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
. 2020 Mar 31;21(7):2399.
doi: 10.3390/ijms21072399.

Programmed Cell Death-1: Programmed Cell Death-Ligand 1 Interaction Protects Human Cardiomyocytes Against T-Cell Mediated Inflammation and Apoptosis Response In Vitro

Affiliations

Programmed Cell Death-1: Programmed Cell Death-Ligand 1 Interaction Protects Human Cardiomyocytes Against T-Cell Mediated Inflammation and Apoptosis Response In Vitro

Woan Ting Tay et al. Int J Mol Sci. .

Abstract

Aim: Immunological checkpoint therapy is considered a powerful method for cancer therapy and acts by re-activating autologous T cells to kill the cancer cell. Myocarditis cases have been reported in cancer patients after immunological therapy; for example, nivolumab treatment is a monoclonal antibody that blocks programmed cell death-1/programmed cell death ligand-1 ligand interaction. This project provided insight into the inflammatory response as a benchmark to investigate the potential cardiotoxic effect of T cell response to the programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) axis in regulating cardiomyocyte injury in vitro.

Methods and results: We investigated cardiomyopathy resulted from the PD-1/PD-L1 axis blockade using the anti-PD-1 antibody in Rockefeller University embryonic stem cells-derived cardiomyocytes (RUES2-CMs) and a melanoma tumor-bearing murine model. We found that nivolumab alone did not induce inflammatory-related proteins, including PD-L1 expression, and did not induce apoptosis, which was contrary to doxorubicin, a cardiotoxic chemotherapy drug. However, nivolumab was able to exacerbate the immune response by increasing cytokine and inflammatory gene expression in RUES2-CMs when co-cultured with CD4+ T lymphocytes and induced apoptosis. This effect was not observed when RUES2-CMs were co-cultured with CD8+ T lymphocytes. The in vivo model showed that the heart function of tumor-bearing mice was decreased after treatment with anti-PD-1 antibody and demonstrated a dilated left ventricle histological examination. The dilated left ventricle was associated with an infiltration of CD4+ and CD8+ T lymphocytes into the myocardium. PD-L1 and inflammatory-associated gene expression were significantly increased in anti-PD-1-treated tumor-bearing mice. Cleaved caspase-3 and mouse plasma cardiac troponin I expressions were increased significantly.

Conclusion: PD-L1 expression on cardiomyocytes suppressed T-cell function. Blockade of PD-1 by nivolumab enhanced cardiomyocyte inflammation and apoptosis through the enhancement of T-cell response towards cardiomyocytes.

Keywords: PD-1; PD-L1; T-lymphocytes; human embryonic stem cell-derived cardiomyocytes; immune-related cardiotoxicity; nivolumab.

PubMed Disclaimer

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Establishment and validation of RUES2 and differentiation of cardiomyocytes in vitro. (A) Schematic of the RUES2-CMs differentiation protocol. (B) Flow cytometric analysis of pluripotent stem cell markers expression: Oct-4, SSEA-4, Nanog, and a positively selected cardiac marker (cTnT). (C) Representative fluorescent images of pluripotent stem cell markers Oct-4 and cardiac markers: α-actinin and cTnT. Scale bar: 50 μm (upper panel); 20 μm (lower panel). (D) PD-L1 expression on RUES2-CMs by flow cytometry and (E) immunofluorescence staining, scale bar: 25 μm. The quantitative result as the right panel. *** p < 0.001 versus control (n = 3), one-way ANOVA, posthoc Bonferroni test. RUES2, Rockefeller University embryonic stem cell line 2; cTnT, cardiac troponin T; Oct4, octamer-binding transcription factor 4; PD-L1, programmed death-ligand 1; RUES2-CMs, Rockefeller University embryonic stem cell line 2-cardiomyocytes.
Figure 2
Figure 2
Effects of nivolumab on cell viability in RUES2-CMs. (A) The effect of nivolumab on cell viability was determined by an MTT assay. Positive control (doxorubicin 5 μg/mL) was provided. *** p < 0.001 versus control (n = 3). (B) Representative images of the TUNEL staining of cardiomyocytes. TUNEL staining was used to detect cell apoptosis (green). The nuclei were counterstained with DAPI (blue). Cyan color represents TUNEL-positive nuclei on merged photos. Positive control (doxorubicin 5 μg/mL) was provided. Scale bar: 50 μm. (C) Representative flow cytometry images of the Annexin V. Positive control (doxorubicin 5 μg/mL) was provided. *** p < 0.001 versus control (n = 3), one-way ANOVA, posthoc Bonferroni test. (D) Representative western blot analysis of caspase3 and inflammation markers—STAT1, NFkB—and the quantitative result (E). Positive control (doxorubicin 5 μg/mL) was provided. *** p < 0.001 versus control (n = 3), one-way ANOVA, posthoc Bonferroni test. Data are shown as the mean ± SD. RUES2-CM, Rockefeller University embryonic stem cell line 2- cardiomyocytes. (F) The PD-L1 expression level in RUES2-CMs analyzed by flow cytometry images. The quantitative result is shown in the right panel. *** p < 0.001 versus control (n = 3), one-way ANOVA, posthoc Bonferroni test.
Figure 3
Figure 3
Validation of PD-1 expression on CD4+ and CD8+ T lymphocytes. (A) Representative flow cytometry results of PD-1 and activation marker CD25 expression on CD4+ and CD8+ T lymphocytes after anti-CD3/CD28 stimulation. PD-1, programmed death-1. (B) Representative micrographs showing immunostained PD-1+ cells (green) on the CD4+ and CD8+ T lymphocytes. Scale bar: 50 μm. (C) Quantification of cTnT+/PD-L1 expression by flow cytometry analysis of levels of cTnT+/PD-L1 expression on RUES2-CMs co-cultured with CD4+ and CD8+ T lymphocytes with nivolumab treatment at different doses. ** p < 0.01, *** p < 0.001 versus RUES2-CMs alone group; # p < 0.05, ### p < 0.001 versus CD4/RUES2-CMs human IgG4 isotype control (w/o co-culture) group (n = 3). (D) Representative western blot analysis on RUES2-CMs co-cultured with CD4+ and CD8+ T lymphocytes containing nivolumab treatment with different doses and quantitative results (EG). * p < 0.05, ** p < 0.01, *** p < 0.001 versus RUES2-CMs alone group; # p < 0.05, ## p < 0.01, ### p < 0.001 versus CD4/RUES2-CMs human IgG4 isotype control (w/o co-culture) group (n=3; & p < 0.05 versus CD8/RUES2-CMs human IgG4 isotype control (w/o co-culture) group; & p < 0.05, versus CD8/RUES2-CMs human IgG4 isotype control (w/o co-culture) group (n = 3), one-way ANOVA, posthoc Bonferroni test. (H) Quantitative result of ELISA assay, showing production of IFN-γ in CD4+ T lymphocytes/RUES2-CMs and CD8+ T lymphocytes/RUES2-CMs co-culture medium. ** p < 0.01, *** p < 0.001 versus CD4+ T lymphocytes alone group; # p < 0.05, ### p < 0.001 versus CD4/RUES2-CMs human IgG4 isotype control (w/o co-culture) group (n = 3).
Figure 4
Figure 4
Effects of anti-PD-1 immunotherapy on tumor growth in tumor-bearing mice. (A) Representative micrographs showing immunostained PD-L1+ cells (green) on the B16-F10 cells. Scale bar: 50 μm. Representative immunohistochemistry staining for IgG (B,C) and PD-L1 (D,E) on mouse heart. The inset box, showing the enlarged pictures for the indicated square area. Scale bars in (B) and (D), 100 μm; scale bars in (C) and (E), 50 μm. Red arrow: PD-L1+ cell. (F) Representative PCR analysis of PD-L1 in B16-F10 cells and mouse heart. PD-L1, programmed death-ligand 1. (G) Representative photograph of xenograft tumors at the end of the experiment. (H) Tumor growth curves for each experiment group. ** p < 0.01, *** p < 0.001 versus rat IgG group (n = 7), two-way ANOVA, posthoc Bonferroni test. Effects of anti-PD-1 immunotherapy on mouse heart function. The quantitative data of fractional shortening (I), and ejection fraction (J) of rat IgG and anti-PD-1-treated group. # p < 0.05 versus rat IgG-treated tumor group (n = 7); # p < 0.05 versus anti-PD-1-treated non-tumor group (n = 7). (K) Representative micrographs showing immunostained CD4+ and CD8+ cells (green) in the myocardium. Scale bar: 1000 μm. (L) Representative western blot analysis and quantitative results (M). *** p < 0.001 versus anti-PD-1-treated non-tumor group; ### p < 0.001 versus rat IgG-treated tumor group (n = 4), two-way ANOVA, posthoc Bonferroni test. Data are shown as the mean ± SD.
Figure 5
Figure 5
Effects of anti-PD-1 immunotherapy on the expression of PD-L1 and inflammation-related markers in tumor-bearing mice. (A) Representative western blot analysis and quantitative results (B). && p < 0.01, ### p < 0.001 versus rat IgG-treated tumor group; *** p < 0.001 versus anti-PD-1-treated non-tumor group; && p < 0.01 versus rat IgG-treated non-tumor group (n = 4), two-way ANOVA, posthoc Bonferroni test. (C) A representative of RT-PCR analysis of IFN-γ and TNF-α in the anti-PD-1-treated non-tumor group and anti-PD-1-treated tumor group. *** p < 0.001 versus anti-PD-1-treated non-tumor group (n = 4), unpaired Student’s t-test. Data are shown as the mean ± SD. (D) Representative western blot analysis of apoptosis markers and quantitative results (E). ## p < 0.01, ### p < 0.001 versus rat IgG-treated tumor group; *** p < 0.001 versus anti-PD-1-treated non-tumor group (n = 4), two-way ANOVA, posthoc Bonferroni test.

References

    1. Nixon N.A., Blais N., Ernst S., Kollmannsberger C., Bebb G., Butler M., Smylie M., Verma S. Current landscape of immunotherapy in the treatment of solid tumours, with future opportunities and challenges. Curr. Oncol. 2018;25:e373–e384. doi: 10.3747/co.25.3840. - DOI - PMC - PubMed
    1. Buchbinder E.I., Desai A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am. J. Clin. Oncol. 2016;39:98–106. doi: 10.1097/COC.0000000000000239. - DOI - PMC - PubMed
    1. Grywalska E., Pasiarski M., Gozdz S., Rolinski J. Immune-checkpoint inhibitors for combating T-cell dysfunction in cancer. Onco Targets Ther. 2018;11:6505–6524. doi: 10.2147/OTT.S150817. - DOI - PMC - PubMed
    1. Tajiri K., Ieda M. Cardiac Complications in Immune Checkpoint Inhibition Therapy. Front. Cardiovasc. Med. 2019;6:3. doi: 10.3389/fcvm.2019.00003. - DOI - PMC - PubMed
    1. Raschi E., Diemberger I., Poluzzi E., De Ponti F. Reporting of immune checkpoint inhibitor-associated myocarditis. Lancet. 2018;392:383. doi: 10.1016/S0140-6736(18)31549-6. - DOI - PubMed