Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2025 Feb:291:117800.
doi: 10.1016/j.ecoenv.2025.117800. Epub 2025 Feb 8.

Nrf2/cyclooxygenase 2 signaling in Cr(VI)-induced carcinogenesis

Affiliations

Nrf2/cyclooxygenase 2 signaling in Cr(VI)-induced carcinogenesis

Lei Zhao et al. Ecotoxicol Environ Saf. 2025 Feb.

Abstract

Long-term exposure to hexavalent chromium [Cr(VI)] has been linked to lung cancer, and cyclooxygenase-2 (COX-2) is a well-known inflammatory factor. However, the role and mechanism of COX-2 in Cr(VI)-induced carcinogenesis are not clear yet. To address this question, we employed a mouse model exposed to Cr(VI) through intranasal instillation of particulate zinc chromate (ZnCrO4) for 12 weeks. Metabolomics and RNA-seq assays revealed enhanced activity of the arachidonic acid (AA)/eicosanoid metabolism pathway in lung tissues from mice exposed to Cr(VI). COX-2, the key enzyme of the AA/eicosanoid pathway, was significantly upregulated in Cr(VI)-exposed lung tissues, as well as in the Cr(VI)-induced transformed (Cr-T) cells compared to parental BEAS-2B (B2B) cells. We then employed multidisciplinary in vitro and in vivo functional assays to characterize the role of COX-2 in Cr(VI)-induced lung cancer. The results indicated that COX-2 functioned as an oncogene to promote the malignant transformation of B2B cells and enhance the proliferation, migration, tumor growth, and angiogenesis of Cr-T cells. Nuclear factor E2-related factor-2 (Nrf2) was identified as a transcription factor for COX-2. Nrf2 was upregulated in response to Cr(VI) exposure and contributed to Cr(VI)-induced lung cancers, in part by upregulating COX-2 expression. Moreover, microRNA-379 (miR-379) was found to target COX-2 to inhibit its expression posttranscriptionally. MiR-379 was downregulated in Cr(VI)-exposed lung tissues and Cr-T cells, and ectopic miR-379 expression reduced Cr-T cell viability and migration, with partial reversal upon COX-2 restoration. In summary, our study revealed the oncogenic role of COX-2 and identified two novel regulatory mechanisms for COX-2 overexpression in Cr(VI)-induced carcinogenesis.

Keywords: COX-2; Carcinogenesis; Cr(VI); Nrf2; miR-379.

PubMed Disclaimer

Conflict of interest statement

Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Figures

Fig. 1
Fig. 1. Activation of the AA/eicosanoid pathway and induction of COX-2 after Cr(VI) exposure.
BALB/cJ mice were exposed to zinc chromate (ZnCrO4) or normal saline by intranasal instillation once per week for 12 weeks. (A) Lung tissues were snap-frozen and subjected to a metabolomics assay. The heat map displays the levels of AA/eicosanoid pathway intermediate metabolites in the lung tissues of the Cr(VI)-treated mice and the control mice. (B) GSEA analysis for the RNA-seq data reveals significant enrichment of the AA/eicosanoid metabolism pathway-related genes among the differentially expressed genes between B2B and Cr-T cells. (C) IHC staining was performed to detect COX-2 in formalin-fixed lung tissues of mice in the Cr(VI) exposure and control groups. Scale bar, 25 μm. (D) COX-2 mRNA expression in the lung tissues was measured using RT-qPCR assay. Ctrl: n=15, Cr(VI) instillation: n=20. (E) The plasma of the mice was separated through density gradient centrifugation, and the COX-2 protein level in the plasma was determined using an ELISA assay. Ctrl: n=15, Cr(VI) instillation: n=20. (F) COX-2 protein expression in B2B and Cr-T cells was assessed using an immunoblotting assay. Data are presented as the mean ± SE of each group of animals. ***, P < 0.001.
Fig. 1
Fig. 1. Activation of the AA/eicosanoid pathway and induction of COX-2 after Cr(VI) exposure.
BALB/cJ mice were exposed to zinc chromate (ZnCrO4) or normal saline by intranasal instillation once per week for 12 weeks. (A) Lung tissues were snap-frozen and subjected to a metabolomics assay. The heat map displays the levels of AA/eicosanoid pathway intermediate metabolites in the lung tissues of the Cr(VI)-treated mice and the control mice. (B) GSEA analysis for the RNA-seq data reveals significant enrichment of the AA/eicosanoid metabolism pathway-related genes among the differentially expressed genes between B2B and Cr-T cells. (C) IHC staining was performed to detect COX-2 in formalin-fixed lung tissues of mice in the Cr(VI) exposure and control groups. Scale bar, 25 μm. (D) COX-2 mRNA expression in the lung tissues was measured using RT-qPCR assay. Ctrl: n=15, Cr(VI) instillation: n=20. (E) The plasma of the mice was separated through density gradient centrifugation, and the COX-2 protein level in the plasma was determined using an ELISA assay. Ctrl: n=15, Cr(VI) instillation: n=20. (F) COX-2 protein expression in B2B and Cr-T cells was assessed using an immunoblotting assay. Data are presented as the mean ± SE of each group of animals. ***, P < 0.001.
Fig. 1
Fig. 1. Activation of the AA/eicosanoid pathway and induction of COX-2 after Cr(VI) exposure.
BALB/cJ mice were exposed to zinc chromate (ZnCrO4) or normal saline by intranasal instillation once per week for 12 weeks. (A) Lung tissues were snap-frozen and subjected to a metabolomics assay. The heat map displays the levels of AA/eicosanoid pathway intermediate metabolites in the lung tissues of the Cr(VI)-treated mice and the control mice. (B) GSEA analysis for the RNA-seq data reveals significant enrichment of the AA/eicosanoid metabolism pathway-related genes among the differentially expressed genes between B2B and Cr-T cells. (C) IHC staining was performed to detect COX-2 in formalin-fixed lung tissues of mice in the Cr(VI) exposure and control groups. Scale bar, 25 μm. (D) COX-2 mRNA expression in the lung tissues was measured using RT-qPCR assay. Ctrl: n=15, Cr(VI) instillation: n=20. (E) The plasma of the mice was separated through density gradient centrifugation, and the COX-2 protein level in the plasma was determined using an ELISA assay. Ctrl: n=15, Cr(VI) instillation: n=20. (F) COX-2 protein expression in B2B and Cr-T cells was assessed using an immunoblotting assay. Data are presented as the mean ± SE of each group of animals. ***, P < 0.001.
Fig. 1
Fig. 1. Activation of the AA/eicosanoid pathway and induction of COX-2 after Cr(VI) exposure.
BALB/cJ mice were exposed to zinc chromate (ZnCrO4) or normal saline by intranasal instillation once per week for 12 weeks. (A) Lung tissues were snap-frozen and subjected to a metabolomics assay. The heat map displays the levels of AA/eicosanoid pathway intermediate metabolites in the lung tissues of the Cr(VI)-treated mice and the control mice. (B) GSEA analysis for the RNA-seq data reveals significant enrichment of the AA/eicosanoid metabolism pathway-related genes among the differentially expressed genes between B2B and Cr-T cells. (C) IHC staining was performed to detect COX-2 in formalin-fixed lung tissues of mice in the Cr(VI) exposure and control groups. Scale bar, 25 μm. (D) COX-2 mRNA expression in the lung tissues was measured using RT-qPCR assay. Ctrl: n=15, Cr(VI) instillation: n=20. (E) The plasma of the mice was separated through density gradient centrifugation, and the COX-2 protein level in the plasma was determined using an ELISA assay. Ctrl: n=15, Cr(VI) instillation: n=20. (F) COX-2 protein expression in B2B and Cr-T cells was assessed using an immunoblotting assay. Data are presented as the mean ± SE of each group of animals. ***, P < 0.001.
Fig. 1
Fig. 1. Activation of the AA/eicosanoid pathway and induction of COX-2 after Cr(VI) exposure.
BALB/cJ mice were exposed to zinc chromate (ZnCrO4) or normal saline by intranasal instillation once per week for 12 weeks. (A) Lung tissues were snap-frozen and subjected to a metabolomics assay. The heat map displays the levels of AA/eicosanoid pathway intermediate metabolites in the lung tissues of the Cr(VI)-treated mice and the control mice. (B) GSEA analysis for the RNA-seq data reveals significant enrichment of the AA/eicosanoid metabolism pathway-related genes among the differentially expressed genes between B2B and Cr-T cells. (C) IHC staining was performed to detect COX-2 in formalin-fixed lung tissues of mice in the Cr(VI) exposure and control groups. Scale bar, 25 μm. (D) COX-2 mRNA expression in the lung tissues was measured using RT-qPCR assay. Ctrl: n=15, Cr(VI) instillation: n=20. (E) The plasma of the mice was separated through density gradient centrifugation, and the COX-2 protein level in the plasma was determined using an ELISA assay. Ctrl: n=15, Cr(VI) instillation: n=20. (F) COX-2 protein expression in B2B and Cr-T cells was assessed using an immunoblotting assay. Data are presented as the mean ± SE of each group of animals. ***, P < 0.001.
Fig. 1
Fig. 1. Activation of the AA/eicosanoid pathway and induction of COX-2 after Cr(VI) exposure.
BALB/cJ mice were exposed to zinc chromate (ZnCrO4) or normal saline by intranasal instillation once per week for 12 weeks. (A) Lung tissues were snap-frozen and subjected to a metabolomics assay. The heat map displays the levels of AA/eicosanoid pathway intermediate metabolites in the lung tissues of the Cr(VI)-treated mice and the control mice. (B) GSEA analysis for the RNA-seq data reveals significant enrichment of the AA/eicosanoid metabolism pathway-related genes among the differentially expressed genes between B2B and Cr-T cells. (C) IHC staining was performed to detect COX-2 in formalin-fixed lung tissues of mice in the Cr(VI) exposure and control groups. Scale bar, 25 μm. (D) COX-2 mRNA expression in the lung tissues was measured using RT-qPCR assay. Ctrl: n=15, Cr(VI) instillation: n=20. (E) The plasma of the mice was separated through density gradient centrifugation, and the COX-2 protein level in the plasma was determined using an ELISA assay. Ctrl: n=15, Cr(VI) instillation: n=20. (F) COX-2 protein expression in B2B and Cr-T cells was assessed using an immunoblotting assay. Data are presented as the mean ± SE of each group of animals. ***, P < 0.001.
Fig. 2
Fig. 2. COX-2 acts as an oncogene to promote Cr(VI)-induced lung cancer.
(A) B2B cells with stable COX-2 overexpression (COX-2 OE) or vector control were generated by lentiviral transduction and puromycin selection. COX-2 protein levels were assessed using an immunoblotting assay. (B) COX-2 overexpressing and control B2B cells were treated with 0.5 μM Cr(VI) for up to 4 months. The colony formation ability of these cells was evaluated at 2 months, 3 months, and 4 months using the soft agar assay. Left: representative images of the soft agar assay; right: quantification of the soft agar assay. Data represent the mean ± SE of six replicate wells at each time point. (C and D) Lentivirus-mediated overexpression of COX-2 was achieved in Cr-T cells. (C) Cell viability was determined by counting cell numbers. (D) A transwell migration assay was conducted to evaluate cell migratory ability. Left panel: representative image of migrated cells; right panel: quantification of migrated cells. Scale bar, 100 μm. (E-H) Cr-T cells stably overexpressing COX-2 or control Cr-T cells were subcutaneously injected into posterior flanks of 6-week-old nude mice at a dose of 2x106 cells per injection. The xenografts were collected and analyzed 18 days after injection. n=8 for each group. (E) Tumor sizes were measured, and the volumes were calculated using the formula: tumor volume = (length × width2) × 0.5. The comparison of the two curves was performed using two-way ANOVA followed by post hoc multiple comparisons. (F) The representative images of the tumors are shown. (G) Measurement of tumor weights. (H) IHC staining for CD31-positive microvessels in xenografts. Left panel: representative images of IHC staining; right panel: quantification of CD31-positive microvessels, which were normalized to the control group. Scale bar, 50 μm. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments unless otherwise specified. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 2
Fig. 2. COX-2 acts as an oncogene to promote Cr(VI)-induced lung cancer.
(A) B2B cells with stable COX-2 overexpression (COX-2 OE) or vector control were generated by lentiviral transduction and puromycin selection. COX-2 protein levels were assessed using an immunoblotting assay. (B) COX-2 overexpressing and control B2B cells were treated with 0.5 μM Cr(VI) for up to 4 months. The colony formation ability of these cells was evaluated at 2 months, 3 months, and 4 months using the soft agar assay. Left: representative images of the soft agar assay; right: quantification of the soft agar assay. Data represent the mean ± SE of six replicate wells at each time point. (C and D) Lentivirus-mediated overexpression of COX-2 was achieved in Cr-T cells. (C) Cell viability was determined by counting cell numbers. (D) A transwell migration assay was conducted to evaluate cell migratory ability. Left panel: representative image of migrated cells; right panel: quantification of migrated cells. Scale bar, 100 μm. (E-H) Cr-T cells stably overexpressing COX-2 or control Cr-T cells were subcutaneously injected into posterior flanks of 6-week-old nude mice at a dose of 2x106 cells per injection. The xenografts were collected and analyzed 18 days after injection. n=8 for each group. (E) Tumor sizes were measured, and the volumes were calculated using the formula: tumor volume = (length × width2) × 0.5. The comparison of the two curves was performed using two-way ANOVA followed by post hoc multiple comparisons. (F) The representative images of the tumors are shown. (G) Measurement of tumor weights. (H) IHC staining for CD31-positive microvessels in xenografts. Left panel: representative images of IHC staining; right panel: quantification of CD31-positive microvessels, which were normalized to the control group. Scale bar, 50 μm. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments unless otherwise specified. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 2
Fig. 2. COX-2 acts as an oncogene to promote Cr(VI)-induced lung cancer.
(A) B2B cells with stable COX-2 overexpression (COX-2 OE) or vector control were generated by lentiviral transduction and puromycin selection. COX-2 protein levels were assessed using an immunoblotting assay. (B) COX-2 overexpressing and control B2B cells were treated with 0.5 μM Cr(VI) for up to 4 months. The colony formation ability of these cells was evaluated at 2 months, 3 months, and 4 months using the soft agar assay. Left: representative images of the soft agar assay; right: quantification of the soft agar assay. Data represent the mean ± SE of six replicate wells at each time point. (C and D) Lentivirus-mediated overexpression of COX-2 was achieved in Cr-T cells. (C) Cell viability was determined by counting cell numbers. (D) A transwell migration assay was conducted to evaluate cell migratory ability. Left panel: representative image of migrated cells; right panel: quantification of migrated cells. Scale bar, 100 μm. (E-H) Cr-T cells stably overexpressing COX-2 or control Cr-T cells were subcutaneously injected into posterior flanks of 6-week-old nude mice at a dose of 2x106 cells per injection. The xenografts were collected and analyzed 18 days after injection. n=8 for each group. (E) Tumor sizes were measured, and the volumes were calculated using the formula: tumor volume = (length × width2) × 0.5. The comparison of the two curves was performed using two-way ANOVA followed by post hoc multiple comparisons. (F) The representative images of the tumors are shown. (G) Measurement of tumor weights. (H) IHC staining for CD31-positive microvessels in xenografts. Left panel: representative images of IHC staining; right panel: quantification of CD31-positive microvessels, which were normalized to the control group. Scale bar, 50 μm. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments unless otherwise specified. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 2
Fig. 2. COX-2 acts as an oncogene to promote Cr(VI)-induced lung cancer.
(A) B2B cells with stable COX-2 overexpression (COX-2 OE) or vector control were generated by lentiviral transduction and puromycin selection. COX-2 protein levels were assessed using an immunoblotting assay. (B) COX-2 overexpressing and control B2B cells were treated with 0.5 μM Cr(VI) for up to 4 months. The colony formation ability of these cells was evaluated at 2 months, 3 months, and 4 months using the soft agar assay. Left: representative images of the soft agar assay; right: quantification of the soft agar assay. Data represent the mean ± SE of six replicate wells at each time point. (C and D) Lentivirus-mediated overexpression of COX-2 was achieved in Cr-T cells. (C) Cell viability was determined by counting cell numbers. (D) A transwell migration assay was conducted to evaluate cell migratory ability. Left panel: representative image of migrated cells; right panel: quantification of migrated cells. Scale bar, 100 μm. (E-H) Cr-T cells stably overexpressing COX-2 or control Cr-T cells were subcutaneously injected into posterior flanks of 6-week-old nude mice at a dose of 2x106 cells per injection. The xenografts were collected and analyzed 18 days after injection. n=8 for each group. (E) Tumor sizes were measured, and the volumes were calculated using the formula: tumor volume = (length × width2) × 0.5. The comparison of the two curves was performed using two-way ANOVA followed by post hoc multiple comparisons. (F) The representative images of the tumors are shown. (G) Measurement of tumor weights. (H) IHC staining for CD31-positive microvessels in xenografts. Left panel: representative images of IHC staining; right panel: quantification of CD31-positive microvessels, which were normalized to the control group. Scale bar, 50 μm. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments unless otherwise specified. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 2
Fig. 2. COX-2 acts as an oncogene to promote Cr(VI)-induced lung cancer.
(A) B2B cells with stable COX-2 overexpression (COX-2 OE) or vector control were generated by lentiviral transduction and puromycin selection. COX-2 protein levels were assessed using an immunoblotting assay. (B) COX-2 overexpressing and control B2B cells were treated with 0.5 μM Cr(VI) for up to 4 months. The colony formation ability of these cells was evaluated at 2 months, 3 months, and 4 months using the soft agar assay. Left: representative images of the soft agar assay; right: quantification of the soft agar assay. Data represent the mean ± SE of six replicate wells at each time point. (C and D) Lentivirus-mediated overexpression of COX-2 was achieved in Cr-T cells. (C) Cell viability was determined by counting cell numbers. (D) A transwell migration assay was conducted to evaluate cell migratory ability. Left panel: representative image of migrated cells; right panel: quantification of migrated cells. Scale bar, 100 μm. (E-H) Cr-T cells stably overexpressing COX-2 or control Cr-T cells were subcutaneously injected into posterior flanks of 6-week-old nude mice at a dose of 2x106 cells per injection. The xenografts were collected and analyzed 18 days after injection. n=8 for each group. (E) Tumor sizes were measured, and the volumes were calculated using the formula: tumor volume = (length × width2) × 0.5. The comparison of the two curves was performed using two-way ANOVA followed by post hoc multiple comparisons. (F) The representative images of the tumors are shown. (G) Measurement of tumor weights. (H) IHC staining for CD31-positive microvessels in xenografts. Left panel: representative images of IHC staining; right panel: quantification of CD31-positive microvessels, which were normalized to the control group. Scale bar, 50 μm. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments unless otherwise specified. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 2
Fig. 2. COX-2 acts as an oncogene to promote Cr(VI)-induced lung cancer.
(A) B2B cells with stable COX-2 overexpression (COX-2 OE) or vector control were generated by lentiviral transduction and puromycin selection. COX-2 protein levels were assessed using an immunoblotting assay. (B) COX-2 overexpressing and control B2B cells were treated with 0.5 μM Cr(VI) for up to 4 months. The colony formation ability of these cells was evaluated at 2 months, 3 months, and 4 months using the soft agar assay. Left: representative images of the soft agar assay; right: quantification of the soft agar assay. Data represent the mean ± SE of six replicate wells at each time point. (C and D) Lentivirus-mediated overexpression of COX-2 was achieved in Cr-T cells. (C) Cell viability was determined by counting cell numbers. (D) A transwell migration assay was conducted to evaluate cell migratory ability. Left panel: representative image of migrated cells; right panel: quantification of migrated cells. Scale bar, 100 μm. (E-H) Cr-T cells stably overexpressing COX-2 or control Cr-T cells were subcutaneously injected into posterior flanks of 6-week-old nude mice at a dose of 2x106 cells per injection. The xenografts were collected and analyzed 18 days after injection. n=8 for each group. (E) Tumor sizes were measured, and the volumes were calculated using the formula: tumor volume = (length × width2) × 0.5. The comparison of the two curves was performed using two-way ANOVA followed by post hoc multiple comparisons. (F) The representative images of the tumors are shown. (G) Measurement of tumor weights. (H) IHC staining for CD31-positive microvessels in xenografts. Left panel: representative images of IHC staining; right panel: quantification of CD31-positive microvessels, which were normalized to the control group. Scale bar, 50 μm. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments unless otherwise specified. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 2
Fig. 2. COX-2 acts as an oncogene to promote Cr(VI)-induced lung cancer.
(A) B2B cells with stable COX-2 overexpression (COX-2 OE) or vector control were generated by lentiviral transduction and puromycin selection. COX-2 protein levels were assessed using an immunoblotting assay. (B) COX-2 overexpressing and control B2B cells were treated with 0.5 μM Cr(VI) for up to 4 months. The colony formation ability of these cells was evaluated at 2 months, 3 months, and 4 months using the soft agar assay. Left: representative images of the soft agar assay; right: quantification of the soft agar assay. Data represent the mean ± SE of six replicate wells at each time point. (C and D) Lentivirus-mediated overexpression of COX-2 was achieved in Cr-T cells. (C) Cell viability was determined by counting cell numbers. (D) A transwell migration assay was conducted to evaluate cell migratory ability. Left panel: representative image of migrated cells; right panel: quantification of migrated cells. Scale bar, 100 μm. (E-H) Cr-T cells stably overexpressing COX-2 or control Cr-T cells were subcutaneously injected into posterior flanks of 6-week-old nude mice at a dose of 2x106 cells per injection. The xenografts were collected and analyzed 18 days after injection. n=8 for each group. (E) Tumor sizes were measured, and the volumes were calculated using the formula: tumor volume = (length × width2) × 0.5. The comparison of the two curves was performed using two-way ANOVA followed by post hoc multiple comparisons. (F) The representative images of the tumors are shown. (G) Measurement of tumor weights. (H) IHC staining for CD31-positive microvessels in xenografts. Left panel: representative images of IHC staining; right panel: quantification of CD31-positive microvessels, which were normalized to the control group. Scale bar, 50 μm. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments unless otherwise specified. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 2
Fig. 2. COX-2 acts as an oncogene to promote Cr(VI)-induced lung cancer.
(A) B2B cells with stable COX-2 overexpression (COX-2 OE) or vector control were generated by lentiviral transduction and puromycin selection. COX-2 protein levels were assessed using an immunoblotting assay. (B) COX-2 overexpressing and control B2B cells were treated with 0.5 μM Cr(VI) for up to 4 months. The colony formation ability of these cells was evaluated at 2 months, 3 months, and 4 months using the soft agar assay. Left: representative images of the soft agar assay; right: quantification of the soft agar assay. Data represent the mean ± SE of six replicate wells at each time point. (C and D) Lentivirus-mediated overexpression of COX-2 was achieved in Cr-T cells. (C) Cell viability was determined by counting cell numbers. (D) A transwell migration assay was conducted to evaluate cell migratory ability. Left panel: representative image of migrated cells; right panel: quantification of migrated cells. Scale bar, 100 μm. (E-H) Cr-T cells stably overexpressing COX-2 or control Cr-T cells were subcutaneously injected into posterior flanks of 6-week-old nude mice at a dose of 2x106 cells per injection. The xenografts were collected and analyzed 18 days after injection. n=8 for each group. (E) Tumor sizes were measured, and the volumes were calculated using the formula: tumor volume = (length × width2) × 0.5. The comparison of the two curves was performed using two-way ANOVA followed by post hoc multiple comparisons. (F) The representative images of the tumors are shown. (G) Measurement of tumor weights. (H) IHC staining for CD31-positive microvessels in xenografts. Left panel: representative images of IHC staining; right panel: quantification of CD31-positive microvessels, which were normalized to the control group. Scale bar, 50 μm. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments unless otherwise specified. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 3
Fig. 3. COX-2 depletion inhibits the cancerous phenotypes of Cr-T cells.
The lentivirus-mediated CRISPR/Cas9 technique was used to achieve COX-2 knockout (KO) in Cr-T cells, while negative control cells were transduced with lentivirus expressing a non-targeting sgRNA. (A) COX-2 protein levels were assessed via immunoblotting assay to validate the knockout. (B) Cell viability was determined by cell counting. (C) The migratory ability of the cells was measured using a transwell migration assay. Left: representative images of migrated cells; right: quantification of the migrated cells. Scale bar, 100 μm. (D-G) Tumor growth assay in nude mice was performed using COX-2-null and control Cr-T cells as above, and the xenografts were harvested 21 days after implantation. n=10 for each group. (D) Tumor sizes were measured at indicated time points, and tumor volumes were calculated and analyzed. The growth curve was plotted and compared with two-way ANOVA followed by post hoc multiple comparisons. (E) The representative images of the tumors are shown. (F) Tumor weights were measured. (G) IHC staining for CD31-positive microvessels in the xenografts. Left panel: representative images of the IHC staining; right panel: quantification of CD31-positive microvessels. The number was normalized to the control group. Scale bar, 50 μm. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 3
Fig. 3. COX-2 depletion inhibits the cancerous phenotypes of Cr-T cells.
The lentivirus-mediated CRISPR/Cas9 technique was used to achieve COX-2 knockout (KO) in Cr-T cells, while negative control cells were transduced with lentivirus expressing a non-targeting sgRNA. (A) COX-2 protein levels were assessed via immunoblotting assay to validate the knockout. (B) Cell viability was determined by cell counting. (C) The migratory ability of the cells was measured using a transwell migration assay. Left: representative images of migrated cells; right: quantification of the migrated cells. Scale bar, 100 μm. (D-G) Tumor growth assay in nude mice was performed using COX-2-null and control Cr-T cells as above, and the xenografts were harvested 21 days after implantation. n=10 for each group. (D) Tumor sizes were measured at indicated time points, and tumor volumes were calculated and analyzed. The growth curve was plotted and compared with two-way ANOVA followed by post hoc multiple comparisons. (E) The representative images of the tumors are shown. (F) Tumor weights were measured. (G) IHC staining for CD31-positive microvessels in the xenografts. Left panel: representative images of the IHC staining; right panel: quantification of CD31-positive microvessels. The number was normalized to the control group. Scale bar, 50 μm. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 3
Fig. 3. COX-2 depletion inhibits the cancerous phenotypes of Cr-T cells.
The lentivirus-mediated CRISPR/Cas9 technique was used to achieve COX-2 knockout (KO) in Cr-T cells, while negative control cells were transduced with lentivirus expressing a non-targeting sgRNA. (A) COX-2 protein levels were assessed via immunoblotting assay to validate the knockout. (B) Cell viability was determined by cell counting. (C) The migratory ability of the cells was measured using a transwell migration assay. Left: representative images of migrated cells; right: quantification of the migrated cells. Scale bar, 100 μm. (D-G) Tumor growth assay in nude mice was performed using COX-2-null and control Cr-T cells as above, and the xenografts were harvested 21 days after implantation. n=10 for each group. (D) Tumor sizes were measured at indicated time points, and tumor volumes were calculated and analyzed. The growth curve was plotted and compared with two-way ANOVA followed by post hoc multiple comparisons. (E) The representative images of the tumors are shown. (F) Tumor weights were measured. (G) IHC staining for CD31-positive microvessels in the xenografts. Left panel: representative images of the IHC staining; right panel: quantification of CD31-positive microvessels. The number was normalized to the control group. Scale bar, 50 μm. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 3
Fig. 3. COX-2 depletion inhibits the cancerous phenotypes of Cr-T cells.
The lentivirus-mediated CRISPR/Cas9 technique was used to achieve COX-2 knockout (KO) in Cr-T cells, while negative control cells were transduced with lentivirus expressing a non-targeting sgRNA. (A) COX-2 protein levels were assessed via immunoblotting assay to validate the knockout. (B) Cell viability was determined by cell counting. (C) The migratory ability of the cells was measured using a transwell migration assay. Left: representative images of migrated cells; right: quantification of the migrated cells. Scale bar, 100 μm. (D-G) Tumor growth assay in nude mice was performed using COX-2-null and control Cr-T cells as above, and the xenografts were harvested 21 days after implantation. n=10 for each group. (D) Tumor sizes were measured at indicated time points, and tumor volumes were calculated and analyzed. The growth curve was plotted and compared with two-way ANOVA followed by post hoc multiple comparisons. (E) The representative images of the tumors are shown. (F) Tumor weights were measured. (G) IHC staining for CD31-positive microvessels in the xenografts. Left panel: representative images of the IHC staining; right panel: quantification of CD31-positive microvessels. The number was normalized to the control group. Scale bar, 50 μm. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 3
Fig. 3. COX-2 depletion inhibits the cancerous phenotypes of Cr-T cells.
The lentivirus-mediated CRISPR/Cas9 technique was used to achieve COX-2 knockout (KO) in Cr-T cells, while negative control cells were transduced with lentivirus expressing a non-targeting sgRNA. (A) COX-2 protein levels were assessed via immunoblotting assay to validate the knockout. (B) Cell viability was determined by cell counting. (C) The migratory ability of the cells was measured using a transwell migration assay. Left: representative images of migrated cells; right: quantification of the migrated cells. Scale bar, 100 μm. (D-G) Tumor growth assay in nude mice was performed using COX-2-null and control Cr-T cells as above, and the xenografts were harvested 21 days after implantation. n=10 for each group. (D) Tumor sizes were measured at indicated time points, and tumor volumes were calculated and analyzed. The growth curve was plotted and compared with two-way ANOVA followed by post hoc multiple comparisons. (E) The representative images of the tumors are shown. (F) Tumor weights were measured. (G) IHC staining for CD31-positive microvessels in the xenografts. Left panel: representative images of the IHC staining; right panel: quantification of CD31-positive microvessels. The number was normalized to the control group. Scale bar, 50 μm. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 3
Fig. 3. COX-2 depletion inhibits the cancerous phenotypes of Cr-T cells.
The lentivirus-mediated CRISPR/Cas9 technique was used to achieve COX-2 knockout (KO) in Cr-T cells, while negative control cells were transduced with lentivirus expressing a non-targeting sgRNA. (A) COX-2 protein levels were assessed via immunoblotting assay to validate the knockout. (B) Cell viability was determined by cell counting. (C) The migratory ability of the cells was measured using a transwell migration assay. Left: representative images of migrated cells; right: quantification of the migrated cells. Scale bar, 100 μm. (D-G) Tumor growth assay in nude mice was performed using COX-2-null and control Cr-T cells as above, and the xenografts were harvested 21 days after implantation. n=10 for each group. (D) Tumor sizes were measured at indicated time points, and tumor volumes were calculated and analyzed. The growth curve was plotted and compared with two-way ANOVA followed by post hoc multiple comparisons. (E) The representative images of the tumors are shown. (F) Tumor weights were measured. (G) IHC staining for CD31-positive microvessels in the xenografts. Left panel: representative images of the IHC staining; right panel: quantification of CD31-positive microvessels. The number was normalized to the control group. Scale bar, 50 μm. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 3
Fig. 3. COX-2 depletion inhibits the cancerous phenotypes of Cr-T cells.
The lentivirus-mediated CRISPR/Cas9 technique was used to achieve COX-2 knockout (KO) in Cr-T cells, while negative control cells were transduced with lentivirus expressing a non-targeting sgRNA. (A) COX-2 protein levels were assessed via immunoblotting assay to validate the knockout. (B) Cell viability was determined by cell counting. (C) The migratory ability of the cells was measured using a transwell migration assay. Left: representative images of migrated cells; right: quantification of the migrated cells. Scale bar, 100 μm. (D-G) Tumor growth assay in nude mice was performed using COX-2-null and control Cr-T cells as above, and the xenografts were harvested 21 days after implantation. n=10 for each group. (D) Tumor sizes were measured at indicated time points, and tumor volumes were calculated and analyzed. The growth curve was plotted and compared with two-way ANOVA followed by post hoc multiple comparisons. (E) The representative images of the tumors are shown. (F) Tumor weights were measured. (G) IHC staining for CD31-positive microvessels in the xenografts. Left panel: representative images of the IHC staining; right panel: quantification of CD31-positive microvessels. The number was normalized to the control group. Scale bar, 50 μm. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 4
Fig. 4. Nrf2 regulates COX-2 expression by directly binding to its promoter region.
(A) BALB/cJ mice were treated as shown in Fig. 1. IHC staining was performed to evaluate Nrf2 expression in the formalin-fixed lung tissues. Scale bar, 25 μm. (B) The correlation between COX-2 and Nrf2 expression in the lungs was evaluated using the Pearson correlation analysis. (C and D) Nrf2 was knocked out in Cr-T cells using the lentiviral CRISPR/Cas9-mediated genome editing technique. The control cells were transduced with lentivirus carrying a non-targeting sgRNA. (C) Nrf2 and COX-2 protein was measured using immunoblotting assay. Quantification of the COX-2 signal was performed using the Image Lab software. The data are presented as the mean ± SE of three independent experiments. (D) COX-2 mRNA in the control and Nrf2-null cells was determined using RT-qPCR assay. The data are presented as the mean ± SE of three independent experiments. (E) Nrf2 expression in B2B cells was induced using a lentivirus-mediated Tet-on system harboring the Nrf2 coding sequence. Control cells were transduced with lentivirus generated from the control vector. Both types of cells were incubated with 1.0 mg/mL tetracycline for 3 days. Nrf2 and COX-2 protein expression was measured by immunoblotting assay. (F) A schematic graph showing the potential binding site of Nrf2 in the promoter region of COX-2. There is an ARE consensus sequence between nucleotides −562 and −572. (G) The ChIP-PCR assay was performed to investigate the binding of Nrf2 to the COX-2 promoter in Cr-T cells, as described in the Materials and Methods section. Left: a representative image of the agarose gel displaying PCR products. COX-2 NT represents the non-target region in the COX-2 promoter without the ARE consensus sequence. Right: quantification of the bands on agarose gel. The data are presented as the mean ± SE of three independent experiments. (H) The interaction between Nrf2 and the COX-2 promoter was analyzed in both control and Nrf2-depleted Cr-T cells using the ChIP-PCR assay. Left: a representative image of PCR products was shown. Right: quantification of the agarose gel bands. The data are presented as the mean ± SE of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 4
Fig. 4. Nrf2 regulates COX-2 expression by directly binding to its promoter region.
(A) BALB/cJ mice were treated as shown in Fig. 1. IHC staining was performed to evaluate Nrf2 expression in the formalin-fixed lung tissues. Scale bar, 25 μm. (B) The correlation between COX-2 and Nrf2 expression in the lungs was evaluated using the Pearson correlation analysis. (C and D) Nrf2 was knocked out in Cr-T cells using the lentiviral CRISPR/Cas9-mediated genome editing technique. The control cells were transduced with lentivirus carrying a non-targeting sgRNA. (C) Nrf2 and COX-2 protein was measured using immunoblotting assay. Quantification of the COX-2 signal was performed using the Image Lab software. The data are presented as the mean ± SE of three independent experiments. (D) COX-2 mRNA in the control and Nrf2-null cells was determined using RT-qPCR assay. The data are presented as the mean ± SE of three independent experiments. (E) Nrf2 expression in B2B cells was induced using a lentivirus-mediated Tet-on system harboring the Nrf2 coding sequence. Control cells were transduced with lentivirus generated from the control vector. Both types of cells were incubated with 1.0 mg/mL tetracycline for 3 days. Nrf2 and COX-2 protein expression was measured by immunoblotting assay. (F) A schematic graph showing the potential binding site of Nrf2 in the promoter region of COX-2. There is an ARE consensus sequence between nucleotides −562 and −572. (G) The ChIP-PCR assay was performed to investigate the binding of Nrf2 to the COX-2 promoter in Cr-T cells, as described in the Materials and Methods section. Left: a representative image of the agarose gel displaying PCR products. COX-2 NT represents the non-target region in the COX-2 promoter without the ARE consensus sequence. Right: quantification of the bands on agarose gel. The data are presented as the mean ± SE of three independent experiments. (H) The interaction between Nrf2 and the COX-2 promoter was analyzed in both control and Nrf2-depleted Cr-T cells using the ChIP-PCR assay. Left: a representative image of PCR products was shown. Right: quantification of the agarose gel bands. The data are presented as the mean ± SE of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 4
Fig. 4. Nrf2 regulates COX-2 expression by directly binding to its promoter region.
(A) BALB/cJ mice were treated as shown in Fig. 1. IHC staining was performed to evaluate Nrf2 expression in the formalin-fixed lung tissues. Scale bar, 25 μm. (B) The correlation between COX-2 and Nrf2 expression in the lungs was evaluated using the Pearson correlation analysis. (C and D) Nrf2 was knocked out in Cr-T cells using the lentiviral CRISPR/Cas9-mediated genome editing technique. The control cells were transduced with lentivirus carrying a non-targeting sgRNA. (C) Nrf2 and COX-2 protein was measured using immunoblotting assay. Quantification of the COX-2 signal was performed using the Image Lab software. The data are presented as the mean ± SE of three independent experiments. (D) COX-2 mRNA in the control and Nrf2-null cells was determined using RT-qPCR assay. The data are presented as the mean ± SE of three independent experiments. (E) Nrf2 expression in B2B cells was induced using a lentivirus-mediated Tet-on system harboring the Nrf2 coding sequence. Control cells were transduced with lentivirus generated from the control vector. Both types of cells were incubated with 1.0 mg/mL tetracycline for 3 days. Nrf2 and COX-2 protein expression was measured by immunoblotting assay. (F) A schematic graph showing the potential binding site of Nrf2 in the promoter region of COX-2. There is an ARE consensus sequence between nucleotides −562 and −572. (G) The ChIP-PCR assay was performed to investigate the binding of Nrf2 to the COX-2 promoter in Cr-T cells, as described in the Materials and Methods section. Left: a representative image of the agarose gel displaying PCR products. COX-2 NT represents the non-target region in the COX-2 promoter without the ARE consensus sequence. Right: quantification of the bands on agarose gel. The data are presented as the mean ± SE of three independent experiments. (H) The interaction between Nrf2 and the COX-2 promoter was analyzed in both control and Nrf2-depleted Cr-T cells using the ChIP-PCR assay. Left: a representative image of PCR products was shown. Right: quantification of the agarose gel bands. The data are presented as the mean ± SE of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 4
Fig. 4. Nrf2 regulates COX-2 expression by directly binding to its promoter region.
(A) BALB/cJ mice were treated as shown in Fig. 1. IHC staining was performed to evaluate Nrf2 expression in the formalin-fixed lung tissues. Scale bar, 25 μm. (B) The correlation between COX-2 and Nrf2 expression in the lungs was evaluated using the Pearson correlation analysis. (C and D) Nrf2 was knocked out in Cr-T cells using the lentiviral CRISPR/Cas9-mediated genome editing technique. The control cells were transduced with lentivirus carrying a non-targeting sgRNA. (C) Nrf2 and COX-2 protein was measured using immunoblotting assay. Quantification of the COX-2 signal was performed using the Image Lab software. The data are presented as the mean ± SE of three independent experiments. (D) COX-2 mRNA in the control and Nrf2-null cells was determined using RT-qPCR assay. The data are presented as the mean ± SE of three independent experiments. (E) Nrf2 expression in B2B cells was induced using a lentivirus-mediated Tet-on system harboring the Nrf2 coding sequence. Control cells were transduced with lentivirus generated from the control vector. Both types of cells were incubated with 1.0 mg/mL tetracycline for 3 days. Nrf2 and COX-2 protein expression was measured by immunoblotting assay. (F) A schematic graph showing the potential binding site of Nrf2 in the promoter region of COX-2. There is an ARE consensus sequence between nucleotides −562 and −572. (G) The ChIP-PCR assay was performed to investigate the binding of Nrf2 to the COX-2 promoter in Cr-T cells, as described in the Materials and Methods section. Left: a representative image of the agarose gel displaying PCR products. COX-2 NT represents the non-target region in the COX-2 promoter without the ARE consensus sequence. Right: quantification of the bands on agarose gel. The data are presented as the mean ± SE of three independent experiments. (H) The interaction between Nrf2 and the COX-2 promoter was analyzed in both control and Nrf2-depleted Cr-T cells using the ChIP-PCR assay. Left: a representative image of PCR products was shown. Right: quantification of the agarose gel bands. The data are presented as the mean ± SE of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 4
Fig. 4. Nrf2 regulates COX-2 expression by directly binding to its promoter region.
(A) BALB/cJ mice were treated as shown in Fig. 1. IHC staining was performed to evaluate Nrf2 expression in the formalin-fixed lung tissues. Scale bar, 25 μm. (B) The correlation between COX-2 and Nrf2 expression in the lungs was evaluated using the Pearson correlation analysis. (C and D) Nrf2 was knocked out in Cr-T cells using the lentiviral CRISPR/Cas9-mediated genome editing technique. The control cells were transduced with lentivirus carrying a non-targeting sgRNA. (C) Nrf2 and COX-2 protein was measured using immunoblotting assay. Quantification of the COX-2 signal was performed using the Image Lab software. The data are presented as the mean ± SE of three independent experiments. (D) COX-2 mRNA in the control and Nrf2-null cells was determined using RT-qPCR assay. The data are presented as the mean ± SE of three independent experiments. (E) Nrf2 expression in B2B cells was induced using a lentivirus-mediated Tet-on system harboring the Nrf2 coding sequence. Control cells were transduced with lentivirus generated from the control vector. Both types of cells were incubated with 1.0 mg/mL tetracycline for 3 days. Nrf2 and COX-2 protein expression was measured by immunoblotting assay. (F) A schematic graph showing the potential binding site of Nrf2 in the promoter region of COX-2. There is an ARE consensus sequence between nucleotides −562 and −572. (G) The ChIP-PCR assay was performed to investigate the binding of Nrf2 to the COX-2 promoter in Cr-T cells, as described in the Materials and Methods section. Left: a representative image of the agarose gel displaying PCR products. COX-2 NT represents the non-target region in the COX-2 promoter without the ARE consensus sequence. Right: quantification of the bands on agarose gel. The data are presented as the mean ± SE of three independent experiments. (H) The interaction between Nrf2 and the COX-2 promoter was analyzed in both control and Nrf2-depleted Cr-T cells using the ChIP-PCR assay. Left: a representative image of PCR products was shown. Right: quantification of the agarose gel bands. The data are presented as the mean ± SE of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 4
Fig. 4. Nrf2 regulates COX-2 expression by directly binding to its promoter region.
(A) BALB/cJ mice were treated as shown in Fig. 1. IHC staining was performed to evaluate Nrf2 expression in the formalin-fixed lung tissues. Scale bar, 25 μm. (B) The correlation between COX-2 and Nrf2 expression in the lungs was evaluated using the Pearson correlation analysis. (C and D) Nrf2 was knocked out in Cr-T cells using the lentiviral CRISPR/Cas9-mediated genome editing technique. The control cells were transduced with lentivirus carrying a non-targeting sgRNA. (C) Nrf2 and COX-2 protein was measured using immunoblotting assay. Quantification of the COX-2 signal was performed using the Image Lab software. The data are presented as the mean ± SE of three independent experiments. (D) COX-2 mRNA in the control and Nrf2-null cells was determined using RT-qPCR assay. The data are presented as the mean ± SE of three independent experiments. (E) Nrf2 expression in B2B cells was induced using a lentivirus-mediated Tet-on system harboring the Nrf2 coding sequence. Control cells were transduced with lentivirus generated from the control vector. Both types of cells were incubated with 1.0 mg/mL tetracycline for 3 days. Nrf2 and COX-2 protein expression was measured by immunoblotting assay. (F) A schematic graph showing the potential binding site of Nrf2 in the promoter region of COX-2. There is an ARE consensus sequence between nucleotides −562 and −572. (G) The ChIP-PCR assay was performed to investigate the binding of Nrf2 to the COX-2 promoter in Cr-T cells, as described in the Materials and Methods section. Left: a representative image of the agarose gel displaying PCR products. COX-2 NT represents the non-target region in the COX-2 promoter without the ARE consensus sequence. Right: quantification of the bands on agarose gel. The data are presented as the mean ± SE of three independent experiments. (H) The interaction between Nrf2 and the COX-2 promoter was analyzed in both control and Nrf2-depleted Cr-T cells using the ChIP-PCR assay. Left: a representative image of PCR products was shown. Right: quantification of the agarose gel bands. The data are presented as the mean ± SE of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 4
Fig. 4. Nrf2 regulates COX-2 expression by directly binding to its promoter region.
(A) BALB/cJ mice were treated as shown in Fig. 1. IHC staining was performed to evaluate Nrf2 expression in the formalin-fixed lung tissues. Scale bar, 25 μm. (B) The correlation between COX-2 and Nrf2 expression in the lungs was evaluated using the Pearson correlation analysis. (C and D) Nrf2 was knocked out in Cr-T cells using the lentiviral CRISPR/Cas9-mediated genome editing technique. The control cells were transduced with lentivirus carrying a non-targeting sgRNA. (C) Nrf2 and COX-2 protein was measured using immunoblotting assay. Quantification of the COX-2 signal was performed using the Image Lab software. The data are presented as the mean ± SE of three independent experiments. (D) COX-2 mRNA in the control and Nrf2-null cells was determined using RT-qPCR assay. The data are presented as the mean ± SE of three independent experiments. (E) Nrf2 expression in B2B cells was induced using a lentivirus-mediated Tet-on system harboring the Nrf2 coding sequence. Control cells were transduced with lentivirus generated from the control vector. Both types of cells were incubated with 1.0 mg/mL tetracycline for 3 days. Nrf2 and COX-2 protein expression was measured by immunoblotting assay. (F) A schematic graph showing the potential binding site of Nrf2 in the promoter region of COX-2. There is an ARE consensus sequence between nucleotides −562 and −572. (G) The ChIP-PCR assay was performed to investigate the binding of Nrf2 to the COX-2 promoter in Cr-T cells, as described in the Materials and Methods section. Left: a representative image of the agarose gel displaying PCR products. COX-2 NT represents the non-target region in the COX-2 promoter without the ARE consensus sequence. Right: quantification of the bands on agarose gel. The data are presented as the mean ± SE of three independent experiments. (H) The interaction between Nrf2 and the COX-2 promoter was analyzed in both control and Nrf2-depleted Cr-T cells using the ChIP-PCR assay. Left: a representative image of PCR products was shown. Right: quantification of the agarose gel bands. The data are presented as the mean ± SE of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 4
Fig. 4. Nrf2 regulates COX-2 expression by directly binding to its promoter region.
(A) BALB/cJ mice were treated as shown in Fig. 1. IHC staining was performed to evaluate Nrf2 expression in the formalin-fixed lung tissues. Scale bar, 25 μm. (B) The correlation between COX-2 and Nrf2 expression in the lungs was evaluated using the Pearson correlation analysis. (C and D) Nrf2 was knocked out in Cr-T cells using the lentiviral CRISPR/Cas9-mediated genome editing technique. The control cells were transduced with lentivirus carrying a non-targeting sgRNA. (C) Nrf2 and COX-2 protein was measured using immunoblotting assay. Quantification of the COX-2 signal was performed using the Image Lab software. The data are presented as the mean ± SE of three independent experiments. (D) COX-2 mRNA in the control and Nrf2-null cells was determined using RT-qPCR assay. The data are presented as the mean ± SE of three independent experiments. (E) Nrf2 expression in B2B cells was induced using a lentivirus-mediated Tet-on system harboring the Nrf2 coding sequence. Control cells were transduced with lentivirus generated from the control vector. Both types of cells were incubated with 1.0 mg/mL tetracycline for 3 days. Nrf2 and COX-2 protein expression was measured by immunoblotting assay. (F) A schematic graph showing the potential binding site of Nrf2 in the promoter region of COX-2. There is an ARE consensus sequence between nucleotides −562 and −572. (G) The ChIP-PCR assay was performed to investigate the binding of Nrf2 to the COX-2 promoter in Cr-T cells, as described in the Materials and Methods section. Left: a representative image of the agarose gel displaying PCR products. COX-2 NT represents the non-target region in the COX-2 promoter without the ARE consensus sequence. Right: quantification of the bands on agarose gel. The data are presented as the mean ± SE of three independent experiments. (H) The interaction between Nrf2 and the COX-2 promoter was analyzed in both control and Nrf2-depleted Cr-T cells using the ChIP-PCR assay. Left: a representative image of PCR products was shown. Right: quantification of the agarose gel bands. The data are presented as the mean ± SE of three independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 5
Fig. 5. Nrf2 promotes Cr(VI)-induced lung cancer through regulating COX-2 expression.
(A) Nrf2 was knocked out in Cr-T cells, as depicted in Fig. 4C. Nrf2 knockout cells (KO#1 and KO#2) were achieved using Nrf2 sgRNA #1 and #2, respectively. Cell viability of control and Nrf2-null Cr-T cells was assessed by cell counting. (B) The migration ability of control and Nrf2-null Cr-T cells was evaluated using a transwell migration assay. Left: representative images of migrated cells; right: quantification of migrated cells. Scale bar, 100 μm. (C-E) Nrf2 KO Cr-T cells were transduced with lentivirus expressing a Tet-on inducible COX-2 coding sequence, while control cells were transduced with a negative control Tet-on lentivirus. Cells were treated with 1.0 μg/mL tetracycline to induce COX-2 expression. (C) COX-2 protein expression in control and COX-2-overexpressing cells was evaluated using an immunoblotting assay. (D) Cell viability was assessed by cell counting. (E) Cell migration ability was evaluated using a transwell migration assay. Left: representative images of migrated cells; right: quantification of migrated cells. Scale bar, 100 μm. Data are presented as the mean ± SE of three independent experiments. **, P < 0.01; ***, P < 0.001.
Fig. 5
Fig. 5. Nrf2 promotes Cr(VI)-induced lung cancer through regulating COX-2 expression.
(A) Nrf2 was knocked out in Cr-T cells, as depicted in Fig. 4C. Nrf2 knockout cells (KO#1 and KO#2) were achieved using Nrf2 sgRNA #1 and #2, respectively. Cell viability of control and Nrf2-null Cr-T cells was assessed by cell counting. (B) The migration ability of control and Nrf2-null Cr-T cells was evaluated using a transwell migration assay. Left: representative images of migrated cells; right: quantification of migrated cells. Scale bar, 100 μm. (C-E) Nrf2 KO Cr-T cells were transduced with lentivirus expressing a Tet-on inducible COX-2 coding sequence, while control cells were transduced with a negative control Tet-on lentivirus. Cells were treated with 1.0 μg/mL tetracycline to induce COX-2 expression. (C) COX-2 protein expression in control and COX-2-overexpressing cells was evaluated using an immunoblotting assay. (D) Cell viability was assessed by cell counting. (E) Cell migration ability was evaluated using a transwell migration assay. Left: representative images of migrated cells; right: quantification of migrated cells. Scale bar, 100 μm. Data are presented as the mean ± SE of three independent experiments. **, P < 0.01; ***, P < 0.001.
Fig. 5
Fig. 5. Nrf2 promotes Cr(VI)-induced lung cancer through regulating COX-2 expression.
(A) Nrf2 was knocked out in Cr-T cells, as depicted in Fig. 4C. Nrf2 knockout cells (KO#1 and KO#2) were achieved using Nrf2 sgRNA #1 and #2, respectively. Cell viability of control and Nrf2-null Cr-T cells was assessed by cell counting. (B) The migration ability of control and Nrf2-null Cr-T cells was evaluated using a transwell migration assay. Left: representative images of migrated cells; right: quantification of migrated cells. Scale bar, 100 μm. (C-E) Nrf2 KO Cr-T cells were transduced with lentivirus expressing a Tet-on inducible COX-2 coding sequence, while control cells were transduced with a negative control Tet-on lentivirus. Cells were treated with 1.0 μg/mL tetracycline to induce COX-2 expression. (C) COX-2 protein expression in control and COX-2-overexpressing cells was evaluated using an immunoblotting assay. (D) Cell viability was assessed by cell counting. (E) Cell migration ability was evaluated using a transwell migration assay. Left: representative images of migrated cells; right: quantification of migrated cells. Scale bar, 100 μm. Data are presented as the mean ± SE of three independent experiments. **, P < 0.01; ***, P < 0.001.
Fig. 5
Fig. 5. Nrf2 promotes Cr(VI)-induced lung cancer through regulating COX-2 expression.
(A) Nrf2 was knocked out in Cr-T cells, as depicted in Fig. 4C. Nrf2 knockout cells (KO#1 and KO#2) were achieved using Nrf2 sgRNA #1 and #2, respectively. Cell viability of control and Nrf2-null Cr-T cells was assessed by cell counting. (B) The migration ability of control and Nrf2-null Cr-T cells was evaluated using a transwell migration assay. Left: representative images of migrated cells; right: quantification of migrated cells. Scale bar, 100 μm. (C-E) Nrf2 KO Cr-T cells were transduced with lentivirus expressing a Tet-on inducible COX-2 coding sequence, while control cells were transduced with a negative control Tet-on lentivirus. Cells were treated with 1.0 μg/mL tetracycline to induce COX-2 expression. (C) COX-2 protein expression in control and COX-2-overexpressing cells was evaluated using an immunoblotting assay. (D) Cell viability was assessed by cell counting. (E) Cell migration ability was evaluated using a transwell migration assay. Left: representative images of migrated cells; right: quantification of migrated cells. Scale bar, 100 μm. Data are presented as the mean ± SE of three independent experiments. **, P < 0.01; ***, P < 0.001.
Fig. 5
Fig. 5. Nrf2 promotes Cr(VI)-induced lung cancer through regulating COX-2 expression.
(A) Nrf2 was knocked out in Cr-T cells, as depicted in Fig. 4C. Nrf2 knockout cells (KO#1 and KO#2) were achieved using Nrf2 sgRNA #1 and #2, respectively. Cell viability of control and Nrf2-null Cr-T cells was assessed by cell counting. (B) The migration ability of control and Nrf2-null Cr-T cells was evaluated using a transwell migration assay. Left: representative images of migrated cells; right: quantification of migrated cells. Scale bar, 100 μm. (C-E) Nrf2 KO Cr-T cells were transduced with lentivirus expressing a Tet-on inducible COX-2 coding sequence, while control cells were transduced with a negative control Tet-on lentivirus. Cells were treated with 1.0 μg/mL tetracycline to induce COX-2 expression. (C) COX-2 protein expression in control and COX-2-overexpressing cells was evaluated using an immunoblotting assay. (D) Cell viability was assessed by cell counting. (E) Cell migration ability was evaluated using a transwell migration assay. Left: representative images of migrated cells; right: quantification of migrated cells. Scale bar, 100 μm. Data are presented as the mean ± SE of three independent experiments. **, P < 0.01; ***, P < 0.001.
Fig. 6
Fig. 6. miR-379 inhibits Cr-T cell viability and migration via directly targeting COX-2.
(A) miR-379 expression was measured in B2B and Cr-T cells using TaqMan real-time PCR assay. (B) Mice were treated with intranasal instillation of saline or Cr(VI) for 12 weeks. miR-379 level in the lung tissues of the mice was measured using the TaqMan assay. Ctrl: n=15, Cr(VI) instillation: n=20. (C) The correlation between COX-2 mRNA expression and miR-379 level in the mouse lung tissues was analyzed using Pearson correlation analysis. (D) A diagram shows the putative seed sequence between miR-379 and 3’-UTR of COX-2 and the wild-type (WT) and mutant (Mut) luciferase reporter constructs. (E) Luciferase reporter assay was performed using HEK293 cells to detect the relative luciferase activities of WT and Mut COX-2 reporters with or without miR-379 overexpression. The pRL-TK Renilla luciferase vector was used as an internal control. The miR-NC was used as a negative control for miR-379 transfection. (F) Cr-T cells were transfected with 50 nM non-targeting control oligos (miR-NC) or miR-379 mimics and incubated for 72 h. COX-2 protein levels were determined using an immunoblotting assay. (G-H) Cr-T cells stably expressing negative control or an inducible COX-2 were transfected with 50 nM miR-NC or miR-379 mimics; the cells were then treated with 1 μg/mL doxycycline for 3 days to restore COX-2 expression. (G) Cell viability was evaluated by counting cell numbers. (H) The migratory ability of cells was evaluated using a transwell migration assay. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments. Scale bar, 100 μm. n.s., not significant; **, p < 0.01; ***, p < 0.001.
Fig. 6
Fig. 6. miR-379 inhibits Cr-T cell viability and migration via directly targeting COX-2.
(A) miR-379 expression was measured in B2B and Cr-T cells using TaqMan real-time PCR assay. (B) Mice were treated with intranasal instillation of saline or Cr(VI) for 12 weeks. miR-379 level in the lung tissues of the mice was measured using the TaqMan assay. Ctrl: n=15, Cr(VI) instillation: n=20. (C) The correlation between COX-2 mRNA expression and miR-379 level in the mouse lung tissues was analyzed using Pearson correlation analysis. (D) A diagram shows the putative seed sequence between miR-379 and 3’-UTR of COX-2 and the wild-type (WT) and mutant (Mut) luciferase reporter constructs. (E) Luciferase reporter assay was performed using HEK293 cells to detect the relative luciferase activities of WT and Mut COX-2 reporters with or without miR-379 overexpression. The pRL-TK Renilla luciferase vector was used as an internal control. The miR-NC was used as a negative control for miR-379 transfection. (F) Cr-T cells were transfected with 50 nM non-targeting control oligos (miR-NC) or miR-379 mimics and incubated for 72 h. COX-2 protein levels were determined using an immunoblotting assay. (G-H) Cr-T cells stably expressing negative control or an inducible COX-2 were transfected with 50 nM miR-NC or miR-379 mimics; the cells were then treated with 1 μg/mL doxycycline for 3 days to restore COX-2 expression. (G) Cell viability was evaluated by counting cell numbers. (H) The migratory ability of cells was evaluated using a transwell migration assay. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments. Scale bar, 100 μm. n.s., not significant; **, p < 0.01; ***, p < 0.001.
Fig. 6
Fig. 6. miR-379 inhibits Cr-T cell viability and migration via directly targeting COX-2.
(A) miR-379 expression was measured in B2B and Cr-T cells using TaqMan real-time PCR assay. (B) Mice were treated with intranasal instillation of saline or Cr(VI) for 12 weeks. miR-379 level in the lung tissues of the mice was measured using the TaqMan assay. Ctrl: n=15, Cr(VI) instillation: n=20. (C) The correlation between COX-2 mRNA expression and miR-379 level in the mouse lung tissues was analyzed using Pearson correlation analysis. (D) A diagram shows the putative seed sequence between miR-379 and 3’-UTR of COX-2 and the wild-type (WT) and mutant (Mut) luciferase reporter constructs. (E) Luciferase reporter assay was performed using HEK293 cells to detect the relative luciferase activities of WT and Mut COX-2 reporters with or without miR-379 overexpression. The pRL-TK Renilla luciferase vector was used as an internal control. The miR-NC was used as a negative control for miR-379 transfection. (F) Cr-T cells were transfected with 50 nM non-targeting control oligos (miR-NC) or miR-379 mimics and incubated for 72 h. COX-2 protein levels were determined using an immunoblotting assay. (G-H) Cr-T cells stably expressing negative control or an inducible COX-2 were transfected with 50 nM miR-NC or miR-379 mimics; the cells were then treated with 1 μg/mL doxycycline for 3 days to restore COX-2 expression. (G) Cell viability was evaluated by counting cell numbers. (H) The migratory ability of cells was evaluated using a transwell migration assay. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments. Scale bar, 100 μm. n.s., not significant; **, p < 0.01; ***, p < 0.001.
Fig. 6
Fig. 6. miR-379 inhibits Cr-T cell viability and migration via directly targeting COX-2.
(A) miR-379 expression was measured in B2B and Cr-T cells using TaqMan real-time PCR assay. (B) Mice were treated with intranasal instillation of saline or Cr(VI) for 12 weeks. miR-379 level in the lung tissues of the mice was measured using the TaqMan assay. Ctrl: n=15, Cr(VI) instillation: n=20. (C) The correlation between COX-2 mRNA expression and miR-379 level in the mouse lung tissues was analyzed using Pearson correlation analysis. (D) A diagram shows the putative seed sequence between miR-379 and 3’-UTR of COX-2 and the wild-type (WT) and mutant (Mut) luciferase reporter constructs. (E) Luciferase reporter assay was performed using HEK293 cells to detect the relative luciferase activities of WT and Mut COX-2 reporters with or without miR-379 overexpression. The pRL-TK Renilla luciferase vector was used as an internal control. The miR-NC was used as a negative control for miR-379 transfection. (F) Cr-T cells were transfected with 50 nM non-targeting control oligos (miR-NC) or miR-379 mimics and incubated for 72 h. COX-2 protein levels were determined using an immunoblotting assay. (G-H) Cr-T cells stably expressing negative control or an inducible COX-2 were transfected with 50 nM miR-NC or miR-379 mimics; the cells were then treated with 1 μg/mL doxycycline for 3 days to restore COX-2 expression. (G) Cell viability was evaluated by counting cell numbers. (H) The migratory ability of cells was evaluated using a transwell migration assay. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments. Scale bar, 100 μm. n.s., not significant; **, p < 0.01; ***, p < 0.001.
Fig. 6
Fig. 6. miR-379 inhibits Cr-T cell viability and migration via directly targeting COX-2.
(A) miR-379 expression was measured in B2B and Cr-T cells using TaqMan real-time PCR assay. (B) Mice were treated with intranasal instillation of saline or Cr(VI) for 12 weeks. miR-379 level in the lung tissues of the mice was measured using the TaqMan assay. Ctrl: n=15, Cr(VI) instillation: n=20. (C) The correlation between COX-2 mRNA expression and miR-379 level in the mouse lung tissues was analyzed using Pearson correlation analysis. (D) A diagram shows the putative seed sequence between miR-379 and 3’-UTR of COX-2 and the wild-type (WT) and mutant (Mut) luciferase reporter constructs. (E) Luciferase reporter assay was performed using HEK293 cells to detect the relative luciferase activities of WT and Mut COX-2 reporters with or without miR-379 overexpression. The pRL-TK Renilla luciferase vector was used as an internal control. The miR-NC was used as a negative control for miR-379 transfection. (F) Cr-T cells were transfected with 50 nM non-targeting control oligos (miR-NC) or miR-379 mimics and incubated for 72 h. COX-2 protein levels were determined using an immunoblotting assay. (G-H) Cr-T cells stably expressing negative control or an inducible COX-2 were transfected with 50 nM miR-NC or miR-379 mimics; the cells were then treated with 1 μg/mL doxycycline for 3 days to restore COX-2 expression. (G) Cell viability was evaluated by counting cell numbers. (H) The migratory ability of cells was evaluated using a transwell migration assay. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments. Scale bar, 100 μm. n.s., not significant; **, p < 0.01; ***, p < 0.001.
Fig. 6
Fig. 6. miR-379 inhibits Cr-T cell viability and migration via directly targeting COX-2.
(A) miR-379 expression was measured in B2B and Cr-T cells using TaqMan real-time PCR assay. (B) Mice were treated with intranasal instillation of saline or Cr(VI) for 12 weeks. miR-379 level in the lung tissues of the mice was measured using the TaqMan assay. Ctrl: n=15, Cr(VI) instillation: n=20. (C) The correlation between COX-2 mRNA expression and miR-379 level in the mouse lung tissues was analyzed using Pearson correlation analysis. (D) A diagram shows the putative seed sequence between miR-379 and 3’-UTR of COX-2 and the wild-type (WT) and mutant (Mut) luciferase reporter constructs. (E) Luciferase reporter assay was performed using HEK293 cells to detect the relative luciferase activities of WT and Mut COX-2 reporters with or without miR-379 overexpression. The pRL-TK Renilla luciferase vector was used as an internal control. The miR-NC was used as a negative control for miR-379 transfection. (F) Cr-T cells were transfected with 50 nM non-targeting control oligos (miR-NC) or miR-379 mimics and incubated for 72 h. COX-2 protein levels were determined using an immunoblotting assay. (G-H) Cr-T cells stably expressing negative control or an inducible COX-2 were transfected with 50 nM miR-NC or miR-379 mimics; the cells were then treated with 1 μg/mL doxycycline for 3 days to restore COX-2 expression. (G) Cell viability was evaluated by counting cell numbers. (H) The migratory ability of cells was evaluated using a transwell migration assay. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments. Scale bar, 100 μm. n.s., not significant; **, p < 0.01; ***, p < 0.001.
Fig. 6
Fig. 6. miR-379 inhibits Cr-T cell viability and migration via directly targeting COX-2.
(A) miR-379 expression was measured in B2B and Cr-T cells using TaqMan real-time PCR assay. (B) Mice were treated with intranasal instillation of saline or Cr(VI) for 12 weeks. miR-379 level in the lung tissues of the mice was measured using the TaqMan assay. Ctrl: n=15, Cr(VI) instillation: n=20. (C) The correlation between COX-2 mRNA expression and miR-379 level in the mouse lung tissues was analyzed using Pearson correlation analysis. (D) A diagram shows the putative seed sequence between miR-379 and 3’-UTR of COX-2 and the wild-type (WT) and mutant (Mut) luciferase reporter constructs. (E) Luciferase reporter assay was performed using HEK293 cells to detect the relative luciferase activities of WT and Mut COX-2 reporters with or without miR-379 overexpression. The pRL-TK Renilla luciferase vector was used as an internal control. The miR-NC was used as a negative control for miR-379 transfection. (F) Cr-T cells were transfected with 50 nM non-targeting control oligos (miR-NC) or miR-379 mimics and incubated for 72 h. COX-2 protein levels were determined using an immunoblotting assay. (G-H) Cr-T cells stably expressing negative control or an inducible COX-2 were transfected with 50 nM miR-NC or miR-379 mimics; the cells were then treated with 1 μg/mL doxycycline for 3 days to restore COX-2 expression. (G) Cell viability was evaluated by counting cell numbers. (H) The migratory ability of cells was evaluated using a transwell migration assay. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments. Scale bar, 100 μm. n.s., not significant; **, p < 0.01; ***, p < 0.001.
Fig. 6
Fig. 6. miR-379 inhibits Cr-T cell viability and migration via directly targeting COX-2.
(A) miR-379 expression was measured in B2B and Cr-T cells using TaqMan real-time PCR assay. (B) Mice were treated with intranasal instillation of saline or Cr(VI) for 12 weeks. miR-379 level in the lung tissues of the mice was measured using the TaqMan assay. Ctrl: n=15, Cr(VI) instillation: n=20. (C) The correlation between COX-2 mRNA expression and miR-379 level in the mouse lung tissues was analyzed using Pearson correlation analysis. (D) A diagram shows the putative seed sequence between miR-379 and 3’-UTR of COX-2 and the wild-type (WT) and mutant (Mut) luciferase reporter constructs. (E) Luciferase reporter assay was performed using HEK293 cells to detect the relative luciferase activities of WT and Mut COX-2 reporters with or without miR-379 overexpression. The pRL-TK Renilla luciferase vector was used as an internal control. The miR-NC was used as a negative control for miR-379 transfection. (F) Cr-T cells were transfected with 50 nM non-targeting control oligos (miR-NC) or miR-379 mimics and incubated for 72 h. COX-2 protein levels were determined using an immunoblotting assay. (G-H) Cr-T cells stably expressing negative control or an inducible COX-2 were transfected with 50 nM miR-NC or miR-379 mimics; the cells were then treated with 1 μg/mL doxycycline for 3 days to restore COX-2 expression. (G) Cell viability was evaluated by counting cell numbers. (H) The migratory ability of cells was evaluated using a transwell migration assay. Data are presented as the mean ± SE of each group of animals or three independent cell line-based experiments. Scale bar, 100 μm. n.s., not significant; **, p < 0.01; ***, p < 0.001.

References

    1. Stoss F, et al. , Health assessment document for chronium. Research Triangle Park, NC: US Environmental Protection Agency, Office of Research and Development, 1983: p. 328.
    1. Shrivastava R, et al. , Effects of chromium on the immune system. FEMS Immunology & Medical Microbiology, 2002. 34(1): p. 1–7. - PubMed
    1. Gibb HJ, et al. , Extended followup of a cohort of chromium production workers. Am J Ind Med, 2015. 58(8): p. 905–13. - PMC - PubMed
    1. Luippold RS, et al. , Lung cancer mortality among chromate production workers. Occup Environ Med, 2003. 60(6): p. 451–7. - PMC - PubMed
    1. DesMarias TL and Costa M, Mechanisms of chromium-induced toxicity. Current Opinion in Toxicology, 2019. 14: p. 1–7. - PMC - PubMed

LinkOut - more resources