Open Access

Antitumor effects of cadmium against diethylnitrosamine‑induced liver tumors in mice

  • Authors:
    • Yu Nie
    • Bo Huang
    • An-Ling Hu
    • Yun-Yan Xu
    • Yan Zou
    • Yun Liu
    • Jie Liu
  • View Affiliations

  • Published online on: November 26, 2021     https://doi.org/10.3892/ol.2021.13151
  • Article Number: 33
  • Copyright: © Nie 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 (Cd) has been reported to exhibit antitumor effects against chemically induced liver tumors. However, the antitumor effects of Cd are not completely understood. Metallotherapy, the use of a toxic metal to attack liver tumors, could be a viable strategy. In the present study, 8‑week old, male, C57BL/6 mice were administered injections of diethylnitrosamine (DEN) (90 mg/kg, and then 50 mg/kg 2 weeks later), followed by liver tumor promotion with carbon tetrachloride. Cadmium chloride was administered in the drinking water (1000 ppm) from 21‑40 weeks after DEN initiation. Body weights were recorded and liver tumor formation was monitored via ultrasound. At the end of experiments, livers were removed, weighed, and the tumor incidence, tumor numbers and tumor size scores were recorded. Liver histology and metallothionein (MT) immunostaining were performed. After DEN injection, animal body weight decreased, and then slowly recovered with time. Cd treatment did not affect animal body weight gain. Ultrasound analysis detected liver tumors 35 weeks after DEN injection, and the mice were necropsied at 40 weeks. Liver/body weight ratios increased in the DEN and DEN + Cd groups. Cd treatment decreased the tumor incidence (71 vs. 17%), tumor numbers (15 vs. 2) and tumor scores (22 vs. 3) when compared with the DEN only group. Histopathology showed hepatocyte degeneration in all groups, and immunohistochemistry showed MT‑deficiency in the liver tumors, while MT staining was intensified in the surrounding tissues. Reverse transcription‑quantitative PCR showed increases in α‑fetoprotein level in DEN‑treated livers, and increases in MT‑2 and tumor necrosis factor α (TNFα) levels in Cd‑treated livers. Thus, it was concluded that Cd is effective in the suppression of DEN‑induced liver tumors, and that the mechanisms may be related to MT‑deficiency in tumors and the induction of TNFα to kill tumor cells.

Introduction

Hepatocellular carcinoma (HCC) is a common and lethal malignancy (1). Due to a late diagnosis and advanced underlying liver cirrhosis, only limited treatment options with marginal clinical benefits are available for affected patients (1,2). Metallotherapeutics, including platinum and other metal-containing antitumor drugs, is a strategy for treating HCC (3).

Cadmium (Cd) is a toxic heavy metal implicated in carcinogenesis (4) and the development of HCC (5). Similar to the use of chemotherapeutics, such as the carcinogenic and toxic metalloid arsenic, a metallotherapeutic agent could be applied as a toxicant to attack the malignancy. Indeed, Cd is also effective against HCC in experimental animals (68) and in cultured liver tumor SMMC cells and their xenografts (9,10). Cd-coordinated supramolecules, such as Cd-pyrithione (11), Cd telluride/Cd sulfide (12,13), Cd-coordinated thiacalix arene tetrasulfate (14), Cd-thiocarbodiazone complex (15), Cd in combination with human second mitochondria-derived activator of caspase (16) and a novel binuclear hydrazone-based Cd(II) complex (17), have shown potential antitumor effects against chemoresistant malignant cells.

HCC is often refractory to chemotherapy and radiotherapy at late stages, and is often associated with the loss of metallothionein (MT), probably due to hypermethylation of the MT genes (1820), and deficiency in MT could make HCC vulnerable to the necrotic effects of Cd, while surrounding normal tissues could be protected through MT induction (6). MTs are small, cysteine-rich cadmium-binding proteins that protect against cadmium toxicity (21).

The present study was initiated in mice to examine the effects of cadmium, administered in drinking water, against HCC formation initiated by diethylnitrosamine (DEN) and promoted by carbon tetrachloride. Small animal ultrasound was employed to monitor tumor formation, immunohistochemistry was used to stain for MT, and the expression levels of the HCC biomarker α- fetoprotein, MT and tumor necrosis factor-α in the liver were determined via reverse transcription-quantitative (RT-q)PCR.

Materials and methods

Reagents

Cadmium chloride (CdCl2), DEN and carbon tetrachloride (CCl4) were obtained from MilliporeSigma. Other reagents were of analytical grade.

Animals

Male C57BL/6 mice (6 weeks old) were purchased from the Animal Center Institute of Surgery Research of the Third Military Medical University (Chongqing, China). Animals were maintained in specific pathogen-free facilities at Zunyi Medical University (Zunyi, China), under a controlled environment (22±1°C, 50±2% humidity and a 12:12 h light:dark cycle) and free access to purified water and standard laboratory chow. Efforts were made to ameliorate distress and harm to animals by daily monitoring and humane treatment. Animals were adequately cared for, and experimental protocols were in compliance with the Animal Management Guidelines of the Chinese Ministry of Health and approved by the Animal Use and Care Committee of Zunyi Medical University (approval no. 2015–01). The study is reported in accordance with ARRIVE guidelines (https://arriveguidelines.org).

Experimental design

After 2 weeks of acclimation, 40 mice (8 weeks of age, weighing 16–19 g) were given the first injection of DEN (90 mg/kg, i.p.). Approximately 80% of mice survived the first DEN injection, and 2 weeks later, the surviving mice were given the second injection of DEN (50 mg/kg, i.p.), according to the protocol (22). At 4 weeks after the initial DEN challenge, mice (12 weeks of age) were orally administered 20% CCl4, 5 ml/kg, twice a week for 4 months in an attempt to promote liver tumors. At 21 weeks after initial DEN initiation, the mice were randomly divided into the DEN (n=14) and DEN + Cd (n=12) group. There was an additional normal control (n=5) group that did not receive any treatment. Cadmium was administered via the drinking water (1000 ppm) as CdCl2 from 21–40 weeks according to the previous literature (7,8). Body weights were monitored weekly, and a single ultrasound examination of liver tumor formation was performed at 35–39 weeks after DEN initiation. At the end of the 40-week experiment, the mice were anesthetized with sodium pentobarbital (65 mg/kg, i.p.) and subsequently sacrificed by decapitation. The mouse livers were harvested, and the liver weights and tumor outcomes were recorded. The treatment process is illustrated in Fig. 1A.

The visible tumors were counted and recorded. The tumor incidence (number of tumor-bearing mice), tumor numbers (total tumors found) and tumor size scores (score of 1, tumor size <1 mm; score of 2, tumor size 1–2 mm; and score of 3, tumor size >2 mm) were recorded.

Ultrasound detection

Liver tumor formation was measured using B-mode ultrasound (Vevo® 2100; Fujifilm VisualSonics, Inc.) with 30-MHz peak frequency linear array transducers (MS400; Fujifilm VisualSonics, Inc.; mean beam frequency range of 22–55 MHz) in a digitized scale as previously described (23). Ultrasound was used to detect tumor incidence and size.

Histopathology

Liver tissues were fixed in 10% buffered formalin for 48 h at room temperature, embedded in paraffin at 60°C and sliced into 3.5-µm-thick sections using a RM2235 microtome (Leica Microsystems GmbH). Sections were deparaffinized in xylene and rehydrated using a gradient of ethanol (100, 95, 85 and 75%). The histological sections were then stained with hematoxylin and eosin at room temperature for 8–10 min and 4–5 sec, respectively. The digitized images of slices were observed via the Olympus image analysis system (light microscope; Olympus Corporation) at ×4 and ×10 magnifications as previously described (24).

Immunohistochemical analysis

Metallothionein (MT) protein retrieval was performed in 10 mmol/l citrate buffer (pH 6.0) at 92°C for 15 min. Endogenous peroxidase was then blocked with 3% H2O2 for 15 min at room temperature. The tissues were then incubated with 10% goat non-immune serum (Invitrogen; Thermo Fisher Scientific, Inc.) for 1 h at room temperature. Subsequently, sections were incubated with MT primary antibodies (cat. no. ab12228, 1:100; Abcam) overnight at 4°C. After washing with PBS for 5 min, slides were incubated with HRP-conjugated anti-mouse secondary antibody (1:1,000; cat. no. A0216; Beyotime Institute of Biotechnology) for 1 h at room temperature following the protocol associated with the SABC Detection System (Beyotime). Finally, sections were stained with DAB and counterstained with hematoxylin for 5 min at room temperature. The digitized images of slices were observed via the LEICA image analysis system (Leica Microsystems GmbH) at ×10 magnification. The expression of proteins was quantitatively measured by Image ProPlus 6.0 to obtain the positive staining-integral optical density/area (density mean). A total of four discontinuous areas were used to analyze the expression levels of protein in a blinded fashion as previously described (24).

RT-qPCR

Total RNA was extracted from tissues with TRIzol® (Takara Biotechnology Co., Ltd.) and reverse transcribed according to the manufacturer's protocol into cDNA using the PrimeScript™ RT reagent kit (Takara Biotechnology Co., Ltd.). qPCR was performed utilizing the iQ™ SYBR Green Supermix (Bio-Rad Laboratories, Inc.). The PCR cycling conditions were 94°C for 3 min, followed by 40 cycles of 94°C for 15 sec, 60°C for 20 sec and 72°C for 40 sec. Data were normalized to β-actin and expressed using the comparative Cq method (23,25,26). Primers for the mouse genes are shown in Table I.

Table I.

Primer sequences for reverse transcription-quantitative PCR.

Table I.

Primer sequences for reverse transcription-quantitative PCR.

NameAccess IDForward primer (5′-3′)Reverse primer (5′-3′)
AFPNM_007423 AGCAGGACTGCTCGAAACAT AGCGAAATGTAGCAGGAGGA
β-actinNM_007393 GATCTGGCACCACACCTTCT GGGGTGTTGAAGGTCTCAAA
MT-2NM_008630 CCGATCTCTCGTCGATCTTC AGGAGCAGCAGCTTTTCTTG
TNF-αNM_013693 TAGCCAGGAGGGAGAACAGA TTTTCTGGAGGGAGATGTGG

[i] AFP, α-fetoprotein; MT, mettalothionein; TNFα, tumor necrosis factor α.

Statistical analysis

Data are presented as the mean ± SE and were analyzed by one-way ANOVA, followed by Ducan's multiple comparison test using SigmaPlot (Systat Sofeware, Inc.) version 14. For tumor incidence, Fisher's exact test was performed for the contingency table, while for tumor number and tumor score, data were analyzed with the Kruskal-Wallis test, followed by Dunn's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Animal survival and body weight

After the initial DEN (90 mg/kg, i.p.) administration, 80% of mice survived within 7 days, with a ~20% reduction in body weight. After the second DEN injection, 95% of mice survived, with a ~15% reduction in body weight. Animal body weights slowly recovered afterwards over time. CCl4 promotion (20%, 5 ml/kg, p.o., twice/week) was started at week 4 post-DEN injection and continued for 4 months. Cd was administered from 21–40 weeks after DEN initiation through the drinking water (1000 ppm) for 19 weeks. All mice survived Cd treatment. Ultrasound was randomly performed on a few mice at week 36 post-DEN injection (15 weeks after Cd treatment), 4 times, until week 40 post-DEN initiation (19 weeks after Cd treatment). The animal body weights after Cd intervention are shown in Fig. 1B.

Ultrasound detection of liver tumor formation

Small animal ultrasound analysis was used to monitor tumor formation 30 weeks after DEN injection, but no tumors were detected. At 36 weeks after DEN injection, liver tumors with small sizes were detected at an ~40% incidence rate. At week 40 after DEN initiation, a 65% tumor incidence rate was recorded. Fig. 2 shows representative ultrasound images of DEN-induced liver tumors at 40 weeks after DEN initiation in control, DEN- and DEN + Cd-treated mice. In the control mice, the ultrasound showed normal liver, while in the DEN-treated mice, the liver density was increased (arrows), indicative of liver tumors. In the DEN + Cd-treated mice, only one mouse showed a detectable tumor with decreased density compared with non-tumor tissues.

Animal liver/body weight ratio

At the end of experiment, the mice were euthanized and their livers were collected and weighed, and the liver/body ratios were calculated. Fig. 3 shows the liver/body weight ratios of the three groups. DEN increased the liver/body weight ratio from 48.5 to 53.4 mg/g, while DEN + Cd further increased the liver/body weight ratio to 55.9 mg/g, which were significantly higher results compared with the controls.

Liver tumor outcome

Representative gross liver images are shown in Fig. 4, where liver tumors were detected in DEN-treated mice, no tumors were found in control mice and only a few tumors were found in DEN + Cd-treated mice.

The tumor outcomes, recorded from blinded observations, are listed in Table II. In total, 10 out of 14 DEN-treated mice had tumors, with a tumor incidence of 71%, and only 2 out of 12 DEN + Cd-treated mice had tumors, with a tumor incidence of 17%; a total of 15 tumors were found in DEN-treated mice, but only 2 tumors in DEN + Cd-treated mice. Some tumors in the DEN-treated mice were large, and the tumor score was 22, while in DEN + Cd-treated mice, the score was 3. The tumor number and tumor score of the DEN + Cd group were statistically significant compared with the DEN group.

Table II.

Tumor outcomes of DEN and DEN + Cd groups.

Table II.

Tumor outcomes of DEN and DEN + Cd groups.

GroupnTumor incidence, n (%)Tumor numberTumor score
Control5000
DEN1410 (71)1522
DEN + Cd122a (17)2a3a

a P<0.05 vs. DEN. DEN, dimethylnitrosamine; Cd, cadmium.

Histology and immunohistochemistry

Fig. 5 shows representative histology and immunochemistry images of the tissues in the different groups. Liver tumors were observed in DEN-treated mice, and hepatocellular degeneration was evident in both DEN- and DEN + Cd-treated mice. In DEN-treated livers, MT staining was not present in the liver tumors, but was strongly present in the tissues surrounding the tumors. DEN + Cd-treated mice exhibited increased intensity of the MT stain.

RT-qPCR analysis of liver gene expression

Fig. 6 shows the expression of liver genes in the control mice (n=5), and in the mice treated with DEN (n=14) and DEN + Cd (n=12), with controls set at 100%. DEN treatment increased the expression of α-fetoprotein (AFP) over 2-fold (242% of control); however, due to huge individual variance, the difference was not statistically significant. DEN + Cd treatment increased the expression to 112% of the control. The expression of MT-2 was slightly decreased by DEN treatment (73% of the control), but significantly increased by DEN + Cd treatment by almost 3-fold (294% of the control). The expression of tumor necrosis factor α (TNFα) was slightly increased by DEN treatment (142% of the control), but significantly increased by DEN + Cd treatment (224% of the control).

Discussion

The present study demonstrated the antitumor effects of cadmium against DEN-initiated and CCl4-promoted HCC, as evidenced by ultrasound imaging, tumor incidence, tumor number and tumor score. Immunohistochemistry showed MT was deficient in HCC, but was induced in the tissues surrounding the tumor. RT-qPCR revealed that the HCC biomarker AFP was increased in HCC, but attenuated by Cd, although the changes were not statistically significant. Cd treatment also induced MT and TNFα in the liver. However, histology showed hepatocyte degeneration in the liver in both DEN- and DEN + Cd-treated groups.

Cadmium has previously been shown to exert antitumor effects against DEN-induced HCC in B6C3F1 mice (68), including the late stage of HCC by inducing tumor necrosis (6). The present study replicated these previous findings in C57/BL mice. Cd exposure is known to produce tumors in the lungs, liver, prostate, pancreas and injection sites (4), and long-term Cd exposure is implicated in HCC development (5). Paradoxically, in an effort to promote DEN-initiated HCC, Cd was unexpectedly found to produce antitumor effects (7). This phenomenon is common for metallo-chemotherapy agents. For example, arsenic is a well-known human carcinogen, but it is also effective against hematological malignancies and certain solid tumors with arsenic-metal complexes (27), and the toxic effects of Cd towards colorectal carcinoma cells (28) might contribute to the antitumor effects of Cd. The use of a toxic metal/metalloid to treat chemo-resistant malignancies could be a strategy in metallo-chemotherapy.

Non-invasive imaging of HCC growth in mice with ultrasound technology (29) would help monitor HCC growth, the treatment efficacy and the treatment duration. We have previously successfully applied ultrasound in cardiovascular studies (23). In the present study, HCC formation could be detected in mice at 4 months after DEN initiation (at 6 months of age), while at 8 months of age, 50% tumor incidence was detected, and at 9 months of age, 65% tumor incidence was detected, which was consistent with the necropsy findings at the end of experiment (at 10 months of age). Tumor promotion with CCl4 in the current study resulted in higher tumor incidence compared with that found in previous studies using Cd alone (7,8). With the aid of ultrasound monitoring, HCC-bearing mice were sacrificed at 40 weeks of age to examine Cd antitumor effects, with findings of decreased incidence (71 vs. 17%), decreased tumor numbers (15 vs. 2) and decreased tumor size scores (22 vs. 3) compared with the DEN-treated group. Ultrasound appears to be a sensitive means to monitor liver tumor development and helped to define the Cd treatment regimen for tumor growth inhibition in the present study.

MTs are small, cysteine-rich cadmium-binding proteins that protect against cadmium toxicity and are encoded by MT isoform genes (21,30). In studies on human HCC, MT-1G was hypermethylated, leading to decreased expression (19), MT-1X and MT-2A tended to decrease with the progression of HCC (20), and at the late stage of HCC, MT-1A, MT-2A and metal regulatory transcription factor 1 levels were decreased (25). In the present study, immunostaining for MT was not detected in HCC, while the surrounding tissues exhibited intense staining for MT. This molecular event could be turned into an advantage for Cd to selectively kill HCC, while saving normal cells that will be protected from Cd cytotoxicity by the MT induction.

AFP is a well-known biomarker of HCC that exhibits increased levels in DEN-treated mouse livers (31). In the present study, the expression of AFP in the Cd-treated group was attenuated, consistent with the antitumor effects of Cd. By contrast, the expression of MT-2 and TNFα in the Cd-treated group was higher compared with that in the DEN model group. Cd is a potent inducer of the MT genes; both MT-1 and MT-2 are coordinately expressed in the mouse liver (21), and increased MT-2 expression could protect against Cd toxicity. Induction of TNFα by Cd plays a dual role in killing tumor cells, as well as in producing liver damage (32).

It should be noted that the antitumor effects of Cd should be carefully assessed to ensure the balance between the benefits and risks from its use. Necrotic effects of Cd on HCC are a desired therapeutic outcome, but Cd-induced liver injury is an undesired toxic effect. In the present study, the antitumor effects of Cd were accompanied by liver damage to various degrees (Figs. 4 and 5). Caution should be taken when using Cd to treat malignancies over a long period of time, and close monitoring of the potential adverse effects to balance efficacy and toxicity is important.

Collectively, the present study demonstrated the antitumor effects of Cd against DEN-induced HCC in C57/B6 mice; the mechanism of action appeared to be related to MT-deficiency in HCC, while normal surrounding tissues were protected by MT. Thus, the targeting of MT-deficiency in HCC could be a potential therapeutic strategy.

Acknowledgements

Not applicable.

Funding

This study was supported by the Guizhou Provincial Science and Technology Program [grant no. QKH(2019)1346], the Science and Technology Talent Support Project of the Educational Department of Guizhou Province [grant no. KY(2018)055], the Innovation Talent Team of Zunyi [grant no. ZSKRC(2019)1], the Innovation Talent Team of Guizhou Science and Technology Department [grant no. QKHPTRC(2020)5007] and the Science and Technology Plan of Zunyi [grant no. ZSKRCPT(2020)7].

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YL, JL and YN conceived the experiment. YN performed the animal studies, including drug administration, body weight recording and necropsy, and qPCR and histology analyses. BH and YYX performed the small animal ultrasound imaging, ALH performed the pathology and immunohistochemistry analyses, and YZ performed the statistical analysis. YL, YN, BH, YYX and ALH confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

All animal care and experimental protocols complied with the Animal Management Guidelines of the Chinese Ministry of Health and were approved by the Animal Use and Care Committee of Zunyi Medical University (Zunyi, China; approval no. 2015-01).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Wörns MA and Galle PR: Future perspectives in hepatocellular carcinoma. Dig Liver Dis. 42 (Suppl 3):S302–S309. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Javan H, Dayyani F and Abi-Jaoudeh N: Therapy in Advanced Hepatocellular Carcinoma. Semin Intervent Radiol. 37:466–474. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Kaluderović GN and Paschke R: Anticancer metallotherapeutics in preclinical development. Curr Med Chem. 18:4738–4752. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Waalkes MP: Cadmium carcinogenesis. Mutat Res. 533:107–120. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Satarug S: Long-term exposure to cadmium in food and cigarette smoke, liver effects and hepatocellular carcinoma. Curr Drug Metab. 13:257–271. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Waalkes MP, Diwan BA, Rehm S, Ward JM, Moussa M, Cherian MG and Goyer RA: Down-regulation of metallothionein expression in human and murine hepatocellular tumors: Association with the tumor-necrotizing and antineoplastic effects of cadmium in mice. J Pharmacol Exp Ther. 277:1026–1033. 1996.PubMed/NCBI

7 

Waalkes MP, Diwan BA, Weghorst CM, Bare RM, Ward JM and Rice JM: Anticarcinogenic effects of cadmium in B6C3F1 mouse liver and lung. Toxicol Appl Pharmacol. 110:327–335. 1991. View Article : Google Scholar : PubMed/NCBI

8 

Waalkes MP, Diwan BA, Weghorst CM, Ward JM, Rice JM, Cherian MG and Goyer RA: Further evidence of the tumor-suppressive effects of cadmium in the B6C3F1 mouse liver and lung: Late stage vulnerability of tumors to cadmium and the role of metallothionein. J Pharmacol Exp Ther. 266:1656–1663. 1993.PubMed/NCBI

9 

Du H, Liu X, Liu Y, Jin M, Huang W and Sun Z: Inhibition effect of cadmium chloride on human hepatocellular carcinoma cells SMMC7721. Chin J Publ Health. 22:194–195. 2006.

10 

Du HJM, Liu Y, Liu X, Wang W and Sun Z: Study on the anti-tumor effect of calcium chloride in vivo. Mod Prev Med. 35:3763–3765. 2008.

11 

Chen X, Wu J, Yang Q, Zhang X, Zhang P, Liao S, He Z, Wang X, Zhao C and Liu J: Cadmium pyrithione suppresses tumor growth in vitro and in vivo through inhibition of proteasomal deubiquitinase. Biometals. 31:29–43. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Xu N, Piao M, Arkin K, Ren L, Zhang J, Hao J, Zheng Y and Shang Q: Imaging of water soluble CdTe/CdS core-shell quantum dots in inhibiting multidrug resistance of cancer cells. Talanta. 201:309–316. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Zhang G, Shi L, Selke M and Wang X: CdTe quantum dots with daunorubicin induce apoptosis of multidrug-resistant human hepatoma HepG2/ADM cells: In vitro and in vivo evaluation. Nanoscale Res Lett. 6:4182011. View Article : Google Scholar : PubMed/NCBI

14 

Zhou X, Koizumi Y, Zhang M, Natsui M, Koyota S, Yamada M, Kondo Y, Hamada F and Sugiyama T: Cadmium-coordinated supramolecule suppresses tumor growth of T-cell leukemia in mice. Cancer Sci. 106:635–641. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Pérez JM, Cerrillo V, Matesanz AI, Millán JM, Navarro P, Alonso C and Souza P: DNA interstrand cross-linking efficiency and cytotoxic activity of novel cadmium(II)-thiocarbodiazone complexes. ChemBioChem. 2:119–123. 2001. View Article : Google Scholar : PubMed/NCBI

16 

Guo C, Li Y, Zhang H, Wang Z, Jin M, Zhang L, An L, Hu G, Liu X, Liu Y, et al: Enhancement of antiproliferative and proapoptotic effects of cadmium chloride combined with hSmac in hepatocellular carcinoma cells. Chemotherapy. 57:27–34. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Bjelogrlić S, Todorović TR, Cvijetić I, Rodić MV, Vujčić M, Marković S, Araškov J, Janović B, Emhemmed F, Muller CD, et al: A novel binuclear hydrazone-based Cd(II) complex is a strong pro-apoptotic inducer with significant activity against 2D and 3D pancreatic cancer stem cells. J Inorg Biochem. 190:45–66. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Jacob ST, Majumder S and Ghoshal K: Suppression of metallothionein-I/II expression and its probable molecular mechanisms. Environ Health Perspect. 110 (Suppl 5):827–830. 2002. View Article : Google Scholar : PubMed/NCBI

19 

Kanda M, Nomoto S, Okamura Y, Nishikawa Y, Sugimoto H, Kanazumi N, Takeda S and Nakao A: Detection of metallothionein 1G as a methylated tumor suppressor gene in human hepatocellular carcinoma using a novel method of double combination array analysis. Int J Oncol. 35:477–483. 2009.PubMed/NCBI

20 

Tao X, Zheng JM, Xu AM, Chen XF and Zhang SH: Downregulated expression of metallothionein and its clinicopathological significance in hepatocellular carcinoma. Hepatol Res. 37:820–827. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Klaassen CD, Liu J and Choudhuri S: Metallothionein: An intracellular protein to protect against cadmium toxicity. Annu Rev Pharmacol Toxicol. 39:267–294. 1999. View Article : Google Scholar : PubMed/NCBI

22 

Yi X, Long L, Yang C, Lu Y and Cheng M: Maotai ameliorates diethylnitrosamine-initiated hepatocellular carcinoma formation in mice. PLoS One. 9:e935992014. View Article : Google Scholar : PubMed/NCBI

23 

Huang B, Hu P, Hu A, Li Y, Shi W, Huang J, Jiang Q, Xu S, Li L and Wu Q: Naringenin attenuates carotid restenosis in rats after balloon injury through its anti-inflammation and anti-oxidative effects via the RIP1-RIP3-MLKL signaling pathway. Eur J Pharmacol. 855:167–174. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Hu A, Huang J, Li S, Gao Y, Wu L, Deng J, Liu J, Gong Q, Li L and Xu S: Involvement of stromal cell-derived factor-1α (SDF-1α), stem cell factor (SCF), fractalkine (FKN) and VEGF in TSG protection against intimal hyperplasia in rat balloon injury. Biomed Pharmacother. 110:887–894. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Li H, Lu YF, Chen H and Liu J: Dysregulation of metallothionein and circadian genes in human hepatocellular carcinoma. Chronobiol Int. 34:192–202. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Fei W, Li C, Tao J, Cai X, Yao W, Ye Y, Zhang Y, Yao Y, Song Q, Li F, et al: Construction of arsenic-metal complexes loaded nanodrugs for solid tumor therapy: A mini review. Int J Pharm. 583:1193852020. View Article : Google Scholar : PubMed/NCBI

28 

Iftode A, Drăghici GA, Macașoi I, Marcovici I, Coricovac DE, Dragoi R, Tischer A, Kovatsi L, Tsatsakis AM, Cretu O, et al: Exposure to cadmium and copper triggers cytotoxic effects and epigenetic changes in human colorectal carcinoma HT-29 cells. Exp Ther Med. 21:1002021. View Article : Google Scholar : PubMed/NCBI

29 

Anton N, Parlog A, Bou About G, Attia MF, Wattenhofer-Donzé M, Jacobs H, Goncalves I, Robinet E, Sorg T and Vandamme TF: Non-invasive quantitative imaging of hepatocellular carcinoma growth in mice by micro-CT using liver-targeted iodinated nano-emulsions. Sci Rep. 7:139352017. View Article : Google Scholar : PubMed/NCBI

30 

Klaassen CD, Liu J and Diwan BA: Metallothionein protection of cadmium toxicity. Toxicol Appl Pharmacol. 238:215–220. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Ghufran H, Azam M, Mehmood A, Butt H and Riazuddin S: Standardization of diethylnitrosamine-induced hepatocellular carcinoma rat model with time based molecular assessment. Exp Mol Pathol. 123:1047152021. 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

Related Articles

Journal Cover

January-2022
Volume 23 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Nie Y, Huang B, Hu A, Xu Y, Zou Y, Liu Y and Liu J: Antitumor effects of cadmium against diethylnitrosamine‑induced liver tumors in mice. Oncol Lett 23: 33, 2022
APA
Nie, Y., Huang, B., Hu, A., Xu, Y., Zou, Y., Liu, Y., & Liu, J. (2022). Antitumor effects of cadmium against diethylnitrosamine‑induced liver tumors in mice. Oncology Letters, 23, 33. https://doi.org/10.3892/ol.2021.13151
MLA
Nie, Y., Huang, B., Hu, A., Xu, Y., Zou, Y., Liu, Y., Liu, J."Antitumor effects of cadmium against diethylnitrosamine‑induced liver tumors in mice". Oncology Letters 23.1 (2022): 33.
Chicago
Nie, Y., Huang, B., Hu, A., Xu, Y., Zou, Y., Liu, Y., Liu, J."Antitumor effects of cadmium against diethylnitrosamine‑induced liver tumors in mice". Oncology Letters 23, no. 1 (2022): 33. https://doi.org/10.3892/ol.2021.13151