Hypertonic saline attenuates the cytokine-induced pro-inflammatory signature in primary human lung epithelia

Sanchayita Mitra, Daran Schiller, Cameron Anderson, Fabia Gamboni, Angelo D'Alessandro, Margeurite Kelher, Christopher C Silliman, Anirban Banerjee, Kenneth L Jones, Sanchayita Mitra, Daran Schiller, Cameron Anderson, Fabia Gamboni, Angelo D'Alessandro, Margeurite Kelher, Christopher C Silliman, Anirban Banerjee, Kenneth L Jones

Abstract

Trauma/hemorrhagic shock is a complex physiological phenomenon that leads to dysregulation of many molecular pathways. For over a decade, hypertonic saline (HTS) has been used as an alternative resuscitation fluid in the setting of trauma/hemorrhagic shock. In addition to restoring circulating volume within the vascular space, studies have shown a positive immunomodulatory effect of HTS. Targeted studies have shown that HTS affects the transcription of several pro-inflammatory cytokines by inhibiting the NF-κB-IκB pathway in model cell lines and rats. However, few studies have been undertaken to assess the unbiased effects of HTS on the whole transcriptome. This study was designed to interrogate the global transcriptional responses induced by HTS and provides insight into the underlying molecular mechanisms and pathways affected by HTS. In this study, RNA sequencing was employed to explore early changes in transcriptional response, identify key mediators, signaling pathways, and transcriptional modules that are affected by HTS in the presence of a strong inflammatory stimulus. Our results suggest that primary human small airway lung epithelial cells (SAECS) exposed to HTS in the presence and absence of a strong pro-inflammatory stimulus exhibit very distinct effects on cellular response, where HTS is highly effective in attenuating cytokine-induced pro-inflammatory responses via mechanisms that involve transcriptional regulation of inflammation which is cell type and stimulus specific. HTS is a highly effective anti-inflammatory agent that inhibits the chemotaxis of leucocytes towards a pro-inflammatory gradient and may attenuate the progression of both the innate and adaptive immune response.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. HTS inhibits the expression of…
Fig 1. HTS inhibits the expression of several key inflammatory mediators.
SAECS in P3 were grown in triplicate to 80% confluence in 12-well dishes, stimulated with TNFα, IL-1β, and IFNγ (cytomix) in the presence and absence of hypertonic saline (HTS) for 4 hrs. RNA was harvested and RNAseq performed. HTS decreases the expression of (a) key chemokines and cytokines, (b) Transporters and (c) Kinases which are important effectors of the inflammatory signaling cascade.
Fig 2. HTS inhibits the expression of…
Fig 2. HTS inhibits the expression of chemokines and cytokines that regulate immune cell trafficking and polarization.
SAECS in P4 were treated with cytomix in the presence and absence of HTS for 4 and 8hrs, and RNA isolated for qPCR analysis. Validation of the RNAseq shows that (a) chemokines that affect T-cell, Dendritic cell, and NK cell interaction/migration, (b) monocyte trafficking, (c) IL-1 family cytokines, and Th17 response are all increased due to cytokine treatment, and attenuated by HTS; whereas HTS-specific genes such as IL-11 are upregulated in response to HTS. (*p

Fig 3. HTS inhibits the transcriptional activation…

Fig 3. HTS inhibits the transcriptional activation of IRF1 and STAT1 dependent genes.

In the…

Fig 3. HTS inhibits the transcriptional activation of IRF1 and STAT1 dependent genes.
In the Ingenuity Pathway Analysis of cytomix vs cytomix+HTS, IRF1 and STAT1 were inferred to regulate the expression of several downstream molecules in this study. (a) 12 of 18 genes have expression consistent with upstream inhibition of IRF1 by HTS (Z-score: -2.564; p = 1.05E-10). (b) 26 of 32 genes have expression consistent with STAT1 inhibition when cytomix is added to cells treated with HTS (Z-score: -4.638; p = 3.85E-16). (c) HTS attenuates cytomix-induced luciferase expression in SAECS transiently transfected with IRF1 and STAT1 reporter expression plasmids treated with cytomix for 4hrs in the presence and absence of HTS. (d) The expression of CCL5, a downstream endogenous target of IRF1 and STAT1, is also attenuated by HTS at 4, 6 and 8hrs as determined by ELISA. (*p<0.05; **p<0.01; ***p<0.001; ****p<0.0001).

Fig 4. HTS inhibits chemotaxis of neutrophils…

Fig 4. HTS inhibits chemotaxis of neutrophils and splenocyte-derived T-cells.

SAECS in P3 were cultured…

Fig 4. HTS inhibits chemotaxis of neutrophils and splenocyte-derived T-cells.
SAECS in P3 were cultured for 24 hrs with cytomix in the presence and absence of HTS and the supernatants were harvested and applied to the bottom chamber of the chemotaxis plate. Chemotaxis of neutrophils and splenocyte derived T cells towards the chemotactic gradient was assayed using Incucyte Zoom imaging system. The results show (a) supernatant from cytomix treated cells elicit maximal migration of neutrophils, and this migration was attenuated in supernatants from cytomix + HTS treated cells. Similarly, in a separate experiment (b), HTS inhibited the migration of splenocyte derived T-cells through the matrigel towards the chemotactic gradient. [solid lines indicate the loss of surface area previously occupied by the neutrophils or splenocytes derived T-cells on the top of membrane as they migrate to the reservoir containing the chemoattractant at the bottom of the membrane; dashed lines indicate p values for pairwise comparison of migration of neutrophils or splenocyte at each time point (control vs HTS, control vs cytomix, control vs cyto-HTS: Fig 4a and Fig 4b main panel; cytomix vs cyto–HTS: Fig 4a and Fig 4b inset). FMLP and CCL5 were used as positive control for the migration assays for neutrophil and splenocyte derived T cells respectively.

Fig 5. HTS inhibits CCL5 expression in…

Fig 5. HTS inhibits CCL5 expression in cytokine-pretreated cells.

SAECS in P3 were co-treated or…

Fig 5. HTS inhibits CCL5 expression in cytokine-pretreated cells.
SAECS in P3 were co-treated or pretreated with cytomix ± HTS for 8hrs and HTS was added at 30, 60, 120, 240, and 360 minutes after cytokine addition. The data shows (a) HTS inhibits the expression of CCL5 mRNA and (b) protein in both pre-treatment and co-treatment, as well as when HTS is applied in incremental time points after cytomix pretreatment. Although maximal inhibition was seen when HTS was applied 30mins or earlier after cytokine pre-incubation, the inhibitory effect remains significant when applied even 6 hrs after initial cytokine challenge. (*p

Fig 6. The anti-inflammatory effects of HTS…

Fig 6. The anti-inflammatory effects of HTS on CCL5 expression are reversible.

SAECS in P4…

Fig 6. The anti-inflammatory effects of HTS on CCL5 expression are reversible.
SAECS in P4 were cultured in the presence of HTS for 4hrs and allowed to rest for 2 and 4hrs in isotonic media before being subjected to a subsequent cytomix challenge. Cells previously treated by HTS can respond to a subsequent cytokine challenge after a minimal recovery period (2-4hrs), with CCL5 levels comparable to initial controls treated with cytomix.
Similar articles
Cited by
References
    1. Tisherman SA, Schmicker RH, Brasel KJ, Bulger EM, Kerby JD, Minei JP, et al. Detailed Description of all Deaths in Both the Shock and Traumatic Brain Injury Hypertonic Saline Trials of the Resuscitation Outcomes Consortium. Ann Surg. 2015;261(3):586–90. 10.1097/SLA.0000000000000837 - DOI - PMC - PubMed
    1. Sauaia A, Moore EE, Johnson JL, Chin TL, Banerjee A, Sperry JL, et al. Temporal trends of postinjury multiple-organ failure: still resource intensive, morbid, and lethal. The journal of trauma and acute care surgery. 2014;76(3):582–92, discussion 92–3. Epub 2014/02/21. 10.1097/TA.0000000000000147 - DOI - PMC - PubMed
    1. Bakowitz M, Bruns B, McCunn M. Acute lung injury and the acute respiratory distress syndrome in the injured patient. Scand J Trauma Resusc Emerg Med. 2012;20:54 Epub 2012/08/14. 10.1186/1757-7241-20-54 - DOI - PMC - PubMed
    1. Nathan C. Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol. 2006;6(3):173–82. http://www.nature.com/nri/journal/v6/n3/suppinfo/nri1785_S1.html. - PubMed
    1. Weigand MA, Horner C, Bardenheuer HJ, Bouchon A. The systemic inflammatory response syndrome. Best Pract Res Clin Anaesthesiol. 2004;18(3):455–75. Epub 2004/06/24. . - PubMed
Show all 65 references
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Fig 3. HTS inhibits the transcriptional activation…
Fig 3. HTS inhibits the transcriptional activation of IRF1 and STAT1 dependent genes.
In the Ingenuity Pathway Analysis of cytomix vs cytomix+HTS, IRF1 and STAT1 were inferred to regulate the expression of several downstream molecules in this study. (a) 12 of 18 genes have expression consistent with upstream inhibition of IRF1 by HTS (Z-score: -2.564; p = 1.05E-10). (b) 26 of 32 genes have expression consistent with STAT1 inhibition when cytomix is added to cells treated with HTS (Z-score: -4.638; p = 3.85E-16). (c) HTS attenuates cytomix-induced luciferase expression in SAECS transiently transfected with IRF1 and STAT1 reporter expression plasmids treated with cytomix for 4hrs in the presence and absence of HTS. (d) The expression of CCL5, a downstream endogenous target of IRF1 and STAT1, is also attenuated by HTS at 4, 6 and 8hrs as determined by ELISA. (*p<0.05; **p<0.01; ***p<0.001; ****p<0.0001).
Fig 4. HTS inhibits chemotaxis of neutrophils…
Fig 4. HTS inhibits chemotaxis of neutrophils and splenocyte-derived T-cells.
SAECS in P3 were cultured for 24 hrs with cytomix in the presence and absence of HTS and the supernatants were harvested and applied to the bottom chamber of the chemotaxis plate. Chemotaxis of neutrophils and splenocyte derived T cells towards the chemotactic gradient was assayed using Incucyte Zoom imaging system. The results show (a) supernatant from cytomix treated cells elicit maximal migration of neutrophils, and this migration was attenuated in supernatants from cytomix + HTS treated cells. Similarly, in a separate experiment (b), HTS inhibited the migration of splenocyte derived T-cells through the matrigel towards the chemotactic gradient. [solid lines indicate the loss of surface area previously occupied by the neutrophils or splenocytes derived T-cells on the top of membrane as they migrate to the reservoir containing the chemoattractant at the bottom of the membrane; dashed lines indicate p values for pairwise comparison of migration of neutrophils or splenocyte at each time point (control vs HTS, control vs cytomix, control vs cyto-HTS: Fig 4a and Fig 4b main panel; cytomix vs cyto–HTS: Fig 4a and Fig 4b inset). FMLP and CCL5 were used as positive control for the migration assays for neutrophil and splenocyte derived T cells respectively.
Fig 5. HTS inhibits CCL5 expression in…
Fig 5. HTS inhibits CCL5 expression in cytokine-pretreated cells.
SAECS in P3 were co-treated or pretreated with cytomix ± HTS for 8hrs and HTS was added at 30, 60, 120, 240, and 360 minutes after cytokine addition. The data shows (a) HTS inhibits the expression of CCL5 mRNA and (b) protein in both pre-treatment and co-treatment, as well as when HTS is applied in incremental time points after cytomix pretreatment. Although maximal inhibition was seen when HTS was applied 30mins or earlier after cytokine pre-incubation, the inhibitory effect remains significant when applied even 6 hrs after initial cytokine challenge. (*p

Fig 6. The anti-inflammatory effects of HTS…

Fig 6. The anti-inflammatory effects of HTS on CCL5 expression are reversible.

SAECS in P4…

Fig 6. The anti-inflammatory effects of HTS on CCL5 expression are reversible.
SAECS in P4 were cultured in the presence of HTS for 4hrs and allowed to rest for 2 and 4hrs in isotonic media before being subjected to a subsequent cytomix challenge. Cells previously treated by HTS can respond to a subsequent cytokine challenge after a minimal recovery period (2-4hrs), with CCL5 levels comparable to initial controls treated with cytomix.
Fig 6. The anti-inflammatory effects of HTS…
Fig 6. The anti-inflammatory effects of HTS on CCL5 expression are reversible.
SAECS in P4 were cultured in the presence of HTS for 4hrs and allowed to rest for 2 and 4hrs in isotonic media before being subjected to a subsequent cytomix challenge. Cells previously treated by HTS can respond to a subsequent cytokine challenge after a minimal recovery period (2-4hrs), with CCL5 levels comparable to initial controls treated with cytomix.

References

    1. Tisherman SA, Schmicker RH, Brasel KJ, Bulger EM, Kerby JD, Minei JP, et al. Detailed Description of all Deaths in Both the Shock and Traumatic Brain Injury Hypertonic Saline Trials of the Resuscitation Outcomes Consortium. Ann Surg. 2015;261(3):586–90. 10.1097/SLA.0000000000000837
    1. Sauaia A, Moore EE, Johnson JL, Chin TL, Banerjee A, Sperry JL, et al. Temporal trends of postinjury multiple-organ failure: still resource intensive, morbid, and lethal. The journal of trauma and acute care surgery. 2014;76(3):582–92, discussion 92–3. Epub 2014/02/21. 10.1097/TA.0000000000000147
    1. Bakowitz M, Bruns B, McCunn M. Acute lung injury and the acute respiratory distress syndrome in the injured patient. Scand J Trauma Resusc Emerg Med. 2012;20:54 Epub 2012/08/14. 10.1186/1757-7241-20-54
    1. Nathan C. Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol. 2006;6(3):173–82. .
    1. Weigand MA, Horner C, Bardenheuer HJ, Bouchon A. The systemic inflammatory response syndrome. Best Pract Res Clin Anaesthesiol. 2004;18(3):455–75. Epub 2004/06/24. .
    1. Matthay MA, Ware LB, Zimmerman GA. The acute respiratory distress syndrome. The Journal of Clinical Investigation. 122(8):2731–40. 10.1172/JCI60331
    1. Manicone AM. Role of the pulmonary epithelium and inflammatory signals in acute lung injury. Expert Rev Clin Immunol. 2009;5(1):63–75. Epub 2009/11/04. 10.1586/177666X.5.1.63
    1. Nydam TL, Moore EE, McIntyre RCJ, Wright FL, Gamboni-Robertson F, Eckels PC, et al. HYPERTONIC SALINE ATTENUATES TNF-α-INDUCED NF-κB ACTIVATION IN PULMONARY EPITHELIAL CELLS. Shock. 2009;31(5):466–72. 10.1097/SHK.0b013e31818ec47d 00024382-200905000-00006.
    1. Wright FL, Gamboni F, Moore EE, Nydam TL, Mitra S, Silliman CC, et al. Hyperosmolarity invokes distinct anti-inflammatory mechanisms in pulmonary epithelial cells: evidence from signaling and transcription layers. PLoS One. 2014;9(12):e114129 Epub 2014/12/06. 10.1371/journal.pone.0114129
    1. Grommes J, Soehnlein O. Contribution of Neutrophils to Acute Lung Injury. Mol Med. 2011;17(3–4):293–307. 10.2119/molmed.2010.00138
    1. Albaiceta GM, Gutierrez-Fernandez A, Garcia-Prieto E, Puente XS, Parra D, Astudillo A, et al. Absence or inhibition of matrix metalloproteinase-8 decreases ventilator-induced lung injury. Am J Respir Cell Mol Biol. 2010;43(5):555–63. Epub 2009/12/10. 10.1165/rcmb.2009-0034OC .
    1. Hastbacka J, Linko R, Tervahartiala T, Varpula T, Hovilehto S, Parviainen I, et al. Serum MMP-8 and TIMP-1 in critically ill patients with acute respiratory failure: TIMP-1 is associated with increased 90-day mortality. Anesth Analg. 2014;118(4):790–8. Epub 2014/03/22. 10.1213/ANE.0000000000000120 .
    1. Hsu AT, Barrett CD, DeBusk GM, Ellson CD, Gautam S, Talmor DS, et al. Kinetics and Role of Plasma Matrix Metalloproteinase-9 Expression in Acute Lung Injury and the Acute Respiratory Distress Syndrome. Shock. 2015;44(2):128–36. Epub 2015/05/27. 10.1097/SHK.0000000000000386 .
    1. Mattox KL, Maningas PA, Moore EE, Mateer JR, Marx JA, Aprahamian C, et al. Prehospital hypertonic saline/dextran infusion for post-traumatic hypotension. The U.S.A. Multicenter Trial. Ann Surg. 1991;213(5):482–91. Epub 1991/05/01.
    1. Cuschieri J, Gourlay D, Garcia I, Jelacic S, Maier RV. Hypertonic preconditioning inhibits macrophage responsiveness to endotoxin. J Immunol. 2002;168(3):1389–96. Epub 2002/01/22. .
    1. Powers KA, Zurawska J, Szaszi K, Khadaroo RG, Kapus A, Rotstein OD. Hypertonic resuscitation of hemorrhagic shock prevents alveolar macrophage activation by preventing systemic oxidative stress due to gut ischemia/reperfusion. Surgery. 2005;137(1):66–74. Epub 2004/12/23. 10.1016/j.surg.2004.05.051 .
    1. Deitch EA, Shi HP, Feketeova E, Hauser CJ, Xu DZ. Hypertonic saline resuscitation limits neutrophil activation after trauma-hemorrhagic shock. Shock. 2003;19(4):328–33. Epub 2003/04/12. .
    1. Bahrami S, Zimmermann K, Szelenyi Z, Hamar J, Scheiflinger F, Redl H, et al. Small-volume fluid resuscitation with hypertonic saline prevents inflammation but not mortality in a rat model of hemorrhagic shock. Shock. 2006;25(3):283–9. Epub 2006/03/23. 10.1097/01.shk.0000208808.03148.ea .
    1. Rizoli SB, Rhind SG, Shek PN, Inaba K, Filips D, Tien H, et al. The immunomodulatory effects of hypertonic saline resuscitation in patients sustaining traumatic hemorrhagic shock: a randomized, controlled, double-blinded trial. Ann Surg. 2006;243(1):47–57. Epub 2005/12/24.
    1. Junger WG, Coimbra R, Liu FC, Herdon-Remelius C, Junger W, Junger H, et al. Hypertonic saline resuscitation: a tool to modulate immune function in trauma patients? Shock. 1997;8(4):235–41. Epub 1997/11/05. .
    1. Wohlauer M, Moore EE, Silliman CC, Fragoso M, Gamboni F, Harr J, et al. NEBULIZED HYPERTONIC SALINE ATTENUATES ACUTE LUNG INJURY FOLLOWING TRAUMA AND HEMORRHAGIC SHOCK. Crit Care Med. 2012;40(9):2647–53. 10.1097/CCM.0b013e3182592006
    1. Bastarache JA, Sebag SC, Grove BS, Ware LB. IFN-γ and TNF-α Act Synergistically to Upregulate Tissue Factor in Alveolar Epithelial Cells. Exp Lung Res. 2011;37(8):509–17. 10.3109/01902148.2011.605512
    1. Kreimeier U, Messmer K. Small-volume resuscitation: from experimental evidence to clinical routine. Advantages and disadvantages of hypertonic solutions. Acta Anaesthesiol Scand. 2002;46(6):625–38. 10.1034/j.1399-6576.2002.460601.x
    1. Wu TD, Nacu S. Fast and SNP-tolerant detection of complex variants and splicing in short reads. Bioinformatics. 2010;26(7):873–81. Epub 2010/02/12. 10.1093/bioinformatics/btq057
    1. Baird NL, Bowlin JL, Cohrs RJ, Gilden D, Jones KL. Comparison of varicella-zoster virus RNA sequences in human neurons and fibroblasts. J Virol. 2014;88(10):5877–80. Epub 2014/03/07. 10.1128/JVI.00476-14
    1. Maycotte P, Jones KL, Goodall ML, Thorburn J, Thorburn A. Autophagy Supports Breast Cancer Stem Cell Maintenance by Regulating IL6 Secretion. Mol Cancer Res. 2015;13(4):651–8. Epub 2015/01/13. 10.1158/1541-7786.MCR-14-0487
    1. Trapnell C, Williams BA, Pertea G, Mortazavi A, Kwan G, van Baren MJ, et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol. 2010;28(5):511–5. Epub 2010/05/04. 10.1038/nbt.1621
    1. Henderson HH, Timberlake KB, Austin ZA, Badani H, Sanford B, Tremblay K, et al. Occupancy of RNA Polymerase II Phosphorylated on Serine 5 (RNAP S5P) and RNAP S2P on Varicella-Zoster Virus Genes 9, 51, and 66 Is Independent of Transcript Abundance and Polymerase Location within the Gene. J Virol. 2015;90(3):1231–43. Epub 2015/11/13. 10.1128/JVI.02617-15
    1. Ciesla DJ, Moore EE, Johnson JL, Burch JM, Cothren CC, Sauaia A. The role of the lung in postinjury multiple organ failure. Surgery. 2005;138(4):749–57; discussion 57–8. Epub 2005/11/05. 10.1016/j.surg.2005.07.020 .
    1. Diamond G, Legarda D, Ryan LK. The innate immune response of the respiratory epithelium. Immunol Rev. 2000;173:27–38. Epub 2000/03/17. .
    1. Gropper MA, Wiener-Kronish J. The epithelium in acute lung injury/acute respiratory distress syndrome. Curr Opin Crit Care. 2008;14(1):11–5. Epub 2008/01/16. 10.1097/MCC.0b013e3282f417a0 .
    1. Angle N, Hoyt DB, Coimbra R, Liu F, Herdon-Remelius C, Loomis W, et al. Hypertonic saline resuscitation diminishes lung injury by suppressing neutrophil activation after hemorrhagic shock. Shock. 1998;9(3):164–70. Epub 1998/04/03. .
    1. Banerjee A, Moore EE, McLaughlin NJ, Lee L, Jones WL, Johnson JL, et al. Hyperosmolarity attenuates TNF-alpha-mediated proinflammatory activation of human pulmonary microvascular endothelial cells. Shock. 2013;39(4):366–72. Epub 2013/02/01. 10.1097/SHK.0b013e3182894016
    1. Sokol CL, Luster AD. The chemokine system in innate immunity. Cold Spring Harb Perspect Biol. 2015;7(5). Epub 2015/01/31. 10.1101/cshperspect.a016303 .
    1. Silva MT. Neutrophils and macrophages work in concert as inducers and effectors of adaptive immunity against extracellular and intracellular microbial pathogens. J Leukoc Biol. 2010;87(5):805–13. 10.1189/jlb.1109767
    1. Kudo M, Ogawa E, Kinose D, Haruna A, Takahashi T, Tanabe N, et al. Oxidative stress induced Interleukin-32 mRNA expression in human bronchial epithelial cells. Respir Res. 2012;13(1):19-. 10.1186/1465-9921-13-19
    1. Su R, Nguyen M-LT, Agarwal MR, Kirby C, Nguyen CP, Ramstein J, et al. Interferon-inducible chemokines reflect severity and progression in sarcoidosis. Respir Res. 2013;14(1):121-. 10.1186/1465-9921-14-121
    1. Tohyama M, Sayama K, Komatsuzawa H, Hanakawa Y, Shirakata Y, Dai X, et al. CXCL16 is a novel mediator of the innate immunity of epidermal keratinocytes. Int Immunol. 2007;19(9):1095–102. 10.1093/intimm/dxm083
    1. Thorley AJ, Goldstraw P, Young A, Tetley TD. Primary Human Alveolar Type II Epithelial Cell CCL20 (Macrophage Inflammatory Protein-3α)–Induced Dendritic Cell Migration. Am J Respir Cell Mol Biol. 2005;32(4):262–7. 10.1165/rcmb.2004-0196OC
    1. Kelly EAB, Koziol-White CJ, Clay KJ, Liu LY, Bates ME, Bertics PJ, et al. Potential Contribution of IL-7 to Allergen-Induced Eosinophilic Airway Inflammation in Asthma. The Journal of Immunology. 2009;182(3):1404–10. 10.4049/jimmunol.182.3.1404
    1. Garlanda C, Dinarello Charles A, Mantovani A. The Interleukin-1 Family: Back to the Future. Immunity. 2013;39(6):1003–18. 10.1016/j.immuni.2013.11.010.
    1. Chan JK, Roth J, Oppenheim JJ, Tracey KJ, Vogl T, Feldmann M, et al. Alarmins: awaiting a clinical response. J Clin Invest. 2012;122(8):2711–9. Epub 2012/08/02. 10.1172/JCI62423
    1. Murphy GEJ, Xu D, Liew FY, McInnes IB. Role of interleukin 33 in human immunopathology. Ann Rheum Dis. 2010;69(Suppl 1):i43–i7. 10.1136/ard.2009.120113
    1. Schmitz J, Owyang A, Oldham E, Song Y, Murphy E, McClanahan TK, et al. IL-33, an Interleukin-1-like Cytokine that Signals via the IL-1 Receptor-Related Protein ST2 and Induces T Helper Type 2-Associated Cytokines. Immunity. 2005;23(5):479–90. 10.1016/j.immuni.2005.09.015.
    1. Manson J, Thiemermann C, Brohi K. Trauma alarmins as activators of damage-induced inflammation. Br J Surg. 2012;99(S1):12–20. 10.1002/bjs.7717
    1. Sun Y, Mozaffarian A, Arnett HA, Dinh H, Trueblood ES, Towne JE. 253: IL-36 induces inflammation and collagen deposition in the lung. Cytokine. 2013;63(3):303 10.1016/j.cyto.2013.06.256.
    1. Iwakura Y, Ishigame H. The IL-23/IL-17 axis in inflammation. J Clin Invest. 2006;116(5):1218–22. 10.1172/JCI28508
    1. Teng MWL, Bowman EP, McElwee JJ, Smyth MJ, Casanova J-L, Cooper AM, et al. IL-12 and IL-23 cytokines: from discovery to targeted therapies for immune-mediated inflammatory diseases. Nat Med. 2015;21(7):719–29. 10.1038/nm.3895
    1. Brunner PM, Glitzner E, Reininger B, Klein I, Stary G, Mildner M, et al. CCL7 contributes to the TNF-alpha-dependent inflammation of lesional psoriatic skin. Exp Dermatol. 2015;24(7):522–8. Epub 2015/04/02. 10.1111/exd.12709 .
    1. Kolls JK, Lindén A. Interleukin-17 Family Members and Inflammation. Immunity. 2004;21(4):467–76. 10.1016/j.immuni.2004.08.018.
    1. Knight D. Leukaemia inhibitory factor (LIF): a cytokine of emerging importance in chronic airway inflammation. Pulm Pharmacol Ther. 2001;14(3):169–76. Epub 2001/07/13. 10.1006/pupt.2001.0282 .
    1. Eklund KK, Niemi K, Kovanen PT. Immune functions of serum amyloid A. Crit Rev Immunol. 2012;32(4):335–48. Epub 2012/12/15. .
    1. Parsons SJ, Parsons JT. Src family kinases, key regulators of signal transduction. Oncogene. 2004;23.
    1. Pazdrak K, Olszewska-Pazdrak B, Stafford S, Garofalo RP, Alam R. Lyn, Jak2, and Raf-1 kinases are critical for the antiapoptotic effect of interleukin 5, whereas only Raf-1 kinase is essential for eosinophil activation and degranulation. J Exp Med. 1998;188.
    1. Weber A, Wasiliew P, Kracht M. Interleukin-1 (IL-1) pathway. Sci Signal. 2010;3(105):cm1 Epub 2010/01/21. 10.1126/scisignal.3105cm1 .
    1. Rawlings JS, Rosler KM, Harrison DA. The JAK/STAT signaling pathway. J Cell Sci. 2004;117(8):1281–3. 10.1242/jcs.00963
    1. Wang Z, Ma LJ, Kang Y, Li X, Zhang XJ. Dickkopf-3 (Dkk3) induces apoptosis in cisplatin-resistant lung adenocarcinoma cells via the Wnt/beta-catenin pathway. Oncol Rep. 2015;33(3):1097–106. Epub 2015/01/13. 10.3892/or.2014.3704 .
    1. Richie JP. Re: CMTM3 Inhibits Human Testicular Cancer Cell Growth through Inducing Cell-Cycle Arrest and Apoptosis. The Journal of Urology. 2015;194(3):706–7. 10.1016/j.juro.2015.06.010.
    1. Mitchell RA, Liao H, Chesney J, Fingerle-Rowson G, Baugh J, David J, et al. Macrophage migration inhibitory factor (MIF) sustains macrophage proinflammatory function by inhibiting p53: Regulatory role in the innate immune response. Proc Natl Acad Sci U S A. 2002;99(1):345–50. 10.1073/pnas.012511599
    1. Davidson G, Niehrs C. Emerging links between CDK cell cycle regulators and Wnt signaling. Trends Cell Biol. 2010;20(8):453–60. 10.1016/j.tcb.2010.05.002.
    1. Gamboni F, Anderson C, Mitra S, Reisz JA, Nemkov T, Dzieciatkowska M, et al. Hypertonic Saline Primes Activation of the p53-p21 Signaling Axis in Human Small Airway Epithelial Cells That Prevents Inflammation Induced by Pro-inflammatory Cytokines. J Proteome Res. 2016;15(10):3813–26. Epub 2016/08/17. 10.1021/acs.jproteome.6b00602 .
    1. Herquel B, Ouararhni K, Khetchoumian K, Ignat M, Teletin M, Mark M, et al. Transcription cofactors TRIM24, TRIM28, and TRIM33 associate to form regulatory complexes that suppress murine hepatocellular carcinoma. Proc Natl Acad Sci U S A. 2011;108(20):8212–7. Epub 2011/05/03. 10.1073/pnas.1101544108
    1. Kramer OH, Knauer SK, Greiner G, Jandt E, Reichardt S, Guhrs KH, et al. A phosphorylation-acetylation switch regulates STAT1 signaling. Genes Dev. 2009;23(2):223–35. Epub 2009/01/28. 10.1101/gad.479209 ;.
    1. Reeves EP, McCarthy C, McElvaney OJ, Vijayan MSN, White MM, Dunlea DM, et al. Inhaled hypertonic saline for cystic fibrosis: Reviewing the potential evidence for modulation of neutrophil signalling and function. World Journal of Critical Care Medicine. 2015;4(3):179–91. 10.5492/wjccm.v4.i3.179
    1. Reeves EP, Williamson M, O’Neill SJ, Greally P, McElvaney NG. Nebulized hypertonic saline decreases IL-8 in sputum of patients with cystic fibrosis. Am J Respir Crit Care Med. 2011;183(11):1517–23. Epub 2011/02/19. 10.1164/rccm.201101-0072OC .

Source: PubMed

3
Suscribir