Role of heparin in pulmonary cell populations in an in-vitro model of acute lung injury

Marta Camprubí-Rimblas, Raquel Guillamat-Prats, Thomas Lebouvier, Josep Bringué, Laura Chimenti, Manuela Iglesias, Carme Obiols, Jessica Tijero, Lluís Blanch, Antonio Artigas, Marta Camprubí-Rimblas, Raquel Guillamat-Prats, Thomas Lebouvier, Josep Bringué, Laura Chimenti, Manuela Iglesias, Carme Obiols, Jessica Tijero, Lluís Blanch, Antonio Artigas

Abstract

Background: In the early stages of acute respiratory distress syndrome (ARDS), pro-inflammatory mediators inhibit natural anticoagulant factors and initiate an increase in procoagulant activity. Previous studies proved the beneficial effects of heparin in pulmonary coagulopathy, which derive from its anticoagulant and anti-inflammatory activities, although it is uncertain whether heparin works. Understanding the specific effect of unfractioned heparin on cell lung populations would be of interest to increase our knowledge about heparin pathways and to treat ARDS.

Methods: In the current study, the effect of heparin was assessed in primary human alveolar macrophages (hAM), alveolar type II cells (hATII), and fibroblasts (hF) that had been injured with LPS.

Results: Heparin did not produce any changes in the Smad/TGFß pathway, in any of the cell types evaluated. Heparin reduced the expression of pro-inflammatory markers (TNF-α and IL-6) in hAM and deactivated the NF-kß pathway in hATII, diminishing the expression of IRAK1 and MyD88 and their effectors, IL-6, MCP-1 and IL-8.

Conclusions: The current study demonstrated that heparin significantly ameliorated the cells lung injury induced by LPS through the inhibition of pro-inflammatory cytokine expression in macrophages and the NF-kß pathway in alveolar cells. Our results suggested that a local pulmonary administration of heparin through nebulization may be able to reduce inflammation in the lung; however, further studies are needed to confirm this hypothesis.

Keywords: Acute Respiratory Distress Syndrome (ARDS); Alveolar cells; Alveolar macrophages; Anticoagulants; Fibroblasts; Inflammation.

Figures

Fig. 1
Fig. 1
Purity of isolated cells. hAM were stained with Diff quick to differentiate macrophages, neutrophils and lymphocytes and also CD68 immunofluorescence. hATII were stained with alkaline phosphatase and also with Surfactant C protein immunofluorescence. hF were stained with Diff Quick and ACTA2 immunofluorescence. Diff quick and alkaline phosphatase images had x400 magnification. Images of Hoechst, CD68, SPC and ACTA2 immunofluorescences had x600 magnification (hAM: human alveolar macrophages; hATII: human alveolar type II cells; hF: human fibroblasts, SPC: surfactant C protein and ACTA2: alpha smooth actin)
Fig. 2
Fig. 2
Human alveolar macrophages gene expression. a Expression of TNF-α, IL-6, IL-8, MCP-1, IRAK-1, MyD88, TGF-β, Smad2 and Smad3 evaluated by q-PCR at 7 h after LPS treatment. Data are expressed mean ± SEM (ΔCt correction was applied using GAPDH as a housekeeping gene and units are relative to the expression of control group) (n = 8 samples per group). b Protein expression for TNF-α, IL-6, IL-8, MCP-1 and TGF-β (n = 4 samples per group). Data are expressed mean ± SEM. *p ≤ 0.05 vs control groups; #p ≤ 0.05 vs LPS group c Immunofluorescence for NF-kß, IRAK-1, MyD88 and Smad2/3 and all the treatments are shown. Magnification is 400x. (ND: non-detectable; LPS: Lipopolysaccharide from Escherichia coli 055:B5 and HEP: unfractionated heparin)
Fig. 3
Fig. 3
Human alveolar type II cells gene expression. a Expression of TNF-α, IL-6, IL-8, MCP-1, IRAK-1, MyD88, TGF-β, Smad2 and Smad3 evaluated by q-PCR at 24 h after LPS treatment. Data are expressed mean ± SEM (ΔCt correction was applied using GAPDH as a housekeeping gene and units are relative to the expression of control group) (n = 8 samples per group). b Protein expression for TNF-α, IL-6, IL-8, MCP-1 and TGF-β (n = 4 samples per group). Data are expressed mean ± SEM. group c Immunofluorescence for NF-kß, IRAK-1, MyD88 and Smad2/3 and all the treatments are shown. Magnification is 400x.*p ≤ 0.05 vs control groups; #p ≤ 0.05 vs LPS group (LPS: Lipopolysaccharide from Escherichia coli 055:B5 and HEP: unfractionated heparin)
Fig. 4
Fig. 4
Human fibroblasts gene expression. a Expression of TNF-α, IL-6, IL-8, MCP-1, IRAK-1, MyD88, TGF-β, Smad2 and Smad3 evaluated by q-PCR at 24 h after LPS treatment. Data are expressed mean ± SEM (ΔCt correction was applied using GAPDH as a housekeeping gene and units are relative to the expression of control group) (n = 8 samples per group). b Protein expression for TNF-α, IL-6, IL-8, MCP-1 and TGF-β (n = 4 samples per group). Data are expressed mean ± SEM group c Immunofluorescence for NF-kß, IRAK-1, MyD88 and Smad2/3 and all the treatments are shown. Magnification is 400x. *p ≤ 0.05 vs control groups; #p ≤ 0.05 vs LPS group (LPS: Lipopolysaccharide from Escherichia coli 055:B5 and HEP: unfractionated heparin)
Fig. 5
Fig. 5
hAM, hATII cells and hF apoptosis/necrosis measured by Annexin V and PI staining with flow cytometry. The proportion of live cells (Annexin V-FITC-/PI-), early apoptotic cells (Annexin V-FITC+/PI-), necrotic cells (Annexin V-FITC-/PI+), late apoptotic/necrotic cells (Annexin V-FITC+/PI+). No changes were observed in LPS treated samples or in LPS + Hep treated ones (hAM:human alveolar macrophages; hATII: human alveolar type II cells; hF: human fibroblasts; FITC: Fluorescein; PI: propidium iodide; LPS: Lipopolysaccharide from Escherichia coli 055:B5 and HEP: unfractionated heparin)

References

    1. Ware LB, Bastarache JA, Wang L. Coagulation and fibrinolysis in human acute lung injury--new therapeutic targets? Keio J Med. 2005;54:142–9. doi: 10.2302/kjm.54.142.
    1. Hofstra J-JH, Juffermans NP, Schultz MJ, Zweers MM. Pulmonary coagulopathy as a new target in lung injury--a review of available pre-clinical models. Curr Med Chem. 2008;15:588–95. doi: 10.2174/092986708783769696.
    1. Choi G, Wolthuis EK, Bresser P, Levi M, van der Poll T, Dzoljic M, et al. Mechanical ventilation with lower tidal volumes and positive end-expiratory pressure prevents alveolar coagulation in patients without lung injury. Anesthesiology. 2006;105:689–95. doi: 10.1097/00000542-200610000-00013.
    1. Finigan JH. The coagulation system and pulmonary endothelial function in acute lung injury. Microvasc Res. 2009;77:35–8. doi: 10.1016/j.mvr.2008.09.002.
    1. dos Santos CC, Slutsky AS. Mechanotransduction, ventilator-induced lung injury and multiple organ dysfunction syndrome. Intensive Care Med. 2000;26:638–42. doi: 10.1007/s001340051217.
    1. Oeckler RA, Hubmayr RD. Ventilator-associated lung injury: a search for better therapeutic targets. Eur Respir J. 2007;30:1216–26. doi: 10.1183/09031936.00104907.
    1. Villar J, Blanco J, Añón JM, Santos-Bouza A, Blanch L, Ambrós A, et al. The ALIEN study: incidence and outcome of acute respiratory distress syndrome in the era of lung protective ventilation. Intensive Care Med. 2011;37:1932–41. doi: 10.1007/s00134-011-2380-4.
    1. Idell S. Anticoagulants for acute respiratory distress syndrome: can they work? Am J Respir Crit Care Med. 2001;164:517–20. doi: 10.1164/ajrccm.164.4.2102095.
    1. Tuinman PR, Dixon B, Levi M, Juffermans NP, Schultz MJ. Nebulized anticoagulants for acute lung injury - a systematic review of preclinical and clinical investigations. Crit Care Lond Engl. 2012;16:R70. doi: 10.1186/cc11325.
    1. Dixon B, Schultz MJ, Hofstra JJ, Campbell DJ, Santamaria JD. Nebulized heparin reduces levels of pulmonary coagulation activation in acute lung injury. Crit Care Lond Engl. 2010;14:445. doi: 10.1186/cc9269.
    1. Glas GJ, Serpa Neto A, Horn J, Cochran A, Dixon B, Elamin EM, et al. Nebulized heparin for patients under mechanical ventilation: an individual patient data meta-analysis. Ann Intensive Care. 2016;6:33. doi: 10.1186/s13613-016-0138-4.
    1. Jaimes F, De La Rosa G, Morales C, Fortich F, Arango C, Aguirre D, et al. Unfractioned heparin for treatment of sepsis: a randomized clinical trial (the HETRASE study) Crit Care Med. 2009;37:1185–96. doi: 10.1097/CCM.0b013e31819c06bc.
    1. Glas GJ, Muller J, Binnekade JM, Cleffken B, Colpaert K, Dixon B, et al. HEPBURN - investigating the efficacy and safety of nebulized heparin versus placebo in burn patients with inhalation trauma: study protocol for a multi-center randomized controlled trial. Trials. 2014;15:91. doi: 10.1186/1745-6215-15-91.
    1. Miller AC, Elamin EM, Suffredini AF. Inhaled anticoagulation regimens for the treatment of smoke inhalation-associated acute lung injury: a systematic review. Crit Care Med. 2014;42:413–9. doi: 10.1097/CCM.0b013e3182a645e5.
    1. Elsharnouby NM, Eid HEA, Abou Elezz NF, Aboelatta YA. Heparin/N-acetylcysteine: an adjuvant in the management of burn inhalation injury: a study of different doses. J Crit Care. 2014;29:182.e1-4. doi: 10.1016/j.jcrc.2013.06.017.
    1. Ledson M, Gallagher M, Hart CA, Walshaw M. Nebulized heparin in Burkholderia cepacia colonized adult cystic fibrosis patients. Eur Respir J. 2001;17:36–8. doi: 10.1183/09031936.01.17100360.
    1. Bendstrup KE, Gram J, Jensen JI. Effect of inhaled heparin on lung function and coagulation in healthy volunteers. Eur Respir J. 2002;19:606–10. doi: 10.1183/09031936.02.00105202.
    1. Li X, Zheng Z, Li X, Ma X. Unfractionated heparin inhibits lipopolysaccharide-induced inflammatory response through blocking p38 MAPK and NF-κB activation on endothelial cell. Cytokine. 2012;60:114–21. doi: 10.1016/j.cyto.2012.06.008.
    1. Fuchs S, Hollins AJ, Laue M, Schaefer UF, Roemer K, Gumbleton M, et al. Differentiation of human alveolar epithelial cells in primary culture: morphological characterization and synthesis of caveolin-1 and surfactant protein-C. Cell Tissue Res. 2003;311:31–45. doi: 10.1007/s00441-002-0653-5.
    1. Witherden IR, Vanden Bon EJ, Goldstraw P, Ratcliffe C, Pastorino U, Tetley TD. Primary human alveolar type II epithelial cell chemokine release: effects of cigarette smoke and neutrophil elastase. Am J Respir Cell Mol Biol. 2004;30:500–9. doi: 10.1165/rcmb.4890.
    1. Johnell M, Elgue G, Larsson R, Larsson A, Thelin S, Siegbahn A. Coagulation, fibrinolysis, and cell activation in patients and shed mediastinal blood during coronary artery bypass grafting with a new heparin-coated surface. J Thorac Cardiovasc Surg. 2002;124:321–32. doi: 10.1067/mtc.2002.122551.
    1. Bouma M, Maessen J, Weerwind P, Dentener M, Fransen E, de Jong D, et al. Release of lipopolysaccharide toxicity-modulating proteins in patients undergoing cardiopulmonary bypass using noncoated and heparin-coated extracorporeal circuits. A clinical pilot study. Chest. 1997;111:577–83. doi: 10.1378/chest.111.3.577.
    1. Ning F, Wang X, Shang L, Wang T, Lv C, Qi Z, et al. Low molecular weight heparin may prevent acute lung injury induced by sepsis in rats. Gene. 2015;557:88–91. doi: 10.1016/j.gene.2014.12.018.
    1. Puig F, Fuster G, Adda M, Blanch L, Farre R, Navajas D, et al. Barrier-protective effects of activated protein C in human alveolar epithelial cells. PLoS One. 2013;8:e56965. doi: 10.1371/journal.pone.0056965.
    1. Zarychanski R, Doucette S, Fergusson D, Roberts D, Houston DS, Sharma S, et al. Early intravenous unfractionated heparin and mortality in septic shock. Crit Care Med. 2008;36:2973–9. doi: 10.1097/CCM.0b013e31818b8c6b.
    1. Suter PM. Nebulised heparin: a new approach to the treatment of acute lung injury? Crit Care Lond Engl. 2008;12:170. doi: 10.1186/cc6947.
    1. Aird WC. The role of the endothelium in severe sepsis and multiple organ dysfunction syndrome. Blood. 2003;101:3765–77. doi: 10.1182/blood-2002-06-1887.
    1. Haeffner-Cavaillon N, Cavaillon JM, Moreau M, Szabó L. Interleukin 1 secretion by human monocytes stimulated by the isolated polysaccharide region of the Bordetella pertussis endotoxin. Mol Immunol. 1984;21:389–95. doi: 10.1016/0161-5890(84)90036-1.
    1. Raymondos K, Martin MU, Schmudlach T, Baus S, Weilbach C, Welte T, et al. Early alveolar and systemic mediator release in patients at different risks for ARDS after multiple trauma. Injury. 2012;43:189–95. doi: 10.1016/j.injury.2011.05.034.
    1. Aggarwal NR, King LS, D’Alessio FR. Diverse macrophage populations mediate acute lung inflammation and resolution. Am J Physiol Lung Cell Mol Physiol. 2014;306:L709–725. doi: 10.1152/ajplung.00341.2013.
    1. Bozza FA, Salluh JI, Japiassu AM, Soares M, Assis EF, Gomes RN, et al. Cytokine profiles as markers of disease severity in sepsis: a multiplex analysis. Crit Care Lond Engl. 2007;11:R49. doi: 10.1186/cc5783.
    1. Mu E, Ding R, An X, Li X, Chen S, Ma X. Heparin attenuates lipopolysaccharide-induced acute lung injury by inhibiting nitric oxide synthase and TGF-β/Smad signaling pathway. Thromb Res. 2012;129:479–85. doi: 10.1016/j.thromres.2011.10.003.
    1. Feng Y, Yang Q, Xu J, Qian G, Liu Y. Effects of HMGB1 on PMN apoptosis during LPS-induced acute lung injury. Exp Mol Pathol. 2008;85:214–22. doi: 10.1016/j.yexmp.2008.09.002.
    1. Puig F, Herrero R, Guillamat-Prats R, Gómez MN, Tijero J, Chimenti L, et al. A new experimental model of acid and endotoxin-induced acute lung injury in rats. Am J Physiol Lung Cell Mol Physiol. 2016. doi: 10.1152/ajplung.00390.2015.

Source: PubMed

3
Sottoscrivi