protrusion of membrane segments and physical interaction between cells flowing in the vase lumen with stromal and parenchymal cells. Between the sinusoids wall and hepatocyte cords, there is the Disse space [37], a space adjacent to the sinusoids that harbors many different cell types in the liver. The very low blood pressure in the sinusoids favors this integrated cellular interaction network, affecting the liver’s biochemical and immunological functions [1]. Although the liver is best known for its primary metabolic functions, the organ is of great importance in the local and systemic immune response [5][38], as depicted below.

4. Perspectives

Today we know many of the biochemical pathways and cell populations that sustain hepatic tolerance, and this knowledge should be translated into practical proposals and alternative treatments for autoimmune or chronic inflammatory diseases, for example. A few initiatives have been proposed, such as the induction of hepatic Treg cells by LSEC to control autoimmune encephalomyelitis (EAE) [39]. Previously, the authors showed that LSECs could induce CD4+ Foxp3+ Treg cells in mice [40]. Subsequently, they injected nanoparticles as carriers of autoimmune peptides in vivo, which were selectively delivered to LSECs. This treatment resulted in antigen-specific Treg cell induction, and these cells could completely and permanently prevent the onset of clinical EAE. Moreover, in mice with established clinical EAE, the treatment for Treg cell induction rapidly and substantially improved muscle paralysis and atonia, whereas the control group deteriorated. Similar results were obtained when antigen-specific Treg cells were expanded using neural autoantigen myelin basic protein (MBP) in the liver [41]. In the case of myelin infusion, antigen-specific Foxp3+ Treg cells exerted their effect by diminishing antigen-bearing inflammatory dendritic cells recruitment to lymph nodes and by impairing their function [42]. The induction of Treg cells in the liver or the periphery is reviewed in [43][44].
Other approaches such as those using antigen-specific immunotherapy [45] and neutralization of immunomodulatory molecules in the liver (reviewed in [46]) are being studied or are already in use. Much remains, however, to be understood about the fascinating pathways and immunological cross-talks hidden in the liver immunophysiology, including after Trypanosoma cruzi infection.

 

References

  1. Grakoui, A.; Crispe, I.N. Presentation of hepatocellular antigens. Cell. Mol. Immunol. 2016, 13, 293–300.
  2. Warren, A.; Le Couteur, D.G.; Fraser, R.; Bowen, D.G.; McCaughan, G.W.; Bertolino, P. Tlymphocytes interact with hepatocytes through fenestrations in murine liver sinusoidal endothelial cells. Hepatology 2006, 44, 1182–1190.
  3. Heymann, F.; Trautwein, C.; Tacke, F. Monocytes and macrophages as cellular targets in liver fibrosis. Inflamm. Allergy Drug Targets 2009, 8, 307–318.
  4. Dunham, R.M.; Thapa, M.; Velazquez, V.M.; Elrod, E.J.; Denning, T.L.; Pulendran, B.; Grakoui, A. Hepatic stellate cells preferentially induce Foxp3+ regulatory T cells by production of retinoic acid. J. Immunol. 2013, 190, 2009–2016.
  5. Crispe, I.N. Liver antigen-presenting cells. J. Hepatol. 2011, 54, 357–365.
  6. Pillarisetty, V.G.; Katz, S.C.; Bleier, J.I.; Shah, A.B.; Dematteo, R.P. Natural killer dendritic cells have both antigen presenting and lytic function and in response to CpG produce IFN-gamma via autocrine IL-12. J. Immunol. 2005, 174, 2612–2618.
  7. Chen, L.; Calomeni, E.; Wen, J.; Ozato, K.; Shen, R.; Gao, J.X. Natural killer dendritic cells are an intermediate of developing dendritic cells. J. Leukoc. Biol. 2007, 81, 1422–1433.
  8. Krueger, P.D.; Kim, T.S.; Sung, S.S.; Braciale, T.J.; Hahn, Y.S. Liver-resident CD103+ dendritic cells prime antiviral CD8+ T cells in situ. J. Immunol. 2015, 194, 3213–3222.
  9. Fisicaro, P.; Valdatta, C.; Massari, M.; Loggi, E.; Biasini, E.; Sacchelli, L.; Cavallo, M.C.; Silini, E.M.; Andreone, P.; Missale, G.; et al. Antiviral intrahepatic T-cell responses can be restored by blocking programmed death-1 pathway in chronic hepatitis B. Gastroenterology 2010, 138, 682–693.
  10. Tzeng, H.T.; Tsai, H.F.; Liao, H.J.; Lin, Y.J.; Chen, L.; Chen, P.J.; Hsu, P.N. PD-1 blockage reverses immune dysfunction and hepatitis B viral persistence in a mouse animal model. PLoS ONE 2012, 7, e39179.
  11. Hou, J.; Zhang, H.; Sun, B.; Karin, M. The immunobiology of hepatocellular carcinoma in humans and mice: Basic concepts and therapeutic implications. J. Hepatol. 2020, 72, 167–182.
  12. Llovet, J.M.; Montal, R.; Sia, D.; Finn, R.S. Molecular therapies and precision medicine for hepatocellular carcinoma. Nat. Rev. Clin. Oncol. 2018, 15, 599–616.
  13. El-Khoueiry, A. The Promise of Immunotherapy in the Treatment of Hepatocellular Carcinoma. Am. Soc. Clin. Oncol. Educ. Book 2017, 37, 311–317.
  14. Umekita, L.F.; Carneiro, S.M.; Sesso, A.; Mota, I. One fate of bloodstream trypomastigote forms of Trypanosoma cruzi after immune clearance: An ultrastructural study. J. Parasitol. 1999, 85, 867–872.
  15. Plata, F.; Wietzerbin, J.; Pons, F.G.; Falcoff, E.; Eisen, H. Synergistic protection by specific antibodies and interferon against infection by Trypanosoma cruzi in vitro. Eur. J. Immunol. 1984, 14, 930–935.
  16. Umekita, L.F.; Takehara, H.A.; Mota, I. Role of the antibody Fc in the immune clearance of Trypanosoma cruzi. Immunol. Lett. 1988, 17, 85–89.
  17. Sardinha, L.R.; Mosca, T.; Elias, R.M.; do Nascimento, R.S.; Gonçalves, L.A.; Bucci, D.Z.; Marinho, C.R.; Penha-Gonçalves, C.; Lima, M.R.; Alvarez, J.M. The liver plays a major role in clearance and destruction of blood trypomastigotes in Trypanosoma cruzi chronically infected mice. PLoS Negl. Trop. Dis. 2010, 4, e578.
  18. Mota, I.; Umekita, L.F. The effect of C3 depletion on the clearance of Trypanosoma cruzi induced by IgG antibodies. Immunol. Lett. 1989, 21, 223–225.
  19. Cestari, I.; Ramirez, M.I. Inefficient complement system clearance of Trypanosoma cruzi metacyclic trypomastigotes enables resistant strains to invade eukaryotic cells. PLoS ONE 2010, 5, e9721.
  20. Tambourgi, D.V.; Kipnis, T.L.; da Silva, W.D.; Joiner, K.A.; Sher, A.; Heath, S.; Hall, B.F.; Ogden, G.B. A partial cDNA clone of trypomastigote decay-accelerating factor (T-DAF), a developmentally regulated complement inhibitor of Trypanosoma cruzi, has genetic and functional similarities to the human complement inhibitor DAF. Infect. Immun. 1993, 61, 3656–3663.
  21. Cestari, I.O.S.; Evans-Osses, I.; Freitas, J.C.; Inal, J.M.; Ramirez, M.I. Complement C2 receptor inhibitor trispanning confers an increased ability to resist complement-mediated lysis in Trypanosoma cruzi. J. Infect. Dis. 2008, 198, 1276–1283.
  22. Atayde, V.D.; Neira, I.; Cortez, M.; Ferreira, D.; Freymüller, E.; Yoshida, N. Molecular basis of non-virulence of Trypanosoma cruzi clone CL-14. Int. J. Parasitol. 2004, 34, 851–860.
  23. Miao, Q.; Ndao, M. Trypanosoma cruzi infection and host lipid metabolism. Mediat. Inflamm. 2014, 2014, 902038.
  24. da Fonseca, L.M.; da Costa, K.M.; Chaves, V.S.; Freire-de-Lima, C.G.; Morrot, A.; Mendonça-Previato, L.; Previato, J.O.; Freire-de-Lima, L. Theft and reception of host cell’s sialic acid: Dynamics of Trypanosoma cruzi trans-sialidases and mucin-like molecules on Chagas’ disease immunomodulation. Front. Immunol. 2019, 10, 164.
  25. Prioli, R.P.; Rosenberg, I.; Pereira, M.E. High- and low-density lipoproteins enhance infection of Trypanosoma cruzi in vitro. Mol. Biochem. Parasitol. 1990, 38, 191–198.
  26. Nagajyothi, F.; Weiss, L.M.; Silver, D.L.; Desruisseaux, M.S.; Scherer, P.E.; Herz, J.; Tanowitz, H.B. Trypanosoma cruzi utilizes the host low density lipoprotein receptor in invasion. PLoS Negl. Trop. Dis. 2011, 5, e953.
  27. Johndrow, C.; Nelson, R.; Tanowitz, H.; Weiss, L.M.; Nagajyothi, F. Trypanosoma cruzi infection results in an increase in intracellular cholesterol. Microbes Infect. 2014, 16, 337–344.
  28. Marra, C.A.; Zaidenberg, A.; de Alaniz, M.J.; Buschiazzo, H. The restoring effect of trifluralin and benznidazole on the abnormal fatty-acid pattern induced by Trypanosoma cruzi in the liver microsomes of infected mice. Ann. Trop. Med. Parasitol. 2002, 96, 249–264.
  29. Onofrio, L.I.; Arocena, A.R.; Paroli, A.F.; Cabalén, M.E.; Andrada, M.C.; Cano, R.C.; Gea, S. Trypanosoma cruzi infection is a potent risk factor for non-alcoholic steatohepatitis enhancing local and systemic inflammation associated with strong oxidative stress and metabolic disorders. PLoS Negl. Trop. Dis. 2015, 9, e0003464.
  30. Sardinha, L.R.; Elias, R.M.; Mosca, T.; Bastos, K.R.; Marinho, C.R.; D’Império Lima, M.R.; Alvarez, J.M. Contribution of NK, NK T, gamma delta T, and alpha beta T cells to the gamma interferon response required for liver protection against Trypanosoma cruzi. Infect. Immun. 2006, 74, 2031–2042.
  31. Meuser-Batista, M.; Vacani-Martins, N.; Cascabulho, C.M.; Beghini, D.G.; Henriques-Pons, A. In the presence of Trypanosoma cruzi antigens, activated peripheral T lymphocytes retained in the liver induce a proinflammatory phenotypic and functional shift in intrahepatic T lymphocyte. J. Leukoc. Biol. 2020, 107, 695–706.
  32. Ravin, H.A.; Rowley, D.; Jenkins, C.; Fine, J. On the absorption of bacterial endotoxin from the gastro-intestinal tract of the normal and shocked animal. J. Exp. Med. 1960, 112, 783–792.
  33. Lumsden, A.B.; Henderson, J.M.; Kutner, M.H. Endotoxin levels measured by a chromogenic assay in portal, hepatic and peripheral venous blood in patients with cirrhosis. Hepatology 1988, 8, 232–236.
  34. Crispe, I.N. The liver as a lymphoid organ. Annu. Rev. Immunol. 2009, 27, 147–163.
  35. Seong, S.Y.; Matzinger, P. Hydrophobicity: An ancient damage-associated molecular pattern that initiates innate immune responses. Nat. Rev. Immunol. 2004, 4, 469–478.
  36. Murphy, K.; Travers, P.; Walport, M. Imunobiologia de Janeway, 7th ed.; Artmed: Porto Alegre, Brazil, 2011.
  37. Wisse, E. An electron microscopic study of the fenestrated endothelial lining of rat liver sinusoids. J. Ultrastruct. Res. 1970, 31, 125–150.
  38. Freudenberg, M.A.; Freudenberg, N.; Galanos, C. Time course of cellular distribution of endotoxin in liver, lungs and kidneys of rats. Br. J. Exp. Pathol. 1982, 63, 56–65.
  39. Carambia, A.; Freund, B.; Schwinge, D.; Bruns, O.T.; Salmen, S.C.; Ittrich, H.; Reimer, R.; Heine, M.; Huber, S.; Waurisch, C.; et al. Nanoparticle-based autoantigen delivery to Treg-inducing liver sinusoidal endothelial cells enables control of autoimmunity in mice. J. Hepatol. 2015, 62, 1349–1356.
  40. Carambia, A.; Freund, B.; Schwinge, D.; Heine, M.; Laschtowitz, A.; Huber, S.; Wraith, D.C.; Korn, T.; Schramm, C.; Lohse, A.W.; et al. TGF-β-dependent induction of CD4+CD25+Foxp3+ Tregs by liver sinusoidal endothelial cells. J. Hepatol. 2014, 61, 594–599.
  41. Lüth, S.; Huber, S.; Schramm, C.; Buch, T.; Zander, S.; Stadelmann, C.; Brück, W.; Wraith, D.C.; Herkel, J.; Lohse, A.W. Ectopic expression of neural autoantigen in mouse liver suppresses experimental autoimmune neuroinflammation by inducing antigen-specific Tregs. J. Clin. Investig. 2008, 118, 3403–3410.
  42. Alissafi, T.; Hatzioannou, A.; Ioannou, M.; Sparwasser, T.; Grün, J.R.; Grützkau, A.; Verginis, P. De novo-induced self-antigen-specific Foxp3+ regulatory T cells impair the accumulation of inflammatory dendritic cells in draining lymph nodes. J. Immunol. 2015, 194, 5812–5824.
  43. Moorman, C.D.; Sohn, S.J.; Phee, H. Emerging Therapeutics for Immune Tolerance: Tolerogenic Vaccines, T cell Therapy, and IL-2 Therapy. Front. Immunol. 2021, 12, 657768.
  44. Zhang, D.; Tu, E.; Kasagi, S.; Zanvit, P.; Chen, Q.; Chen, W. Manipulating regulatory T cells: A promising strategy to treat autoimmunity. Immunotherapy 2015, 7, 1201–1211.
  45. Richardson, N.; Ng, S.T.H.; Wraith, D.C. Antigen-Specific Immunotherapy for Treatment of Autoimmune Liver Diseases. Front. Immunol. 2020, 11, 1586.
  46. Esfahani, K.; Roudaia, L.; Buhlaiga, N.; Del Rincon, S.V.; Papneja, N.; Miller, W.H. A review of cancer immunotherapy: From the past, to the present, to the future. Curr. Oncol. 2020, 27, S87–S97.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 396
Revisions: 2 times (View History)
Update Date: 03 Sep 2021
1000/1000