#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

MIF Contributes to Associated Immunopathogenicity Development


Uncontrolled inflammation is a major contributor to pathogenicity development during many chronic parasitic infections, including African trypanosome infections. Hence, therapies should aim at re-establishing the balance between pro- and anti-inflammatory responses to reduce tissue damage. Our experiments uncovered that macrophage migration inhibitory factor (MIF) plays a pivotal role in trypanosomiasis-associated pathogenicity development. Hereby, MIF-deficient and neutralizing anti-MIF antibody-treated wild type (WT) T. brucei-infected mice exhibited decreased inflammatory responses, reduced liver damage and anemia (i.e. the most prominent pathogenicity features) compared to WT control mice. The reduced tissue damage coincided with reduced infiltration of pathogenic monocytic cells and neutrophils, whereby neutrophil-derived MIF contributed more significantly than monocyte-derived MIF to tissue damage. MIF also promoted anemia development by suppressing red blood cell production and enhancing their clearance. The clinical significance of these findings follows from human genetic data indicating that low-expression (protective) MIF alleles are enriched in Africans. The current findings therefore offer promise for human translation and open the possibility of assessing MIF levels or MIF genotype as an indication of an individual's risk for severe trypanosomiasis. Furthermore, given the unmet medical need of African trypanosomiasis affecting millions of people, these findings highlight MIF as a potential new therapeutic target for treatment of trypanosomiasis-associated pathogenicity.


Vyšlo v časopise: MIF Contributes to Associated Immunopathogenicity Development. PLoS Pathog 10(9): e32767. doi:10.1371/journal.ppat.1004414
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004414

Souhrn

Uncontrolled inflammation is a major contributor to pathogenicity development during many chronic parasitic infections, including African trypanosome infections. Hence, therapies should aim at re-establishing the balance between pro- and anti-inflammatory responses to reduce tissue damage. Our experiments uncovered that macrophage migration inhibitory factor (MIF) plays a pivotal role in trypanosomiasis-associated pathogenicity development. Hereby, MIF-deficient and neutralizing anti-MIF antibody-treated wild type (WT) T. brucei-infected mice exhibited decreased inflammatory responses, reduced liver damage and anemia (i.e. the most prominent pathogenicity features) compared to WT control mice. The reduced tissue damage coincided with reduced infiltration of pathogenic monocytic cells and neutrophils, whereby neutrophil-derived MIF contributed more significantly than monocyte-derived MIF to tissue damage. MIF also promoted anemia development by suppressing red blood cell production and enhancing their clearance. The clinical significance of these findings follows from human genetic data indicating that low-expression (protective) MIF alleles are enriched in Africans. The current findings therefore offer promise for human translation and open the possibility of assessing MIF levels or MIF genotype as an indication of an individual's risk for severe trypanosomiasis. Furthermore, given the unmet medical need of African trypanosomiasis affecting millions of people, these findings highlight MIF as a potential new therapeutic target for treatment of trypanosomiasis-associated pathogenicity.


Zdroje

1. BarrettMP, BurchmoreRJ, StichA, LazzariJO, FraschAC, et al. (2003) The trypanosomiases. Lancet 362: 1469–1480.

2. d'IeterenGD, AuthieE, WissocqN, MurrayM (1998) Trypanotolerance, an option for sustainable livestock production in areas at risk from trypanosomosis. Rev Sci Tech 17: 154–175.

3. StijlemansB, VankrunkelsvenA, CaljonG, BockstalV, GuilliamsM, et al. (2010) The central role of macrophages in trypanosomiasis-associated anemia: rationale for therapeutical approaches. Endocr Metab Immune Disord Drug Targets 10: 71–82.

4. NaessensJ (2006) Bovine trypanotolerance: A natural ability to prevent severe anaemia and haemophagocytic syndrome? Int J Parasitol 36: 521–528.

5. StijlemansB, VankrunkelsvenA, BrysL, MagezS, De BaetselierP (2008) Role of iron homeostasis in trypanosomiasis-associated anemia. Immunobiology 213: 823–835.

6. WeissG, GoodnoughLT (2005) Anemia of chronic disease. N Engl J Med 352: 1011–1023.

7. BosschaertsT, GuilliamsM, StijlemansB, De BaetselierP, BeschinA (2009) Understanding the role of monocytic cells in liver inflammation using parasite infection as a model. Immunobiology 214: 737–747.

8. MagezS, StijlemansB, BaralT, De BaetselierP (2002) VSG-GPI anchors of African trypanosomes: their role in macrophage activation and induction of infection-associated immunopathology. Microbes Infect 4: 999–1006.

9. StijlemansB, BaralTN, GuilliamsM, BrysL, KorfJ, et al. (2007) A glycosylphosphatidylinositol-based treatment alleviates trypanosomiasis-associated immunopathology. J Immunol 179: 4003–4014.

10. CalandraT, RogerT (2003) Macrophage migration inhibitory factor: a regulator of innate immunity. Nat Rev Immunol 3: 791–800.

11. LarsonDF, HorakK (2006) Macrophage migration inhibitory factor: controller of systemic inflammation. Crit Care 10: 138.

12. AyoubS, HickeyMJ, MorandEF (2008) Mechanisms of disease: macrophage migration inhibitory factor in SLE, RA and atherosclerosis. Nat Clin Pract Rheumatol 4: 98–105.

13. Rosado JdeD, Rodriguez-SosaM (2011) Macrophage migration inhibitory factor (MIF): a key player in protozoan infections. Int J Biol Sci 7: 1239–1256.

14. BernhagenJ, KrohnR, LueH, GregoryJL, ZerneckeA, et al. (2007) MIF is a noncognate ligand of CXC chemokine receptors in inflammatory and atherogenic cell recruitment. Nat Med 13: 587–596.

15. CalandraT, BernhagenJ, MetzCN, SpiegelLA, BacherM, et al. (1995) MIF as a glucocorticoid-induced modulator of cytokine production. Nature 377: 68–71.

16. MitchellRA, LiaoH, ChesneyJ, Fingerle-RowsonG, BaughJ, et al. (2002) Macrophage migration inhibitory factor (MIF) sustains macrophage proinflammatory function by inhibiting p53: regulatory role in the innate immune response. Proc Natl Acad Sci U S A 99: 345–350.

17. MagezS, TruyensC, MerimiM, RadwanskaM, StijlemansB, et al. (2004) P75 tumor necrosis factor-receptor shedding occurs as a protective host response during African trypanosomiasis. J Infect Dis 189: 527–539.

18. GuilliamsM, MovahediK, BosschaertsT, VandenDriesscheT, ChuahMK, et al. (2009) IL-10 dampens TNF/inducible nitric oxide synthase-producing dendritic cell-mediated pathogenicity during parasitic infection. J Immunol 182: 1107–1118.

19. DaryadelA, GrifoneRF, SimonHU, YousefiS (2006) Apoptotic neutrophils release macrophage migration inhibitory factor upon stimulation with tumor necrosis factor-alpha. J Biol Chem 281: 27653–27661.

20. SocolovskyM, NamH, FlemingMD, HaaseVH, BrugnaraC, et al. (2001) Ineffective erythropoiesis in Stat5a(−/−)5b(−/−) mice due to decreased survival of early erythroblasts. Blood 98: 3261–3273.

21. LiuJ, ZhangJ, GinzburgY, LiH, XueF, et al. (2013) Quantitative analysis of murine terminal erythroid differentiation in vivo: novel method to study normal and disordered erythropoiesis. Blood 121: e43–49.

22. NishimuraK, NakayaH, NakagawaH, MatsuoS, OhnishiY, et al. (2011) Differential effects of Trypanosoma brucei gambiense and Trypanosoma brucei brucei on rat macrophages. J Parasitol 97: 48–54.

23. CalandraT, BernhagenJ, MitchellRA, BucalaR (1994) The macrophage is an important and previously unrecognized source of macrophage migration inhibitory factor. J Exp Med 179: 1895–1902.

24. BosschaertsT, GuilliamsM, StijlemansB, MoriasY, EngelD, et al. (2010) Tip-DC development during parasitic infection is regulated by IL-10 and requires CCL2/CCR2, IFN-gamma and MyD88 signaling. PLoS Pathog 6: e1001045.

25. GregoryJL, MorandEF, McKeownSJ, RalphJA, HallP, et al. (2006) Macrophage migration inhibitory factor induces macrophage recruitment via CC chemokine ligand 2. J Immunol 177: 8072–8079.

26. LinM, CarlsonE, DiaconuE, PearlmanE (2007) CXCL1/KC and CXCL5/LIX are selectively produced by corneal fibroblasts and mediate neutrophil infiltration to the corneal stroma in LPS keratitis. J Leukoc Biol 81: 786–792.

27. OkaM, NagasawaH, ItoY, HimenoK (1989) Granulocyte-macrophage colony-stimulating activity in the serum of mice stimulated with homogenates of Trypanosoma gambiense. Clin Exp Immunol 78: 285–291.

28. KlebanoffSJ (2005) Myeloperoxidase: friend and foe. J Leukoc Biol 77: 598–625.

29. MarquesPE, AmaralSS, PiresDA, NogueiraLL, SorianiFM, et al. (2012) Chemokines and mitochondrial products activate neutrophils to amplify organ injury during mouse acute liver failure. Hepatology 56: 1971–1982.

30. NaessensJ, KitaniH, NakamuraY, YagiY, SekikawaK, et al. (2005) TNF-alpha mediates the development of anaemia in a murine Trypanosoma brucei rhodesiense infection, but not the anaemia associated with a murine Trypanosoma congolense infection. Clin Exp Immunol 139: 405–410.

31. StijlemansB, VankrunkelsvenA, BrysL, RaesG, MagezS, et al. (2010) Scrutinizing the mechanisms underlying the induction of anemia of inflammation through GPI-mediated modulation of macrophage activation in a model of African trypanosomiasis. Microbes Infect 12: 389–399.

32. NishimuraK, NakayaH, NakagawaH, MatsuoS, OhnishiY, et al. (2011) Effect of Trypanosoma brucei brucei on erythropoiesis in infected rats. J Parasitol 97: 88–93.

33. McDevittMA, XieJ, ShanmugasundaramG, GriffithJ, LiuA, et al. (2006) A critical role for the host mediator macrophage migration inhibitory factor in the pathogenesis of malarial anemia. J Exp Med 203: 1185–1196.

34. SoniS, BalaS, GwynnB, SahrKE, PetersLL, et al. (2006) Absence of erythroblast macrophage protein (Emp) leads to failure of erythroblast nuclear extrusion. J Biol Chem 281: 20181–20189.

35. Angelillo-ScherrerA, BurnierL, LambrechtsD, FishRJ, TjwaM, et al. (2008) Role of Gas6 in erythropoiesis and anemia in mice. J Clin Invest 118: 583–596.

36. De MuylderG, DaulouedeS, LecordierL, UzureauP, MoriasY, et al. (2013) A Trypanosoma brucei kinesin heavy chain promotes parasite growth by triggering host arginase activity. PLoS Pathog 9: e1003731.

37. RennerP, RogerT, CalandraT (2005) Macrophage migration inhibitory factor: gene polymorphisms and susceptibility to inflammatory diseases. Clin Infect Dis 41 Suppl 7S513–519.

38. ZhongXB, LengL, BeitinA, ChenR, McDonaldC, et al. (2005) Simultaneous detection of microsatellite repeats and SNPs in the macrophage migration inhibitory factor (MIF) gene by thin-film biosensor chips and application to rural field studies. Nucleic Acids Res 33: e121.

39. Fingerle-RowsonG, PetrenkoO, MetzCN, ForsthuberTG, MitchellR, et al. (2003) The p53-dependent effects of macrophage migration inhibitory factor revealed by gene targeting. Proc Natl Acad Sci U S A 100: 9354–9359.

40. CalandraT, EchtenacherB, RoyDL, PuginJ, MetzCN, et al. (2000) Protection from septic shock by neutralization of macrophage migration inhibitory factor. Nat Med 6: 164–170.

41. RaesG, BrysL, DahalBK, BrandtJ, GrootenJ, et al. (2005) Macrophage galactose-type C-type lectins as novel markers for alternatively activated macrophages elicited by parasitic infections and allergic airway inflammation. J Leukoc Biol 77: 321–327.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 9
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

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

Všetky kurzy
Prihlásenie
Zabudnuté heslo

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

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#