Nasal Lipopolysaccharide Challenge and Cytokine Measurement Reflects Innate Mucosal Immune Responsiveness

Jaideep Dhariwal, Jeremy Kitson, Reema E Jones, Grant Nicholson, Tanushree Tunstall, Ross P Walton, Grace Francombe, Jane Gilbert, Andrew J Tan, Robert Murdoch, Onn Min Kon, Peter J Openshaw, Trevor T Hansel, Jaideep Dhariwal, Jeremy Kitson, Reema E Jones, Grant Nicholson, Tanushree Tunstall, Ross P Walton, Grace Francombe, Jane Gilbert, Andrew J Tan, Robert Murdoch, Onn Min Kon, Peter J Openshaw, Trevor T Hansel

Abstract

Background: Practical methods of monitoring innate immune mucosal responsiveness are lacking. Lipopolysaccharide (LPS) is a component of the cell wall of Gram negative bacteria and a potent activator of Toll-like receptor (TLR)-4. To measure LPS responsiveness of the nasal mucosa, we administered LPS as a nasal spray and quantified chemokine and cytokine levels in mucosal lining fluid (MLF).

Methods: We performed a 5-way cross-over, single blind, placebo-controlled study in 15 healthy non-atopic subjects (n = 14 per protocol). Doses of ultrapure LPS (1, 10, 30 or 100μg/100μl) or placebo were administered by a single nasal spray to each nostril. Using the recently developed method of nasosorption with synthetic adsorptive matrices (SAM), a series of samples were taken. A panel of seven cytokines/chemokines were measured by multiplex immunoassay in MLF. mRNA for intercellular cell adhesion molecule-1 (ICAM-1) was quantified from nasal epithelial curettage samples taken before and after challenge.

Results: Topical nasal LPS was well tolerated, causing no symptoms and no visible changes to the nasal mucosa. LPS induced dose-related increases in MLF levels of IL-1β, IL-6, CXCL8 (IL-8) and CCL3 (MIP-1α) (AUC at 0.5 to 10h, compared to placebo, p<0.05 at 30 and 100μg LPS). At 100μg LPS, IL-10, IFN-α and TNF-α were also increased (p<0.05). Dose-related changes in mucosal ICAM-1 mRNA were also seen after challenge, and neutrophils appeared to peak in MLF at 8h. However, 2 subjects with high baseline cytokine levels showed prominent cytokine and chemokine responses to relatively low LPS doses (10μg and 30μg LPS).

Conclusions: Topical nasal LPS causes dose-dependent increases in cytokines, chemokines, mRNA and cells. However, responsiveness can show unpredictable variations, possibly because baseline innate tone is affected by environmental factors. We believe that this new technique will have wide application in the study of the innate immune responses of the respiratory mucosa.

Key messages: Ultrapure LPS was used as innate immune stimulus in a human nasal challenge model, with serial sampling of nasal mucosal lining fluid (MLF) by nasosorption using a synthetic absorptive matrix (SAM), and nasal curettage of mucosal cells. A dose response could be demonstrated in terms of levels of IL-1β, IL-6, CXCL8 and CCL3 in MLF, as well as ICAM-1 mRNA in nasal curettage specimens, and levels of neutrophils in nasal lavage. Depending on higher baseline levels of inflammation, there were occasional magnified innate inflammatory responses to LPS.

Trial registration: Clinical Trials.gov NCT02284074.

Conflict of interest statement

Competing Interests: This study was partly funded by Glaxo SmithKline, the employer of JK, REJ, GF, JG and RM. In the past 5 years TTH has been Principal Investigator for respiratory clinical studies carried out at Imperial College, that have been sponsored by Pharmaceutical Companies: Novartis, Merck, GlaxoSmithKline (GSK) and Dainippon-Sumitomo. TTH has received lecture fees from Novartis, Merck, GSK and Dainippon-Sumitomo. TTH is involved in setting up a spin-off company with Imperial Innovations called Mucosal Diagnostics, and has received support to attend Respiratory Conferences from Boehringer Ingelheim. There are no patents, products in development or marketed products to declare. This does not alter the authors' adherence to all the PLOS ONE policies on sharing data and materials, as detailed online in the guide for authors.

Figures

Fig 1. Consort Flow Chart.
Fig 1. Consort Flow Chart.
Fig 2. Nasal lipopolysaccharide challenge study design…
Fig 2. Nasal lipopolysaccharide challenge study design and procedures.
14 subjects completed the entire protocol (per protocol), while 1 subject only received nasal challenges at placebo and LPS. TNSS–total nasal symptom score. AE–adverse events
Fig 3. Median levels of chemokines and…
Fig 3. Median levels of chemokines and cytokines (pg/ml) in nasal mucosal lining fluid (MLF) are shown after nasal spray challenge with 4 different doses of LPS and a placebo (see key below).
Lower limits of detection (LLOD) for cytokines and chemokines were as follows: IL-1β (5.0pg/ml), IL-6 (5.0pg/ml), CXCL8/IL-8 (10.0pg/ml), CCL3/MIP-1α (5.0pg/ml), CCL2/MCP-1 (25.0pg/ml), IL-10 (5.0pg/ml). Time -0.5h refers to nasosorption performed 30 min prior to topical nasal challenge, and this varies considerably between individuals due to variations in individual’s microbial flora and mucosal immune responses. Nasal lavage was performed after nasosorption at -0.5h, but prior to nasal challenge, in order to partially wash the nose free of baseline inflammatory microbes and mediators, but causes detectable levels of inflammatory mediators at 0.5h to decrease markedly from -0.5h. There is a tendency for nasosorption levels of mediators to gradually increase from 0.5h to 10h in the placebo arm, but levels of inflammatory cytokines and chemokines after LPS are generally higher.
Fig 4. Levels of interleukins (IL) in…
Fig 4. Levels of interleukins (IL) in nasosorption eluates presented as area under the curve (AUC) for times 0.5-10h in relation to topical nasal challenge (n = 14 or 15).
AUC is derived from from 0.5 since the baseline sample at -0.5h is pre-nasal lavage and is highly variable due to difference in individuals in terms of nasal microbes and inflammatory mediators. A) IL-1β as medians with quartiles; B) IL-6 as medians with quartiles; C) IL-1β AUC data for individual subjects; D) IL- AUC data for individual subjects. Note the idiosyncratic large responses of subject 2009 after nasal challenge with 10μg LPS, and subject 3101 after nasal challenge with 30μg LPS, these responses being too large to be presented on the y axis.
Fig 5. Idiosyncratic large nasal responses to…
Fig 5. Idiosyncratic large nasal responses to lipopolysaccharide (LPS) on 2 occasions.
Raw data for levels of chemokines and cytokines (pg/ml) in nasal mucosal lining fluid (MLF). Data is presented for individual subjects: A. Subject 2009 after nasal challenge with 10μg LPS. B. Subject 3101 after nasal challenge with 30μg LPS. Note that baseline levels of cytokines and chemokines at -0.5h are markedly raised, and come down to only a small extent after nasal lavage at 0.5h. It is suggested that these individuals may have had a non-symptomatic alteration in their nasal microbial flora prior to LPS challenge on these opccasions: possibly the individuals had a subclinical viral infection or bacterial colonisation.
Fig 6. Nasal lavage leukocyte counts (medians…
Fig 6. Nasal lavage leukocyte counts (medians expressed as cells/ml) following nasal LPS challenge.
Cells were counted in a modified Neubauer chamber, and a leukocyte differential performed on a stained cytospin observed under a light microscope.
Fig 7. ICAM-1 mRNA abundance within nasal…
Fig 7. ICAM-1 mRNA abundance within nasal curettage samples after nasal LPS challenge, Data is shown as means with 95% confidence intervals.
Relative abundance of expression with Reverse Transcriptase quantitative Polymerase Chain Reaction (RT-qPCR) using normalized data and (glyceraldehyde-3-phosphate dehydrogenase) (GAPDH) as a housekeeper gene (see Methods).

References

    1. O'Byrne PM, Gauvreau GM, Brannan JD (2009) Provoked models of asthma: what have we learnt? Clin Exp Allergy 39: 181–192. CEA3172 [pii];10.1111/j.1365-2222.2008.03172.x
    1. Holgate ST (2012) Innate and adaptive immune responses in asthma. Nat Med 18: 673–683. nm.2731 [pii];10.1038/nm.2731
    1. Pollard AJ, Savulescu J, Oxford J, Hill AV, Levine MM, Lewis DJ, Read RC, Graham DY, Sun W, Openshaw P, Gordon SB (2012) Human microbial challenge: the ultimate animal model. Lancet Infect Dis 12: 903–905. S1473-3099(12)70292-X [pii];10.1016/S1473-3099(12)70292-X
    1. Yan N, Chen ZJ (2012) Intrinsic antiviral immunity. Nat Immunol 13: 214–222. ni.2229 [pii];10.1038/ni.2229
    1. Holt PG, Sly PD (2012) Viral infections and atopy in asthma pathogenesis: new rationales for asthma prevention and treatment. Nat Med 18: 726–735. nm.2768 [pii];10.1038/nm.2768
    1. Hansel TT, Johnston SL, Openshaw PJ (2013) Microbes and mucosal immune responses in asthma. Lancet. S0140-6736(12)62202-8 [pii];10.1016/S0140-6736(12)62202-8
    1. Kawai T, Akira S (2011) Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 34: 637–650. S1074-7613(11)00190-7 [pii];10.1016/j.immuni.2011.05.006
    1. Netea MG, Wijmenga C, O'Neill LA (2012) Genetic variation in Toll-like receptors and disease susceptibility. Nat Immunol 13: 535–542. ni.2284 [pii];10.1038/ni.2284
    1. Simpson A, Martinez FD (2010) The role of lipopolysaccharide in the development of atopy in humans. Clin Exp Allergy 40: 209–223. CEA3391 [pii];10.1111/j.1365-2222.2009.03391.x
    1. Imai Y, Kuba K, Neely GG, Yaghubian-Malhami R, Perkmann T, van LG, Ermolaeva M, Veldhuizen R, Leung YH, Wang H, Liu H, Sun Y, Pasparakis M, Kopf M, Mech C, Bavari S, Peiris JS, Slutsky AS, Akira S, Hultqvist M, Holmdahl R, Nicholls J, Jiang C, Binder CJ, Penninger JM (2008) Identification of oxidative stress and Toll-like receptor 4 signaling as a key pathway of acute lung injury. Cell 133: 235–249. S0092-8674(08)00340-1 [pii];10.1016/j.cell.2008.02.043
    1. Shirey KA, Lai W, Scott AJ, Lipsky M, Mistry P, Pletneva LM, Karp CL, McAlees J, Gioannini TL, Weiss J, Chen WH, Ernst RK, Rossignol DP, Gusovsky F, Blanco JC, Vogel SN (2013) The TLR4 antagonist Eritoran protects mice from lethal influenza infection. Nature. nature12118 [pii];10.1038/nature12118
    1. Trompette A, Divanovic S, Visintin A, Blanchard C, Hegde RS, Madan R, Thorne PS, Wills-Karp M, Gioannini TL, Weiss JP, Karp CL (2009) Allergenicity resulting from functional mimicry of a Toll-like receptor complex protein. Nature 457: 585–588. nature07548 [pii];10.1038/nature07548
    1. Hammad H, Chieppa M, Perros F, Willart MA, Germain RN, Lambrecht BN (2009) House dust mite allergen induces asthma via Toll-like receptor 4 triggering of airway structural cells. Nat Med 15: 410–416. nm.1946 [pii];10.1038/nm.1946
    1. Braun-Fahrlander C, Riedler J, Herz U, Eder W, Waser M, Grize L, Maisch S, Carr D, Gerlach F, Bufe A, Lauener RP, Schierl R, Renz H, Nowak D, von ME (2002) Environmental exposure to endotoxin and its relation to asthma in school-age children. N Engl J Med 347: 869–877. 10.1056/NEJMoa020057347/12/869 [pii].
    1. Heederik D, von ME (2012) Does diversity of environmental microbial exposure matter for the occurrence of allergy and asthma? J Allergy Clin Immunol 130: 44–50. S0091-6749(12)00255-2 [pii];10.1016/j.jaci.2012.01.067
    1. Pillay J, Ramakers BP, Kamp VM, Lo ATL, Lam SW, Hietbrink F, Leenen LP, Tool AT, Pickkers P, Koenderman L (2010) Functional heterogenecity and differential priming of circulating neutrophils in human experimental endotoxemia. J Leukoc Biol 88: 10.1189/jlb.1209793
    1. Hernandez ML, Herbst M, Lay JC, Alexis NE, Brickey WJ, Ting JP, Zhou H, Peden DB (2012) Atopic asthmatic patients have reduced airway inflammatory cell recruitment after inhaled endotoxin challenge compared with healthy volunteers. J Allergy Clin Immunol 130: 869–876. S0091-6749(12)00864-0 [pii];10.1016/j.jaci.2012.05.026
    1. Schaumann F, Muller M, Braun A, Luettig B, Peden DB, Hohlfeld JM, Krug N (2008) Endotoxin augments myeloid dendritic cell influx into the airways in patients with allergic asthma. Am J Respir Crit Care Med 177: 1307–1313. 200706-870OC [pii];10.1164/rccm.200706-870OC
    1. Peden DB, Tucker K, Murphy P, Newlin-Clapp L, Boehlecke B, Hazucha M, Bromberg P, Reed W (1999) Eosinophil influx to the nasal airway after local, low-level LPS challenge in humans. J Allergy Clin Immunol 104: 388–394.
    1. Danuser B, Rebsamen H, Weber C, Krueger H (2000) Lipopolysaccharide-induced nasal cytokine response: a dose-response evaluation. Eur Arch Otorhinolaryngol 257: 527–532.
    1. Nita I, Hollander C, Westin U, Janciauskiene S-M (2005) Prolastin, a pharmaceutical preparation of purified human alpha1-antitrypsin, blocks endotoxin-mediated cytokine release. Respiratory Research 6: 1–11.
    1. Doreswamy V, Alexis NE, Zhou H, Peden DB (2011) Nasal PMN response to repeated challenge with endotoxin in healthy volunteers. Inhal Toxicol 23: 142–147. 10.3109/08958378.2011.553247
    1. Ekman AK, Fransson M, Rydberg C, Adner M, Cardell LO (2009) Nasal challenge with LPS stimulates the release of macrophage inflammatory protein 1alpha. Int Arch Allergy Immunol 149: 154–160. 000189199 [pii];10.1159/000189199
    1. Virtala R, Ekman AK, Jansson L, Westin U, Cardell LO (2012) Airway inflammation evaluated in a human nasal lipopolysaccharide challenge model by investigating the effect of a CXCR2 inhibitor. Clin Exp Allergy 42: 590–596. 10.1111/j.1365-2222.2011.03921.x
    1. Bachar O, Gustafsson J, Jansson L, Adner M, Cardell LO (2007) Lipopolysaccharide administration to the allergic nose contributes to lower airway inflammation. Clin Exp Allergy 37: 1773–1780. CEA2842 [pii];10.1111/j.1365-2222.2007.02842.x
    1. Ekman AK, Virtala R, Fransson M, Adner M, Benson M, Jansson L, Cardell LO (2012) Systemic Up-Regulation of TLR4 Causes Lipopolysaccharide-Induced Augmentation of Nasal Cytokine Release in Allergic Rhinitis. Int Arch Allergy Immunol 159: 6–14. 000335196 [pii];10.1159/000335196
    1. Folsgaard NV, Chawes BL, Rasmussen MA, Bischoff AL, Carson CG, Stokholm J, Pedersen L, Hansel TT, Bonnelykke K, Brix S, Bisgaard H (2011) Maternal atopic skewing of the neonatal nasal cytokine signature. Am J Resp Crit Care Med.
    1. Chawes BL, Edwards MJ, Shamji B, Walker C, Nicholson GC, Tan AJ, Folsgaard NV, Bonnelykke K, Bisgaard H, Hansel TT (2010) A novel method for assessing unchallenged levels of mediators in nasal epithelial lining fluid. J Allergy Clin Immunol 125: 1387–1389. S0091-6749(10)00172-7 [pii];10.1016/j.jaci.2010.01.039
    1. Nicholson GC, Kariyawasam HH, Tan AJ, Hohlfeld JM, Quinn D, Walker C, Rodman D, Westwick J, Jurcevic S, Kon OM, Barnes PJ, Krug N, Hansel TT (2011) The effects of an anti-IL-13 mAb on cytokine levels and nasal symptoms following nasal allergen challenge. J Allergy Clin Immunol 128: 800–807. S0091-6749(11)00770-6 [pii];10.1016/j.jaci.2011.05.013
    1. Scadding GW, Calderon MA, Bellido V, Koed GK, Nielsen NC, Lund K, Togias A, Phippard D, Turka LA, Hansel TT, Durham SR, Wurtzen PA (2012) Optimisation of grass pollen nasal allergen challenge for assessment of clinical and immunological outcomes. J Immunol Methods 384: 25–32. S0022-1759(12)00174-3 [pii];10.1016/j.jim.2012.06.013
    1. Dreskin SC, Dale SN, Foster SM, Martin D, Buchmeier A, Nelson HS (2002) Measurement of changes in mRNA for IL-5 in noninvasive scrapings of nasal epithelium taken from patients undergoing nasal allergen challenge. J Immunol Methods 268: 189–195. S0022175902002065 [pii].
    1. Proud D, Turner RB, Winther B, Wiehler S, Tiesman JP, Reichling TD, Juhlin KD, Fulmer AW, Ho BY, Walanski AA, Poore CL, Mizoguchi H, Jump L, Moore ML, Zukowski CK, Clymer JW (2008) Gene expression profiles during in vivo human rhinovirus infection: insights into the host response. Am J Respir Crit Care Med 178: 962–968. 200805-670OC [pii];10.1164/rccm.200805-670OC
    1. Wang SZ, Ma FM, Zhao JD (2012) Expressions of nuclear factor-kappa B p50 and p65 and their significance in the up-regulation of intercellular cell adhesion molecule-1 mRNA in the nasal mucosa of allergic rhinitis patients. Eur Arch Otorhinolaryngol. 10.1007/s00405-012-2136-y
    1. Greiff L, Pipkorn U, Alkner U, Persson CG (1990) The 'nasal pool' device applies controlled concentrations of solutes on human nasal airway mucosa and samples its surface exudations/secretions. Clin Exp Allergy 20: 253–259.
    1. Virtala R, Ekman AK, Jansson L, Westin U, Cardell LO (2012) Airway inflammation evaluated in a human nasal lipopolysaccharide challenge model by investigating the effect of a CXCR2 inhibitor. Clin Exp Allergy 42: 590–596. 10.1111/j.1365-2222.2011.03921.x
    1. Fransson M, Adner M, Uddman R, Cardell LO (2007) Lipopolysaccharide-induced down-regulation of uteroglobin in the human nose. Acta Otolaryngol 127: 285–291. 771160953 [pii];10.1080/00016480600801340
    1. Riechelmann H, Deutschle T, Friemel E, Gross HJ, Bachem M (2003) Biological markers in nasal secretions. Eur Respir J 21: 600–605.
    1. Erin EM, Zacharasiewicz AS, Nicholson GC, Higgins LA, Tan AJ, Williams TJ, Murdoch RD, Durham SR, Barnes PJ, Hansel TT (2005) Topical corticosteroids inhibit IL-4, IL-5, and IL-13 levels in nasal secretions following local allergen challenge. Clin Exp Allergy 35: 1608–1614.
    1. Hentschel J, Muller U, Doht F, Fischer N, Boer K, Sonnemann J, Hipler C, Hunniger K, Kurzai O, Markert UR, Mainz JG (2014) Influences of nasal lavage collection-, processing- and storage methods on inflammatory markers—evaluation of a method for non-invasive sampling of epithelial lining fluid in cystic fibrosis and other respiratory diseases. J Immunol Methods 404: 41–51. S0022-1759(13)00367-0 [pii];10.1016/j.jim.2013.12.003
    1. Habibzay M, Saldana JI, Goulding J, Lloyd CM, Hussell T (2012) Altered regulation of Toll-like receptor responses impairs antibacterial immunity in the allergic lung. Mucosal Immunol 5: 524–534. mi201228 [pii];10.1038/mi.2012.28
    1. Tan AM, Chen HC, Pochard P, Eisenbarth SC, Herrick CA, Bottomly HK (2010) TLR4 signaling in stromal cells is critical for the initiation of allergic Th2 responses to inhaled antigen. J Immunol 184: 3535–3544. jimmunol.0900340 [pii];10.4049/jimmunol.0900340

Source: PubMed

3
購読する