Summary

Activation and Measurement of NLRP3 Inflammasome Activity Using IL-1β in Human Monocyte-derived Dendritic Cells

Published: May 22, 2014
doi:

Summary

Dendritic cells (DCs) secrete IL-1β in response to TLR8 recognition of synthetic purine, R848, followed by NLRP3 inflammasome activation with nigericin, therefore, IL-1β can be used to measure NLRP3 inflammasome activity. Intracellular cytokine staining, immunoblotting, and ELISA are used to accurately measure NLRP3 inflammasome priming and activation via IL-1β expression.

Abstract

Inflammatory processes resulting from the secretion of Interleukin (IL)-1 family cytokines by immune cells lead to local or systemic inflammation, tissue remodeling and repair, and virologic control1,2 . Interleukin-1β is an essential element of the innate immune response and contributes to eliminate invading pathogens while preventing the establishment of persistent infection1-5.

Inflammasomes are the key signaling platform for the activation of interleukin 1 converting enzyme (ICE or Caspase-1). The NLRP3 inflammasome requires at least two signals in DCs to cause IL-1β secretion6. Pro-IL-1β protein expression is limited in resting cells; therefore a priming signal is required for IL-1β transcription and protein expression. A second signal sensed by NLRP3 results in the formation of the multi-protein NLRP3 inflammasome. The ability of dendritic cells to respond to the signals required for IL-1β secretion can be tested using a synthetic purine, R848, which is sensed by TLR8 in human monocyte derived dendritic cells (moDCs) to prime cells, followed by activation of the NLRP3 inflammasome with the bacterial toxin and potassium ionophore, nigericin.

Monocyte derived DCs are easily produced in culture and provide significantly more cells than purified human myeloid DCs. The method presented here differs from other inflammasome assays in that it uses in vitro human, instead of mouse derived, DCs thus allowing for the study of the inflammasome in human disease and infection.

Introduction

Activation of innate immune system is required to steer adaptive immune responses during infection, disease, and vaccination7. Dendritic cells are the most potent antigen presenting cells of the innate immune system; they are specialized for uptake of antigens, migration to lymph nodes, and activation of naïve CD4+ and cytolytic CD8+ T-cells8-10. To enable rapid pathogen detection the innate immune system utilizes numerous germline encoded pattern recognition receptors (PRR) that recognize conserved pathogen derived motifs or host derived markers of cell stress and damage. Toll like receptors (TLRs) are membrane bound pattern recognition receptors that recognize certain extracellular phagocytized pathogen associated molecular patterns (PAMPs) and danger associated molecular patterns (DAMPs). By contrast nod like receptors (NLRs) are cytosolic and respond to a diverse range of PAMPs and DAMPs. Nod like receptors represent a second line of defense against pathogens that evade cell surface and endocytic PRRs. The interaction of pathogen derived, or “danger” associated, factors with TLR and NLR ligands leads to a state of DC maturation resulting in increased DC interaction with other immune cells and promotion of T cell and natural killer cell activation11.

Interleukin-1β is a crucial component of the host defense against infection. Upon recognition of a microorganism, the highly proinflammatory cytokine, IL-1β, is secreted and functions as a chemo attractant and activator of innate and adaptive immune cells. In vivo IL-1β is largely responsible for the acute phase response including fever and inflammatory cytokine synthesis12.

Most NLRs contain a C terminal leucine rich repeat domain that is thought to function in ligand sensing, a central nucleotide binding domain (NACHT) that is important for NLRP3 oligomerization, and an N terminal effector domain (PYD in NLRP3) that mediates signal transduction to downstream targets through protein protein interactions. The NLRP3 protein defines the most intensely studied inflammasome complex. This protein is a member of the NLR family and has the ability to form a multi molecular protein complex composed of NLRP3, the adaptor protein PYCARD (also known as ASC), and ICE. Upon inflammasome activation PYCARD binds to NLRP3 N terminal domains and recruits ICE via caspase activation and recruitment domain (CARD) domains. Interleukin-1 converting enzyme is initially generated as a zymogen containing a CARD motif at its N-terminus. Inflammasome formation results in bringing two ICE molecules sufficiently close to induce their autocatalytic activation. The inflammasome complex is necessary for activating ICE thus allowing it to convert cytoplasmic pro-IL-1β to mature cytokine.

Successful secretion of IL-1β in DCs requires sensing of two different and independent danger signals. First, TLR sensing of PAMPs, DAMPs, or cytokine signaling (TNFα or IL-1β) causes an upregulation of cytoplasmic pro-IL-1β protein expression. A second, often different, signal is required for inflammasome complex formation upstream of ICE maturation. A few inflammasome stimulating signals include bacterial membrane pore forming toxins (such as nigericin), lysosomal disrupting crystals (such as monosodium urate crystals, MSU), and extracellular ATP. The upstream mechanism leading to NLRP3 inflammasome activation by these diverse activators is unclear. Studies investigating signaling upstream of inflammasome formation proposes that intracellular events, such as induction of hypokalemia or reactive oxygen species (ROS) indirectly activate the inflammasome13-28.

Amongst the different viral activators of the NLRP3 inflammasome is influenza, which provides both the primary and secondary signal required for IL-1β secretion3,29-33 . Using mouse NLRP3 knockout models it was found that IL-1β secretion in DCs is NLRP3 dependent32. Additionally, NLRP3 knockout mice attracted fewer leukocytes to the site of infection and experienced higher mortality2,5. Two recent papers suggest a mechanism for NLRP3 inflammasome activation during Influenza virus infection; first, priming through recognition of viral RNA by TLR7 or TLR8 (depending on TLR expression of the responding cell) or through sensing of commensal bacteria by other TLRs to induce cytoplasmic pro-IL-1β expression, followed by a second signal, activation of NLRP3 inflammasome formation by viral ion channel protein M2 on the trans Golgi network33,34. In the latter step, triggering of the NLRP3 inflammasome is accomplished by disturbance of the intracellular ionic milieu leading to ROS production, which is, simply, sensed by NLPR3 as a signal to form the inflammasome. However, the precise mechanism of inflammasome activation upstream of ICE activity during Influenza infection still remains unclear.

This work describes a technique valuable for studying the NLRP3 inflammasome in human moDCs that can be used as a foundation for further investigation of the pathway underlying DC based IL-1β secretion in response to TLR8 ligation with R848 followed by activation of the inflammasome by a well known activator of NLRP3, nigericin. Variations of this method can be used with other cell types including, but not limited to: monocytes, macrophages, other DC subsets, and epithelial cells.

Protocol

Ethics Statement: Research samples are obtained and stored for research with donors’ consent. All samples should be coded or anonymized prior to use. This protocol follows the guidelines of our Institutional Review Board. 1. Differentiation of Human Peripheral Blood Monocytes into Monocyte Derived Dendritic Cells. Note: Human buffy coats serve as the source of human peripheral blood cells (PBMCs) and were obtained from the New York Blood Center (New York, NY). B…

Representative Results

These techniques measure TLR8 priming with R848. Intracellular cytokine staining for pro-IL-1β allows for microscopy and FACs readouts from CD14-CD11c+ moDCs. Both techniques can be quantified relative to a non primed, or resting, cell control as well as an isotype control (Figures 1 and 2). Percent of pro-IL-1β+ staining cells is multiplied by the geometric median of this population to provide the median fluorescent intensity (…

Discussion

Inflammatory cytokines are integral in steering the innate and adaptive immune response to fight viral infection. Secreted IL-1β has been shown to increase during Influenza infection3,43,44 . The precise mechanisms by which these cytokines are processed in response to viral recognition in human dendritic cells are not fully understood. Myeloid DC isolation kits are expensive and time consuming. Isolation kits and FAC sorting may unintentionally stress or activat…

Offenlegungen

The authors have nothing to disclose.

Acknowledgements

The authors would like to acknowledge Olivier Manches, Ph.D., Davor Frleta, Ph.D., and Meagan O’Brien, M.D. for their support and feedback. This research was supported by the National Institute of Allergy and Infectious Disease and completed with funding from NIH grants Ruth L. Kirschstein National Research Service Awards for Individual Predoctoral Fellowships (F31) to Promote Diversity in Health-Related Research (AI089030) and RO1 (AI081848).

Materials

Name of Reagent/ Equipment Company Catalog Number Comments/Description
IL-4 R&D
GM-CSF Genzyme NDC 58468-0180-2 We acquire this item through our local pharmacy with a prescription
RPMI 1640 with L-glutamine Cellgro 10-040-CV
Peripheral blood mononuclear cells New York Blood Center PBMCs were isolated from the blood of healthy donors
12-well tissue culture plates Sigma-Aldrich 3516
96-well round bottom tissue culture plates Sigma-Aldrich 3799
α-IL-1β-FITC R&D IC201F
FITC isotype control Miltenyi Biotec 130-092-213
α-β-Tubulin Santa Cruz SC-9014
α-IL-1β R&D mab201
PVDF Immobilon-FL membrane Millipore IPFL00010
gradient 4-12 % polycrylamide gel Bio Rad 161-1159
laemmli sample buffer Bio Rad 161-0737
BSA Equitech Bio Inc 30% solution sterile/filtered
PFA Electron Microscopy Sciences 15710 16% solution
human inflammatory cytokine bead array kit BD 551811
nigericin Invivogen tlrl-nig
R848 3M Corp.
α-CD14 BD 340436
α-CD11c BD 555392
β-mercaptoethanol Sigma-Aldrich M6250-10ML
TBS On site stock room
Tween-20 Sigma-Aldrich P2287-100mL
Nunc EasYFlask 225cm2, Filter Cap, 70mL working volume, 30/Cs Thermo Scientific 159933
20 μM Sterile Disposable Filter Units Thermo Scientific 569-0020
Gentamicin Invitrogen 15750060
Hepes Invitrogen 15630080
goat α-mouse IRDye 800CW Licor 926-32210
donkey α-rabbit IRDye 680RD Licor 926-68073
Spectra multicolor broad range protein ladder Thermo Scientific 26634
Tris Glycine SDS 10x Bio Rad 1610732
Tris Glycine 10x Bio Rad 161-0734
Methanol – 4L Fisher Scientific A433P-4
Prolong Gold antifade Reagent with DAPI Life Technologies P-36931
8 chamber polystyrene vessel tissue culture treated glass slide BD Falcon 354108
Poly-L-Lysine Sigma P4707

Referenzen

  1. Durrant, D. M., Robinette, M. L., Klein, R. S. IL-1R1 is required for dendritic cell-mediated T cell reactivation within the CNS during West Nile virus encephalitis. The Journal of Experimental Medicine. 210, 503-516 (2013).
  2. Thomas, P. G., et al. The intracellular sensor NLRP3 mediates key innate and healing responses to influenza A virus via the regulation of caspase-1. Immunity. 30, 566-575 (2009).
  3. Schmitz, N., Kurrer, M., Bachmann, M. F., Kopf, M. Interleukin-1 is responsible for acute lung immunopathology but increases survival of respiratory influenza virus infection. Journal of Virology. 79, 6441-6448 (2005).
  4. Pang, I. K., Ichinohe, T., Iwasaki, A. IL-1R signaling in dendritic cells replaces pattern-recognition receptors in promoting CD8(+) T cell responses to influenza A virus. Nat Immunol. 14, 246-253 (2013).
  5. Allen, I. C., et al. The NLRP3 inflammasome mediates in vivo innate immunity to influenza A virus through recognition of viral RNA. Immunity. 30, 556-565 (2009).
  6. Bauernfeind, F. G., et al. Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J Immunol. 183, 787-791 (2009).
  7. Zabel, F., Kundig, T. M., Bachmann, M. F. Virus-induced humoral immunity: on how B cell responses are initiated. Current Opinion in Virology. , (2013).
  8. Steinman, R. M. Lasker Basic Medical Research Award. Dendritic cells: versatile controllers of the immune system. Nature Medicine. 13, 1155-1159 (2007).
  9. Bhardwaj, N., et al. Influenza virus-infected dendritic cells stimulate strong proliferative and cytolytic responses from human CD8+ T cells. The Journal of Clinical Investigation. 94, 797-807 (1994).
  10. Sheng, K. C., Day, S., Wright, M. D., Stojanovska, L., Apostolopoulos, V. Enhanced Dendritic Cell-Mediated Antigen-Specific CD4+ T Cell Responses: IFN-Gamma Aids TLR Stimulation. Journal of Drug Delivery. 2013, (2013).
  11. Pohl, C., Shishkova, J., Schneider-Schaulies, S. Viruses and dendritic cells: enemy mine. Cellular Microbiology. 9, 279-289 (2007).
  12. Dinarello, C. A. Cytokines as mediators in the pathogenesis of septic shock. Curr Top Microbiol Immunol. 216, 133-165 (1996).
  13. Petrilli, V., et al. Activation of the NALP3 inflammasome is triggered by low intracellular potassium concentration. Cell Death and Differentiation. 14, 1583-1589 (2007).
  14. Hussen, J., Duvel, A., Koy, M., Schuberth, H. J. Inflammasome activation in bovine monocytes by extracellular ATP does not require the purinergic receptor P2X7. Developmental and Comparative Immunology. 38, 312-320 (2012).
  15. Rajamaki, K., et al. Extracellular Acidosis Is a Novel Danger Signal Alerting Innate Immunity via the NLRP3 Inflammasome. The Journal of Biological Chemistry. 288, 13410-13419 (2013).
  16. Ayna, G., et al. ATP release from dying autophagic cells and their phagocytosis are crucial for inflammasome activation in macrophages. PLoS One. , (2012).
  17. Vyleta, M. L., Wong, J., Magun, B. E. Suppression of ribosomal function triggers innate immune signaling through activation of the NLRP3 inflammasome. PLoS One. 7, (2012).
  18. Lacroix-Lamande, S., et al. Downregulation of the Na/K-ATPase pump by leptospiral glycolipoprotein activates the NLRP3 inflammasome. J Immunol. 188, 2805-2814 (2012).
  19. Segovia, J., et al. TLR2/MyD88/NF-kappaB pathway, reactive oxygen species, potassium efflux activates NLRP3/ASC inflammasome during respiratory syncytial virus infection. PLoS One. 7, (2012).
  20. Hamon, M. A., Cossart, P. K. K+ efflux is required for histone H3 dephosphorylation by Listeria monocytogenes listeriolysin O and other pore-forming toxins. Infection and Immunity. 79, 2839-2846 (2011).
  21. Schorn, C., et al. Sodium overload and water influx activate the NALP3 inflammasome. The Journal of Biological Chemistry. 286, 35-41 (2011).
  22. Said-Sadier, N., Padilla, E., Langsley, G., Ojcius, D. M. Aspergillus fumigatus stimulates the NLRP3 inflammasome through a pathway requiring ROS production and the Syk tyrosine kinase. PLoS One. 5, (2010).
  23. Arlehamn, C. S., Petrilli, V., Gross, O., Tschopp, J., Evans, T. J. The role of potassium in inflammasome activation by bacteria. The Journal of Biological Chemistry. 285, 10508-10518 (2010).
  24. Chu, J., et al. Cholesterol-dependent cytolysins induce rapid release of mature IL-1beta from murine macrophages in a NLRP3 inflammasome and cathepsin B-dependent manner. Journal of Leukocyte Biology. 86, 1227-1238 (2009).
  25. Silverman, W. R., et al. The pannexin 1 channel activates the inflammasome in neurons and astrocytes. The Journal of Biological Chemistry. 284, 18143-18151 (2009).
  26. Piccini, A., et al. ATP is released by monocytes stimulated with pathogen-sensing receptor ligands and induces IL-1beta and IL-18 secretion in an autocrine way. Proc Natl Acad Sci U S A. 105, 8067-8072 (2008).
  27. Wickliffe, K. E., Leppla, S. H., Moayeri, M. Anthrax lethal toxin-induced inflammasome formation and caspase-1 activation are late events dependent on ion fluxes and the proteasome. Cellular Microbiology. 10, 332-343 (2008).
  28. Franchi, L., Kanneganti, T. D., Dubyak, G. R., Nunez, G. Differential requirement of P2X7 receptor and intracellular K+ for caspase-1 activation induced by intracellular and extracellular bacteria. The Journal of Biological Chemistry. 282, 18810-18818 (2007).
  29. Owen, D. M., Gale, M. Fighting the flu with inflammasome signaling. Immunity. 30, 476-478 (2009).
  30. Pang, I. K., Iwasaki, A. Inflammasomes as mediators of immunity against influenza virus. Trends in Immunology. 32, 34-41 (2011).
  31. Kanneganti, T. D., et al. Critical role for Cryopyrin/Nalp3 in activation of caspase-1 in response to viral infection and double-stranded RNA. The Journal of Biological Chemistry. 281, 36560-36568 (2006).
  32. Ichinohe, T., Lee, H. K., Ogura, Y., Flavell, R., Iwasaki, A. Inflammasome recognition of influenza virus is essential for adaptive immune responses. The Journal of Experimental Medicine. 206, 79-87 (2009).
  33. Ichinohe, T., Pang, I. K., Iwasaki, A. Influenza virus activates inflammasomes via its intracellular M2 ion channel. Nat Immunol. 11, 404-410 (2010).
  34. Ichinohe, T., et al. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc Natl Acad Sci U S A. 108, 5354-5359 (2011).
  35. O’Neill, D. W., Bhardwaj, N. Differentiation of peripheral blood monocytes into dendritic cells. Current Protocols in Immunology. 22, (2005).
  36. Sabado, R. L., Miller, E., Spadaccia, M., Vengco, I., Hasan, F., Bhardwaj, N. Preparation of Tumor Antigen-loaded Mature Dendritic Cells for Immunotherapy. J. Vis. Exp. (78), (2013).
  37. Rubartelli, A., Cozzolino, F., Talio, M., Sitia, R. A novel secretory pathway for interleukin-1 beta, a protein lacking a signal sequence. The EMBO Journal. 9, 1503-1510 (1990).
  38. Ritchie, H., Booth, N. A. Secretion of plasminogen activator inhibitor 2 by human peripheral blood monocytes occurs via an endoplasmic reticulum-golgi-independent pathway. Experimental Cell Research. 242, 439-450 (1998).
  39. Lamoreaux, L., Roederer, M., Koup, R. Intracellular cytokine optimization and standard operating procedure. Nature Protocols. 1, 1507-1516 (2006).
  40. He, H., Courtney, A. N., Wieder, E., Sastry, K. J. Multicolor Flow Cytometry Analyses of Cellular Immune Response in Rhesus Macaques. J Vis Exp. 38, (2010).
  41. Mahmood, T., Yang, P. C. Western blot: technique, theory, and trouble shooting. North American journal of Medical Sciences. 4, 429-434 (2012).
  42. Alegria-Schaffer, A., Lodge, A., Vattem, K. Performing and optimizing Western blots with an emphasis on chemiluminescent detection. Methods in Enzymology. 463, 573-599 (2009).
  43. Hennet, T., Ziltener, H. J., Frei, K., Peterhans, E. A kinetic study of immune mediators in the lungs of mice infected with influenza A virus. J Immunol. 149, 932-939 (1992).
  44. Pirhonen, J., Sareneva, T., Kurimoto, M., Julkunen, I., Matikainen, S. Virus infection activates IL-1 beta and IL-18 production in human macrophages by a caspase-1-dependent pathway. J Immunol. 162, 7322-7329 (1999).
  45. Anderson, J., Gustafsson, K., Himoudi, N. Licensing of killer dendritic cells in mouse and humans: functional similarities between IKDC and human blood gammadelta T-lymphocytes. Journal of Immunotoxicology. 9, 259-266 (2012).
  46. Waithman, J., Mintern, J. D. Dendritic cells and influenza A virus infection. Virulence. 3, 603-608 (2012).
check_url/de/51284?article_type=t

Play Video

Diesen Artikel zitieren
Fernandez, M. V., Miller, E. A., Bhardwaj, N. Activation and Measurement of NLRP3 Inflammasome Activity Using IL-1β in Human Monocyte-derived Dendritic Cells. J. Vis. Exp. (87), e51284, doi:10.3791/51284 (2014).

View Video