Hyperosmolarity invokes distinct anti-inflammatory mechanisms in pulmonary epithelial cells: evidence from signaling and transcription layers

Franklin L Wright, Fabia Gamboni, Ernest E Moore, Trevor L Nydam, Sanchayita Mitra, Christopher C Silliman, Anirban Banerjee, Franklin L Wright, Fabia Gamboni, Ernest E Moore, Trevor L Nydam, Sanchayita Mitra, Christopher C Silliman, Anirban Banerjee

Abstract

Hypertonic saline (HTS) has been used intravenously to reduce organ dysfunction following injury and as an inhaled therapy for cystic fibrosis lung disease. The role and mechanism of HTS inhibition was explored in the TNFα and IL-1β stimulation of pulmonary epithelial cells. Hyperosmolar (HOsm) media (400 mOsm) inhibited the production of select cytokines stimulated by TNFα and IL-1β at the level of mRNA translation, synthesis and release. In TNFα stimulated A549 cells, HOsm media inhibited I-κBα phosphorylation, NF-κB translocation into the nucleus and NF-κB nuclear binding. In IL-1β stimulated cells HOsm inhibited I-κBα phosphorylation without affecting NF-κB translocation or nuclear binding. Incubation in HOsm conditions inhibited both TNFα and IL-1β stimulated nuclear localization of interferon response factor 1 (IRF-1). Additional transcription factors such as AP-1, Erk-1/2, JNK and STAT-1 were unaffected by HOsm. HTS and sorbitol supplemented media produced comparable outcomes in all experiments, indicating that the effects of HTS were mediated by osmolarity, not by sodium. While not affecting MAPK modules discernibly in A549 cells, both HOsm conditions inhibit IRF-1 against TNFα or IL-1β, but inhibit p65 NF-kB translocation only against TNFα but not IL-1β. Thus, anti-inflammatory mechanisms of HTS/HOsm appear to disrupt cytokine signals at distinct intracellular steps.

Conflict of interest statement

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

Figures

Figure 1. NaCl or sorbitol HOsmolarity (HOsm)inhibit…
Figure 1. NaCl or sorbitol HOsmolarity (HOsm)inhibit chemokine secretion similarly, depending on stimulus.
RANTES (A) and IP-10 (B) release was inhibited by both HTS and SOR when stimulated by TNFα or IL-1β. HTS and SOR inhibited MCP-1 (C) release only when stimulated by TNFα but not by IL-1β. HOsm did not inhibit IL-6 (D) release with either stimulus. No significant differences were seen between HTS and SOR groups. Chemokine production was measured at 18 hours when treated with TNFα or IL-1β in the presence of 400 mOsm HTS or Sorbitol.
Figure 2. HOsm inhibits TNFα or IL-1β…
Figure 2. HOsm inhibits TNFα or IL-1β stimulated mRNA for RANTES (A), IP-10 (B) and MCP-1 (C) chemokines in parallel with overall chemokine release levels.
mRNA levels were measured following stimulation for 6 hours and normalized to constitutive GAPDH production after treatment with TNFα or IL-1β in the presence of 400 mOsm HTS or Sorbitol. No significant differences were seen between any of the HTS and SOR groups.
Figure 3. HOsm inhibits translocation of NF-κB…
Figure 3. HOsm inhibits translocation of NF-κB into the nucleus induced by TNFα stimulation but not by IL-1β stimulation.
Indirect immunofluorescent staining was used to examine the intracellular location of the p65 subunit of NF-κB following 30 minutes of TNFα or IL-1β stimulation in the presence of isotonic (upper panels) or 400 mOsm HTS (SOR group not shown but appeared similar to HTS group). Cell structure is depicted in red (WGA glycoprotein stain), nuclei in blue (DAPI) and NF-κB in green. NF-kB translocation into the nucleus is seen in TNFα, IL-1β treated cells in isotonic medium. Under hyperosmolar conditions, translocation (thick arrows) stimulated by TNFα (but not IL-1β) is diminished (thin arrows). This figure is representative of three separate experiments. The bottom panel shows that HOsm does not significantly affect cell viability. Viabilty of pulmonary epithelial cells measured by MTT assay (absorbance) at 18 hours in the presence of 400 mOsm HTS or SOR. Cells were killed by heat treatment, at 90°Cfor 30 minutes prior to assay.
Figure 4. HOsm inhibits NF-κB activation by…
Figure 4. HOsm inhibits NF-κB activation by TNFα but not IL-1β stimulation.
The time course of NF-κB consensus binding of the p65 subunit in nuclear fraction of cells stimulated by TNFα (A) or IL-1β (B) in the presence of 400 mOsm HTS or SOR (n = 5). Activation assayed by DNA binding to consensus sequence; * p

Figure 5. HOsm inhibits phosphorylation of I-κBα…

Figure 5. HOsm inhibits phosphorylation of I-κBα induced by both TNFα and IL-1β.

Phosphorylation of…

Figure 5. HOsm inhibits phosphorylation of I-κBα induced by both TNFα and IL-1β.
Phosphorylation of I-κBα is inhibited at 10 minutes in cells stimulated with TNFα or IL-1β (10 ng/ml) by either 400 mOsm HTS or SOR treatment. The immunoblot is representative of five separate experiments. LC represent the loading control obtained by Ponceau-S stain of the nitro cellulose membranes. Phosphoprotein ELISA time course of cells stimulated by TNFα (B) or IL-1β (C) (both 10 ng/ml), in the presence of 400 mOsm HTS or Sorbitol; normalized for phosphoprotein/total protein levels. Phosphorylation of I-κBα initially diminished with either TNFα (B) or IL-1β (C) stimulation when treated with HOsm; however, I-κBα phosphorylation increases at 30 minutes with HOsm over isotonic treatment with either stimulus; * p

Figure 6. HOsm inhibits IRF-1 localization to…

Figure 6. HOsm inhibits IRF-1 localization to the nucleus with either TNFα or IL-1β stimulation.

Figure 6. HOsm inhibits IRF-1 localization to the nucleus with either TNFα or IL-1β stimulation.
IRF-1 in the nuclear extract of cells stimulated by TNFα or IL-1β in the presence of 400 mOsm HTS or sorbitol was examined by immunoblotting (A), figure is representative of three separate experiments. The gels were digitized and quantitated for nuclear IRF-1 after TNFα (A) or IL-1β stimulation (C); * p

Figure 7. HOsm does not significantly affect…

Figure 7. HOsm does not significantly affect cell volume.

Top left panel shows the mean…

Figure 7. HOsm does not significantly affect cell volume.
Top left panel shows the mean total cell volume of pulmonary epithelial cells measured by immunofluorescent microscopy is not changed in the presence of 400 mOsm HTS or SOR after 18 hours of incubation (n = 3). The panel on top right shows mean (± sem) forward scatter of pulmonary epithelial cells measured by flow cytometry is also unchanged in the presence of isotonic and 400, 500, and 600 mOsm solutions of media plus HTS, after 18 hours of incubation (n = 3).
All figures (7)
Similar articles
Cited by
References
    1. Cuschieri J, Gourlay D, Garcia I, Jelacic S, Maier RV (2002) Hypertonic preconditioning inhibits macrophage responsiveness to endotoxin. J Immunol 168:1389–1396. - PubMed
    1. Junger WG, Coimbra R, Liu FC, Herdon-Remelius C, Junger W, et al. (1997) Hypertonic saline resuscitation: a tool to modulate immune function in trauma patients? Shock 8:235–241. - PubMed
    1. Rizoli SB, Rhind SG, Shek PN, Inaba K, Filips D, et al. (2006) The immunomodulatory effects of hypertonic saline resuscitation in patients sustaining traumatic hemorrhagic shock: a randomized, controlled, double-blinded trial. Ann Surg 243:47–57. - PMC - PubMed
    1. Mattox KL, Maningas PA, Moore EE, Mateer JR, Marx JA, et al. (1991) Prehospital hypertonic saline/dextran infusion for post-traumatic hypotension. The U.S.A. Multicenter Trial. Ann Surg 213:482–491. - PMC - PubMed
    1. Bulger EM, Jurkovich GJ, Nathens AB, Copass MK, Hanson S, et al. (2008) Hypertonic resuscitation of hypovolemic shock after blunt trauma: a randomized controlled trial. Arch Surg 143:139–148 discussion 149. - PubMed
Show all 52 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 5. HOsm inhibits phosphorylation of I-κBα…
Figure 5. HOsm inhibits phosphorylation of I-κBα induced by both TNFα and IL-1β.
Phosphorylation of I-κBα is inhibited at 10 minutes in cells stimulated with TNFα or IL-1β (10 ng/ml) by either 400 mOsm HTS or SOR treatment. The immunoblot is representative of five separate experiments. LC represent the loading control obtained by Ponceau-S stain of the nitro cellulose membranes. Phosphoprotein ELISA time course of cells stimulated by TNFα (B) or IL-1β (C) (both 10 ng/ml), in the presence of 400 mOsm HTS or Sorbitol; normalized for phosphoprotein/total protein levels. Phosphorylation of I-κBα initially diminished with either TNFα (B) or IL-1β (C) stimulation when treated with HOsm; however, I-κBα phosphorylation increases at 30 minutes with HOsm over isotonic treatment with either stimulus; * p

Figure 6. HOsm inhibits IRF-1 localization to…

Figure 6. HOsm inhibits IRF-1 localization to the nucleus with either TNFα or IL-1β stimulation.

Figure 6. HOsm inhibits IRF-1 localization to the nucleus with either TNFα or IL-1β stimulation.
IRF-1 in the nuclear extract of cells stimulated by TNFα or IL-1β in the presence of 400 mOsm HTS or sorbitol was examined by immunoblotting (A), figure is representative of three separate experiments. The gels were digitized and quantitated for nuclear IRF-1 after TNFα (A) or IL-1β stimulation (C); * p

Figure 7. HOsm does not significantly affect…

Figure 7. HOsm does not significantly affect cell volume.

Top left panel shows the mean…

Figure 7. HOsm does not significantly affect cell volume.
Top left panel shows the mean total cell volume of pulmonary epithelial cells measured by immunofluorescent microscopy is not changed in the presence of 400 mOsm HTS or SOR after 18 hours of incubation (n = 3). The panel on top right shows mean (± sem) forward scatter of pulmonary epithelial cells measured by flow cytometry is also unchanged in the presence of isotonic and 400, 500, and 600 mOsm solutions of media plus HTS, after 18 hours of incubation (n = 3).
All figures (7)
Similar articles
Cited by
References
    1. Cuschieri J, Gourlay D, Garcia I, Jelacic S, Maier RV (2002) Hypertonic preconditioning inhibits macrophage responsiveness to endotoxin. J Immunol 168:1389–1396. - PubMed
    1. Junger WG, Coimbra R, Liu FC, Herdon-Remelius C, Junger W, et al. (1997) Hypertonic saline resuscitation: a tool to modulate immune function in trauma patients? Shock 8:235–241. - PubMed
    1. Rizoli SB, Rhind SG, Shek PN, Inaba K, Filips D, et al. (2006) The immunomodulatory effects of hypertonic saline resuscitation in patients sustaining traumatic hemorrhagic shock: a randomized, controlled, double-blinded trial. Ann Surg 243:47–57. - PMC - PubMed
    1. Mattox KL, Maningas PA, Moore EE, Mateer JR, Marx JA, et al. (1991) Prehospital hypertonic saline/dextran infusion for post-traumatic hypotension. The U.S.A. Multicenter Trial. Ann Surg 213:482–491. - PMC - PubMed
    1. Bulger EM, Jurkovich GJ, Nathens AB, Copass MK, Hanson S, et al. (2008) Hypertonic resuscitation of hypovolemic shock after blunt trauma: a randomized controlled trial. Arch Surg 143:139–148 discussion 149. - PubMed
Show all 52 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 6. HOsm inhibits IRF-1 localization to…
Figure 6. HOsm inhibits IRF-1 localization to the nucleus with either TNFα or IL-1β stimulation.
IRF-1 in the nuclear extract of cells stimulated by TNFα or IL-1β in the presence of 400 mOsm HTS or sorbitol was examined by immunoblotting (A), figure is representative of three separate experiments. The gels were digitized and quantitated for nuclear IRF-1 after TNFα (A) or IL-1β stimulation (C); * p

Figure 7. HOsm does not significantly affect…

Figure 7. HOsm does not significantly affect cell volume.

Top left panel shows the mean…

Figure 7. HOsm does not significantly affect cell volume.
Top left panel shows the mean total cell volume of pulmonary epithelial cells measured by immunofluorescent microscopy is not changed in the presence of 400 mOsm HTS or SOR after 18 hours of incubation (n = 3). The panel on top right shows mean (± sem) forward scatter of pulmonary epithelial cells measured by flow cytometry is also unchanged in the presence of isotonic and 400, 500, and 600 mOsm solutions of media plus HTS, after 18 hours of incubation (n = 3).
All figures (7)
Figure 7. HOsm does not significantly affect…
Figure 7. HOsm does not significantly affect cell volume.
Top left panel shows the mean total cell volume of pulmonary epithelial cells measured by immunofluorescent microscopy is not changed in the presence of 400 mOsm HTS or SOR after 18 hours of incubation (n = 3). The panel on top right shows mean (± sem) forward scatter of pulmonary epithelial cells measured by flow cytometry is also unchanged in the presence of isotonic and 400, 500, and 600 mOsm solutions of media plus HTS, after 18 hours of incubation (n = 3).

References

    1. Cuschieri J, Gourlay D, Garcia I, Jelacic S, Maier RV (2002) Hypertonic preconditioning inhibits macrophage responsiveness to endotoxin. J Immunol 168:1389–1396.
    1. Junger WG, Coimbra R, Liu FC, Herdon-Remelius C, Junger W, et al. (1997) Hypertonic saline resuscitation: a tool to modulate immune function in trauma patients? Shock 8:235–241.
    1. Rizoli SB, Rhind SG, Shek PN, Inaba K, Filips D, et al. (2006) The immunomodulatory effects of hypertonic saline resuscitation in patients sustaining traumatic hemorrhagic shock: a randomized, controlled, double-blinded trial. Ann Surg 243:47–57.
    1. Mattox KL, Maningas PA, Moore EE, Mateer JR, Marx JA, et al. (1991) Prehospital hypertonic saline/dextran infusion for post-traumatic hypotension. The U.S.A. Multicenter Trial. Ann Surg 213:482–491.
    1. Bulger EM, Jurkovich GJ, Nathens AB, Copass MK, Hanson S, et al. (2008) Hypertonic resuscitation of hypovolemic shock after blunt trauma: a randomized controlled trial. Arch Surg 143:139–148 discussion 149.
    1. Wohlauer M, Moore EE, Silliman CC, Fragoso M, Gamboni F, et al. (2012) Nebulized hypertonic saline attenuates acute lung injury following trauma and hemorrhagic shock via inhibition of matrix metalloproteinase-13. Crit Care Med 40:2647–2653.
    1. Ciesla DJ, Moore EE, Biffl WL, Gonzalez RJ, Silliman CC (2001) Hypertonic saline attenuation of the neutrophil cytotoxic response is reversed upon restoration of normotonicity and reestablished by repeated hypertonic challenge. Surgery 129:567–575.
    1. Powers KA, Zurawska J, Szaszi K, Khadaroo RG, Kapus A, et al. (2005) Hypertonic resuscitation of hemorrhagic shock prevents alveolar macrophage activation by preventing systemic oxidative stress due to gut ischemia/reperfusion. Surgery 137:66–74.
    1. Magor BG, Magor KE (2001) Evolution of effectors and receptors of innate immunity. Dev Comp Immunol 25:651–682.
    1. Weber A, Wasiliew P, Kracht M (2010) Interleukin-1 (IL-1) pathway. Sci Signal 3:cm1.
    1. Bhatia M, Moochhala S (2004) Role of inflammatory mediators in the pathophysiology of acute respiratory distress syndrome. Journal of Pathology 202:145–156.
    1. Goodman RB, Pugin J, Lee JS, Matthay MA (2003) Cytokine-mediated inflammation in acute lung injury. Cytokine Growth Factor Rev 14:523–535.
    1. Jarrar D, Kuebler JF, Rue LW 3rd, Matalon S, Wang P, et al. (2002) Alveolar macrophage activation after trauma-hemorrhage and sepsis is dependent on NF-kappaB and MAPK/ERK mechanisms. Am J Physiol Lung Cell Mol Physiol 283:L799–805.
    1. Park WY, Goodman RB, Steinberg KP, Ruzinski JT, Radella F 2nd, et al. (2001) Cytokine balance in the lungs of patients with acute respiratory distress syndrome. Am J Respir Crit Care Med 164:1896–1903.
    1. Ganter MT, Roux J, Miyazawa B, Howard M, Frank JA, et al. (2008) Interleukin-1beta causes acute lung injury via alphavbeta5 and alphavbeta6 integrin-dependent mechanisms. Circ Res 102:804–812.
    1. Sato H, Kasai K, Tanaka T, Kita T, Tanaka N (2008) Role of tumor necrosis factor-alpha and interleukin-1beta on lung dysfunction following hemorrhagic shock in rats. Med Sci Monit 14:BR79–87.
    1. Nydam TL, Moore EE, McIntyre RC Jr, Wright FL, Gamboni-Robertson F, et al. (2009) Hypertonic saline attenuates TNF-alpha-induced NF-kappaB activation in pulmonary epithelial cells. Shock 31:466–472.
    1. Solt LA, Madge LA, Orange JS, May MJ (2007) Interleukin-1-induced NF-kappaB activation is NEMO-dependent but does not require IKKbeta. J Biol Chem 282:8724–8733.
    1. Zhao T, Yang L, Sun Q, Arguello M, Ballard DW, et al. (2007) The NEMO adaptor bridges the nuclear factor-kappaB and interferon regulatory factor signaling pathways. Nat Immunol 8:592–600.
    1. Ghosh S, Hayden MS (2008) New regulators of NF-kappaB in inflammation. Nat Rev Immunol 8:837–848.
    1. Shultz DB, Rani MR, Fuller JD, Ransohoff RM, Stark GR (2009) Roles of IKK-beta, IRF1, and p65 in the activation of chemokine genes by interferon-gamma. J Interferon Cytokine Res 29:817–824.
    1. Abraham E (2003) Neutrophils and acute lung injury. Crit Care Med 31:S195–199.
    1. Held HD, Boettcher S, Hamann L, Uhlig S (2001) Ventilation-induced chemokine and cytokine release is associated with activation of nuclear factor-kappaB and is blocked by steroids. Am J Respir Crit Care Med 163:711–716.
    1. Solt LA, Madge LA, May MJ (2009) NEMO-binding domains of both IKKalpha and IKKbeta regulate IkappaB kinase complex assembly and classical NF-kappaB activation. J Biol Chem 284:27596–27608.
    1. Heinemeyer T, Chen X, Karas H, Kel AE, Kel OV, et al. (1999) Expanding the TRANSFAC database towards an expert system of regulatory molecular mechanisms. Nucleic Acids Res 27:318–322.
    1. Werner T, Fessele S, Maier H, Nelson PJ (2003) Computer modeling of promoter organization as a tool to study transcriptional coregulation. FASEB J 17:1228–1237.
    1. Ciesla DJ, Moore EE, Zallen G, Biffl WL, Silliman CC (2000) Hypertonic saline attenuation of polymorphonuclear neutrophil cytotoxicity: timing is everything. J Trauma 48:388–395.
    1. Benjamini Y, Hochberg Y (1995) Controlling the False Discovery Rate: A practical and powerful approach to multiple testing. Journal of the Royal Statistical Society 57:289–300.
    1. Belperio JA, Keane MP, Burdick MD, Londhe V, Xue YY, et al. (2002) Critical role for CXCR2 and CXCR2 ligands during the pathogenesis of ventilator-induced lung injury. J Clin Invest 110:1703–1716.
    1. Levitt JE, Gould MK, Ware LB, Matthay MA (2009) The Pathogenetic and Prognostic Value of Biologic Markers in Acute Lung Injury. J Intensive Care Med.
    1. Matthay MA (2008) Treatment of acute lung injury: clinical and experimental studies. Proc Am Thorac Soc 5:297–299.
    1. Ziegler EC, Ghosh S (2005) Regulating inducible transcription through controlled localization. Sci STKE 2005:re6.
    1. Hacker H, Karin M (2006) Regulation and function of IKK and IKK-related kinases. Sci STKE 2006:re13.
    1. Ananko EA, Kondrakhin YV, Merkulova TI, Kolchanov NA (2007) Recognition of interferon-inducible sites, promoters, and enhancers. BMC Bioinformatics 8:56.
    1. Banerjee A, Moore EE, McLaughlin NJ, Lee L, Jones WL, et al. (2013) Hyperosmolarity attenuates TNF-alpha-mediated proinflammatory activation of human pulmonary microvascular endothelial cells. Shock 39:366–372.
    1. Eckels PC, Banerjee A, Moore E, McLaughlin NJ, Gries L, et al. (2009) Amantadine Inhibits Platelet-Activating Factor Induced Clathrin-Mediated Endocytosis in Human Neutrophils. Am J Physiol Cell Physiol.
    1. Gundersen Y, Ruud TE, Krohn CD, Sveen O, Lyngstadaas SP, et al. (2010) Impact of hypertonic saline on the release of selected cytokines after stimulation with LPS or peptidoglycan in ex vivo whole blood from healthy humans. Shock 34:450–454.
    1. Waskiewicz AJ, Cooper JA (1995) Mitogen and stress response pathways: MAP kinase cascades and phosphatase regulation in mammals and yeast. Curr Opin Cell Biol 7:798–805.
    1. Shapiro L, Dinarello CA (1997) Hyperosmotic stress as a stimulant for proinflammatory cytokine production. Exp Cell Res 231:354–362.
    1. Hayden MS, Ghosh S (2008) Shared principles in NF-kappaB signaling. Cell 132:344–362.
    1. Chen LF, Greene WC (2004) Shaping the nuclear action of NF-kappaB. Nat Rev Mol Cell Biol 5:392–401.
    1. Li W, Zheng S, Tang C, Zhu Y, Wang X (2007) JNK-AP-1 pathway involved in interleukin-1beta-induced calcitonin gene-related peptide secretion in human type II alveolar epithelial cells. Peptides 28:1252–1259.
    1. Shukla A, Hashiguchi N, Chen Y, Coimbra R, Hoyt DB, et al. (2004) Osmotic regulation of cell function and possible clinical applications. Shock 21:391–400.
    1. Buttmann M, Berberich-Siebelt F, Serfling E, Rieckmann P (2007) Interferon-beta is a potent inducer of interferon regulatory factor-1/2-dependent IP-10/CXCL10 expression in primary human endothelial cells. J Vasc Res 44:51–60.
    1. Yarilina A, Park-Min KH, Antoniv T, Hu X, Ivashkiv LB (2008) TNF activates an IRF1-dependent autocrine loop leading to sustained expression of chemokines and STAT1-dependent type I interferon-response genes. Nat Immunol 9:378–387.
    1. dos Santos CC, Han B, Andrade CF, Bai X, Uhlig S, et al. (2004) DNA microarray analysis of gene expression in alveolar epithelial cells in response to TNFalpha, LPS, and cyclic stretch. Physiol Genomics 19:331–342.
    1. Rose CE Jr, Sung SS, Fu SM (2003) Significant involvement of CCL2 (MCP-1) in inflammatory disorders of the lung. Microcirculation 10:273–288.
    1. Fehrenbach H (2001) Alveolar epithelial type II cell: defender of the alveolus revisited. Respir Res 2:33–46.
    1. Henriquet C, Gougat C, Combes A, Lazennec G, Mathieu M (2007) Differential regulation of RANTES and IL-8 expression in lung adenocarcinoma cells. Lung Cancer 56:167–174.
    1. Ochi H, Masuda J, Gimbrone MA (2002) Hyperosmotic stimuli inhibit VCAM-1 expression in cultured endothelial cells via effects on interferon regulatory factor-1 expression and activity. Eur J Immunol 32:1821–1831.
    1. Guggino WB (1999) Cystic fibrosis and the salt controversy. Cell 96:607–610.
    1. Tarran R, Donaldson S, Boucher RC (2007) Rationale for hypertonic saline therapy for cystic fibrosis lung disease. Semin Respir Crit Care Med 28:295–302.

Source: PubMed

3
Suscribir