Next Article in Journal
The Degradation Process of Typical Neonicotinoid Insecticides in Tidal Streams in Subtropical Cities: A Case Study of the Wuchong Stream, South China
Next Article in Special Issue
Association between Residential Exposure to Air Pollution and Incident Coronary Heart Disease Is Not Mediated by Leukocyte Telomere Length: A UK Biobank Study
Previous Article in Journal
Coupling Environmental Whole Mixture Toxicity Screening with Unbiased RNA-Seq Reveals Site-Specific Biological Responses in Zebrafish
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Commentary

On the Need for Human Studies of PM Exposure Activation of the NLRP3 Inflammasome

1
Department of Public Health Sciences, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
2
Institute for Community Research, Hartford, CT 06106, USA
3
Somerville Transportation Equity Partnership, Somerville, MA 02145, USA
*
Author to whom correspondence should be addressed.
Toxics 2023, 11(3), 202; https://doi.org/10.3390/toxics11030202
Submission received: 12 December 2022 / Revised: 5 February 2023 / Accepted: 16 February 2023 / Published: 21 February 2023

Abstract

:
Particulate matter air pollution is associated with blood inflammatory biomarkers, however, the biological pathways from exposure to periferal inflammation are not well understood. We propose that the NLRP3 inflammasome is likely stimulated by ambient particulate matter, as it is by some other particles and call for more research into this pathway.

Research that we have helped lead, focused on associations of ultrafine particles (UFPs; particles with an aerodynamic diameter of < 100 nm) with inflammatory biomarkers in peripheral blood, has evolved naturally into an interest in research on inflammatory mechanisms [1,2,3]. In research on both UFP exposure and in the broader literature on particulate matter (PM) generally, especially on PM2.5 (PM of < 2.5 um), there are many reports of associations with biomarkers of inflammation, such as C-reactive protein (CRP) and interleukin-6 (IL-6) [4,5,6]. This has led to low-level, chronic systemic inflammation becoming a leading hypothesis for the underlying mechanism by which PM influence the development of adverse cardiovascular outcomes [7] as well as respiratory and neurological diseases [8].
The pathways that drive inflammatory responses are relatively well-elucidated (Figure 1). Because IL-6 promotes increases in CRP, we have long seen that associations of UFPs with both biomarkers in peripheral blood are quite similar. While our research teams have not studied the earlier steps in this pathway for UFPs, it is known that IL-6 levels are, in turn, stimulated by interleukin-1beta (IL-1beta), which has been activated by inflammasomes [9].
The NLRP3 inflammasome (nucleotide-binding oligomerization domain-like receptor (NLR) family pyrin domain-containing 3) is of special interest because it has been established that silica, uric acid, cholesterol crystals, and other particles stimulate NLRP3, leading to inflammation [11,12,13]. Since these forms of PM activate inflammation via NLRP3, it is possible that inflammation generated by ambient PM also acts through NLRP3. While there are many other inflammasomes, they respond highly specifically to infectious organisms.
Thus, we hypothesize that UFP, PM2.5, and other ambient combustion PM are acting through NLRP3 to generate what is often called “sterile inflammation”; that is, inflammation without infectious agents, such as viruses or bacteria. The specific hypothesis would be that UFPs somehow activate NLRP3, which then generates IL-1beta and IL-18, leading to the production of IL-6 and, subsequently, CRP, both of which many have found to be associated with combustion PM. The resulting inflammation could then be associated with a wide range of diseases, including those that are cardiovascular, respiratory, and neurological.
In recent years, there has been increased laboratory-based research on PM and the activation of NLRP3, mostly in China. A cursory review of the literature turns up many relevant studies. A more systematic review would be warranted.
Numerous studies in cell lines have reported the activation of NLRP3 and associated cytokines by PM2.5 [14,15,16,17,18,19,20] and, in one study, PM10 [21]. There has also been a similar output of studies on PM2.5 inducing NLRP3 in animal and rodent models [9,22,23,24,25,26,27,28]. A smaller number of reports suggested that there were factors that could reverse the activation of NLRP3 by PM [29,30]. Studies on UFPs are much rarer so far, with one study that we found reporting that black carbon nano PM could activate NLRP3 [31].
It is notable how recently these studies appeared, and it is likely that many more are in the pipeline. It is also critical to consider that, to our knowledge, the epidemiology of PM, and especially UFPs, effects on NLRP3 in humans is virtually non-existent.
Given the sparseness of data on human populations, it is interesting to consider the research of Ridker and colleagues, who explored the benefits of suppressing IL-1beta production with canakinumab in a large, randomized clinical trial. They found reduced cardiovascular events in the treatment arms independent of and separate from lipid reductions by medications such as statins; however, they also found meaningful levels of residual inflammation even after treatment [32,33].
They suggest, plausibly in our opinion, that it might be necessary to inhibit inflammation at an earlier stage than IL-1beta, which would be at the level of inflammasomes; for PM, that would mean the NLRP3 inflammasome. Since their findings point to a critical role of inflammasomes in illness secondary to chronic inflammation, we suggest that there is a need for further epidemiological research on PM, including UFPs, and NLRP3 activation in human populations.
A deeper understanding of the pathway by which PM generates a low burn of inflammation would be useful in multiple ways: First, it would add to the science behind the mechanisms by which PM cause adverse health outcomes, further cementing the causal nature of the association for PM2.5 and establishing a causal case for UFPs. Second, as with Ridker’s work on canakinumab as a therapeutic agent, it might be that inhibitors of NLRP3 activation could be protective against adverse effects of breathing in ambient PM.
One possibility is that some traditional Chinese medicines could be protective. There is considerable evidence that these medicines act through NLRP3 [34]. More than a dozen heat-clearing and detoxifying herbs, such as Scutellaria baicalensis, Coptis chinensis, Flos lonicerae, Forsythia suspensa, Radix isatidis, and Houttuynia cordata, have been reported as having anti-inflammatory and antimicrobial functions [35,36]. Besides being ingested as traditional medicine, anti-inflammatory herbs are also commonly made in easily accessible tea bags (e.g., Flos lonicerae, Radix isatidis, gingko, and ginseng) or found in the kitchen (e.g., ginger, Portulaca oleracea, cinnamon, red peppers, orange peel, and Houttuynia cordata), being used very frequently but not necessarily being perceived as herbal medicine [37,38].
In addition, some studies show that acupuncture may also have an anti-inflammatory effect [39,40,41,42]. The conscious use of traditional medicine for healing and wellness, or unconscious cultural practices in daily life, may play a protective role. This possibility is supported by data that we collected from Asian immigrants, which suggest reduced immune responses to UFP exposure [5,43].
Ultimately, greater knowledge about how PM activates the NLRP3 inflammasome could help us understand which fractions of PM are the most toxic, which would, in turn, suggest priorities for reducing sources of exposure.

Author Contributions

D.B. took the lead on writing, incorporating lessons from his leadership of multiple studies on UFPs and health in near-highway populations. J.L. reviewed and approved the manuscript, as well as writing the section on traditional Chinese medicine. W.Z. brought the biology of the NLRP3 inflammasome to the attention of the research teams, helped us understand the relevant literature, and reviewed as well as approved this manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work is supported by NIEHS (R01-ES026980 and R01-ES030289).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank the rest of the members of our research teams over the last 15 years, too many to name them all, without whom we would not have our current understanding that led us to see the need for this line of research.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lane, K.J.; Scammell, M.K.; Levy, J.I.; Fuller, C.H.; Parambi, R.; Zamore, W.; Mwamburi, M.; Brugge, D. Positional error and time-activity patterns in near-highway proximity studies: An exposure misclassification analysis. Environ. Health 2013, 12, 75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Walker, D.I.; Lane, K.J.; Liu, K.; Uppal, K.; Patton, A.P.; Durant, J.L.; Jones, D.P.; Brugge, D.; Pennell, K.D. Metabolomic assessment of exposure to near-highway ultrafine particles. J. Expo. Sci. Environ. Epidemiol. 2019, 29, 469–483. [Google Scholar] [CrossRef] [PubMed]
  3. Nephew, B.C.; Nemeth, A.; Hudda, N.; Beamer, G.; Mann, P.; Petitto, J.; Cali, R.; Febo, M.; Kulkarni, P.; Poirier, G.; et al. Traffic-related particulate matter affects behavior, inflammation, and neural integrity in a developmental rodent model. Environ. Res. 2020, 183, 109242. [Google Scholar] [CrossRef] [PubMed]
  4. Liu, Q.; Gu, X.; Deng, F.; Mu, L.; Baccarelli, A.A.; Guo, X.; Wu, S. Ambient particulate air pollution and circulating C-reactive protein level: A systematic review and meta-analysis. Int. J. Hyg. Environ. Health 2019, 222, 756–764. [Google Scholar] [CrossRef] [PubMed]
  5. Lane, K.J.; Levy, J.I.; Scammell, M.K.; Peters, J.L.; Patton, A.P.; Reisner, E.; Lowe, L.; Zamore, W.; Durant, J.L.; Brugge, D. Association of modeled long-term personal exposure to ultrafine particles with inflammatory and coagulation biomarkers. Environ. Int. 2016, 92, 173–182. [Google Scholar] [CrossRef] [Green Version]
  6. Lane, K.J.; Levy, J.I.; Patton, A.P.; Durant, J.L.; Zamore, W.; Brugge, D. Relationship between traffic-related air pollution and inflammation biomarkers using structural equation modelling. Sci. Total Environ. 2023, 870, 161874. [Google Scholar] [CrossRef]
  7. Brook, R.D.; Rajagopalan, S.; Pope III, C.A.; Brook, J.R.; Bhatnagar, A.; Diez-Roux, A.V.; Holguin, F.; Hong, Y.; Luepker, R.V.; Mittleman, M.A. Particulate matter air pollution and cardiovascular disease: An update to the scientific statement from the American Heart Association. Circulation 2010, 121, 2331–2378. [Google Scholar] [CrossRef] [Green Version]
  8. Cohen, A.J.; Brauer, M.; Burnett, R.; Anderson, H.R.; Frostad, J.; Estep, K.; Balakrishnan, K.; Brunekreef, B.; Dandona, L.; Dandona, R. Estimates and 25-year trends of the global burden of disease attributable to ambient air pollution: An analysis of data from the Global Burden of Diseases Study 2015. Lancet 2017, 389, 1907–1918. [Google Scholar] [CrossRef] [Green Version]
  9. Duan, S.; Wang, N.; Huang, L.; Zhao, Y.; Shao, H.; Jin, Y.; Zhang, R.; Li, C.; Wu, W.; Wang, J. NLRP3 inflammasome activation is associated with PM2. 5-induced cardiac functional and pathological injury in mice. Environ. Toxicol. 2019, 34, 1246–1254. [Google Scholar] [CrossRef]
  10. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 inflammasome: An overview of mechanisms of activation and regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [Green Version]
  11. Swanson, K.V.; Deng, M.; Ting, J.P.-Y. The NLRP3 inflammasome: Molecular activation and regulation to therapeutics. Nat. Rev. Immunol. 2019, 19, 477–489. [Google Scholar] [CrossRef] [PubMed]
  12. Leso, V.; Fontana, L.; Iavicoli, I. Nanomaterial exposure and sterile inflammatory reactions. Toxicol. Appl. Pharmacol. 2018, 355, 80–92. [Google Scholar] [CrossRef] [PubMed]
  13. Kim, S.-K.; Choe, J.-Y.; Park, K.-Y. Anti-inflammatory effect of artemisinin on uric acid-induced NLRP3 inflammasome activation through blocking interaction between NLRP3 and NEK7. Biochem. Biophys. Res. Commun. 2019, 517, 338–345. [Google Scholar] [CrossRef] [PubMed]
  14. Gao, L.; Qin, J.X.; Shi, J.Q.; Jiang, T.; Wang, F.; Xie, C.; Gao, Q.; Zhi, N.; Dong, Q.; Guan, Y.T. Fine particulate matter exposure aggravates ischemic injury via NLRP3 inflammasome activation and pyroptosis. CNS Neurosci. Ther. 2022, 28, 1045–1058. [Google Scholar] [CrossRef] [PubMed]
  15. Niu, L.; Li, L.; Xing, C.; Luo, B.; Hu, C.; Song, M.; Niu, J.; Ruan, Y.; Sun, X.; Lei, Y. Airborne particulate matter (PM2. 5) triggers cornea inflammation and pyroptosis via NLRP3 activation. Ecotoxicol. Environ. Saf. 2021, 207, 111306. [Google Scholar] [CrossRef] [PubMed]
  16. Tien, C.-P.; Chen, C.-H.; Lin, W.-Y.; Liu, C.-S.; Liu, K.-J.; Hsiao, M.; Chang, Y.-C.; Hung, S.-C. Ambient particulate matter attenuates Sirtuin1 and augments SREBP1-PIR axis to induce human pulmonary fibroblast inflammation: Molecular mechanism of microenvironment associated with COPD. Aging 2019, 11, 4654. [Google Scholar] [CrossRef] [PubMed]
  17. Xu, F.; Qiu, X.; Hu, X.; Shang, Y.; Pardo, M.; Fang, Y.; Wang, J.; Rudich, Y.; Zhu, T. Effects on IL-1β signaling activation induced by water and organic extracts of fine particulate matter (PM2.5) in vitro. Environ. Pollut. 2018, 237, 592–600. [Google Scholar] [CrossRef]
  18. Zheng, R.; Tao, L.; Jian, H.; Chang, Y.; Cheng, Y.; Feng, Y.; Zhang, H. NLRP3 inflammasome activation and lung fibrosis caused by airborne fine particulate matter. Ecotoxicol. Environ. Saf. 2018, 163, 612–619. [Google Scholar] [CrossRef]
  19. Zheng, R.; Zhang, J.; Han, X.; Wu, Y.; Yan, J.; Song, P.; Wang, Y.; Wu, X.; Zhang, H. Particulate matter aggravates Alzheimer’s disease by activating the NLRP3 inflammasome to release ASC specks. Environ. Sci. Nano 2021, 8, 2177–2190. [Google Scholar] [CrossRef]
  20. Zhong, Y.; Wang, Y.; Zhang, C.; Hu, Y.; Sun, C.; Liao, J.; Wang, G. Identification of long non-coding RNA and circular RNA in mice after intra-tracheal instillation with fine particulate matter. Chemosphere 2019, 235, 519–526. [Google Scholar] [CrossRef]
  21. Chan, Y.L.; Wang, B.; Chen, H.; Ho, K.F.; Cao, J.; Hai, G.; Jalaludin, B.; Herbert, C.; Thomas, P.S.; Saad, S. Pulmonary inflammation induced by low-dose particulate matter exposure in mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 2019, 317, L424–L430. [Google Scholar] [CrossRef] [PubMed]
  22. Du, X.; Jiang, S.; Zeng, X.; Zhang, J.; Pan, K.; Zhou, J.; Xie, Y.; Kan, H.; Song, W.; Sun, Q. Air pollution is associated with the development of atherosclerosis via the cooperation of CD36 and NLRP3 inflammasome in ApoE-/-mice. Toxicol. Lett. 2018, 290, 123–132. [Google Scholar] [CrossRef] [PubMed]
  23. Du, X.; Jiang, S.; Zeng, X.; Zhang, J.; Pan, K.; Song, L.; Zhou, J.; Kan, H.; Sun, Q.; Zhao, J. Fine particulate matter-induced cardiovascular injury is associated with NLRP3 inflammasome activation in Apo E-/-mice. Ecotoxicol. Environ. Saf. 2019, 174, 92–99. [Google Scholar] [CrossRef] [PubMed]
  24. Jiang, J.; Ding, S.; Zhang, G.; Dong, Y. Ambient particulate matter exposure plus a high-fat diet exacerbate renal injury by activating the NLRP3 inflammasome and TGF-β1/Smad2 signaling pathway in mice. Ecotoxicol. Environ. Saf. 2022, 238, 113571. [Google Scholar] [CrossRef] [PubMed]
  25. Jiang, J.; Zhang, G.; Yu, M.; Gu, J.; Zheng, Y.; Sun, J.; Ding, S. Quercetin improves the adipose inflammatory response and insulin signaling to reduce “real-world” particulate matter-induced insulin resistance. Environ. Sci. Pollut. Res. 2022, 29, 2146–2157. [Google Scholar] [CrossRef] [PubMed]
  26. Li, M.; Hua, Q.; Shao, Y.; Zeng, H.; Liu, Y.; Diao, Q.; Zhang, H.; Qiu, M.; Zhu, J.; Li, X. Circular RNA circBbs9 promotes PM2. 5-induced lung inflammation in mice via NLRP3 inflammasome activation. Environ. Int. 2020, 143, 105976. [Google Scholar] [CrossRef]
  27. Li, J.; Zhang, Y.; Zhang, L.; An, Z.; Song, J.; Wang, C.; Ma, Y.; Gu, Q.; Luo, Q.; Yang, W. Fine particulate matter exposure exacerbated nasal mucosal damage in allergic rhinitis mice via NLRP3 mediated pyroptosis. Ecotoxicol. Environ. Saf. 2021, 228, 112998. [Google Scholar] [CrossRef]
  28. Song, L.; Lei, L.; Jiang, S.; Pan, K.; Zeng, X.; Zhang, J.; Zhou, J.; Xie, Y.; Zhou, L.; Dong, C. NLRP3 inflammasome is involved in ambient PM2. 5-related metabolic disorders in diabetic model mice but not in wild-type mice. Inhal. Toxicol. 2021, 33, 260–267. [Google Scholar] [CrossRef]
  29. Ding, S.; Wang, H.; Wang, M.; Bai, L.; Yu, P.; Wu, W. Resveratrol alleviates chronic “real-world” ambient particulate matter-induced lung inflammation and fibrosis by inhibiting NLRP3 inflammasome activation in mice. Ecotoxicol. Environ. Saf. 2019, 182, 109425. [Google Scholar] [CrossRef]
  30. Lee, C.-W.; Chi, M.-C.; Hsu, L.-F.; Yang, C.-M.; Hsu, T.-H.; Chuang, C.-C.; Lin, W.-N.; Chu, P.-M.; Lee, I.-T. Carbon monoxide releasing molecule-2 protects against particulate matter-induced lung inflammation by inhibiting TLR2 and 4/ROS/NLRP3 inflammasome activation. Mol. Immunol. 2019, 112, 163–174. [Google Scholar] [CrossRef]
  31. Zhou, L.; Li, P.; Zhang, M.; Han, B.; Chu, C.; Su, X.; Li, B.; Kang, H.; Ning, J.; Zhang, B. Carbon black nanoparticles induce pulmonary fibrosis through NLRP3 inflammasome pathway modulated by miR-96 targeted FOXO3a. Chemosphere 2020, 241, 125075. [Google Scholar] [CrossRef] [PubMed]
  32. Ridker, P.M.; MacFadyen, J.G.; Thuren, T.; Libby, P. Residual inflammatory risk associated with interleukin-18 and interleukin-6 after successful interleukin-1β inhibition with canakinumab: Further rationale for the development of targeted anti-cytokine therapies for the treatment of atherothrombosis. Eur. Heart J. 2020, 41, 2153–2163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Ridker, P.M.; MacFadyen, J.G.; Everett, B.M.; Libby, P.; Thuren, T.; Glynn, R.J.; Kastelein, J.; Koenig, W.; Genest, J.; Lorenzatti, A. Relationship of C-reactive protein reduction to cardiovascular event reduction following treatment with canakinumab: A secondary analysis from the CANTOS randomised controlled trial. Lancet 2018, 391, 319–328. [Google Scholar] [CrossRef]
  34. Özenver, N.; Efferth, T. Phytochemical inhibitors of the NLRP3 inflammasome for the treatment of inflammatory diseases. Pharmacol. Res. 2021, 170, 105710. [Google Scholar] [CrossRef]
  35. Wang, Q.; Su, C.-P.; Zhang, H.-M.; Ren, Y.-L.; Wang, W.; Guo, S.-Z. Anti-inflammatory mechanism of heat-clearing and detoxifying Chinese herbs. China J. Chin. Mater. Med. 2018, 43, 3787–3794. [Google Scholar] [CrossRef]
  36. Muluye, R.A.; Bian, Y.; Alemu, P.N. Anti-inflammatory and antimicrobial effects of heat-clearing Chinese herbs: A current review. J. Tradit. Complement. Med. 2014, 4, 93–98. [Google Scholar] [CrossRef] [Green Version]
  37. Pan, M.-H.; Chiou, Y.-S.; Tsai, M.-L.; Ho, C.-T. Anti-inflammatory activity of traditional Chinese medicinal herbs. J. Tradit. Complement. Med. 2011, 1, 8–24. [Google Scholar] [CrossRef] [Green Version]
  38. Ichikawa, H.; Wang, X.; Konishi, T. Role of component herbs in antioxidant activity of shengmai san—A traditional Chinese medicine formula preventing cerebral oxidative damage in rat. Am. J. Chin. Med. 2003, 31, 509–521. [Google Scholar] [CrossRef]
  39. Zijlstra, F.J.; Van den Berg-de Lange, I.; Huygen, F.J.; Klein, J. Anti-inflammatory actions of acupuncture. Mediat. Inflamm. 2003, 12, 59–69. [Google Scholar] [CrossRef]
  40. McDonald, J.L.; Cripps, A.W.; Smith, P.K.; Smith, C.A.; Xue, C.C.; Golianu, B. The anti-inflammatory effects of acupuncture and their relevance to allergic rhinitis: A narrative review and proposed model. Evid. Based Complement. Altern. Med. 2013, 2013, 591796. [Google Scholar] [CrossRef] [Green Version]
  41. Kavoussi, B.; Ross, B.E. The neuroimmune basis of anti-inflammatory acupuncture. Integr. Cancer Ther. 2007, 6, 251–257. [Google Scholar] [CrossRef] [PubMed]
  42. Kim, S.K.; Bae, H. Acupuncture and immune modulation. Auton. Neurosci. 2010, 157, 38–41. [Google Scholar] [CrossRef] [PubMed]
  43. Corlin, L.; Woodin, M.; Thanikachalam, M.; Lowe, L.; Brugge, D. Evidence for the healthy immigrant effect in older Chinese immigrants: A cross-sectional study. BMC Public Health 2014, 14, 603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Pathways for NLRP3 inflammasome activation, illustrating priming (left) and activation (right). The interaction of particulate matter with these pathways is included in the upper-right corner (reproduced from Kelley et al., 2019 [10]).
Figure 1. Pathways for NLRP3 inflammasome activation, illustrating priming (left) and activation (right). The interaction of particulate matter with these pathways is included in the upper-right corner (reproduced from Kelley et al., 2019 [10]).
Toxics 11 00202 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Brugge, D.; Li, J.; Zamore, W. On the Need for Human Studies of PM Exposure Activation of the NLRP3 Inflammasome. Toxics 2023, 11, 202. https://doi.org/10.3390/toxics11030202

AMA Style

Brugge D, Li J, Zamore W. On the Need for Human Studies of PM Exposure Activation of the NLRP3 Inflammasome. Toxics. 2023; 11(3):202. https://doi.org/10.3390/toxics11030202

Chicago/Turabian Style

Brugge, Doug, Jianghong Li, and Wig Zamore. 2023. "On the Need for Human Studies of PM Exposure Activation of the NLRP3 Inflammasome" Toxics 11, no. 3: 202. https://doi.org/10.3390/toxics11030202

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop