#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Identification of Host-Targeted Small Molecules That Restrict Intracellular Growth


Mycobacterium tuberculosis remains a significant threat to global health. Macrophages are the host cell for M. tuberculosis infection, and although bacteria are able to replicate intracellularly under certain conditions, it is also clear that macrophages are capable of killing M. tuberculosis if appropriately activated. The outcome of infection is determined at least in part by the host-pathogen interaction within the macrophage; however, we lack a complete understanding of which host pathways are critical for bacterial survival and replication. To add to our understanding of the molecular processes involved in intracellular infection, we performed a chemical screen using a high-content microscopic assay to identify small molecules that restrict mycobacterial growth in macrophages by targeting host functions and pathways. The identified host-targeted inhibitors restrict bacterial growth exclusively in the context of macrophage infection and predominantly fall into five categories: G-protein coupled receptor modulators, ion channel inhibitors, membrane transport proteins, anti-inflammatories, and kinase modulators. We found that fluoxetine, a selective serotonin reuptake inhibitor, enhances secretion of pro-inflammatory cytokine TNF-α and induces autophagy in infected macrophages, and gefitinib, an inhibitor of the Epidermal Growth Factor Receptor (EGFR), also activates autophagy and restricts growth. We demonstrate that during infection signaling through EGFR activates a p38 MAPK signaling pathway that prevents macrophages from effectively responding to infection. Inhibition of this pathway using gefitinib during in vivo infection reduces growth of M. tuberculosis in the lungs of infected mice. Our results support the concept that screening for inhibitors using intracellular models results in the identification of tool compounds for probing pathways during in vivo infection and may also result in the identification of new anti-tuberculosis agents that work by modulating host pathways. Given the existing experience with some of our identified compounds for other therapeutic indications, further clinically-directed study of these compounds is merited.


Vyšlo v časopise: Identification of Host-Targeted Small Molecules That Restrict Intracellular Growth. PLoS Pathog 10(2): e32767. doi:10.1371/journal.ppat.1003946
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003946

Souhrn

Mycobacterium tuberculosis remains a significant threat to global health. Macrophages are the host cell for M. tuberculosis infection, and although bacteria are able to replicate intracellularly under certain conditions, it is also clear that macrophages are capable of killing M. tuberculosis if appropriately activated. The outcome of infection is determined at least in part by the host-pathogen interaction within the macrophage; however, we lack a complete understanding of which host pathways are critical for bacterial survival and replication. To add to our understanding of the molecular processes involved in intracellular infection, we performed a chemical screen using a high-content microscopic assay to identify small molecules that restrict mycobacterial growth in macrophages by targeting host functions and pathways. The identified host-targeted inhibitors restrict bacterial growth exclusively in the context of macrophage infection and predominantly fall into five categories: G-protein coupled receptor modulators, ion channel inhibitors, membrane transport proteins, anti-inflammatories, and kinase modulators. We found that fluoxetine, a selective serotonin reuptake inhibitor, enhances secretion of pro-inflammatory cytokine TNF-α and induces autophagy in infected macrophages, and gefitinib, an inhibitor of the Epidermal Growth Factor Receptor (EGFR), also activates autophagy and restricts growth. We demonstrate that during infection signaling through EGFR activates a p38 MAPK signaling pathway that prevents macrophages from effectively responding to infection. Inhibition of this pathway using gefitinib during in vivo infection reduces growth of M. tuberculosis in the lungs of infected mice. Our results support the concept that screening for inhibitors using intracellular models results in the identification of tool compounds for probing pathways during in vivo infection and may also result in the identification of new anti-tuberculosis agents that work by modulating host pathways. Given the existing experience with some of our identified compounds for other therapeutic indications, further clinically-directed study of these compounds is merited.


Zdroje

1. DereticV, SinghS, MasterS, HarrisJ, RobertsE, et al. (2006) Mycobacterium tuberculosis inhibition of phagolysosome biogenesis and autophagy as a host defence mechanism. Cellular Microbiology 8: 719–727.

2. MalikZA, IyerSS, KusnerDJ (2001) Mycobacterium tuberculosis Phagosomes Exhibit Altered Calmodulin-Dependent Signal Transduction: Contribution to Inhibition of Phagosome-Lysosome Fusion and Intracellular Survival in Human Macrophages. The Journal of Immunology 166: 3392–3401.

3. JayachandranR, SundaramurthyV, CombaluzierB, MuellerP, KorfH, et al. (2007) Survival of Mycobacteria in Macrophages Is Mediated by Coronin 1-Dependent Activation of Calcineurin. Cell 130: 37–50.

4. DivangahiM, DesjardinsD, Nunes-AlvesC, RemoldHG, BeharSM (2010) Eicosanoid pathways regulate adaptive immunity to Mycobacterium tuberculosis. Nat Immunol 11: 751–758.

5. ChenM, DivangahiM, GanH, ShinDSJ, HongS, et al. (2008) Lipid mediators in innate immunity against tuberculosis: opposing roles of PGE2 and LXA4 in the induction of macrophage death. The Journal of Experimental Medicine 205: 2791–2801.

6. MartinCJ, BootyMG, RosebrockTR, Nunes-AlvesC, DesjardinsDM, et al. (2012) Efferocytosis Is an Innate Antibacterial Mechanism. Cell Host & Microbe 12: 289–300.

7. ShinD-M, JeonB-Y, LeeH-M, JinHS, YukJ-M, et al. (2010) Mycobacterium tuberculosis Eis Regulates Autophagy, Inflammation, and Cell Death through Redox-dependent Signaling. PLoS Pathog 6: e1001230.

8. MasterSS, RampiniSK, DavisAS, KellerC, EhlersS, et al. (2008) Mycobacterium tuberculosis Prevents Inflammasome Activation. Cell Host & Microbe 3: 224–232.

9. PeyronP, VaubourgeixJ, PoquetY, LevillainF, BotanchC, et al. (2008) Foamy macrophages from tuberculous patients' granulomas constitute a nutrient-rich reservoir for M. tuberculosis persistence. PLoS Pathogens 4: e1000204.

10. KumarD, NathL, KamalMA, VarshneyA, JainA, et al. (2010) Genome-wide Analysis of the Host Intracellular Network that Regulates Survival of Mycobacterium tuberculosis. Cell 140: 731–743.

11. JayaswalS, KamalMA, DuaR, GuptaS, MajumdarT, et al. (2010) Identification of Host-Dependent Survival Factors for Intracellular Mycobacterium tuberculosis through an siRNA Screen. PLoS Pathog 6: e1000839.

12. KuijlC, SavageNDL, MarsmanM, TuinAW, JanssenL, et al. (2007) Intracellular bacterial growth is controlled by a kinase network around PKB/AKT1. Nature 450: 725–U710.

13. 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.

14. StanleySA, HungDT (2009) Chemical Tools for Dissecting Bacterial Physiology and Virulence. Biochemistry 48: 8776–8786.

15. ChristopheT, JacksonM, JeonHK, FenisteinD, Contreras-DominguezM, et al. (2009) High Content Screening Identifies Decaprenyl-Phosphoribose 2′ Epimerase as a Target for Intracellular Antimycobacterial Inhibitors. PLoS Pathog 5: e1000645.

16. PetheK, BifaniP, JangJ, KangS, ParkS, et al. (2013) Discovery of Q203, a potent clinical candidate for the treatment of tuberculosis. Nat Med advance online publication.

17. SundaramurthyV, BarsacchiR, SamusikN, MarsicoG, GilleronJ, et al. (2013) Integration of Chemical and RNAi Multiparametric Profiles Identifies Triggers of Intracellular Mycobacterial Killing. Cell Host & Microbe 13: 129–142.

18. StanleySA, GrantSS, KawateT, IwaseN, ShimizuM, et al. (2012) Identification of Novel Inhibitors of M. tuberculosis Growth Using Whole Cell Based High-Throughput Screening. ACS Chemical Biology 7: 1377–1384.

19. CarpenterAE, JonesTR, LamprechtMR, ClarkeC, KangIH, et al. (2006) CellProfiler: image analysis software for identifying and quantifying cell phenotypes. Genome Biology 7.

20. WeiX, GaoL, ZhangX, QianH, RowanK, et al. (2013) Introducing Bayesian Thinking to High-Throughput Screening for False-Negative Rate Estimation. Journal of Biomolecular Screening

21. IlougaPE, HesterkampT (2012) On the Prediction of Statistical Parameters in High-Throughput Screening Using Resampling Techniques. Journal of Biomolecular Screening 17: 705–712.

22. KeaneJ, GershonS, WiseRP, Mirabile-LevensE, KasznicaJ, et al. (2001) Tuberculosis Associated with Infliximab, a Tumor Necrosis Factor-alpha ìNeutralizing Agent. New England Journal of Medicine 345: 1098–1104.

23. GardamMA, KeystoneEC, MenziesR, MannersS, SkameneE, et al. (2003) Anti-tumour necrosis factor agents and tuberculosis risk: mechanisms of action and clinical management. The Lancet Infectious Diseases 3: 148–155.

24. GlosliH, Stray-PedersenA, BrunAC, HoltmonLW, TonjumT, et al. (2008) Infections Due to Various Atypical Mycobacteria in a Norwegian Multiplex Family with Dominant Interferon-gamma Receptor Deficiency. Clinical Infectious Diseases 46: e23–e27.

25. BekkerL-G, FreemanS, MurrayPJ, RyffelB, KaplanG (2001) TNF-alpha Controls Intracellular Mycobacterial Growth by Both Inducible Nitric Oxide Synthase-Dependent and Inducible Nitric Oxide Synthase-Independent Pathways. The Journal of Immunology 166: 6728–6734.

26. Warner-SchmidtJL, VanoverKE, ChenEY, MarshallJJ, GreengardP (2011) Antidepressant effects of selective serotonin reuptake inhibitors (SSRIs) are attenuated by antiinflammatory drugs in mice and humans. Proceedings of the National Academy of Sciences 108: 9262–9267.

27. HarrisJ, KeaneJ (2010) How tumour necrosis factor blockers interfere with tuberculosis immunity. Clinical & Experimental Immunology 161: 1–9.

28. NapierRJ, RafiW, CheruvuM, PowellKR, ZaunbrecherMA, et al. (2011) Imatinib-Sensitive Tyrosine Kinases Regulate Mycobacterial Pathogenesis and Represent Therapeutic Targets against Tuberculosis. Cell Host & Microbe 10: 475–485.

29. BrunsH, StegelmannF, FabriM, D√∂hnerK, van ZandbergenG, et al. (2012) Abelson Tyrosine Kinase Controls Phagosomal Acidification Required for Killing of Mycobacterium tuberculosis in Human Macrophages. The Journal of Immunology 189: 4069–4078.

30. ZhaoZ, LeisterWH, RobinsonRG, BarnettSF, Defeo-JonesD, et al. (2005) Discovery of 2,3,5-trisubstituted pyridine derivatives as potent Akt1 and Akt2 dual inhibitors. Bioorganic & Medicinal Chemistry Letters 15: 905–909.

31. BarnettSF, Defeo-JonesD, FuS, HancockPJ, HaskellKM, et al. (2005) Identification and characterization of pleckstrin-homology-domain-dependent and isoenzyme-specific Akt inhibitors. Biochem J 385: 399–408.

32. AlessiDRA, M.; Caudwell, B.; Cron, P.; Morrice, N.; Cohen, P.; HemmingsBA (1996) Mechanism of activation of protein kinase B by insulin and IGF-1. EMBO J 15: 6541–6551.

33. WongS, WitteON (2004) The BCR-ABL Story: Bench to Bedside and Back. Annual Review of Immunology 22: 247–306.

34. WongS, McLaughlinJ, ChengD, ZhangC, ShokatKM, et al. (2004) Sole BCR-ABL inhibition is insufficient to eliminate all myeloproliferative disorder cell populations. Proceedings of the National Academy of Sciences of the United States of America 101: 17456–17461.

35. EierhoffT, HrinciusER, RescherU, LudwigS, EhrhardtC (2010) The Epidermal Growth Factor Receptor (EGFR) Promotes Uptake of Influenza A Viruses (IAV) into Host Cells. PLoS Pathog 6: e1001099.

36. LiuK, GualanoRC, HibbsML, AndersonGP, BozinovskiS (2008) Epidermal Growth Factor Receptor Signaling to Erk1/2 and STATs Control the Intensity of the Epithelial Inflammatory Responses to Rhinovirus Infection. Journal of Biological Chemistry 283: 9977–9985.

37. YanF, CaoH, ChaturvediR, KrishnaU, HobbsSS, et al. (2009) Epidermal Growth Factor Receptor Activation Protects Gastric Epithelial Cells From Helicobacter pylori-Induced Apoptosis. Gastroenterology 136: 1297–1307.e1293.

38. LynchT (2003) Clinical benefit in NSCLC: the evidence for gefitinib (Iressa, ZD1839). European Journal of Cancer Supplements 1: 17–22.

39. SpanglerJB, NeilJR, AbramovitchS, YardenY, WhiteFM, et al. (2010) Combination antibody treatment down-regulates epidermal growth factor receptor by inhibiting endosomal recycling. Proceedings of the National Academy of Sciences 107: 13252–13257.

40. LeeMRD (2005) C (2005) MAP kinase p38 inhibitors: Clinical results and an intimate look at their interactions with p38 alpha protein. Current Medicinal Chemistry 12: 2979–2994.

41. WebberJL, ToozeSA (2010) Coordinated regulation of autophagy by p38-alpha MAPK through mAtg9 and p38IP. EMBO J 29: 27–40.

42. SubbianS, TsenovaL, O'BrienP, YangG, KooM-S, et al. (2011) Phosphodiesterase-4 Inhibition Alters Gene Expression and Improves Isoniazid-Mediated Clearance of Mycobacterium tuberculosis in Rabbit Lungs. PLoS Pathog 7: e1002262.

43. GuptaS, SalamN, SrivastavaV, SinglaR, BeheraD, et al. (2009) Voltage Gated Calcium Channels Negatively Regulate Protective Immunity to Mycobacterium tuberculosis. PLoS ONE 4: e5305.

44. AdamsKN, TakakiK, ConnollyLE, WiedenhoftH, WingleeK, et al. (2011) Drug Tolerance in Replicating Mycobacteria Mediated by a Macrophage-Induced Efflux Mechanism. Cell 145: 39–53.

45. GuptaST, Sandeep (2013) Almeida, DeepakV (2013) Maiga, MariamaC (2013) Ammerman, NicoleC (2013) Bishai, WilliamR (2013) Acceleration of tuberculosis treatment by adjunctive therapy with verapamil as an efflux inhibitor. AJRCCM 10.1164/rccm.201304-0650OC.

46. SinghV, JamwalS, JainR, VermaP, GokhaleR, et al. (2012) Mycobacterium tuberculosis-Driven Targeted Recalibration of Macrophage Lipid Homeostasis Promotes the Foamy Phenotype. Cell Host & Microbe 12: 669–681.

47. de las Casas-EngelM, Dominguez-SotoA, Sierra-FilardiE, BragadoR, NietoC, et al. (2013) Serotonin Skews Human Macrophage Polarization through HTR2B and HTR7. The Journal of Immunology 190: 2301–2310.

48. GaskillPC, Loreto (2012) Eugenin, EliseoA (2012) Berman, JoanW (2012) Characterization and function of the human macrophage dopaminergic system: implications for CNS disease and drug abuse. Journal of Neuroimflammation 9: 203–216.

49. IzeboudCM, JAJ; Monshouwer, M; van Miert, ASJPAM; WitkampRF (1999) Participation of beta-Adrenergic Receptors on Marophages in Modulation of LPS-Induced Cytokine Release. Journal of Receptors and Signal Transduction 19: 191–202.

50. SternbergEM, TrialJ, ParkerCW (1986) Effect of serotonin on murine macrophages: suppression of Ia expression by serotonin and its reversal by 5-HT2 serotonergic receptor antagonists. The Journal of Immunology 137: 276–282.

51. Cloez-TayaraniI, Petit-BertronA-F, VentersHD, CavaillonJ-M (2003) Differential effect of serotonin on cytokine production in lipopolysaccharide-stimulated human peripheral blood mononuclear cells: involvement of 5-hydroxytryptamine2A receptors. International Immunology 15: 233–240.

52. TobinDM, RocaFJ, OhSF, McFarlandR, VickeryTW, et al. (2012) Host Genotype-Specific Therapies Can Optimize the Inflammatory Response to Mycobacterial Infections. Cell 148: 434–446.

53. RocaFJ, RamakrishnanL (2013) TNF Dually Mediates Resistance and Susceptibility to Mycobacteria via Mitochondrial Reactive Oxygen Species. Cell 153: 521–534.

54. KooM-S, MancaC, YangG, O'BrienP, SungN, et al. (2011) Phosphodiesterase 4 Inhibition Reduces Innate Immunity and Improves Isoniazid Clearance of Mycobacterium tuberculosis in the Lungs of Infected Mice. PLoS ONE 6: e17091.

55. ShubinH, LambertRE, HeikenCA, SokmensuerA, GlaskinA (1959) Steroid therapy and tuberculosis. Journal of the American Medical Association 170: 1885–1890.

56. CritchleyJA, YoungF, OrtonL, GarnerP (2013) Corticosteroids for prevention of mortality in people with tuberculosis: a systematic review and meta-analysis. The Lancet Infectious Diseases 13: 223–237.

57. WallisRS (2005) Reconsidering Adjuvant Immunotherapy for Tuberculosis. Clinical Infectious Diseases 41: 201–208.

58. SkerryC, HarperJ, KlunkM, BishaiWR, JainSK (2012) Adjunctive TNF Inhibition with Standard Treatment Enhances Bacterial Clearance in a Murine Model of Necrotic TB Granulomas. PLoS ONE 7: e39680.

59. MaigaM, AgarwalN, AmmermanNC, GuptaR, GuoH, et al. (2012) Successful Shortening of Tuberculosis Treatment Using Adjuvant Host-Directed Therapy with FDA-Approved Phosphodiesterase Inhibitors in the Mouse Model. PLoS ONE 7: e30749.

60. SchwegmannA, BrombacherF (2008) Host-Directed Drug Targeting of Factors Hijacked by Pathogens. Science Signaling 1: re8.

61. KorfH, Vander BekenS, RomanoM, SteffensenKR, StijlemansB, et al. (2009) Liver X receptors contribute to the protective immune response against Mycobacterium tuberculosis in mice. The Journal of Clinical Investigation 119: 1626–1637.

62. LeeM, LeeJ, CarrollMW, ChoiH, MinS, et al. (2012) Linezolid for Treatment of Chronic Extensively Drug-Resistant Tuberculosis. New England Journal of Medicine 367: 1508–1518.

63. StanleySA, KawateT, IwaseN, ShimizuM, ClatworthyAE, et al. (2013) Diarylcoumarins inhibit mycolic acid biosynthesis and kill Mycobacterium tuberculosis by targeting FadD32. Proceedings of the National Academy of Sciences 110: 11565–11570.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 2
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#