#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Hypoxia Inducible Factor Signaling Modulates Susceptibility to Mycobacterial Infection via a Nitric Oxide Dependent Mechanism


Tuberculosis is a current major world-health problem, exacerbated by the causative pathogen, Mycobacterium tuberculosis (Mtb), becoming increasingly resistant to conventional antibiotic treatment. Mtb is able to counteract the bactericidal mechanisms of leukocytes to survive intracellularly and develop a niche permissive for proliferation and dissemination. Understanding of the pathogenesis of mycobacterial infections such as tuberculosis (TB) remains limited, especially for early infection and for reactivation of latent infection. Signaling via hypoxia inducible factor α (HIF-α) transcription factors has previously been implicated in leukocyte activation and host defence. We have previously shown that hypoxic signaling via stabilization of Hif-1α prolongs the functionality of leukocytes in the innate immune response to injury. We sought to manipulate Hif-α signaling in a well-established Mycobacterium marinum (Mm) zebrafish model of TB to investigate effects on the host's ability to combat mycobacterial infection. Stabilization of host Hif-1α, both pharmacologically and genetically, at early stages of Mm infection was able to reduce the bacterial burden of infected larvae. Increasing Hif-1α signaling enhanced levels of reactive nitrogen species (RNS) in neutrophils prior to infection and was able to reduce larval mycobacterial burden. Conversely, decreasing Hif-2α signaling enhanced RNS levels and reduced bacterial burden, demonstrating that Hif-1α and Hif-2α have opposing effects on host susceptibility to mycobacterial infection. The antimicrobial effect of Hif-1α stabilization, and Hif-2α reduction, were demonstrated to be dependent on inducible nitric oxide synthase (iNOS) signaling at early stages of infection. Our findings indicate that induction of leukocyte iNOS by stabilizing Hif-1α, or reducing Hif-2α, aids the host during early stages of Mm infection. Stabilization of Hif-1α therefore represents a potential target for therapeutic intervention against tuberculosis.


Vyšlo v časopise: Hypoxia Inducible Factor Signaling Modulates Susceptibility to Mycobacterial Infection via a Nitric Oxide Dependent Mechanism. PLoS Pathog 9(12): e32767. doi:10.1371/journal.ppat.1003789
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003789

Souhrn

Tuberculosis is a current major world-health problem, exacerbated by the causative pathogen, Mycobacterium tuberculosis (Mtb), becoming increasingly resistant to conventional antibiotic treatment. Mtb is able to counteract the bactericidal mechanisms of leukocytes to survive intracellularly and develop a niche permissive for proliferation and dissemination. Understanding of the pathogenesis of mycobacterial infections such as tuberculosis (TB) remains limited, especially for early infection and for reactivation of latent infection. Signaling via hypoxia inducible factor α (HIF-α) transcription factors has previously been implicated in leukocyte activation and host defence. We have previously shown that hypoxic signaling via stabilization of Hif-1α prolongs the functionality of leukocytes in the innate immune response to injury. We sought to manipulate Hif-α signaling in a well-established Mycobacterium marinum (Mm) zebrafish model of TB to investigate effects on the host's ability to combat mycobacterial infection. Stabilization of host Hif-1α, both pharmacologically and genetically, at early stages of Mm infection was able to reduce the bacterial burden of infected larvae. Increasing Hif-1α signaling enhanced levels of reactive nitrogen species (RNS) in neutrophils prior to infection and was able to reduce larval mycobacterial burden. Conversely, decreasing Hif-2α signaling enhanced RNS levels and reduced bacterial burden, demonstrating that Hif-1α and Hif-2α have opposing effects on host susceptibility to mycobacterial infection. The antimicrobial effect of Hif-1α stabilization, and Hif-2α reduction, were demonstrated to be dependent on inducible nitric oxide synthase (iNOS) signaling at early stages of infection. Our findings indicate that induction of leukocyte iNOS by stabilizing Hif-1α, or reducing Hif-2α, aids the host during early stages of Mm infection. Stabilization of Hif-1α therefore represents a potential target for therapeutic intervention against tuberculosis.


Zdroje

1. KoulA, ArnoultE, LounisN, GuillemontJ, AndriesK (2011) The challenge of new drug discovery for tuberculosis. Nature 469: 483–490.

2. PodinovskaiaM, LeeW, CaldwellS, RussellDG (2012) Infection of macrophages with Mycobacterium tuberculosis induces global modifications to phagosomal function. Cell Microbiol 15: 843–59.

3. DannenbergAMJr (1993) Immunopathogenesis of pulmonary tuberculosis. Hosp Pract (Off Ed) 28: 51–58.

4. FlynnJL, ChanJ, LinPL (2011) Macrophages and control of granulomatous inflammation in tuberculosis. Mucosal Immunol 4: 271–278.

5. FriedrichN, HagedornM, Soldati-FavreD, SoldatiT (2012) Prison break: pathogens' strategies to egress from host cells. Microbiol Mol Biol Rev 76: 707–720.

6. BoshoffHI, BarryCE3rd (2005) Tuberculosis - metabolism and respiration in the absence of growth. Nat Rev Microbiol 3: 70–80.

7. ViaLE, LinPL, RaySM, CarrilloJ, AllenSS, et al. (2008) Tuberculous granulomas are hypoxic in guinea pigs, rabbits, and nonhuman primates. Infect Immun 76: 2333–2340.

8. EpsteinAC, GleadleJM, McNeillLA, HewitsonKS, O'RourkeJ, et al. (2001) C. elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell 107: 43–54.

9. BruickRK, McKnightSL (2001) A conserved family of prolyl-4-hydroxylases that modify HIF. Science 294: 1337–1340.

10. MahonPC, HirotaK, SemenzaGL (2001) FIH-1: a novel protein that interacts with HIF-1alpha and VHL to mediate repression of HIF-1 transcriptional activity. Genes Dev 15: 2675–2686.

11. WengerRH (2002) Cellular adaptation to hypoxia: O2-sensing protein hydroxylases, hypoxia-inducible transcription factors, and O2-regulated gene expression. FASEB J 16: 1151–1162.

12. CramerT, YamanishiY, ClausenBE, ForsterI, PawlinskiR, et al. (2003) HIF-1alpha is essential for myeloid cell-mediated inflammation. Cell 112: 645–657.

13. PeyssonnauxC, DattaV, CramerT, DoedensA, TheodorakisEA, et al. (2005) HIF-1alpha expression regulates the bactericidal capacity of phagocytes. J Clin Invest 115: 1806–1815.

14. WalmsleySR, ChilversER, WhyteMK (2009) Hypoxia. Hypoxia, hypoxia inducible factor and myeloid cell function. Arthritis Res Ther 11: 219.

15. NizetV, JohnsonRS (2009) Interdependence of hypoxic and innate immune responses. Nat Rev Immunol 9: 609–617.

16. ElksPM, van EedenFJ, DixonG, WangX, Reyes-AldasoroCC, et al. (2011) Activation of hypoxia-inducible factor-1alpha (Hif-1alpha) delays inflammation resolution by reducing neutrophil apoptosis and reverse migration in a zebrafish inflammation model. Blood 118: 712–722.

17. AnandRJ, GribarSC, LiJ, KohlerJW, BrancaMF, et al. (2007) Hypoxia causes an increase in phagocytosis by macrophages in a HIF-1alpha-dependent manner. J Leukoc Biol 82: 1257–1265.

18. SantorielloC, ZonLI (2012) Hooked! Modeling human disease in zebrafish. J Clin Invest 122: 2337–2343.

19. SwaimLE, ConnollyLE, VolkmanHE, HumbertO, BornDE, et al. (2006) Mycobacterium marinum infection of adult zebrafish causes caseating granulomatous tuberculosis and is moderated by adaptive immunity. Infect Immun 74: 6108–6117.

20. ParikkaM, HammarenMM, HarjulaSK, HalfpennyNJ, OksanenKE, et al. (2012) Mycobacterium marinum causes a latent infection that can be reactivated by gamma irradiation in adult zebrafish. PLoS Pathog 8: e1002944.

21. BergRD, RamakrishnanL (2012) Insights into tuberculosis from the zebrafish model. Trends in molecular medicine 18: 689–690.

22. AlibaudL, RomboutsY, TrivelliX, BurguiereA, CirilloSL, et al. (2011) A Mycobacterium marinum TesA mutant defective for major cell wall-associated lipids is highly attenuated in Dictyostelium discoideum and zebrafish embryos. Mol Microbiol 80: 919–934.

23. StoopEJ, SchipperT, HuberSK, NezhinskyAE, VerbeekFJ, et al. (2011) Zebrafish embryo screen for mycobacterial genes involved in the initiation of granuloma formation reveals a newly identified ESX-1 component. Dis Model Mech 4: 526–536.

24. van der VaartM, van SoestJJ, SpainkHP, MeijerAH (2013) Functional analysis of a zebrafish myd88 mutant identifies key transcriptional components of the innate immune system. Disease models & mechanisms 6: 841–54.

25. SanthakumarK, JudsonEC, ElksPM, McKeeS, ElworthyS, et al. (2012) A zebrafish model to study and therapeutically manipulate hypoxia signaling in tumorigenesis. Cancer Res 72: 4017–4027.

26. ManothamK, TanakaT, OhseT, KojimaI, MiyataT, et al. (2005) A biologic role of HIF-1 in the renal medulla. Kidney Int 67: 1428–1439.

27. LinkeS, StojkoskiC, KewleyRJ, BookerGW, WhitelawML, et al. (2004) Substrate requirements of the oxygen-sensing asparaginyl hydroxylase factor-inhibiting hypoxia-inducible factor. J Biol Chem 279: 14391–14397.

28. RojasDA, Perez-MunizagaDA, CentaninL, AntonelliM, WappnerP, et al. (2007) Cloning of hif-1alpha and hif-2alpha and mRNA expression pattern during development in zebrafish. Gene Expr Patterns 7: 339–345.

29. SummersgillJT, PowellLA, BusterBL, MillerRD, RamirezJA (1992) Killing of Legionella pneumophila by nitric oxide in gamma-interferon-activated macrophages. J Leukoc Biol 52: 625–629.

30. ForlenzaM, ScharsackJP, KachamakovaNM, Taverne-ThieleAJ, RomboutJH, et al. (2008) Differential contribution of neutrophilic granulocytes and macrophages to nitrosative stress in a host-parasite animal model. Mol Immunol 45: 3178–3189.

31. van der VaartM, SpainkHP, MeijerAH (2012) Pathogen recognition and activation of the innate immune response in zebrafish. Adv Hematol 2012: 159807.

32. HallC, FloresMV, StormT, CrosierK, CrosierP (2007) The zebrafish lysozyme C promoter drives myeloid-specific expression in transgenic fish. BMC Dev Biol 7: 42.

33. EllettF, PaseL, HaymanJW, AndrianopoulosA, LieschkeGJ (2011) mpeg1 promoter transgenes direct macrophage-lineage expression in zebrafish. Blood 117: e49–56.

34. HallCJ, FloresMV, OehlersSH, SandersonLE, LamEY, et al. (2012) Infection-responsive expansion of the hematopoietic stem and progenitor cell compartment in zebrafish is dependent upon inducible nitric oxide. Cell Stem Cell 10: 198–209.

35. LepillerS, LaurensV, BouchotA, HerbomelP, SolaryE, et al. (2007) Imaging of nitric oxide in a living vertebrate using a diamino-fluorescein probe. Free Radic Biol Med 43: 619–627.

36. ClayH, VolkmanHE, RamakrishnanL (2008) Tumor necrosis factor signaling mediates resistance to mycobacteria by inhibiting bacterial growth and macrophage death. Immunity 29: 283–294.

37. NorthTE, GoesslingW, PeetersM, LiP, CeolC, et al. (2009) Hematopoietic stem cell development is dependent on blood flow. Cell 137: 736–748.

38. ChristiansenB, WellendorphP, Brauner-OsborneH (2006) Known regulators of nitric oxide synthase and arginase are agonists at the human G-protein-coupled receptor GPRC6A. Br J Pharmacol 147: 855–863.

39. RodaJM, SumnerLA, EvansR, PhillipsGS, MarshCB, et al. (2011) Hypoxia-inducible factor-2alpha regulates GM-CSF-derived soluble vascular endothelial growth factor receptor 1 production from macrophages and inhibits tumor growth and angiogenesis. J Immunol 187: 1970–1976.

40. TakedaN, O'DeaEL, DoedensA, KimJW, WeidemannA, et al. (2010) Differential activation and antagonistic function of HIF-{alpha} isoforms in macrophages are essential for NO homeostasis. Genes Dev 24: 491–501.

41. KuijlC, SavageND, MarsmanM, TuinAW, JanssenL, et al. (2007) Intracellular bacterial growth is controlled by a kinase network around PKB/AKT1. Nature 450: 725–730.

42. TobinDM, RocaFJ, OhSF, McFarlandR, VickeryTW, et al. (2012) Host genotype-specific therapies can optimize the inflammatory response to mycobacterial infections. Cell 148: 434–446.

43. TailleuxL, WaddellSJ, PelizzolaM, MortellaroA, WithersM, et al. (2008) Probing host pathogen cross-talk by transcriptional profiling of both Mycobacterium tuberculosis and infected human dendritic cells and macrophages. PLoS One 3: e1403.

44. MasakiT, QuJ, Cholewa-WaclawJ, BurrK, RaaumR, et al. (2013) Reprogramming Adult Schwann Cells to Stem Cell-like Cells by Leprosy Bacilli Promotes Dissemination of Infection. Cell 152: 51–67.

45. EhrtS, SchnappingerD, BekiranovS, DrenkowJ, ShiS, et al. (2001) Reprogramming of the macrophage transcriptome in response to interferon-gamma and Mycobacterium tuberculosis: signaling roles of nitric oxide synthase-2 and phagocyte oxidase. J Exp Med 194: 1123–1140.

46. TobinDM, VaryJCJr, RayJP, WalshGS, DunstanSJ, et al. (2010) The lta4h locus modulates susceptibility to mycobacterial infection in zebrafish and humans. Cell 140: 717–730.

47. PeyssonnauxC, BoutinAT, ZinkernagelAS, DattaV, NizetV, et al. (2008) Critical role of HIF-1alpha in keratinocyte defense against bacterial infection. The Journal of investigative dermatology 128: 1964–1968.

48. OkumuraCY, HollandsA, TranDN, OlsonJ, DaheshS, et al. (2012) A new pharmacological agent (AKB-4924) stabilizes hypoxia inducible factor-1 (HIF-1) and increases skin innate defenses against bacterial infection. Journal of molecular medicine 90: 1079–1089.

49. EvansTJ, ButteryLD, CarpenterA, SpringallDR, PolakJM, et al. (1996) Cytokine-treated human neutrophils contain inducible nitric oxide synthase that produces nitration of ingested bacteria. Proc Natl Acad Sci U S A 93: 9553–9558.

50. ClayH, DavisJM, BeeryD, HuttenlocherA, LyonsSE, et al. (2007) Dichotomous role of the macrophage in early Mycobacterium marinum infection of the zebrafish. Cell Host Microbe 2: 29–39.

51. YangCT, CambierCJ, DavisJM, HallCJ, CrosierPS, et al. (2012) Neutrophils exert protection in the early tuberculous granuloma by oxidative killing of mycobacteria phagocytosed from infected macrophages. Cell Host Microbe 12: 301–312.

52. LoweDM, RedfordPS, WilkinsonRJ, O'GarraA, MartineauAR (2012) Neutrophils in tuberculosis: friend or foe? Trends Immunol 33: 14–25.

53. LieschkeGJ, OatesAC, CrowhurstMO, WardAC, LaytonJE (2001) Morphologic and functional characterization of granulocytes and macrophages in embryonic and adult zebrafish. Blood 98: 3087–3096.

54. EiserichJP, HristovaM, CrossCE, JonesAD, FreemanBA, et al. (1998) Formation of nitric oxide-derived inflammatory oxidants by myeloperoxidase in neutrophils. Nature 391: 393–397.

55. MissonP, van den BruleS, BarbarinV, LisonD, HuauxF (2004) Markers of macrophage differentiation in experimental silicosis. J Leukoc Biol 76: 926–932.

56. KeithB, JohnsonRS, SimonMC (2012) HIF1alpha and HIF2alpha: sibling rivalry in hypoxic tumour growth and progression. Nature reviews Cancer 12: 9–22.

57. Nusslein-Volhard C DR (2002) Zebrafish: A Practical Approach. Oxford: Oxford University Press.

58. RenshawSA, LoynesCA, TrushellDM, ElworthyS, InghamPW, et al. (2006) A transgenic zebrafish model of neutrophilic inflammation. Blood 108: 3976–3978.

59. van der SarAM, SpainkHP, ZakrzewskaA, BitterW, MeijerAH (2009) Specificity of the zebrafish host transcriptome response to acute and chronic mycobacterial infection and the role of innate and adaptive immune components. Mol Immunol 46: 2317–2332.

60. CuiC, BenardEL, KanwalZ, StockhammerOW, van der VaartM, et al. (2011) Infectious disease modeling and innate immune function in zebrafish embryos. Methods Cell Biol 105: 273–308.

61. BenardEL, van der SarAM, EllettF, LieschkeGJ, SpainkHP, et al. (2012) Infection of zebrafish embryos with intracellular bacterial pathogens. J Vis Exp 61: pii: 3781.

62. ReddMJ, CooperL, WoodW, StramerB, MartinP (2004) Wound healing and inflammation: embryos reveal the way to perfect repair. Philos Trans R Soc Lond B Biol Sci 359: 777–784.

63. LoynesCA, MartinJS, RobertsonA, TrushellDM, InghamPW, et al. (2010) Pivotal Advance: Pharmacological manipulation of inflammation resolution during spontaneously resolving tissue neutrophilia in the zebrafish. J Leukoc Biol 87: 203–212.

64. BurgessA, VigneronS, BrioudesE, LabbeJC, LorcaT, et al. (2010) Loss of human Greatwall results in G2 arrest and multiple mitotic defects due to deregulation of the cyclin B-Cdc2/PP2A balance. Proc Natl Acad Sci U S A 107: 12564–12569.

65. KwanKM, FujimotoE, GrabherC, MangumBD, HardyME, et al. (2007) The Tol2kit: a multisite gateway-based construction kit for Tol2 transposon transgenesis constructs. Dev Dyn 236: 3088–3099.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2013 Číslo 12
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#