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
. 2024 Jun 24;14(1):14545.
doi: 10.1038/s41598-024-65447-w.

Effect of apigetrin in pseudo-SARS-CoV-2-induced inflammatory and pulmonary fibrosis in vitro model

Affiliations

Effect of apigetrin in pseudo-SARS-CoV-2-induced inflammatory and pulmonary fibrosis in vitro model

Hengmin Han et al. Sci Rep. .

Abstract

SARS-CoV-2 has become a global public health problem. Acute respiratory distress syndrome (ARDS) is the leading cause of death due to the SARS-CoV-2 infection. Pulmonary fibrosis (PF) is a severe and frequently reported COVID-19 sequela. In this study, an in vitro model of ARDS and PF caused by SARS-CoV-2 was established in MH-S, THP-1, and MRC-5 cells using pseudo-SARS-CoV-2 (PSCV). Expression of proinflammatory cytokines (IL-6, IL-1β, and TNF-α) and HIF-1α was increased in PSCV-infected MH-S and THP-1 cells, ARDS model, consistent with other profiling data in SARS-CoV-2-infected patients have been reported. Hypoxia-inducible factor-1 alpha (HIF-1α) siRNA and cobalt chloride were tested using this in vitro model. HIF-1α knockdown reduces inflammation caused by PSCV infection in MH-S and THP-1 cells and lowers elevated levels of CTGF, COLA1, and α-SMA in MRC-5 cells exposed to CPMSCV. Furthermore, apigetrin, a glycoside bioactive dietary flavonoid derived from several plants, including Crataegus pinnatifida, which is reported to be a HIF-1α inhibitor, was tested in this in vitro model. Apigetrin significantly reduced the increased inflammatory cytokine (IL-6, IL-1β, and TNF-α) expression and secretion by PSCV in MH-S and THP-1 cells. Apigetrin inhibited the binding of the SARS-CoV-2 spike protein RBD to the ACE2 protein. An in vitro model of PF induced by SARS-CoV-2 was produced using a conditioned medium of THP-1 and MH-S cells that were PSCV-infected (CMPSCV) into MRC-5 cells. In a PF model, CMPSCV treatment of THP-1 and MH-S cells increased cell growth, migration, and collagen synthesis in MRC-5 cells. In contrast, apigetrin suppressed the increase in cell growth, migration, and collagen synthesis induced by CMPSCV in THP-1 and MH-S MRC-5 cells. Also, compared to control, fibrosis-related proteins (CTGF, COLA1, α-SMA, and HIF-1α) levels were over two-fold higher in CMPSV-treated MRC-5 cells. Apigetrin decreased protein levels in CMPSCV-treated MRC-5 cells. Thus, our data suggest that hypoxia-inducible factor-1 alpha (HIF-1α) might be a novel target for SARS-CoV-2 sequela therapies and apigetrin, representative of HIF-1alpha inhibitor, exerts anti-inflammatory and PF effects in PSCV-treated MH-S, THP-1, and CMPVSC-treated MRC-5 cells. These findings indicate that HIF-1α inhibition and apigetrin would have a potential value in controlling SARS-CoV-2-related diseases.

Keywords: Acute respiratory distress syndrome (ARDS); Apigetrin; Hypoxia-inducible factor-1α (HIF-1α); Pulmonary fibrosis (PF); SARS-CoV-2.

PubMed Disclaimer

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Evaluation of in vitro ARDS model. MH-S (A) and THP-1 cells (B) were infected with PSCV for 24 h. Cell lysates were prepared and subjected to western blotting to analyze the expression of Spike1, HIF-1α, IL-6, IL-1β, TNF-α, and β-actin. The results are presented as the mean ± SD of three independent experiments. **p < 0.01, and ***p < 0.001 (compared to normal control).
Figure 2
Figure 2
Evaluation of in vitro PF model. MRC-5 cells were treated with CMPSCV from MH-S (A) or THP-1 (B) cells for 24 h. Cell lysates were prepared and subjected to western blotting to analyze the CTGF, COLA1, α-SMA, HIF-1α, and β-actin expressions. The results are presented as the mean ± SD of three independent experiments. **p < 0.01, and ***p < 0.001 (compared to normal control).
Figure 3
Figure 3
Effect of HIF-1α siRNA and CoCl2 in PSCV-infected MH-S or THP-1 cells and CPMSCV-exposure MRC-5 cells. MH-S (A) and THP-1 cells (B) were transfected with HIF-1α siRNA for 48 h and were incubated in the presence or absence of PSCV for 24 h. PSCV-infected MH-S (C) and THP-1 cells (D) were treated with CoCl2 for 24 h. Cell lysates were prepared and subjected to western blotting to analyze the Spike1, HIF-1α, IL-6, IL-1β, TNF-α, and β-actin expressions. The results are presented as the mean ± SD of three independent experiments. ##p < 0.01, ###p < 0.001 (compared to normal control), **p < 0.01, and ***p < 0.001 (compared to PSCV-infected control). MRC-5 cells were transfected with HIF-1α siRNA for 48 h and were incubated in the presence or absence of PSCV-treated MH-S (E) or THP-1 cells (F) CMPSCV for 24 h. Cell lysates were prepared and subjected to western blotting to analyze the CTGF, COLA1, α-SMA, HIF-1α, and β-actin expressions. The results are presented as the mean ± SD of three independent experiments. ##p < 0.01, ###p < 0.001 (compared to normal control), **p < 0.01, and ***p < 0.001 (compared to CMPSCV-infected control).
Figure 4
Figure 4
Evaluation of apigetrin on spike protein expression and binding to S protein RBD in PSCV-infected MH-S and THP-1 cells. (A) The SARS-CoV-2 inhibitor screening kit was used to test the ability of apigetrin to bind to SARS-CoV-2 S protein RBD. The results are presented as the mean ± SD of three independent experiments. ***p < 0.001 (compared to normal control). MH-S (B) and THP-1 cells (C) were treated with PSCV and apigetrin for 24 h. The cells were fixed in 100% methanol, exposed to the diluted Spike 1 antibody, and incubated with the goat anti-rabbit IgG. The reacted cells were incubated with VECTASHIELD antifade mounting medium with DAPI, and the image was captured using a fluorescence microscope. a–c means in a row by different superscripts are significantly different by LSD (least significant difference).
Figure 5
Figure 5
Anti-inflammatory effect of apigetrin in PSCV-infected MH-S and THP-1 cells. MH-S (A) and THP-1 cells (B) were infected with PSCV and apigetrin for 24 h. Cell lysates were prepared and subjected to western blotting to analyze the Spike1, HIF-1α, IL-6, IL-1β, TNF-α, and β-actin expressions. IL-6, IL-1β, and TNF-α concentrations in the serum samples of MH-S (CE), and THP-1 (FH) cells were determined by ELISA. The results are presented as the mean ± SD of three independent experiments. ##p < 0.01, ###p < 0.001 (compared to normal control), **p < 0.01, and ***p < 0.001 (compared to PSCV-infected control).
Figure 6
Figure 6
Effect of apigetrin on pulmonary fibrosis in CMPSCV-treated MRC-5 cell model. MRC-5 cells were treated with apigetrin in MH-S (A,B) or THP-1 cells (D,E) CMPSCV for 24 h. CELLOMAX kit was used to measure cell proliferation. a–c means in a row by different superscripts are significantly different by LSD (least significant difference). After treatment the cells were resolved in 3.5% formaldehyde, permeabilized by methanol, and stained by crystal violet. Picro-Sirius Red Stain Kit was used to stain collagen fibers and the image was captured using a light microscope (B,E). a–d means in a row by different superscripts are significantly different by LSD (least significant difference). MRC-5 cells were treated with apigetrin in MH-S (C) or THP-1 cells (F) CMPSCV for 24 h. Cell lysates were prepared and subjected to western blotting to analyze the CTGF, COLA1, α-SMA, HIF-1α, and β-actin expressions. The results are presented as the mean ± SD of three independent experiments at p < 0.05. ##p < 0.01, ###p < 0.001 (compared to normal control), **p < 0.01, and ***p < 0.001 (compared to CMPSCV-infected control).

Similar articles

References

    1. World Health Organization. Coronavirus disease (COVID-19). https://www.who.int/health-topics/coronavirus#tab=tab_1 (2023).
    1. Ye Q, Wang B, Mao J. Cytokine storm in COVID-19 and treatment. J. Infect. 2020;80(6):607–613. doi: 10.1016/j.jinf.2020.03.037. - DOI - PMC - PubMed
    1. Grasselli G, et al. Pathophysiology of COVID-19-associated acute respiratory distress syndrome—Authors' reply. Lancet Respir. Med. 2021;9(1):e5–e6. doi: 10.1016/S2213-2600(20)30525-7. - DOI - PMC - PubMed
    1. Fara A, et al. Cytokine storm and COVID-19: A chronicle of pro-inflammatory cytokines. Open Biol. 2020;10(9):200160. doi: 10.1098/rsob.200160. - DOI - PMC - PubMed
    1. Ukwenya VO, et al. The impact of deranged glucose metabolism and diabetes in the pathogenesis and prognosis of the novel SARS-CoV-2: A systematic review of literature. Curr. Diabetes Rev. 2022;18(3):101–112. doi: 10.2174/1573399817666210806104349. - DOI - PubMed

MeSH terms

Substances