Skip to main content
Log in

Therapeutic treatment of dietary docosahexaenoic acid for particle-induced pulmonary inflammation in Balb/c mice

  • Original Research Paper
  • Published:
Inflammation Research Aims and scope Submit manuscript

Abstract

Objective and design

The omega-3 polyunsaturated fatty acid docosahexaenoic acid (DHA) has been reported to suppress inflammation. Pulmonary inflammation can be directly linked to exposure of various occupational and man-made particles leading to pulmonary diseases. Therapeutic treatments are lacking for particle-induced pulmonary inflammation. These studies evaluated DHA as a therapeutic treatment for semi-acute and chronic particle-induced pulmonary inflammation.

Methods

Balb/c mice were oropharyngeal instilled with hydrophobic multi-walled carbon nanotube (MWCNT) or hydrophilic crystalline silica (SiO2) either as one instillation (semi-acute) or once a week for 4 weeks (chronic). One week later, the mice were placed on either a control or 1% DHA-containing diet for 3 weeks (semi-acute) or 12 weeks (chronic). Mice were assessed for inflammatory signaling within the lung lavage fluid, impact on phagolysosomal membrane permeability, shifts of macrophage phenotype gene expression (M1, M2a, M2b, and M2c), and pulmonary histopathology.

Results

DHA increased pulmonary inflammatory markers and lung pathology when mice were exposed to SiO2. There were trending decreases of inflammatory markers for MWCNT-exposed mice with DHA treatment, however, mostly not statistically significant.

Conclusion

The anti-inflammatory benefits of DHA treatment depend upon the type of inflammatory particle, magnitude of inflammation, and duration of treatment.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10

Similar content being viewed by others

Availability of data and materials

Data and material will be provided by the authors if requested.

References

  1. Calder PC. Omega-3 fatty acids and inflammatory processes: from molecules to man. Biochem Soc Trans. 2017;45:1105–15. https://doi.org/10.1042/BST20160474.

    Article  CAS  PubMed  Google Scholar 

  2. Molendi-Coste O, Legry V, Leclercq IA. Why and how meet n-3 PUFA dietary recommendations? Gastroenterol Res Pract. 2011. https://doi.org/10.1155/2011/364040.

    Article  PubMed  Google Scholar 

  3. Swanson D, Block R, Mousa SA. Omega-3 fatty acids EPA and DHA: health benefits throughout life. Adv Nutr. 2012;3:1–7. https://doi.org/10.3945/an.111.000893.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Bates MA, Brandenberger C, Langohr II, Kumagai K, Lock AL, Harkema JR, Holian A, Pestka JJ. Silica-triggered autoimmunity in lupus-prone mice blocked by docosahexaenoic acid consumption. PLoS ONE. 2016;11(8):e0160622. https://doi.org/10.1371/journal.pone.0160622.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Li X-Y, Hao L, Liu Y-H, Chen C-Y, Pai VJ, Kang JX. Protection against fine particle-induced pulmonary and systemic inflammation by omega-3 polyunsaturated fatty acids. Biochim Biophys Acta. 2017;1861:577–84. https://doi.org/10.1016/j.bbagen.2016.12.018.

    Article  CAS  Google Scholar 

  6. Forum of International Respiratory Societies. The global impact of respiratory disease. 2nd ed. Sheffield: European Respiratory Society; 2017.

    Google Scholar 

  7. Wong J, Magun BE, Wood LJ. Lung inflammation caused by inhaled toxicants: a review. Int J Chronic Obstr Pulm Dis. 2016;11:1391–401. https://doi.org/10.2147/COPD.S106009.

    Article  CAS  Google Scholar 

  8. Lam C, James JT, McCluskey R, Arepalli S, Hunter RL. A review of carbon nanotube toxicity and assessment of potential occupational and environmental health risks. Crit Rev Toxicol. 2006;36:189–217. https://doi.org/10.1080/10408440600570233.

    Article  CAS  PubMed  Google Scholar 

  9. Ray JL, Holian A. Sex differences in the inflammatory immune response to multi-walled carbon nanotubes and crystalline silica. Inhal Toxicol. 2019;31(7):285–97. https://doi.org/10.1080/08958378.2019.1669743.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Pollard KM. Silica, silicosis, and autoimmunity. Front Immunol. 2016. https://doi.org/10.3389/fimmu.2016.00097.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Pavan C, Santalucia R, Leinardi R, Fabbiani M, Yakoub Y, Fubini B, et al. Nearly free surface silanols are the critical molecular moieties that initiate the toxicity of silica particles. Proc Natl Acad Sci. 2020;117(45):27836–46. https://doi.org/10.1073/pnas.2008006117.

    Article  CAS  PubMed  Google Scholar 

  12. Watanabe H, Numata K, Ito T, Takagi K, Matsukawa A. Innate immune response in TH1- and TH2-dominant mouse strains. Shock. 2004;22(5):460–6. https://doi.org/10.1097/01.shk.0000142249.08135.e9.

    Article  CAS  PubMed  Google Scholar 

  13. Limjunyawong N, Craig JM, Lagassé HAD, Scott AL, Mitzner W. Experimental progressive emphysema in BALB/cJ mice as a model for chronic alveolar destruction in humans. Am J Physiol Lung Cell Mol Physiol. 2015;309:L662–76. https://doi.org/10.1152/ajplung.00214.2015.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Sahu N, Morales JL, Fowell D, August A. Modeling susceptibility versus resistance in allergic airway disease reveals regulation by Tec kinase Itk. PLoS ONE. 2010;5(6):e11348. https://doi.org/10.1371/journal.pone.0011348.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hamilton RF, Buford M, Xiang C, Wu N, Holian A. NLRP3 inflammasome activation in murine alveolar macrophages and related lung pathology is associated with MWCNT nickel contamination. Inhal Toxicol. 2012;24(14):995–1008. https://doi.org/10.3109/08958378.2012.745633.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Thakur SA, Hamilton R, Pikkarainen T, Holian A. Differential binding of inorganic particles to MARCO. Toxicol Sci. 2009;107(1):238–46. https://doi.org/10.1093/toxsci/kfn210.

    Article  CAS  PubMed  Google Scholar 

  17. EFSA Nda Panel (EFSA Panel on Dietetic Products, Nutrition and Allergies). Scientific Opinion on the extension of use for DHA and EPA-rich algal oil from Schizochytrium sp as a novel food ingredient. Eur Food Saf Auth J. 2014;12(10):3843. https://doi.org/10.2903/j.efsa.2014.3843.

    Article  CAS  Google Scholar 

  18. Jessop F, Hamilton RF, Rhoderick JF, Fletcher P, Holian A. Phagolysosome acidification is required for silica and engineered nanoparticle-induced lysosome membrane permeabilization and resultant NLRP3 inflammasome activity. Toxicol Appl Pharmacol. 2017;318:58–68. https://doi.org/10.1016/j.taap.2017.01.012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Jessop F, Holian A. Extracellular HMGB1 regulates multi-walled carbon nanotube-induced inflammation in vivo. Nanotoxicology. 2015;9(3):365–72. https://doi.org/10.3109/17435390.2014.933904.

    Article  CAS  PubMed  Google Scholar 

  20. Burmeister R, Rhoderick JF, Holian A. Prevention of crystalline silica-induced inflammation by the anti-malarial hydroxychloroquine. Inhal Toxicol. 2019;31(7):274–84. https://doi.org/10.1080/08958378.2019.1668091.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Fletcher P, Hamilton RF, Rhoderick JF, Pestka JJ, Holian A. Docosahexaenoic acid impacts macrophage phenotype subsets and phagolysosomal membrane permeability with particle exposure. J Toxicol Environ Health Part A. 2020. https://doi.org/10.1080/15287394.2020.1842826.

    Article  Google Scholar 

  22. Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140:821–32. https://doi.org/10.1016/j.cell.2010.01.040.

    Article  CAS  Google Scholar 

  23. Strowig T, Henao-Mejia J, Elinav E, Flavell R. Inflammasomes in health and disease. Nature. 2012;481:278–86. https://doi.org/10.1038/nature10759.

    Article  CAS  PubMed  Google Scholar 

  24. Labonte AC, Tosello-Trampont A-C, Hahn YS. The role of macrophage polarization in infectious and inflammatory diseases. Mol Cells. 2014;37(4):275–85. https://doi.org/10.14348/molcells.2014.2374.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Italiani P, Boraschi D. From monocytes to M1/M2 macrophages: phenotypical vs. functional differentiation. Front Immunol. 2014. https://doi.org/10.3389/fimmu.2014.00514.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Byrne AJ, Mathie SA, Gregory LG, Lloyd CM. Pulmonary macrophages: Key players in the innate defence of the airways. Thorax. 2015;70:1189–96. https://doi.org/10.1136/thoraxjnl-2015-207020.

    Article  PubMed  Google Scholar 

  27. Saifuddin N, Raziah AZ, Junizah AR. Carbon nanotubes: a review on structure and their interaction with proteins. J Chem. 2013. https://doi.org/10.1155/2013/676815.

    Article  Google Scholar 

  28. Dyachenko AG, Borysenko MV, Pakhovchyshyn SV. Hydrophilic/hydrophobic properties of silica surfaces modified with metal oxides and polydimethylsiloxane. Adsorpt Sci Technol. 2004;22(6):511–6. https://doi.org/10.1260/0263617042879546.

    Article  CAS  Google Scholar 

  29. Turk HF, Chapkin RS. Membrane lipid raft organization is uniquely modified by n-3 polyunsaturated fatty acids. Prostaglandins Leukot Essent Fatty Acids. 2013;88(1):43–7. https://doi.org/10.1016/j.plefa.2012.03.008.

    Article  CAS  PubMed  Google Scholar 

  30. Wassall SR, Leng X, Canner SW, Pennington ER, Kinnun JJ, Cavazos AT, Dadoo S, Johnson D, Heberle FA, Katsaras J. Shaikh SR Docosahexaenoic acid regulates the formation of lipid rafts: a unified view from experiment and simulation. Biochim Biophys Acta Biomembr. 2018;1860(10):1985–93. https://doi.org/10.1016/j.bbamem.2018.04.016.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Stunault MI, Bories G, Guinamard RR, Ivanov S. Metabolism plays a key role during macrophage activation. Mediat Inflamm. 2018. https://doi.org/10.1155/2018/2426138.

    Article  Google Scholar 

  32. Li R, Qiu X, Xu F, Lin Y, Fang Y, Zhu T. Macrophage-mediated effects of airborne fine particulate matter (PM2.5) on hepatocyte insulin resistance in vitro. ACS Omega. 2016;1:736–43. https://doi.org/10.1021/acsomega.6b00135.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Finucane OM, Sugrue J, Rubio-Araiz A, Guillot-Sestier M-V, Lynch MA. The NLRP3 inflammasome modulates glycolysis by increasing PFKFB3 in an IL-1β-dependent manner in macrophages. Sci Rep. 2019;9:4034. https://doi.org/10.1038/s41598-019-40619-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ali M, Heyob K, Rogers LK. DHA suppresses primary macrophage inflammatory responses via notch 1/ jagged 1 signaling. Sci Rep. 2016;6:22276. https://doi.org/10.1038/srep22276.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Benmoussa K, Garaude J, Acín-Pérez R. How mitochondrial metabolism contributes to macrophage phenotype and functions. J Mol Biol. 2018;430:3906–21. https://doi.org/10.1016/j.jmb.2018.07.003.

    Article  CAS  PubMed  Google Scholar 

  36. Shen L, Yang Y, Ou T, Key C-CC, Tong SH, Zhu X, et al. Dietary PUFAs attenuate NLRP3 inflammasome activation via enhancing macrophage autophagy. J Lipid Res. 2017;58:1808–21. https://doi.org/10.1194/jlr.M075879.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Saitoh T, Fujita N, Jang MH, Uematsu S, Yang B-G, Akira S, et al. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1β production. Nature. 2008;456:264–8. https://doi.org/10.1038/nature07383.

    Article  CAS  PubMed  Google Scholar 

  38. Williams-Bey Y, Boularan C, Vural A, Huang N-N, Hwang I-Y, Shan-Shi C, Kehrl JH. Omega-3 Free fatty acids suppress macrophage inflammasome activation by inhibiting NF-κB activation and enhancing autophagy. PLoS ONE. 2014;9(6):e97957. https://doi.org/10.1371/journal.pone.0097957.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kawano A, Ariyoshi W, Yoshioka Y, Hikiji H, Nishihara T, Okinaga T. Docosahexaenoic acid enhances M2 macrophage polarization via the p38 signaling pathway and autophagy. J Cell Biochem. 2019;120:12604–17. https://doi.org/10.1002/jcb.28527.

    Article  CAS  PubMed  Google Scholar 

  40. Jessop F, Hamilton RF, Rhoderick JF, Shaw PK, Holian A. Autophagy deficiency in macrophages enhances NLRP3 inflammasome activity and chronic lung disease following silica exposure. Toxicol Appl Pharmacol. 2016;309:101–10. https://doi.org/10.1016/j.taap.2016.08.029.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors would like to thank the technical support from the CEHS Core Facilities: Inhalation and Pulmonary Physiology Core, Molecular Histology and Fluorescence Imaging Core, and the Fluorescence Cytometry Core. A special thank you to: Dr. Joanna Kreitinger at Dermaxon and Dr. Sarjubhai Patel at FYR Diagnostics for use of their 384-well CFX Maestro’s; Lou Herritt, and Pamela Shaw within the CEHS Core facilities; UM’s Laboratory Animal Resources technicians and facility; and Iheanyi Amadi for help with lung airway thickness analysis.

Funding

Paige Fletcher was supported by the Ruth L. Kirschstein NRSA Pre-doctoral Fellowship from the National Institute of Environmental Health Sciences (F31 ES028100). This research was supported by grants from the National Institute of Environmental Health Sciences (R01 ES023209 and R01 ES027353) and National Institute of General Medical Sciences (P30 GM103338). The content within is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. Paige Fletcher was awarded one of QIAGEN’s featured young scientists of the month (November 2018) where she received QIAGEN products that contributed to this research.

Author information

Authors and Affiliations

Authors

Contributions

PF designed and carried out the in vivo studies, analyzed the in vivo and ex vivo studies, and performed statistical analysis. PF wrote the first draft of the manuscript. RFH setup the ex vivo studies, assisted in lung pathology scoring, and provided statistical advice. JFR assisted with mRNA quantification by qPCR and provided qPCR advice. BP and MB assisted PF with the in vivo studies. JJP assisted PF with logistics of the in vivo studies and supplied the DHA microalgal oil within the diets. AH assisted PF with overall study design and coordination. All authors contributed to furthering the manuscript’s drafts and approved the final manuscript.

Corresponding author

Correspondence to Paige Fletcher.

Ethics declarations

Conflict of interest

The authors have no conflicts of interest or competing interests to declare.

Ethical approval

The animal use protocol (035-16AHCEHS-062816) was approved by the University of Montana Institutional Animal Care and Use Committee for all mouse studies described within this manuscript. The mice are maintained in microisolation containers within the BSL-2 Laboratory Animal Resources facility at the University of Montana in the accordance with the Guide for the Care and Use of Laboratory Animals. The animal care facility at the University of Montana is staffed with full-time veterinarians that are AAALAC accredited. Mice were monitored on a daily basis along with during/after exposure to particles. Mice were anesthetized with isoflurane before particle or vehicle control instillations so as not to use any restraints or cause distress. All procedures within these studies caused minimal discomfort to the mice; however, in any cases where it was deemed that the mice were in pain or distress (adverse body weight, abnormal activity, poor grooming, abnormal posture) the animal was humanely euthanized.

Additional information

Responsible Editor: John Di Battista.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (PDF 805 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fletcher, P., Hamilton, R.F., Rhoderick, J.F. et al. Therapeutic treatment of dietary docosahexaenoic acid for particle-induced pulmonary inflammation in Balb/c mice. Inflamm. Res. 70, 359–373 (2021). https://doi.org/10.1007/s00011-021-01443-4

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00011-021-01443-4

Keywords

Navigation