#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Exposure-Dependent Control of Malaria-Induced Inflammation in Children


Malaria remains a major cause of disease and death worldwide. When mosquitoes infect people with malaria parasites for the first time, the parasite rapidly multiplies in the blood and the body responds by producing molecules that cause inflammation and fever, and sometimes the infection progresses to life-threatening disease. However, in regions where people are repeatedly infected with malaria parasites, most infections do not cause fever and parasites often do not multiply uncontrollably. For example, in Mali where this study was conducted, children are infected with malaria parasites ≥100 times/year but only get malaria fever ∼2 times/year and often manage to control parasite numbers in the blood. To understand these observations we collected immune cells from the blood of healthy children before the malaria season and 7 days after malaria fever. We simulated malaria infection at these time points by exposing the immune cells to malaria parasites in a test-tube. We found that re-exposing immune cells to parasites after malaria fever results in reduced expression of molecules that cause fever and enhanced expression of molecules involved in parasite killing. These findings help explain how the immune system prevents fever and controls malaria parasite growth in children who are repeatedly infected with malaria parasites.


Vyšlo v časopise: Exposure-Dependent Control of Malaria-Induced Inflammation in Children. PLoS Pathog 10(4): e32767. doi:10.1371/journal.ppat.1004079
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004079

Souhrn

Malaria remains a major cause of disease and death worldwide. When mosquitoes infect people with malaria parasites for the first time, the parasite rapidly multiplies in the blood and the body responds by producing molecules that cause inflammation and fever, and sometimes the infection progresses to life-threatening disease. However, in regions where people are repeatedly infected with malaria parasites, most infections do not cause fever and parasites often do not multiply uncontrollably. For example, in Mali where this study was conducted, children are infected with malaria parasites ≥100 times/year but only get malaria fever ∼2 times/year and often manage to control parasite numbers in the blood. To understand these observations we collected immune cells from the blood of healthy children before the malaria season and 7 days after malaria fever. We simulated malaria infection at these time points by exposing the immune cells to malaria parasites in a test-tube. We found that re-exposing immune cells to parasites after malaria fever results in reduced expression of molecules that cause fever and enhanced expression of molecules involved in parasite killing. These findings help explain how the immune system prevents fever and controls malaria parasite growth in children who are repeatedly infected with malaria parasites.


Zdroje

1. CollinsWE, JefferyGM (1999) A retrospective examination of sporozoite- and trophozoite-induced infections with Plasmodium falciparum: development of parasitologic and clinical immunity during primary infection. Am J Trop Med Hyg 61: 4–19.

2. DayNP, HienTT, SchollaardtT, LocPP, ChuongLV, et al. (1999) The prognostic and pathophysiologic role of pro- and antiinflammatory cytokines in severe malaria. J Infect Dis 180: 1288–1297.

3. LykeKE, BurgesR, CissokoY, SangareL, DaoM, et al. (2004) Serum levels of the proinflammatory cytokines interleukin-1 beta (IL-1beta), IL-6, IL-8, IL-10, tumor necrosis factor alpha, and IL-12(p70) in Malian children with severe Plasmodium falciparum malaria and matched uncomplicated malaria or healthy controls. Infect Immun 72: 5630–5637.

4. WaltherM, WoodruffJ, EdeleF, JeffriesD, TongrenJE, et al. (2006) Innate immune responses to human malaria: heterogeneous cytokine responses to blood-stage Plasmodium falciparum correlate with parasitological and clinical outcomes. J Immunol 177: 5736–5745.

5. UrbanBC, IngR, StevensonMM (2005) Early interactions between blood-stage plasmodium parasites and the immune system. Curr Top Microbiol Immunol 297: 25–70.

6. LanghorneJ, NdunguFM, SponaasAM, MarshK (2008) Immunity to malaria: more questions than answers. Nat Immunol 9: 725–732.

7. KwiatkowskiD, HillAV, SambouI, TwumasiP, CastracaneJ, et al. (1990) TNF concentration in fatal cerebral, non-fatal cerebral, and uncomplicated Plasmodium falciparum malaria. Lancet 336: 1201–1204.

8. GrauGE, TaylorTE, MolyneuxME, WirimaJJ, VassalliP, et al. (1989) Tumor necrosis factor and disease severity in children with falciparum malaria. N Engl J Med 320: 1586–1591.

9. Molineaux L (1985) The impact of parasitic diseases and their control on mortality, with emphasis on malaria and Africa. In: Vallin J, Lopez AD, editors. Health Policy, Social Policy and Mortality Prospects. Liege: Ordina. pp. 13–44.

10. BaliraineFN, AfraneYA, AmenyaDA, BonizzoniM, MengeDM, et al. (2009) High prevalence of asymptomatic plasmodium falciparum infections in a highland area of western Kenya: a cohort study. J Infect Dis 200: 66–74.

11. BottiusE, GuanzirolliA, TrapeJF, RogierC, KonateL, et al. (1996) Malaria: even more chronic in nature than previously thought; evidence for subpatent parasitaemia detectable by the polymerase chain reaction. Trans R Soc Trop Med Hyg 90: 15–19.

12. McGregorIA, GillesHM, WaltersJH, DaviesAH, PearsonFA (1956) Effects of heavy and repeated malarial infections on Gambian infants and children; effects of erythrocytic parasitization. Br Med J 2: 686–692.

13. PortugalS, PierceSK, CromptonPD (2013) Young lives lost as B cells falter: what we are learning about antibody responses in malaria. J Immunol 190: 3039–3046.

14. RileyEM, WahlS, PerkinsDJ, SchofieldL (2006) Regulating immunity to malaria. Parasite Immunol 28: 35–49.

15. SintonJA (1938) Immunity or Tolerance in Malarial Infections: (Section of Comparative Medicine). Proc R Soc Med 31: 1298–1302.

16. BoutlisCS, YeoTW, AnsteyNM (2006) Malaria tolerance–for whom the cell tolls? Trends Parasitol 22: 371–377.

17. SchofieldL, HewittMC, EvansK, SiomosMA, SeebergerPH (2002) Synthetic GPI as a candidate anti-toxic vaccine in a model of malaria. Nature 418: 785–789.

18. BoutlisCS, RileyEM, AnsteyNM, de SouzaJB (2005) Glycosylphosphatidylinositols in malaria pathogenesis and immunity: potential for therapeutic inhibition and vaccination. Curr Top Microbiol Immunol 297: 145–185.

19. LiC, CorralizaI, LanghorneJ (1999) A defect in interleukin-10 leads to enhanced malarial disease in Plasmodium chabaudi chabaudi infection in mice. Infect Immun 67: 4435–4442.

20. WenischC, ParschalkB, NarztE, LooareesuwanS, GraningerW (1995) Elevated serum levels of IL-10 and IFN-gamma in patients with acute Plasmodium falciparum malaria. Clin Immunol Immunopathol 74: 115–117.

21. DodooD, OmerFM, ToddJ, AkanmoriBD, KoramKA, et al. (2002) Absolute levels and ratios of proinflammatory and anti-inflammatory cytokine production in vitro predict clinical immunity to Plasmodium falciparum malaria. J Infect Dis 185: 971–979.

22. OmerFM, RileyEM (1998) Transforming growth factor beta production is inversely correlated with severity of murine malaria infection. J Exp Med 188: 39–48.

23. WaltherM, TongrenJE, AndrewsL, KorbelD, KingE, et al. (2005) Upregulation of TGF-beta, FOXP3, and CD4+CD25+ regulatory T cells correlates with more rapid parasite growth in human malaria infection. Immunity 23: 287–296.

24. HeymanA, BeesonPB (1949) Influence of various disease states upon the febrile response to intravenous injection of typhoid bacterial pyrogen; with particular reference to malaria and cirrhosis of the liver. J Lab Clin Med 34: 1400–1403.

25. RubensteinM, MulhollandJH, JefferyGM, WolffSM (1965) Malaria Induced Endotoxin Tolerance. Proc Soc Exp Biol Med 118: 283–287.

26. WestMA, HeagyW (2002) Endotoxin tolerance: a review. Crit Care Med 30: S64–73.

27. FosterSL, HargreavesDC, MedzhitovR (2007) Gene-specific control of inflammation by TLR-induced chromatin modifications. Nature 447: 972–978.

28. del FresnoC, Garcia-RioF, Gomez-PinaV, Soares-SchanoskiA, Fernandez-RuizI, et al. (2009) Potent phagocytic activity with impaired antigen presentation identifying lipopolysaccharide-tolerant human monocytes: demonstration in isolated monocytes from cystic fibrosis patients. J Immunol 182: 6494–6507.

29. SpencePJ, LanghorneJ (2012) T cell control of malaria pathogenesis. Curr Opin Immunol 24: 444–448.

30. TranTM, SamalB, KirknessE, CromptonPD (2012) Systems immunology of human malaria. Trends Parasitol 28: 248–257.

31. CromptonPD, TraoreB, KayentaoK, DoumboS, OngoibaA, et al. (2008) Sickle Cell Trait Is Associated with a Delayed Onset of Malaria: Implications for Time-to-Event Analysis in Clinical Studies of Malaria. J Infect Dis 198: 1265–1275.

32. OchielDO, AwandareGA, KellerCC, HittnerJB, KremsnerPG, et al. (2005) Differential regulation of beta-chemokines in children with Plasmodium falciparum malaria. Infect Immun 73: 4190–4197.

33. KrishnegowdaG, HajjarAM, ZhuJ, DouglassEJ, UematsuS, et al. (2005) Induction of proinflammatory responses in macrophages by the glycosylphosphatidylinositols of Plasmodium falciparum: cell signaling receptors, glycosylphosphatidylinositol (GPI) structural requirement, and regulation of GPI activity. J Biol Chem 280: 8606–8616.

34. ShioMT, EisenbarthSC, SavariaM, VinetAF, BellemareMJ, et al. (2009) Malarial hemozoin activates the NLRP3 inflammasome through Lyn and Syk kinases. PLoS Pathog 5: e1000559.

35. ParrocheP, LauwFN, GoutagnyN, LatzE, MonksBG, et al. (2007) Malaria hemozoin is immunologically inert but radically enhances innate responses by presenting malaria DNA to Toll-like receptor 9. Proc Natl Acad Sci U S A 104: 1919–1924.

36. SharmaS, DeOliveiraRB, KalantariP, ParrocheP, GoutagnyN, et al. (2011) Innate immune recognition of an AT-rich stem-loop DNA motif in the Plasmodium falciparum genome. Immunity 35: 194–207.

37. GriffithJW, SunT, McIntoshMT, BucalaR (2009) Pure Hemozoin is inflammatory in vivo and activates the NALP3 inflammasome via release of uric acid. J Immunol 183: 5208–5220.

38. ChaussabelD, QuinnC, ShenJ, PatelP, GlaserC, et al. (2008) A modular analysis framework for blood genomics studies: application to systemic lupus erythematosus. Immunity 29: 150–164.

39. SharifO, KnappS (2008) From expression to signaling: roles of TREM-1 and TREM-2 in innate immunity and bacterial infection. Immunobiology 213: 701–713.

40. CouperKN, BlountDG, RileyEM (2008) IL-10: the master regulator of immunity to infection. J Immunol 180: 5771–5777.

41. PrabhakarU, ConwayTM, MurdockP, MooneyJL, ClarkS, et al. (2005) Correlation of protein and gene expression profiles of inflammatory proteins after endotoxin challenge in human subjects. DNA Cell Biol 24: 410–431.

42. FinneyOC, RileyEM, WaltherM (2010) Regulatory T cells in malaria–friend or foe? Trends Immunol 31: 63–70.

43. O'GarraA, VieiraP (2007) T(H)1 cells control themselves by producing interleukin-10. Nat Rev Immunol 7: 425–428.

44. WeissGE, TraoreB, KayentaoK, OngoibaA, DoumboS, et al. (2010) The Plasmodium falciparum-specific human memory B cell compartment expands gradually with repeated malaria infections. PLoS Pathog 6: e1000912.

45. CromptonPD, KayalaMA, TraoreB, KayentaoK, OngoibaA, et al. (2010) A prospective analysis of the Ab response to Plasmodium falciparum before and after a malaria season by protein microarray. Proc Natl Acad Sci U S A 107: 6958–6963.

46. SmithT, FelgerI, TannerM, BeckHP (1999) Premunition in Plasmodium falciparum infection: insights from the epidemiology of multiple infections. Trans R Soc Trop Med Hyg 93(Suppl 1): 59–64.

47. HansenDS, SchofieldL (2010) Natural regulatory T cells in malaria: host or parasite allies? PLoS Pathog 6: e1000771.

48. MarshK, ForsterD, WaruiruC, MwangiI, WinstanleyM, et al. (1995) Indicators of life-threatening malaria in African children. N Engl J Med 332: 1399–1404.

49. SchofieldL, GrauGE (2005) Immunological processes in malaria pathogenesis. Nat Rev Immunol 5: 722–735.

50. GuptaS, SnowRW, DonnellyCA, MarshK, NewboldC (1999) Immunity to non-cerebral severe malaria is acquired after one or two infections. Nat Med 5: 340–343.

51. AvrilM, TripathiAK, BrazierAJ, AndisiC, JanesJH, et al. (2012) A restricted subset of var genes mediates adherence of Plasmodium falciparum-infected erythrocytes to brain endothelial cells. Proc Natl Acad Sci U S A 109: E1782–1790.

52. ClaessensA, AdamsY, GhumraA, LindergardG, BuchanCC, et al. (2012) A subset of group A-like var genes encodes the malaria parasite ligands for binding to human brain endothelial cells. Proc Natl Acad Sci U S A 109: E1772–1781.

53. LavstsenT, TurnerL, SagutiF, MagistradoP, RaskTS, et al. (2012) Plasmodium falciparum erythrocyte membrane protein 1 domain cassettes 8 and 13 are associated with severe malaria in children. Proc Natl Acad Sci U S A 109: E1791–1800.

54. TurnerL, LavstsenT, BergerSS, WangCW, PetersenJE, et al. (2013) Severe malaria is associated with parasite binding to endothelial protein C receptor. Nature 498: 502–505.

55. TakeuchiO, AkiraS (2010) Pattern recognition receptors and inflammation. Cell 140: 805–820.

56. HaydenMS, GhoshS (2008) Shared principles in NF-kappaB signaling. Cell 132: 344–362.

57. DavisBK, WenH, TingJP (2011) The inflammasome NLRs in immunity, inflammation, and associated diseases. Annu Rev Immunol 29: 707–735.

58. BuckleyJM, WangJH, RedmondHP (2006) Cellular reprogramming by gram-positive bacterial components: a review. J Leukoc Biol 80: 731–741.

59. ZhongB, MaHY, YangQ, GuFR, YinGQ, et al. (2008) Decrease in toll-like receptors 2 and 4 in the spleen of mouse with endotoxic tolerance. Inflamm Res 57: 252–259.

60. MantovaniA, BonecchiR, LocatiM (2006) Tuning inflammation and immunity by chemokine sequestration: decoys and more. Nat Rev Immunol 6: 907–918.

61. FinneyOC, NwakanmaD, ConwayDJ, WaltherM, RileyEM (2009) Homeostatic regulation of T effector to Treg ratios in an area of seasonal malaria transmission. Eur J Immunol 39: 1288–1300.

62. WaltherM, JeffriesD, FinneyOC, NjieM, EbonyiA, et al. (2009) Distinct roles for FOXP3 and FOXP3 CD4 T cells in regulating cellular immunity to uncomplicated and severe Plasmodium falciparum malaria. PLoS Pathog 5: e1000364.

63. MinigoG, WoodberryT, PieraKA, SalwatiE, TjitraE, et al. (2009) Parasite-dependent expansion of TNF receptor II-positive regulatory T cells with enhanced suppressive activity in adults with severe malaria. PLoS Pathog 5: e1000402.

64. GerosaF, NisiiC, RighettiS, MiccioloR, MarchesiniM, et al. (1999) CD4(+) T cell clones producing both interferon-gamma and interleukin-10 predominate in bronchoalveolar lavages of active pulmonary tuberculosis patients. Clin Immunol 92: 224–234.

65. JankovicD, KullbergMC, FengCG, GoldszmidRS, CollazoCM, et al. (2007) Conventional T-bet(+)Foxp3(−) Th1 cells are the major source of host-protective regulatory IL-10 during intracellular protozoan infection. J Exp Med 204: 273–283.

66. AndersonCF, OukkaM, KuchrooVJ, SacksD (2007) CD4(+)CD25(−)Foxp3(−) Th1 cells are the source of IL-10-mediated immune suppression in chronic cutaneous leishmaniasis. J Exp Med 204: 285–297.

67. NylenS, MauryaR, EidsmoL, ManandharKD, SundarS, et al. (2007) Splenic accumulation of IL-10 mRNA in T cells distinct from CD4+CD25+ (Foxp3) regulatory T cells in human visceral leishmaniasis. J Exp Med 204: 805–817.

68. JagannathanP, Eccles-JamesI, BowenK, NankyaF, AumaA, et al. (2014) IFNgamma/IL-10 Co-producing Cells Dominate the CD4 Response to Malaria in Highly Exposed Children. PLoS Pathog 10: e1003864.

69. MarshK, KinyanjuiS (2006) Immune effector mechanisms in malaria. Parasite Immunol 28: 51–60.

70. O'SheaJJ, PaulWE (2010) Mechanisms underlying lineage commitment and plasticity of helper CD4+ T cells. Science 327: 1098–1102.

71. LeitnerWW, KrzychU (1997) Plasmodium falciparum malaria blood stage parasites preferentially inhibit macrophages with high phagocytic activity. Parasite Immunol 19: 103–110.

72. UrbanBC, RobertsDJ (2002) Malaria, monocytes, macrophages and myeloid dendritic cells: sticking of infected erythrocytes switches off host cells. Curr Opin Immunol 14: 458–465.

73. McGilvrayID, SerghidesL, KapusA, RotsteinOD, KainKC (2000) Nonopsonic monocyte/macrophage phagocytosis of Plasmodium falciparum-parasitized erythrocytes: a role for CD36 in malarial clearance. Blood 96: 3231–3240.

74. SerghidesL, KainKC (2002) Mechanism of protection induced by vitamin A in falciparum malaria. Lancet 359: 1404–1406.

75. BiswasSK, Lopez-CollazoE (2009) Endotoxin tolerance: new mechanisms, molecules and clinical significance. Trends Immunol 30: 475–487.

76. FosterSL, MedzhitovR (2009) Gene-specific control of the TLR-induced inflammatory response. Clin Immunol 130: 7–15.

77. QuinnEM, WangJ, RedmondHP (2012) The emerging role of microRNA in regulation of endotoxin tolerance. J Leukoc Biol 91: 721–727.

78. KleinnijenhuisJ, QuintinJ, PreijersF, JoostenLA, IfrimDC, et al. (2012) Bacille Calmette-Guerin induces NOD2-dependent nonspecific protection from reinfection via epigenetic reprogramming of monocytes. Proc Natl Acad Sci U S A 109: 17537–17542.

79. QuintinJ, SaeedS, MartensJH, Giamarellos-BourboulisEJ, IfrimDC, et al. (2012) Candida albicans infection affords protection against reinfection via functional reprogramming of monocytes. Cell Host Microbe 12: 223–232.

80. TranTM, LiS, DoumboS, DoumtabeD, HuangCY, et al. (2013) An intensive longitudinal cohort study of Malian children and adults reveals no evidence of acquired immunity to Plasmodium falciparum infection. Clin Infect Dis 57: 40–47.

81. ChaussabelD, PascualV, BanchereauJ (2010) Assessing the human immune system through blood transcriptomics. BMC Biol 8: 84.

82. HviidL, TheanderTG, AbdulhadiNH, Abu-ZeidYA, BayoumiRA, et al. (1991) Transient depletion of T cells with high LFA-1 expression from peripheral circulation during acute Plasmodium falciparum malaria. Eur J Immunol 21: 1249–1253.

83. CollinsWE, JefferyGM (1999) A retrospective examination of secondary sporozoite- and trophozoite-induced infections with Plasmodium falciparum: development of parasitologic and clinical immunity following secondary infection. Am J Trop Med Hyg 61: 20–35.

84. Mackey-LawrenceNM, GuoX, SturdevantDE, VirtanevaK, HernandezMM, et al. (2013) Effect of the Leptin Receptor Q223R Polymorphism on the Host Transcriptome following Infection with Entamoeba histolytica. Infect Immun 81: 1460–1470.

85. Artavanis-TsakonasK, RileyEM (2002) Innate immune response to malaria: rapid induction of IFN-gamma from human NK cells by live Plasmodium falciparum-infected erythrocytes. J Immunol 169: 2956–2963.

86. RoedererM, NozziJL, NasonMX (2011) SPICE: Exploration and analysis of post-cytometric complex multivariate datasets. Cytometry A 79: 167–174.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 4
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#