#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Infection Causes Distinct Epigenetic DNA Methylation Changes in Host Macrophages


The L.
donovani parasite causes visceral leishmaniasis, a tropical, neglected disease with an estimated number of 500,000 cases worldwide. Current drug treatments have toxic side effects, lead to drug resistance, and an effective vaccine is not available. The parasite has a complex life cycle residing within different host environments including the gut of a sand fly and immune cells of the mammalian host. Alteration of host cell gene expression including signaling pathways has been shown to be a major strategy to evade host cell immune response and thus enables the Leishmania parasite to survive, replicate and persist in its host cells. Recently it was demonstrated that intracellular pathogens such as viruses and bacteria are able to manipulate epigenetic processes, thereby perhaps facilitating their intracellular survival. Using an unbiased genome-wide DNA methylation approach, we demonstrate here that an intracellular parasite can alter host cell DNA methylation patterns resulting in altered gene expression possibly to establish disease. Thus DNA methylation changes in host cells upon infection might be a common strategy among intracellular pathogens for their uncontrolled replication and dissemination.


Vyšlo v časopise: Infection Causes Distinct Epigenetic DNA Methylation Changes in Host Macrophages. PLoS Pathog 10(10): e32767. doi:10.1371/journal.ppat.1004419
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004419

Souhrn

The L.
donovani parasite causes visceral leishmaniasis, a tropical, neglected disease with an estimated number of 500,000 cases worldwide. Current drug treatments have toxic side effects, lead to drug resistance, and an effective vaccine is not available. The parasite has a complex life cycle residing within different host environments including the gut of a sand fly and immune cells of the mammalian host. Alteration of host cell gene expression including signaling pathways has been shown to be a major strategy to evade host cell immune response and thus enables the Leishmania parasite to survive, replicate and persist in its host cells. Recently it was demonstrated that intracellular pathogens such as viruses and bacteria are able to manipulate epigenetic processes, thereby perhaps facilitating their intracellular survival. Using an unbiased genome-wide DNA methylation approach, we demonstrate here that an intracellular parasite can alter host cell DNA methylation patterns resulting in altered gene expression possibly to establish disease. Thus DNA methylation changes in host cells upon infection might be a common strategy among intracellular pathogens for their uncontrolled replication and dissemination.


Zdroje

1. MurrayHW, BermanJD, DaviesCR, SaraviaNG (2005) Advances in leishmaniasis. Lancet 366: 1561–1577.

2. MoradinN, DescoteauxA (2012) Leishmania promastigotes: building a safe niche within macrophages. Front Cell Infect Microbiol 2: 121.

3. DograN, WarburtonC, McMasterWR (2007) Leishmania major abrogates gamma interferon-induced gene expression in human macrophages from a global perspective. Infect Immun 75: 3506–3515.

4. LiuD, UzonnaJE (2012) The early interaction of Leishmania with macrophages and dendritic cells and its influence on the host immune response. Front Cell Infect Microbiol 2: 83.

5. BhardwajS, SrivastavaN, SudanR, SahaB (2010) Leishmania interferes with host cell signaling to devise a survival strategy. J Biomed Biotechnol 2010: 109189.

6. GregoryDJ, OlivierM (2005) Subversion of host cell signalling by the protozoan parasite Leishmania. Parasitology 130 Suppl: S27–35.

7. ShioMT, HassaniK, IsnardA, RalphB, ContrerasI, et al. (2012) Host cell signalling and leishmania mechanisms of evasion. J Trop Med 2012: 819512.

8. KhavariDA, SenGL, RinnJL (2010) DNA methylation and epigenetic control of cellular differentiation. Cell Cycle 9: 3880–3883.

9. GhoshS, YatesAJ, FruhwaldMC, MiecznikowskiJC, PlassC, et al. (2010) Tissue specific DNA methylation of CpG islands in normal human adult somatic tissues distinguishes neural from non-neural tissues. Epigenetics 5: 527–538.

10. BirdA (2007) Perceptions of epigenetics. Nature 447: 396–398.

11. IoshikhesIP, ZhangMQ (2000) Large-scale human promoter mapping using CpG islands. Nat Genet 26: 61–63.

12. WeberM, HellmannI, StadlerMB, RamosL, PaaboS, et al. (2007) Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nat Genet 39: 457–466.

13. JonesMJ, FejesAP, KoborMS (2013) DNA methylation, genotype and gene expression: who is driving and who is along for the ride? Genome Biol 14: 126.

14. JjingoD, ConleyAB, YiSV, LunyakVV, JordanIK (2012) On the presence and role of human gene-body DNA methylation. Oncotarget 3: 462–474.

15. LamLL, EmberlyE, FraserHB, NeumannSM, ChenE, et al. (2012) Factors underlying variable DNA methylation in a human community cohort. Proc Natl Acad Sci U S A 109 Suppl 2: 17253–17260.

16. MinarovitsJ (2009) Microbe-induced epigenetic alterations in host cells: the coming era of patho-epigenetics of microbial infections. A review. Acta Microbiol Immunol Hung 56: 1–19.

17. Gomez-DiazE, JordaM, PeinadoMA, RiveroA (2012) Epigenetics of host-pathogen interactions: the road ahead and the road behind. PLoS Pathog 8: e1003007.

18. LangC, HildebrandtA, BrandF, OpitzL, DihaziH, et al. (2012) Impaired chromatin remodelling at STAT1-regulated promoters leads to global unresponsiveness of Toxoplasma gondii-infected macrophages to IFN-gamma. PLoS Pathog 8: e1002483.

19. Silmon de MonerriNC, KimK (2014) Pathogens Hijack the Epigenome: A New Twist on Host-Pathogen Interactions. Am J Pathol 184: 897–911.

20. BibikovaM, BarnesB, TsanC, HoV, KlotzleB, et al. (2011) High density DNA methylation array with single CpG site resolution. Genomics 98: 288–295.

21. G.K S (2005) Limma: linear models for microarray data. In: Gentleman R. CV, Dudoit S., Irizarry R. and Huber W., editor. Bioinformatics using R and Bioconductor. New York: Springer. pp. 397–420.

22. Huang daW, ShermanBT, LempickiRA (2009) Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4: 44–57.

23. Huang daW, ShermanBT, LempickiRA (2009) Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res 37: 1–13.

24. MillJ, HeijmansBT (2013) From promises to practical strategies in epigenetic epidemiology. Nat Rev Genet 14: 585–594.

25. OlivierM, BaimbridgeKG, ReinerNE (1992) Stimulus-response coupling in monocytes infected with Leishmania. Attenuation of calcium transients is related to defective agonist-induced accumulation of inositol phosphates. J Immunol 148: 1188–1196.

26. ShadabM, AliN (2011) Evasion of Host Defence by Leishmania donovani: Subversion of Signaling Pathways. Mol Biol Int 2011: 343961.

27. AudersetF, SchusterS, CoutazM, KochU, DesgrangesF, et al. (2012) Redundant Notch1 and Notch2 signaling is necessary for IFNgamma secretion by T helper 1 cells during infection with Leishmania major. PLoS Pathog 8: e1002560.

28. JaramilloM, GomezMA, LarssonO, ShioMT, TopisirovicI, et al. (2011) Leishmania repression of host translation through mTOR cleavage is required for parasite survival and infection. Cell Host Microbe 9: 331–341.

29. PinheiroNFJr, HermidaMD, MacedoMP, MengelJ, BaficaA, et al. (2006) Leishmania infection impairs beta 1-integrin function and chemokine receptor expression in mononuclear phagocytes. Infect Immun 74: 3912–3921.

30. RabhiI, RabhiS, Ben-OthmanR, RascheA, DaskalakiA, et al. (2012) Transcriptomic signature of Leishmania infected mice macrophages: a metabolic point of view. PLoS Negl Trop Dis 6: e1763.

31. Garcia-GarciaJC, BaratNC, TrembleySJ, DumlerJS (2009) Epigenetic silencing of host cell defense genes enhances intracellular survival of the rickettsial pathogen Anaplasma phagocytophilum. PLoS Pathog 5: e1000488.

32. PaschosK, AlldayMJ (2010) Epigenetic reprogramming of host genes in viral and microbial pathogenesis. Trends Microbiol 18: 439–447.

33. Koch-NolteF, KernstockS, Mueller-DieckmannC, WeissMS, HaagF (2008) Mammalian ADP-ribosyltransferases and ADP-ribosylhydrolases. Front Biosci 13: 6716–6729.

34. PriveC, DescoteauxA (2000) Leishmania donovani promastigotes evade the activation of mitogen-activated protein kinases p38, c-Jun N-terminal kinase, and extracellular signal-regulated kinase-1/2 during infection of naive macrophages. Eur J Immunol 30: 2235–2244.

35. GhoshS, BhattacharyyaS, SirkarM, SaGS, DasT, et al. (2002) Leishmania donovani suppresses activated protein 1 and NF-kappaB activation in host macrophages via ceramide generation: involvement of extracellular signal-regulated kinase. Infect Immun 70: 6828–6838.

36. RogerT, LugrinJ, Le RoyD, GoyG, MombelliM, et al. (2011) Histone deacetylase inhibitors impair innate immune responses to Toll-like receptor agonists and to infection. Blood 117: 1205–1217.

37. HirschDS, PironeDM, BurbeloPD (2001) A new family of Cdc42 effector proteins, CEPs, function in fibroblast and epithelial cell shape changes. J Biol Chem 276: 875–883.

38. LermM, HolmA, SeironA, SarndahlE, MagnussonKE, et al. (2006) Leishmania donovani requires functional Cdc42 and Rac1 to prevent phagosomal maturation. Infect Immun 74: 2613–2618.

39. LieblD, GriffithsG (2009) Transient assembly of F-actin by phagosomes delays phagosome fusion with lysosomes in cargo-overloaded macrophages. J Cell Sci 122: 2935–2945.

40. RodriguezNE, Gaur DixitU, AllenLA, WilsonME (2011) Stage-specific pathways of Leishmania infantum chagasi entry and phagosome maturation in macrophages. PLoS One 6: e19000.

41. (2008) R Development Core Team. R: A language and environment for statistical computing.

42. DuP, KibbeWA, LinSM (2008) lumi: a pipeline for processing Illumina microarray. Bioinformatics 24: 1547–1548.

43. DuP, ZhangX, HuangCC, JafariN, KibbeWA, et al. (2010) Comparison of Beta-value and M-value methods for quantifying methylation levels by microarray analysis. BMC Bioinformatics 11: 587.

44. SmythGK (2004) Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol 3: Article3.

45. PriceME, CottonAM, LamLL, FarreP, EmberlyE, et al. (2013) Additional annotation enhances potential for biologically-relevant analysis of the Illumina Infinium HumanMethylation450 BeadChip array. Epigenetics Chromatin 6: 4.

46. SchmittgenTD, LivakKJ (2008) Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc 3: 1101–1108.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 10
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#