Tuberc Respir Dis.  2017 Jul;80(3):247-254. 10.4046/trd.2017.80.3.247.

ACN9 Regulates the Inflammatory Responses in Human Bronchial Epithelial Cells

Affiliations
  • 1Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea. pulmo2@kangwon.ac.kr
  • 2Environmental Health Center, Kangwon National University Hospital, Chuncheon, Korea.
  • 3Department of Pathology, Kangwon National University School of Medicine, Chuncheon, Korea.

Abstract

BACKGROUND
Airway epithelial cells are the first line of defense, against pathogens and environmental pollutants, in the lungs. Cellular stress by cadmium (Cd), resulting in airway inflammation, is assumed to be directly involved in tissue injury, linked to the development of lung cancer, and chronic obstructive pulmonary disease (COPD). We had earlier shown that ACN9 (chromosome 7q21), is a potential candidate gene for COPD, and identified significant interaction with smoking, based on genetic studies. However, the role of ACN9 in the inflammatory response, in the airway cells, has not yet been reported.
METHODS
We first checked the anatomical distribution of ACN9 in lung tissues, using mRNA in situ hybridization, and immunohistochemistry. Gene expression profiling in bronchial epithelial cells (BEAS-2B), was performed, after silencing ACN9. We further tested the roles of ACN9, in the intracellular mechanism, leading to Cd-induced production, of proinflammatory cytokines in BEAS-2B.
RESULTS
ACN9 was localized in lymphoid, and epithelial cells, of human lung tissues. ACN9 silencing, led to differential expression of 216 genes. Pathways of sensory perception to chemical stimuli, and cell surface receptor-linked signal transduction, were significantly enriched. ACN9 silencing, further increased the expression of proinflammatory cytokines, in BEAS-2B after Cd exposure.
CONCLUSION
Our findings suggest, that ACN9 may have a role, in the inflammatory response in the airway.

Keyword

Succinate Dehydrogenase; Cadmium; Pulmonary Disease, Chronic Obstructive; Gene Expression; Inflammation

MeSH Terms

Cadmium
Cytokines
Environmental Pollutants
Epithelial Cells*
Gene Expression
Gene Expression Profiling
Humans*
Immunohistochemistry
In Situ Hybridization
Inflammation
Lung
Lung Neoplasms
Pulmonary Disease, Chronic Obstructive
RNA, Messenger
Signal Transduction
Smoke
Smoking
Succinate Dehydrogenase
Cadmium
Cytokines
Environmental Pollutants
RNA, Messenger
Smoke
Succinate Dehydrogenase

Figure

  • Figure 1 ACN9 localization in human lung tissues. (A) In situ hybridization showing ACN9 mRNA localized in human lung tissues. Paraffin sections were rehydrated and incubated with an anti-ACN9 or scramble probe as negative control. Results are normalized to anti-PPIB probe (internal control) used. (B) Protein-level expression of ACN9 in the liver and lung tissues was analyzed using immunohistochemistry. Data shown is representative of at least two independent experiments.

  • Figure 2 Heatmaps for representative gene sets dysregulated by cadmium (Cd)-treated ACN9-silenced cells. Effect of Cd and siACN9 on the expression of ACN9 mRNA level. (A) BEAS-2B cells transfected with siACN9 (15 nM) compared to the control scrambled siRNA. (B) BEAS-2B cells (2×105 cells per well for quantitative polymerase chain reaction) were treated with the indicated concentrations of Cd (10 µM) for 24 or 48 hours. For quantitative real time polymerase chain reaction (qRT-PCR), statistical evaluation was performed using Student's t test. Data shown is representative of at least three independent experiments. (C) Comparison of gene expression patterns obtained from cDNA microarray and qRT-PCR with ACN9 knockdown in Cd-treated bronchial epithelial cells. Heat map and bars indicate the magnitude of gene expression changes. (D) Numbers of differentially expressed genes in the ACN9-silenced BEAS-2B cells with/without Cd. (E) Gene ontology analysis showing Cd and ACN9 silencing responsive relationships in terms of biological processes. *p≤0.05, **p≤0.01, and ***p≤0.001 were considered significant. Mock: transfection reagent alone; siNC: small interference RNA of non-targeting control; siACN91-3: small interference RNA for ACN9 at 10 nM, 15 nM, and 25 nM.

  • Figure 3 ACN9-silenced cells drive inflammatory responses. BEAS-2B cells were pretreated for 24 hours with small interference RNA for ACN9 (siACN9; 15 nM) after being stimulated with cadmium (Cd) (10 µM). (A) mRNA was extracted from total cell lysates and analyzed by quantitative polymerase chain reaction for proinflammatory cytokines. (B) Supernatants were analyzed by enzyme-linked immunosorbent assay for interleukin 6 (IL-6). (C) Whole-cell lysates were analyzed by Western blotting for proinflammatory cytokine (IL-1β) and p38/mitogen-activated protein kinase (MAPK) signal pathway and normalized by β-actin. (D) The protein expression levels of phosphorylated JNK, ERK, and p38 MAPK were calculated by normalized to total ERK, and IL-1β and IL-1RA were normalized by β-actin. For enzyme-linked immunosorbent assay and quantitative real time polymerase chain reaction, statistical evaluation was conducted, using Student's t test *p≤0.05, **p≤0.01, and ***p≤0.001 were considered significant. Data shown is representative of at least three independent experiments. TNF-α: tumor necrosis factor α; COX2: cyclooxygenase 2; iNOS: inducible nitric oxide synthase; MMP9: matrix metalloproteinase 9; siNC: small interference RNA of non-targeting control.


Reference

1. Mathers CD, Loncar D. Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med. 2006; 3:e442.
2. Fabbri LM. Smoking, not COPD, as the disease. N Engl J Med. 2016; 374:1885–1886.
3. Peacock JL, Anderson HR, Bremner SA, Marston L, Seemungal TA, Strachan DP, et al. Outdoor air pollution and respiratory health in patients with COPD. Thorax. 2011; 66:591–596.
4. Lamprecht B, McBurnie MA, Vollmer WM, Gudmundsson G, Welte T, Nizankowska-Mogilnicka E, et al. COPD in never smokers: results from the population-based burden of obstructive lung disease study. Chest. 2011; 139:752–763.
5. Lag M, Rodionov D, Ovrevik J, Bakke O, Schwarze PE, Refsnes M. Cadmium-induced inflammatory responses in cells relevant for lung toxicity: expression and release of cytokines in fibroblasts, epithelial cells and macrophages. Toxicol Lett. 2010; 193:252–260.
6. Kirschvink N, Martin N, Fievez L, Smith N, Marlin D, Gustin P. Airway inflammation in cadmium-exposed rats is associated with pulmonary oxidative stress and emphysema. Free Radic Res. 2006; 40:241–250.
7. Bertin G, Averbeck D. Cadmium: cellular effects, modifications of biomolecules, modulation of DNA repair and genotoxic consequences (a review). Biochimie. 2006; 88:1549–1559.
8. Hassan F, Xu X, Nuovo G, Killilea DW, Tyrrell J, Da Tan C, et al. Accumulation of metals in GOLD4 COPD lungs is associated with decreased CFTR levels. Respir Res. 2014; 15:69.
9. Kim WJ, Lim MN, Hong Y, Silverman EK, Lee JH, Jung BH, et al. Association of lung function genes with chronic obstructive pulmonary disease. Lung. 2014; 192:473–480.
10. Dennis RA, McCammon MT. ACN9 is a novel protein of gluconeogenesis that is located in the mitochondrial intermembrane space. Eur J Biochem. 1999; 261:236–243.
11. Na U, Yu W, Cox J, Bricker DK, Brockmann K, Rutter J, et al. The LYR factors SDHAF1 and SDHAF3 mediate maturation of the iron-sulfur subunit of succinate dehydrogenase. Cell Metab. 2014; 20:253–266.
12. Zhu GZ, Zhang M, Kou CZ, Ni YH, Ji CB, Cao XG, et al. Effects of Lyrm1 knockdown on mitochondrial function in 3 T3-L1 murine adipocytes. J Bioenerg Biomembr. 2012; 44:225–232.
13. Haack TB, Madignier F, Herzer M, Lamantea E, Danhauser K, Invernizzi F, et al. Mutation screening of 75 candidate genes in 152 complex I deficiency cases identifies pathogenic variants in 16 genes including NDUFB9. J Med Genet. 2012; 49:83–89.
14. Lim SC, Friemel M, Marum JE, Tucker EJ, Bruno DL, Riley LG, et al. Mutations in LYRM4, encoding iron-sulfur cluster biogenesis factor ISD11, cause deficiency of multiple respiratory chain complexes. Hum Mol Genet. 2013; 22:4460–4473.
15. Dick DM, Aliev F, Wang JC, Saccone S, Hinrichs A, Bertelsen S, et al. A Systematic single nucleotide polymorphism screen to fine-map alcohol dependence genes on chromosome 7 identifies association with a novel susceptibility gene ACN9. Biol Psychiatry. 2008; 63:1047–1053.
16. Cloonan SM, Choi AM. Mitochondria: commanders of innate immunity and disease? Curr Opin Immunol. 2012; 24:32–40.
17. Ghezzi D, Goffrini P, Uziel G, Horvath R, Klopstock T, Lochmuller H, et al. SDHAF1, encoding a LYR complex-II specific assembly factor, is mutated in SDH-defective infantile leukoencephalopathy. Nat Genet. 2009; 41:654–656.
18. Hopps E, Noto D, Caimi G, Averna MR. A novel component of the metabolic syndrome: the oxidative stress. Nutr Metab Cardiovasc Dis. 2010; 20:72–77.
19. Wang Y, Fang J, Leonard SS, Rao KM. Cadmium inhibits the electron transfer chain and induces reactive oxygen species. Free Radic Biol Med. 2004; 36:1434–1443.
20. Heinzmann A, Thoma C, Dietrich H, Deichmann KA. Identification of common polymorphisms in the mitochondrial genome. Allergy. 2003; 58:830–831.
21. Kim SR, Kim DI, Kim SH, Lee H, Lee KS, Cho SH, et al. NLRP3 inflammasome activation by mitochondrial ROS in bronchial epithelial cells is required for allergic inflammation. Cell Death Dis. 2014; 5:e1498.
22. Kang MJ, Shadel GS. A mitochondrial perspective of chronic obstructive pulmonary disease pathogenesis. Tuberc Respir Dis. 2016; 79:207–213.
23. Bowler RP, Barnes PJ, Crapo JD. The role of oxidative stress in chronic obstructive pulmonary disease. COPD. 2004; 1:255–277.
24. Koedrith P, Kim HL, Seo YR. Integrative toxicogenomics-based approach to risk assessment of heavy metal mixtures/complexes: strategies and challenges. Mol Cell Toxicol. 2015; 11:265–276.
25. Mills E, O'Neill LA. Succinate: a metabolic signal in inflammation. Trends Cell Biol. 2014; 24:313–320.
26. Tannahill GM, Curtis AM, Adamik J, Palsson-McDermott EM, McGettrick AF, Goel G, et al. Succinate is an inflammatory signal that induces IL-1beta through HIF-1alpha. Nature. 2013; 496:238–242.
27. Gimenez-Roqueplo AP, Favier J, Rustin P, Rieubland C, Kerlan V, Plouin PF, et al. Functional consequences of a SDHB gene mutation in an apparently sporadic pheochromocytoma. J Clin Endocrinol Metab. 2002; 87:4771–4774.
28. Dahia PL, Ross KN, Wright ME, Hayashida CY, Santagata S, Barontini M, et al. A HIF1alpha regulatory loop links hypoxia and mitochondrial signals in pheochromocytomas. PLoS Genet. 2005; 1:72–80.
29. Hobert JA, Mester JL, Moline J, Eng C. Elevated plasma succinate in PTEN, SDHB, and SDHD mutation-positive individuals. Genet Med. 2012; 14:616–619.
Full Text Links
  • TRD
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr