Open Access

Effect of cadmium on the expression levels of interleukin-1α and interleukin-10 cytokines in human lung cells

  • Authors:
    • Caroline Odewumi
    • Lekan M. Latinwo
    • Andre Sinclair
    • Veera L.D. Badisa
    • Ahkinyala Abdullah
    • Ramesh B. Badisa
  • View Affiliations

  • Published online on: September 10, 2015     https://doi.org/10.3892/mmr.2015.4316
  • Pages: 6422-6426
  • Copyright: © Odewumi et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cadmium is an environmentally hazardous metal, which causes toxicity in humans. Inhalation of cigarette smoke and industrial fumes containing cadmium are sources of cadmium exposure. It is responsible for the malfunction of various organs, leading to disease particularly in the lungs, liver and kidneys. In the present study, the effect of cadmium chloride (CdCl2) on cell viability, and the expression levels of interleukin (IL)‑1α and IL‑10 cytokines at various concentrations and incubation durations were assessed in MRC‑9 human normal lung and A549 human lung cancer cells to elucidate the mechanism of cadmium toxicity. Cell viability was measured using a crystal violet dye binding assay. The expression levels of the cytokines were measured by cytokine specific enzyme‑linked immunosorbent assay kits. The viability assay results revealed higher sensitivity of the A549 lung cancer cells to CdCl2 compared with the normal MRC‑9 lung cells. In the normal MRC‑9 lung cells, higher expression levels of the cytokines were observed at the lowest CdCl2 concentration at a shorter exposure time compared with the lung cancer cells. Higher levels of the cytokines were observed in the A549 lung cancer cells at all other times and concentrations compared with the MRC‑9 cells, indicating higher levels of inflammation. The cytokine levels were reduced at higher CdCl2 concentrations and longer exposure durations, demonstrating the toxic effect of cadmium. The results indicated that CdCl2 affected the expression levels of the cytokines and led to cytotoxicity in human lung cells, and suggested that compounds which reduce inflammation may prevent cadmium toxicity.

Introduction

Cadmium is a hazardous environmental pollutant with economic value, however no known biological function. It is an industrial toxicant, which has been classified as a type I carcinogen (1,2). It is a highly reactive metal and complexes with ligands to form different compounds, which affect numerous biological molecules and organs (3). A significant quantity of cadmium is introduced into the environment through anthropogenic activities, including copper and nickel smelting, electroplating, galvanizing, nickel-cadmium battery production, welding, phosphate fertilizers, sewage sludge and cigarette smoke (3,4). The toxicokinetics of cadmium depend on the form of cadmium, the dose, the time of exposure and the accumulation in the affected organ (5,6). Cadmium can be observed in all organs of the body; however, the majority accumulates in the lungs, liver and kidney (7,8). Although the level of cadmium exposure to humans is low, it can accumulate and remain in the system for 15–30 years (911), which is responsible for the toxicity in various organs. Cadmium molecular toxicity has been associated with various diseases, including tumor formation.

Our previous study investigated cadmium toxicity and the protective effect of different antioxidant or chelating compounds, which reduce the toxic effect of cadmium in liver cells (1215). Inhalation is one of the predominant causes of cadmium exposure in humans and it has been reported to cause chronic inflammation and is responsible for various lung diseases (1619). Cytokines are the predominant mediators of inflammation. Interleukin (IL)-1α is an important pro-inflammatory cytokine, which regulates the expression levels of other cytokines and chemokines (2022). Conversely, cells respond to various toxic insults by secreting different anti-inflammatory cytokines, including IL-10, for defense and repair mechanisms (23).

The aim of the present study was to measure the expression levels of the IL-1α and IL-10 cytokines and to determine the viability of normal and cancerous human lung cells treated with various concentrations of cadmium chloride (CdCl2) for different incubation periods in order to elucidate the mechanism of cadmium toxicity. The IL-1α and IL-10 cytokines were selected as they were significantly upregulated following treatment with 75 µM CdCl2 after 24 h in our previous study using human A549 cancer cells (24).

Materials and methods

Chemicals

The F12 K medium, penicillin/streptomycin antibiotic solution (100×), fetal bovine serum (FBS), trypsin-EDTA solution (1X), amphotericin B (1,000×), phosphate-buffered saline without calcium and magnesium, CdCl2, 25% glutaraldehyde and crystal violet were purchased from Sigma-Aldrich (St. Louis, MO, USA). The human IL-10 (cat. no. ELH-IL10–001) and human IL-1α (cat. no. ELH-IL1alpha-001) enzyme-linked immunosorbent assay (ELISA) kits were purchased from Ray Biotech, Inc. (Norcross, GA, USA).

Maintenance of the cell lines

The human MRC-9 normal lung (cat. no. CCL-212) and human A549 lung cancer (cat. no. CCL-185) cell lines were purchased from the American Type Culture Collection (Manassas, VA, USA). The supplied frozen cells were cultured, according to the manufacturer's instructions. The cells were grown in 10 ml minimum essential medium (American Type Culture Collection) (MRC-9) or F12K (A549) medium, containing 100 U/ml penicillin, 100 µg/ml streptomycin, 0.025 µg/ml amphotericin B and 10% FBS in T-75 cm2 tissue culture flasks at 37°C in a 5% CO2 incubator (Nuaire Co., Plymouth, MN, USA).

Crystal violet viability test

The viability test was performed as previously reported (25). Briefly, to investigate the effect of CdCl2 on the viability of the cells, ~1×105 A549 lung cancer cells or 5×104 normal MRC-9 lung cells were plated into each well of a 24-well tissue culture plate and allowed to stabilize overnight in a 5% CO2 incubator at 37°C. The cells were treated with 0, 25, 50, 75, 100, 125, 150 or 200 µM CdCl2 in a final volume of 1 ml in triplicate wells and were subsequently incubated for 24 h at 37°C in a 5% CO2 incubator. Following incubation, the viability of the cells was measured. The median lethal dose (LD50) value was calculated from the LD50 graph where the two lines meet, according to a previous study (26).

Preparation of cell extracts

A total of ~3.9×106 A549 lung cancer cells or 1.95×106 normal MRC-9 lung cells were plated into T-75 cm2 flasks in complete medium. Each cell line was treated with 0, 50, 100 or 150 µM CdCl2 in triplicate flasks for various durations (0, 6, 12, 18 or 24 h) and were incubated at 37°C at 5% CO2. At the end of each treatment period, the cells were trypsinized and centrifuged at 1,350 × g for 5 min. The cells were lysed in 1 ml of 50 mM potassium phosphate (pH 7.0) lysis buffer, containing 0.1% Triton X-100, by homogenization in a vial on ice three times for 10 sec using a polytron homogenizer (Pro Scientific, Inc., Oxford, CT, USA). The homogenate was subsequently transferred to an eppendorf tube and centrifuged at 12,740 × g for 10 min at 4°C to remove the lysed cell membrane debris. The supernatants were transferred into fresh tubes and the cell lysates were stored at −20°C for cytokine analysis.

Protein estimation

The protein concentration of the cell lysates were determined using a Bicinchoninic Acid Protein Assay kit (Pierce Biotechnology, Inc., Rockford, IL, USA) with bovine serum albumin as a standard protein. The protein standards and working reagents were prepared, according to the manufacturer's instructions. In triplicate eppendorf tubes, 25 µl standard or lysate sample were added. The working reagent (500 µl) was added to each tube, incubated at 37°C for 30 min and was subsequently measured at 562 nm in a Beckman spectrophotometer (Beckman Coulter, Inc., Fullerton, CA, USA). The concentration of the lysate samples were determined using the standard curve.

ELISA

The human IL-1α and IL-10 ELISA kits were purchased from Ray Biotech Inc. and used for the quantitative measurement of the expression levels of the cytokines in human lung cells. The ELISA was performed according to the manufacturer's instructions. The intensity of the color in the 96-well plate was measured at 450 nm using a plate reader (EL 800; Bio-Tek Instruments, Inc., Winooski, VT, USA).

Statistical analysis

The viability and cytokine assay results are presented as the mean ± standard deviation (n=3). All CdCl2-treated cell data are presented as a percentage value compared with the untreated control cells (100%). The data were analyzed for significance by Dunnet's multiple comparison test, using GraphPad Prism software version 3.0 (GraphPad Software Inc., San Diego, CA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of CdCl2 on the cell viability of human MRC-9 normal lung and A549 cancer cells

The viability of the normal and cancerous lung cells exposed to various CdCl2 concentrations was demonstrated to be dose-dependent (Fig. 1). The viability of the normal lung MRC-9 cells was reduced by 6, 11, 11, 44, 55, 60 and 75% (Fig. 1A), while the viability of the A594 lung cancer cells was decreased by 13, 36, 42, 58, 65, 76 and 81% (Fig. 1B) following 24 h treatment with 25, 50, 75, 100, 125, 150 and 200 µM CdCl2, respectively. The significant toxic effect of CdCl2 was observed at 25 µM CdCl2 in the A549 lung cancer cells, while the MRC-9 normal lung cells exhibited a significant decrease in viability at 50 µM CdCl2. The LD50 of CdCl2 was revealed to be 87.5 µM in the A549 cells and 112.5 µM in the MRC-9 cells. These results demonstrated the toxic effect of CdCl2 on normal human lung cells and lung cancer cells.

Effect of CdCl2 concentration and treatment duration on the expression levels of IL-1α and IL-10 cytokines in human MRC-9 normal lung cells

The human MRC-9 normal lung cells were treated with different concentrations of CdCl2 (0, 50, 100 or 150 µM) for various durations (6, 12, 18 or 24 h), and the expression levels of IL-1α and IL-10 cytokines were measured. The cells treated with 50 µM CdCl2 for 6 h demonstrated the maximum expression levels of each cytokine (Fig. 2). The cytokine levels decreased with increasing concentration and duration (Fig. 2). The results clearly demonstrated that normal MRC-9 lung cells responded to the toxic effect of CdCl2 with high expression levels of the cytokines in the lysate at early time points and high concentrations and longer exposure durations of CdCl2 demonstrated toxic effects on the expression levels of the cytokines.

Effect of the concentration of CdCl2 and duration on the expression level of IL-1α and IL-10 cytokines in human A549 lung cancer cells

As shown in Fig. 3, the expression levels of IL-1α and IL-10 cytokines in the A549 lung cancer cells treated with 0, 50, 100 or 150 µM CdCl2 concentrations for 6, 12, 18 or 24 h were detected. The lung cancer cells treated with 50 µM CdCl2 for 12 h demonstrated the maximum expression of each cytokine, and the expression levels decreased as the duration of exposure and concentration of CdCl2 increased (Fig. 3). The results clearly demonstrated the response of the A549 lung cancer cells to the toxic effect of CdCl2, with high expression of cytokines at high concentrations and longer exposure durations of CdCl2, demonstrating the toxic effect on the expression of cytokines.

Discussion

The widespread industrial usage of cadmium presents a health risk directly and indirectly to humans and other living organisms. Industrial waste fumes, burning of fossil fuels and cigarette smoke are the predominant direct sources of cadmium exposure to humans. It is estimated that 90% of the inhaled cadmium particles are absorbed by lung tissue and cause pulmonary damage, emphysema and lung cancer (1619). Organisms respond to xeno-biotics via inflammation and it is initiated through various signaling molecules, including cytokines, in the cells. To date, few investigations into the expression levels of the cytokines, IL-1α (pro-inflammatory) and IL-10 (anti-inflammatory), have been reported (27). Therefore, elucidating the expression levels of IL-1α and IL-10, and the cell viability at various CdCl2 concentrations and incubation durations in human MRC-9 normal lung and A549 lung cancer cells may provide an understanding of how lung cells response to cadmium toxicity.

The results indicated that A549 lung cancer cells were observed to be more sensitive to CdCl2 compared with the MRC-9 normal lung cells. The difference in their sensitivities was reflected in their LD50 values. The LD50 of CdCl2 for the normal MRC-9 lung cells was 112.5 µM, whereas the LD50 of the lung cancer cells was 87.5 µM (Fig. 1). A previous study demonstrated that carcinomas contain significantly less metallothionein compared with their corresponding normal cells (28). This may be one of the contributing factors for the higher sensitivity of cancer cells to cadmium toxicity, however, further investigation is required to prove this hypothesis.

Normal lung cells and the lung cancer cells revealed high expression levels of the IL-1α and IL-10 cytokines following treatment with CdCl2 (Figs. 1 and 2). In a previous study (29), a significant increase in IL-1α and IL-10 cytokines was observed in the tested cells, when human-derived bronchial epithelium was exposed to tobacco smoke components. Furthermore, other previous studies have revealed that cells release pro-inflammatory cytokines during inflammation, to activate the cytokine network and the secretion of anti-inflammatory cytokines (30,31). The higher expression of cytokines, which lead to inflammation, may be responsible for the later cytotoxic effects in the cells, which lead to a decrease in viability, as observed in the present study (Fig. 1). This is supported by a previous study (32) demonstrating that high levels of pro-inflammatory cytokines due to cadmium exposure cause pathological conditions in a biological system.

When the cytokine levels were compared in the normal lung cells, a higher expression of pro-inflammatory cytokine, IL-1α, was observed compared with the anti-inflammatory cytokine, IL-10, at 6 h exposure (Fig. 2). A high level of IL-1α cytokine in the cells exposed to xeno-biotics has been reported to inhibit the expression of metallothionein protein (33). In that previous study, IL-1α inhibited the mRNA expression of metallothionein in endometrial stromal cells and amniotic cells treated with CdCl2. Based on the above report, the present study hypothesized that increased levels of IL-1α may reduce the protein expression of metallothionein, which in turn leads to the increase in unbound CdCl2 in the cell, which may now damage biological molecules, including proteins, as shown by the decreased expression of IL-1α and IL-10 observed in the present study (Fig. 2).

The expression pattern of the IL-1α and IL-10 cytokines from the A549 cancer lung cells was different compared with the normal lung cells. The maximum levels of the IL-1α and IL-10 cytokines were observed at a later period (12 h, Fig. 3) in the lung cancer cells compared with the normal cells (6 h, Fig. 2). In addition, it was also observed that each cytokine was highly expressed in cancer cells compared with the normal cells treated with CdCl2 (Figs. 2 and 3). A similar observation was observed when the cytokine levels of patients with cancer were compared with the cytokine levels of normal individuals (34). The delayed expression and higher expression levels of the cytokines observed in cancer cells may be as a result of the malfunction of the cell regulatory mechanisms commonly observed in cancer cells.

Lung cancer cells demonstrated higher expression of the anti-inflammatory cytokine, IL-10, compared with the pro-inflammatory cytokine, IL-1α (Fig. 3). This was consistent with our previously reported findings (24) and another previous study (35). IL-10 acts as anti-inflammatory cytokine and is also responsible for cell death. IL-10 cytokine has been demonstrated to decrease the translocation of nuclear factor-κB, which is important in increasing apoptotic markers, which later leads to apoptotic cell death (31). Therefore, the higher levels of IL-10 cytokine in the A549 lung cancer cells may be one of the reasons for the higher cytotoxicity caused by CdCl2 (Fig. 1).

In the present study, higher cadmium concentrations (100 and 150 µM) decreased the expression levels of the cytokines in the normal and cancer cells (Figs. 2 and 3). Higher concentrations of cadmium induced higher levels of reactive oxygen species, which degrade macromolecules, including proteins and DNA (12,13,36). The low expression levels of the IL-1α and IL-10 cytokines in each cell line may also be linked to the toxic effect of unbound cadmium in the cells, as a result of the lack of metallothionein protein. This result is consistent with a previous study, which reported inhibition of the expression of IL-1α in rat hepatocytes following treatment with high concentrations of CdCl2 (32). In addition, longer exposure durations may also be a factor in causing higher toxicity, as decreased expression of cytokines were observed with increased incubation durations in the present study (Figs. 2 and 3). The longer exposure duration led to the accumulation of cadmium inside of the cell, which in turn causes decreased cytokine levels leading to cell death.

In conclusion, the viability result revealed that human A549 lung cancer cells exhibited higher sensitivity to CdCl2 compared with the normal MRC-9 lung cells. Furthermore, the cells demonstrated a differential expression of the cytokines in response to CdCl2. The maximum cytokine levels were observed in the normal MRC-9 lung cells at an early incubation time (6 h) compared with the lung cancer cells (12 h), demonstrating an early immune response of normal lung cells. The present study clearly demonstrated the effect of CdCl2 on the expression levels of cytokines in lung cells and suggested that compounds, which activate the cytokines and reduce inflammation, may prevent cadmium toxicity.

Acknowledgments

This study was supported by the FAMU Title III, Department of Education (grant no. DOEHBGIPO31B40108–08) and the RCMI, National Institute of Health (grant nos. G12RR03020 and G12D007582).

References

1 

International agency for research on cancer (IARC): Monographs on the Evaluation of the Carcinogenic Risks to Humans, Beryllium, Cadmium, Mercury and Exposures in the Glass Manufacturing industry. IARC Scientific Publications; Lyon, France: pp. 119–238. 1993

2 

Goering PL, Waalkes MP and Klaassen CD: Toxicology of cadmium. Toxicology of metals: Biochemical Aspects. Goyer RA and Cherias M: Handb Exp Pharmacol. 15:189–214. 1995. View Article : Google Scholar

3 

Agency for Toxic Substances Disease Registry (ATSDR): Toxicological Profile for Cadmium. Atlanta, GA: U.S. Department of Health and Human Services, Public Health Service; pp. 1–512. 2012

4 

Joseph P: Mechanisms of cadmium carcinogenesis. Toxicol Appl Pharmacol. 238:272–279. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Chin TA and Templeton DM: Protective elevations of glutathione and metallothionein in cadmium-exposed mesangial cells. Toxicology. 77:145–156. 1993. View Article : Google Scholar : PubMed/NCBI

6 

Bridges CC and Zalups RK: Molecular and ionic mimicry and the transport of toxic metals. Toxicol Appl Pharmacol. 204:274–308. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Habeebu SS, Liu J and Klaassen CD: Cadmium-induced apoptosis in mouse liver. Toxicol Appl Pharmacol. 149:203–209. 1998. View Article : Google Scholar : PubMed/NCBI

8 

Rikans LE and Yamano T: Mechanisms of cadmium-mediated acute hepatotoxicity. J Biochem Mol Toxicol. 14:110–117. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Jin T, Lu J and Nordberg M: Toxicokinetics and biochemistry of cadmium with special emphasis on the role of metallothionein. Neurotoxicology. 19:529–535. 1998.PubMed/NCBI

10 

Waalkes MP, Anver M and Diwan BA: Carcinogenic effects of cadmium in the noble (NBL/Cr) rat: Induction of pituitary, testicular and injection site tumors and intraepithelial proliferative lesions of the dorsolateral prostate. Toxicol Sci. 52:154–161. 1999. View Article : Google Scholar

11 

Boujelben M, Ghorbel F, Vincent C, Makni-Ayadi F, Guermazi F, Croute F and El-Feki A: Lipid peroxidation and HSP72/73 expression in rat following cadmium chloride administration: Interactions of magnesium supplementation. Exp Toxicol Pathol. 57:437–443. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Ikediobi CO, Badisa VL, Ayuk-Takem LT, Latinwo LM and West J: Response of antioxidant enzymes and redox metabolites to cadmium-induced oxidative stress in CRL-1439 normal rat liver cells. Int J Mol Med. 14:87–92. 2004.PubMed/NCBI

13 

Badisa VL, Latinwo LM, Odewumi CO, Ikediobi CO, Badisa RB, Brooks-Walter A, Lambert AT and Nwoga J: Cytotoxicity and stress gene microarray analysis in cadmium-exposed CRL-1439 normal rat liver cells. Int J Mol Med. 22:213–219. 2008.PubMed/NCBI

14 

Odewumi CO, Badisa VL, Le UT, Latinwo LM, Ikediobi CO, Badisa RB and Darling-Reed SF: Protective effects of N-acetylcysteine against cadmium-induced damage in cultured rat normal liver cells. Int J Mol Med. 27:243–248. 2011. View Article : Google Scholar

15 

Odewumi CO, Buggs R, Badisa VL, Latinwo LM, Badisa RB, Ikediobi CO, Darling-Reed SF and Owens MA: Mitigative action of monoisoamy 1-2,3-dimercaptosuccinate (MiADMS) against cadmium-induced damage in cultured rat normal liver cells. Toxicol In Vitro. 25:1733–1739. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Navarro Silvera SA and Rohan TE: Trace elements and cancer risk: A review of the epidemiologic evidence. Cancer Causes Control. 18:7–27. 2007. View Article : Google Scholar

17 

Straif K, Benbrahim-Tallaa L, Baan R, Grosse Y, Secretan B, El Ghissassi F, Bouvard V, Guha N, Freeman C, Galichet L, et al WHO International Agency for Research on Cancer Monograph Working Group: A review of human Carcinogens part C: Metals, arsenic, dusts and fibres. Lancet Oncol. 10:453–454. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Kundu S, Sengupta S and Bhattacharyya A: EGFR upregulates inflammatory and proliferative responses in human lung adenocarcinoma cell line (A549), induced by lower dose of cadmium chloride. Inhal Toxicol. 23:339–348. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Rogalska J, Pilat-Marcinkiewicz B and Brzóska MM: Protective effect of zinc against cadmium hepatotoxicity depends on this bioelement intake and level of cadmium exposure: A study in a rat model. Chem Biol Interact. 193:191–203. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Shimabukuro DW, Sawa T and Gropper MA: Injury and repair in lung and airways. Crit. Care Med. 31(8 Suppl): S524–S531. 2003. View Article : Google Scholar : PubMed/NCBI

21 

Grivennikov SI, Kuprash DV, Liu ZG and Nedospasov SA: Intracellular signals and events activated by cytokines of the tumor necrosis factor superfamily: From simple paradigms to complex mechanisms. Int Rev Cytol. 252:129–161. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Barksby HE, Lea SR, Preshaw PM and Taylor JJ: The expanding family of interleukin-1 cytokines and their role in destructive inflammatory disorders. Clin Exp Immunol. 149:217–225. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Trandem K, Jin Q, Weiss KA, James BR, Zhao J and Perlman S: Virally expressed interleukin-10 ameliorates acute encephalomyelitis and chronic demyelination in coronavirus-infected mice. J Virol. 85:6822–6831. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Odewumi CO, Fils-Aime S, Badisa VL, Latinwo LM, Ruden ML, Ikediobi C and Badisa RB: Chemoprotective effect of monoisoamyl 2, 3-dimercaptosuccinate (MiADMS) on cytokines expression in cadmium chloride treated human lung cells. Environ Toxicol. 30:704–711. 2015. View Article : Google Scholar

25 

Badisa RB, Tzakou O, Couladis M and Pilarinou E: Cytotoxic activities of some Greek Labiatae herbs. Phytother Res. 17:472–476. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Ipsen J and Feigl P: Bancroft's Introduction to Biostatistics. Second Edition. Harper and Row; New York: pp. 1641970

27 

Krocova Z, Macela A, Kroca M and Hernychova L: The immu-nomodulatory effect(s) of lead and cadmium on the cells of immune system in vitro. Toxicol In Vitro. 14:33–40. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Janssen AM, van Duijn W, Kubben FJ, Griffioen G, Lamers CB, van Krieken JH, van de Velde CJ and Verspaget HW: Prognostic significance of metallothionein in human gastrointestinal cancer. Clin Cancer Res. 8:1889–1896. 2002.PubMed/NCBI

29 

Balharry D, Sexton K and BéruBé KA: An in vitro approach to assess the toxicity of inhaled tobacco smoke components: Nicotine, cadmium, formaldehyde and urethane. Toxicology. 244:66–76. 2008. View Article : Google Scholar

30 

Bergman M, Djaldetti M, Salman H and Bessler H: Inflammation and colorectal cancer: Does aspirin affect the interaction between cancer and immune cells? Inflammation. 34:22–28. 2011. View Article : Google Scholar

31 

Romoser AA, Chen PL, Berg JM, Seabury C, Ivanov I, Criscitiello MF and Sayes CM: Quantum dots trigger immunomodulation of the NFκB pathway in human skin cells. Mol Immunol. 48:1349–1359. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Kayama F, Yoshida T, Elwell MR and Luster MI: Role of tumor necrosis factor-alpha in cadmium-induced hepatotoxicity. Toxicol Appl Pharmacol. 131:224–234. 1995. View Article : Google Scholar : PubMed/NCBI

33 

Kawano Y, Furukawa Y, Kawano Y, Abe W, Hirakawa T and Narahara H: Cadmium chloride induces the expression of metallothionein mRNA by endometrial stromal cells and amnion-derived (WISH) cells. Gynecol Obstet Invest. 71:240–244. 2011. View Article : Google Scholar

34 

Katsumata N, Eguchi K, Fukuda M, Yamamoto N, Ohe Y, Oshita F, Tamura T, Shinkai T and Saijo N: Serum levels of cytokines in patients with untreated primary lung cancer. Clin Cancer Res. 2:553–559. 1996.PubMed/NCBI

35 

Bermúdez-Morales VH, Peralta-Zaragoza O, Alcocer-González JM, Moreno J and Madrid-Marina V: IL-10 expression is regulated by HPV E2 protein in cervical cancer cells. Mol Med Report. 4:369–375. 2011.

36 

Thévenod F and Lee WK: Cadmium and cellular signaling cascades: Interactions between cell death and survival pathways. Arch Toxicol. 87:1743–1786. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2015
Volume 12 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Odewumi C, Latinwo LM, Sinclair A, Badisa VL, Abdullah A and Badisa RB: Effect of cadmium on the expression levels of interleukin-1α and interleukin-10 cytokines in human lung cells. Mol Med Rep 12: 6422-6426, 2015
APA
Odewumi, C., Latinwo, L.M., Sinclair, A., Badisa, V.L., Abdullah, A., & Badisa, R.B. (2015). Effect of cadmium on the expression levels of interleukin-1α and interleukin-10 cytokines in human lung cells. Molecular Medicine Reports, 12, 6422-6426. https://doi.org/10.3892/mmr.2015.4316
MLA
Odewumi, C., Latinwo, L. M., Sinclair, A., Badisa, V. L., Abdullah, A., Badisa, R. B."Effect of cadmium on the expression levels of interleukin-1α and interleukin-10 cytokines in human lung cells". Molecular Medicine Reports 12.5 (2015): 6422-6426.
Chicago
Odewumi, C., Latinwo, L. M., Sinclair, A., Badisa, V. L., Abdullah, A., Badisa, R. B."Effect of cadmium on the expression levels of interleukin-1α and interleukin-10 cytokines in human lung cells". Molecular Medicine Reports 12, no. 5 (2015): 6422-6426. https://doi.org/10.3892/mmr.2015.4316