Skip to main content

Potential role of PIM1 inhibition in the treatment of SARS-CoV-2 infection

Abstract

Background

SARS-CoV-2 infection involves disturbing multiple molecular pathways related to immunity and cellular functions. PIM1 is a serine/threonine-protein kinase found to be involved in the pathogenesis of several viral infections. One PIM1 substrate, Myc, was reported to interact with TMPRSS2, which is crucial for SARS-CoV-2 cell entry. PIM1 inhibitors were reported to have antiviral activity through multiple mechanisms related to immunity and proliferation. This study aimed to evaluate the antiviral activity of 2-pyridone PIM1 inhibitor against SARS-CoV-2 and its potential role in hindering the progression of COVID-19. It also aimed to assess PIM1 inhibitor’s effect on the expression of several genes of Notch signaling and Wnt pathways. In vitro study was conducted on Vero-E6 cells infected by SARS-CoV-2 “NRC-03-nhCoV” virus. Protein–protein interaction of the study genes was assessed to evaluate their relation to cell proliferation and immunity. The effect of 2-pyridone PIM1 inhibitor treatment on viral load and mRNA expression of target genes was assessed at three time points.

Results

Treatment with 2-pyridone PIM1 inhibitor showed potential antiviral activity against SARS-CoV-2 (IC50 of 37.255 µg/ml), significantly lowering the viral load. Functional enrichments of the studied genes include negative regulation of growth rate, several biological processes involved in cell proliferation, and Interleukin-4 production, with interleukin-6 as a predicted functional partner. These results suggest an interplay between study genes with relation to cell proliferation and immunity. Following in vitro SARS-CoV-2 infection, Notch pathway genes, CTNNB1, SUMO1, and TDG, were found to be overexpressed compared to uninfected cells. Treatment with 2-pyridone PIM1 inhibitor significantly lowers the expression levels of study genes, restoring Notch1 and BCL9 to the control level while decreasing Notch2 and CTNNB1 below control levels.

Conclusion

2-pyridone PIM1 inhibitor could hinder cellular entry of SARS-CoV-2 and modulate several pathways implicated in immunity, suggesting a potential benefit in the development of anti-SARS-CoV-2 therapeutic approach.

Background

Coronavirus disease 2019 (COVID-19) is an emerging disease with significant morbidities and mortalities, caused by SARS-CoV-2 virus [1]. COVID-19 patients showed acute and long-term complications that could progress to serious illness especially in aged and immunocompromised patients [2].

SARS-CoV-2 entry routes were found to be through interaction of its spike protein (S-protein) with angiotensin-converting enzyme 2 (ACE2) receptors on cell membrane/transmembrane protease, Serine 2 (TMPRSS2). TMPRSS2 is responsible for proteolytic cleavage and activation of the S-protein of SARS-CoV-2, leading to fusion of the viral particles with cell membrane and viral entry [3]. Endosomal pathway is another route of SARS-CoV-2 entry via Cathepsin B, which is an endosomal cysteine protease. Cathepsin B releases the virus in cytoplasm through a mechanism similar to that of TMPRSS2 [3], although it works independently from TMPRSS2 [4]. Drugs targeting these routs could hinder viral entry and decrease viral load in infected cells.

Serine/threonine-protein kinase PIM-1 (PIM1) is an active serine/threonine kinase which plays a crucial role in cell survival via hindering apoptosis, among several biological processes [5]. PIM1 was also reported to be involved in host response to many viral infections, such as hepatitis B, Epstein-Barr, and human papilloma viral infections. Recently, it has been found that EV-A71 infection elevated both the mRNA and protein levels of PIM1. The elevated PIM promoted EV-A71 replication through enhancing IRES activity and blocking AUF1 cytosol translocation counteracting its antiviral property [6]. In addition, PIM1 was reported to promote Zika virus replication by suppressing host cells’ natural immunity through downregulating phosphorylation of both STAT1 and STAT2 which are essential for cellular antiviral response [7]. Hence, targeting PIM1 using PIM1 inhibitors could be effective in combating virus infection. The implication of PIM1 in immunosuppression during chronic viral infections was also reported. PIM1 was found to be highly expressed in suppressive neutrophils and had a crucial role in maintaining their survival and function. Thus, PIM1 inhibition mitigated suppressive neutrophils-mediated immunosuppression, with consequent increased CD8 T-cell function and improved viral control [8].

Previous studies had highlighted the interplay between PIM1 and Myc, which is a transcription factor known to be a PIM1 substrate [9]. Several studies had pointed out some interaction between Myc and TMPRSS2 [10, 11]. However, on a mechanistic level, the interaction of PIM1 and its substrate, Myc, with TMPRSS2 is not clear yet.

Notch signaling has been reported to facilitate the infectivity of many viruses including Epstein-Barr virus, the human papillomavirus (HPV), hepatitis B virus (HBV), and hepatitis C virus (HCV). In SARS-CoV-2, it was found that Notch1 can indirectly enhance viral entry through inducing FURIN expression, the protease responsible for exposing the fusion sequences of the viral S-protein, facilitating viral particle fusion with cell membrane [12]. Furthermore, the implication of the Notch pathway and downstream genes in many COVID-19 events as cytokines storm, hypoxic response, and coagulopathic response has been reported [13].

The interaction between PIM kinases and Notch signaling pathways has been studied. It was reported that PIM kinases induced Notch1 activity via phosphorylation, promoting its signaling along with other signaling pathways such as the Wnt pathway [14]. PIM1 inhibitors could modulate molecular functions of PIM1. One of them is a 2-pyridone compound (6-amino-4-(3,4-dimethoxyphenyl)-1-(2-ethylphenyl)-2-oxo-1,2-dihydropyridine-3,5-dicarbonitrile), named 2-pyridone PIM1 inhibitor hereafter, had shown significant PIM1 inhibition with minor toxicity risk in vitro [15].

This study aimed to evaluate the antiviral activity of 2-pyridone PIM1 inhibitor against SARS-CoV-2 and its potential role in hindering the progression of COVID-19. It also aims to assess PIM1 inhibitor’s effect on Notch signaling pathway, Wnt pathway, and some key genes involved in SARS-CoV-2 pathogenesis.

Methods

2-Pyridone PIM1 inhibitor synthesis and preparation

It is described previously [15].

Cytotoxicity and antiviral activity

Vero-E6 cells were sustained in DMEM (Dulbecco’s Modified Eagle’s Medium) containing 1% penicillin/streptomycin mixture and 10% fetal bovine serum (FBS) in a humidified incubator at 37 °C and 5% CO2. SARS-CoV-2 (NRC-03-nhCoV) virus [16] was propagated and titrated as described previously [17].

Cytotoxicity (CC 50 ) assay

Half-maximal cytotoxic concentration (CC50) of 2-pyridone PIM1 inhibitor was assessed in VERO-E6 cells using crystal violet as described previously [18]. Briefly, cells were seeded in 96-well plates as 100 µL/well with a density of 3 × 105 cells/ml. Plates were incubated for 24 h in a 5% CO2 humidified incubator at 37 °C. After the incubation period, cells were treated with various concentrations of the compound in triplicates. After 72 h, the supernatant was discarded, and cell monolayers were fixed (using 10% formaldehyde for 1 h at room temperature), dried, and stained with crystal violet on a bench rocker (50 µl of 0.1% solution, for 20 min at room temperature). After washing and drying the stained cell monolayers, methanol (200 µl/well for 20 min) was used to dissolve the crystal violet on a bench rocker at room temperature. Crystal violet solution absorbance was measured at λmax 570 nm using a plate reader. The CC50 of 2-pyridone PIM1 inhibitor was calculated as the concentration required to induce cytotoxicity by 50% in the treated cells relative to the virus control.

Inhibitory concentration 50 (IC50) determination

The IC50 value for 2-pyridone PIM1 inhibitor was determined as described previously [17], with slight modifications. Briefly, VERO-E6 cells were seeded in tissue culture plates (2.4 × 104/well) and incubated overnight in a humidified incubator at 37 °C, 5% CO2. The cell monolayers were then washed using 1 × PBS. An aliquot of the SARS-CoV-2 virus containing 100 TCID50 was incubated with serial dilutions of 2-pyridone PIM1 inhibitor and kept at 37 °C for 1 h. The VERO-E6 cells were treated with the virus alone or virus/2-pyridone PIM1 inhibitor and incubated at 37 °C in a total volume of 200 µl per well along with uninfected untreated cells as a control and untreated infected cells as a virus control. After incubation in a 5% CO2 incubator for 72 h at 37 °C, cells were fixed using 10% paraformaldehyde for 20 min and stained with 0.5% crystal violet in dH2O for 15 min at room temperature. Absolute methanol (100 μl/well) was used to dissolve crystal violet. Crystal violet solution absorbance was measured at λmax 570 nm using a plate reader. The IC50 of the compound is the concentration required to reduce the virus-induced cytopathic effect (CPE) by 50%, relative to the virus control.

Protein–protein interaction prediction

GeneMANIA prediction tool [19] and STRING [20] were used to predict the interaction between the study genes and to evaluate the protein–protein interaction (PPI) enrichment. VirusMINT (http://mint.bio.uniroma2.it/virusmint/RRID:SCR_005987) was used for multivariate interaction between host and viral proteins. Gene list input in STRING and VirusMINT was PIM1, Notch1, Notch2, CTNNB1, CDC42, PSEN1, BCL9, ACTR2, TDG, SUMO1, CDC34, and IL-6. p-value < 0.05 and false discovery rate (FDR) < 0.05 were considered significant.

Gene expression assay

Cell treatment

Five-hundred TCID50 of SARS-CoV-2 were used to infect VERO E6 cells. Cells with and without treatment with 2-pyridone PIM1 inhibitor were collected in 1 × PBS pellets by centrifugation at time intervals of 6, 12, and 24 h post-infection. Control cells are uninfected untreated cells.

qPCR

Total RNA was extracted from treated and untreated cells (at three time points: 6, 12, and 24 h post-infection) using RNeasy Mini Kit, following the manufacturer’s instructions. The extracted mRNA was reverse-transcribed into cDNA using RevertAid First-Strand cDNA Synthesis Kit (Thermo Scientific USA). Primers were designed for relative quantification of the study genes (Notch1, 2, CTNNB1, PSEN1, CDC42, BCL9, ACTR2, SUMO1, and TDG), using Primer3 software (https://primer3.ut.ee/, RRID:SCR_003139). SYBR Green qPCR Master Mix (Thermo Scientific, USA) was used to prepare the reaction mixture with GAPDH as a housekeeping gene. Technical replicates were prepared for the three time point samples (6, 12, and 24 h post-infection). Results were expressed using the 2−ΔΔCt method. Primers and annealing temperature Ta for each are listed in Table 1. qPCR cycling conditions were 95 °C for 10 min followed by 40 cycles (95 °C for 15 s, Ta for 30 s, and 72 °C for 40 s).

Statistical analysis

Two-tailed independent Student’s t-test for parametric data was used to assess the significant differences between the values of the control and treatment groups where p-value < 0.05 was considered significant. Experiments and assays were performed in biological and technical replicates.

Results

Antiviral activity

The CC50 of 2-pyridone PIM1 inhibitor was 1734 µg/ml that showed a potential antiviral activity against SARS-CoV-2 with an IC50 of 37.255 µg/ml (Fig. 1). SARS-CoV-2 viral load in VERO-E6 cells treated with 2-pyridone PIM1 inhibitor was found to be considerably low compared to non-treated SARS-CoV-2-infected VERO-E6 cells.

Fig. 1
figure 1

Cytotoxicity and in vitro anti-SARS-CoV-2 activity of A and B, respectively. A Cytotoxicity of 2-pyridone in Vero E6 cells as measured by MTT assay. B Dose-inhibition curves against SARS-CoV-2 “NRC-03-nhCoV” virus in Vero E6 cells. Cytotoxic concentration 50 (CC50) and inhibitory concentration 50 (IC50) values were calculated using nonlinear regression analysis of GraphPad Prism software (version 5.01) by plotting log inhibitor versus normalized response (variable slope)

Protein–protein interaction prediction

Types of interactions between studied genes are displayed in Fig. 2A, showing physical interaction (47.15%) between the study genes and several genes involved in immunity including IL-6 and multiple pathway interactions. PIM1 was shown to have a significant genetic interaction with BCL9 (p-value: 0.0004), ACTR2 (p-value: 0.0007), and CDC42 effector protein (p-value: 0.0006) along with co-expression with PSEN1 (p-value: 0.0153) and TDG (p-value: 0.016) (supplementary file 1).

Fig. 2
figure 2

Protein–protein interaction and functional prediction. A Protein–protein interaction network of PIM-1 with the study genes, showing prominent physical interaction (dark pink lines), pathways (light blue lines), and co-expression (violet lines). B Protein functional enrichment network of the study genes. C Significant interactions of the study genes with viral proteins, assessed through VirusMINT database

Functional enrichments of the studied genes (network PPI enrichment p-value: 0.00134, Fig. 2B) include the following:

  1. 1.

    Negative regulation of growth rate (GO: 0,045,967/FDR 0.00091)

  2. 2.

    Several biological processes involved in cardioblast proliferation and heart development (GO: 0,003,264/FDR 0.00016, GO: 0,003,266/FDR0.0027, GO: 0,003,161/FDR 0.0041, GO: 0,061,311/FDR 0.00043, and GO: 0,061,314/FDR 0.0009)

  3. 3.

    Positive regulation of neuroblast, astrocytes, and glial cells proliferation (GO: 0,002,052/FDR 0.0084, GO: 0,048,710/FDR 0.0104, GO: 0,060,251/FDR0.0119)

  4. 4.

    Interleukin-4 production (GO:0,032,633/FDR 0.0027)

Protein network of the study genes showed that the predicted functional partners are interleukin-6; (IL-6) and ubiquitin-conjugating enzyme E2 R1 (CDC34) (Fig. 2B, Supplementary file 2).

Multivariate interactions prediction of the study proteins with viral proteins showed significant interaction between the studied protein list and SARS-CoV proteins (Fig. 2C).

Effect of SARS-Cov-2 and 2-pyridone PIM1 inhibitor on some Notch signaling and Notch downstream genes

SARS-CoV-2 infection resulted in a significant overexpression of Notch1, Notch2, CTNNB1, CDC42, and PSEN1 after 24 h of infection. No significant change in expression levels of all genes was observed at time points 6 and 12 h post-infection, compared to control. Treatment with 2-pyridone PIM1 inhibitor was able to significantly downregulate Notch1, Notch2, CTNNB1, and CDC42 in SARS-CoV-2-infected cells at 24 h post-infection (p-value < 0.001). The expression of Notch1 was normalized, while the expression levels of Notch2 and CTNNB1 were below those of the control cells. The expression levels of CDC42, although significantly decreased, remained above those of the control cells. On the other hand, the change in PSEN1 expression levels was insignificant after 2-pyridone PIM1 inhibitor treatment compared to untreated infected cells (Fig. 3).

Fig. 3
figure 3

Expression of Notch signaling and downstream genes after 24 h of SARS-CoV-2 infection in vitro. Expression levels are shown relative to control, with and without 2-pyridone PIM1 inhibitor treatment

The expression levels of BCL9 and ACTR2, which are Notch-signaling downstream genes, were assessed. SARS-CoV-2 infection caused overexpression of BCL9 and underexpression of ACTR2 after 24 h of infection of the cells. 2-pyridone PIM1 inhibitor-treated cells showed significantly lower levels of both genes (p-value < 0.0001). BCL9 levels were restored to levels close to those observed in control cells, while ACTR2 levels were lower in treated cells compared to both control cells and infected untreated cells (Fig. 3).

Effect of SARS-CoV-2 and 2-pyridone PIM1 inhibitor on TDG and SUMO 1

SARS-CoV-2 infection resulted in a significant overexpression of SUMO1 and TDG after 24 h but insignificant compared to control cells at 6 h post-infection. At the 12-h time point post-infection, TDG expression showed significant overexpression compared to control cells, while SUMO1 remained unchanged.

Treatment of SARS-CoV-2-infected cells with 2-pyridone PIM1 inhibitor caused significant downregulation of TDG and SUMO1 expressions below expression levels in control cells (p-value < 0.0001) compared to untreated SARS-CoV-2-infected cells (Fig. 4).

Fig. 4
figure 4

Expression of TDG and SUMO1 after 24 h of SARS-CoV-2 infection in vitro. Expression levels are shown relative to control, with and without 2-pyridone PIM1 inhibitor treatment

Discussion

The development of specific antiviral agents for SARS-CoV-2 is still a major concern of many studies. Immunomodulation, protease inhibition, and targeting the viral RNA-dependent RNA polymerase (RdRp) are potential targets for anti-coronavirus agents [21]. PPI functional enrichment prediction of the present study genes showed significant enrichment of genes related to immunity, such as interleukin-4 production, with IL-6 and CDC34 as predicted functional partners. Notch1 was previously reported to positively regulate IL-6 production in macrophages, which in turn amplifies the Notch signaling leading to further production of IL-6 — through positive feedback mechanism. Also, Notch signaling promotes the production of inflammatory Th1/Th17 cytokines in T-helper cells [12].

PPI functional enrichment prediction suggested a role of the study genes in growth and proliferation of different types of cells including cardiac and CNS cells. However, the role of these genes in cardiac and neuronal complications of COVID-19 and long COVID needs further investigations. Host–pathogen interaction analysis by VirusMINT database showed a significant interaction between coronaviruses and the current study genes. These results suggest that the interplay between the study genes and SARS-CoV-2 might help in evaluating new treatment approaches Table 1.

Table 1 Primer sequences of target genes

PIM1 kinase was reported to be highly expressed in the bronchial epithelium. Inhibition of PIM1 kinase was shown to reduce viral replication and viral particles release in cultured PBECs through induction of apoptosis during viral infection [5]. The present study provided evidence of the role of PIM1 inhibition in lowering the viral load. SARS-CoV-2- infected cells treated with 2-pyridone PIM1 inhibitor showed lower viral load compared to untreated infected cells, possibly through the effect of PIM1 inhibition on TMPRSS2 through the PIM1 kinase substrate, Myc. However, understanding the mechanism of this effect needs further investigation.

The initial viral load is not necessarily associated with the severity of symptoms or duration of disease [22]. Hence, hindering viral entry is not sufficient to prevent COVID-19 progression. Studying the effect of 2-pyridone PIM1 inhibitor on pathways implicated in immunity could provide a better idea about its therapeutic effect and mechanism of action.

To assess the effect of PIM1 inhibition on the molecular pathogenesis of SARS-CoV-2, expression of some Notch pathway genes, Wnt/β-catenin signaling pathway, TDG, and SUMOylation, was studied in the current study.

The significant overexpression of Notch1 induced by SARS-CoV-2 infection was normalized in cells treated with 2-pyridone PIM1 inhibitor. For Notch2, the significant overexpression induced by SARS-CoV-2 infection was reduced in cells with 2-pyridone PIM1 inhibitor below the control levels.

Notch pathway has a crucial role in the activation and the differentiation of innate and adaptive immune cells [12].

In SARS-Cov2 infection, a previous study using a computational model reported that the proteins interacting with SARS-CoV-2 RNA 5′-region were associated with Notch2 receptor signaling [23]. In addition, Notch signaling indirectly promotes viral entry through Furin induction by Notch1 [12]. Furthermore, the involvement of Notch signaling in cytokines storm, coagulopathy, and hypoxic events accompanied SARS-CoV-2 infection has been reported [13].

Hence, the ability of 2-pyridone PIM1 inhibitor to normalize the expression of Notch 1 and reduce the expression of Notch 2 below the control levels could provide a mechanism by which 2-pyridone PIM1 inhibitor could be effective in treatment of COVID-19. Previous studies reported that inhibition of Notch signaling pathway can hinder SARS-CoV-2 infection, decrease inflammatory responses, and help alveolar regeneration [24].

Another pathway implicated in host–pathogen interaction is the Wnt/β-catenin signaling pathway. Several previous studies reported the crucial role of Wnt/β-catenin signaling pathway in many viral infections including Flavivirus [25], influenza virus [26], and herpes simplex virus 1 [27]. All those studies reported that the activation of Wnt/β-catenin signaling during viral infection disturbs cellular homeostasis and provides a pathogenic environment that induces viral replication. Also, they reported that inhibition of Wnt/β-catenin signaling repressed viral replication and had strong antiviral effect.

In the present study, CTNNB1 was overexpressed after SARS-CoV-2 infection, in agreement with previous studies [28, 29]. ِAfter 2-pyridone PIM1 inhibitor treatment, CTNNB1 expression was significantly decreased, even below its levels in control cells. The explanation and potential effects of this downregulation need further studies. During SARS-CoV-2 infection, Wnt/β-catenin pathway was reported to be upregulated in association with TGF-b and STAT pathway leading to stimulate immune signaling, increase inflammatory cytokines production, and promote pulmonary fibrosis. Thus, Wnt/β-catenin signaling inhibition could negatively modulate SARS-CoV-2 infection [28].

Being one of the Rho family GTPases, CDC42 plays a vital role in the regulation of actin cytoskeleton, vesicle trafficking, and cell–cell adhesion. Thus, several viruses including Ebola virus, respiratory syncytial virus, and coronavirus hijack CDC42 to facilitate viral cell entry and subsequent nuclear infiltration [30]. Also, Kolyvushko et al. (2020) identified that the activation of CDC42 by EHV-1 is required for stabilization of tubulin to promote intracellular virus transport and cell-to-cell virus spread. They also found that the chemical inhibition of CDC42 resulted in precluding virus trafficking to the nucleus of infected cells [31]. In the current study, CDC42 was upregulated in SARS-CoV-2-infected cells. Treatment with 2-pyridone PIM1 inhibitor was able to downregulate its level, yet it remained higher than the control levels. In line with our results, a previous study showed alteration in expression of CDC42 in nasopharyngeal swabs of COVID‐19 compared with non‐COVID‐19 patients. Also, they found downregulation of CDC42 expression upon treatment of human lung carcinoma A549 cells with Rho GTPases inhibitor as atorvastatin [32]. The role of CDC42 in COVID-19 progression remains unclear. However, an in vitro study performed on Vero E6 cells confirmed that SARS-CoV-2 depended on CDC42 as one of different signaling pathways involved in viral cell entry and cell–cell fusion mechanisms [33].

In the current study, Notch signaling downstream genes, BCL9 and ACTR2, were also assessed. SARS-CoV-2 infection caused overexpression of BCL9 and underexpression of ACTR2 in infected cells after 24 h post-infection. Treatment with 2-pyridone PIM1 inhibitor was able to restore BCL9 expression to a level near that of control cells, while the level of ACTR2 expression was lower in the treated cells than both in control cells and infected untreated cells. Previously, a strong interaction between BCL9 and the Wnt/β-catenin pathway was reported. BCL9 acts as a transcriptional coactivator of CTNNB1 protein via binding to its N-terminus [34]. Thus, BCL9 knockdown strongly inhibits the transcriptional activity of CTNNB1 and also the expression of various downstream genes in the Wnt/β-catenin pathway [35].

ACTR2 gene codes for ARP2 protein which has a vital role in actin polymerization, cell shape, and motility. Also, ARP2 has a crucial role in RSV spread through formation of filopodia [36]. Recently, it was suggested that ARP2 plays a role in host–pathogen interaction in SARS-CoV-2 infection. Moreover, inhibition of viral RNA expression was reported upon pharmacological inhibition of ARP2/3 complex [37]. Collectively, the studied Notch pathway genes were increased with increased viral load after SARS-CoV-2 infection, with the exception of ACTR2 as discussed above. Treatment with 2-pyridone PIM1 inhibitor decreased the viral load and was able to significantly reverse the effect of SARS-CoV-2 infection on the Notch pathway genes. As detailed above, Notch pathway was previously shown to interact with many pathogens including SARS-CoV-2, modulating their infectivity and viral entry [13]. Hence, results of the current study suggest that the 2-pyridone PIM1 inhibitor could hinder SARS-CoV-2 infectivity via modulating its interaction with Notch pathway.

In the present study, SARS-CoV-2-infected cells showed a significant overexpression of SUMO1 and TDG. Treatment with 2-pyridone PIM1 inhibitor significantly downregulated both genes to levels even below their levels in control cells.

SUMOylation pathway was reported to have a potential role in regulating cell homeostasis during viral infection. Various viruses were found to be able to hijack host cells’ SUMO pathways to promote virus replication and pathogenesis [38]. Also, SUMOylation was reported to play a vital role in regulating innate immunity through altering interferon production and inhibiting NF-κB transcription [39].

As the 2-pyridone PIM1 inhibitor significantly shifted SUMO1 levels below control levels, inhibition of SUMO1 could be a part of the 2-pyridone PIM1 inhibitor antiviral activity. As reported by [38], alteration in expression of genes regulating SUMO pathways was found in COVID-19 patients, suggesting SUMO pathway as a target for anti-coronavirus therapy. In addition, the interaction between the N-protein of SARS-CoV and SUMO machinery was documented. Thus, the interaction between SARS-CoV-2 and SUMO is possible due to significant similarity of the N-proteins among coronaviruses [40].

SUMOylation was reported to affect the substrate binding capacity and catalytic activity of TDG enzyme [41]. TDG was found to be upregulated in the current study in SARS-CoV-2-infected cells. 2-pyridone PIM1 inhibitor decreased its levels below the control levels. The ability of SUMO1 to regulate TDG activity could explain the TDG overexpression after SARS-CoV-2 infection [42]. Another study revealed that human TDG could be manipulated by SARS-CoV2 during the genome organization leading to the appearance of mutated strains with subsequent increase in the viral transmission rate and pathogenicity [43].

Conclusion

Treatment with 2-pyridone PIM1 inhibitor could hinder SARS-CoV-2 cell entry and lower the viral load. In addition, 2-pyridone PIM1 inhibitor could significantly lower the expression levels of Notch pathway genes, CTNNB1, SUMO1, and TDG, which were overexpressed by SARS-CoV-2 infection. Thus, 2-pyridone PIM1 inhibitor could affect SARS-CoV-2 pathogenesis through modulation of several pathways implicated in immunity, suggesting its potential use in the development of anti-SARS-CoV-2 therapeutic approach.

Availability of data and materials

All data generated or analyzed during this study are included in this published article and its supplementary information files.

Abbreviations

COVID-19:

Coronavirus disease 2019

S-protein:

Spike protein

ACE2:

Angiotensin-converting enzyme 2

TMPRSS2:

Transmembrane protease, serine 2

PIM1:

Serine/threonine-protein kinase pim-1

HPV:

Human papillomavirus

HBV:

Hepatitis B virus

HCV:

Hepatitis C virus

DMEM:

Dulbecco’s Modified Eagle’s Medium

FBS:

Fetal bovine serum

CC50 :

Half-maximal cytotoxic concentration

IC50 :

Inhibitory concentration

CPE:

Cytopathic effect

PPI:

Protein–protein interaction

FDR:

False discovery rate

IL-6:

Interleukin-6

References

  1. Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, Qiu Y, Wang J, Liu Y, Wei Y, Xia J, Yu T, Zhang X, Zhang L (2020) Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet 395:507–513. https://doi.org/10.1016/s0140-6736(20)30211-7.

    Article  Google Scholar 

  2. Hembram P (2021) An outline of SARS-CoV-2 pathogenesis and the complement cascade of immune system. Bulletin of the National Research Centre 45. https://doi.org/10.1186/s42269-021-00582-2.

  3. Jackson CB, Farzan M, Chen B, Choe H (2021) Mechanisms of SARS-CoV-2 entry into cells. Nat Rev Mol Cell Biol 23:3–20. https://doi.org/10.1038/s41580-021-00418-x.

    Article  Google Scholar 

  4. Padmanabhan P, Desikan R, Dixit NM (2020) Targeting TMPRSS2 and Cathepsin B/L together may be synergistic against SARS-CoV-2 infection. PLOS Computational Biology 16:e1008461. https://doi.org/10.1371/journal.pcbi.1008461.

  5. de Vries M, Smithers NP, Howarth PH, Nawijn MC, Davies DE (2015) Inhibition of Pim1 kinase reduces viral replication in primary bronchial epithelial cells. Eur Respir J 45:1745–1748. https://doi.org/10.1183/09031936.00206514.

    Article  Google Scholar 

  6. Zhou F, Wan Q, Lu J, Chen Y, Lu G, He M-L (2019) Pim1 impacts Enterovirus A71 replication and represents a potential target in antiviral therapy. iScience 19:715–727 . https://doi.org/10.1016/j.isci.2019.08.008.

  7. Zhou F, Wan Q, Chen Y, Chen S, He M (2021) PIM1 kinase facilitates Zika virus replication by suppressing host cells’ natural immunity. Signal Transduction and Targeted Therapy 6: https://doi.org/10.1038/s41392-021-00539-x.

  8. Volberding PJ, Xin G, Kasmani MY, Khatun A, Brown AK, Nguyen C, Stancill JS, Martinez E, Corbett JA, Cui W (2021) Suppressive neutrophils require PIM1 for metabolic fitness and survival during chronic viral infection. Cell Reports 35:109160. https://doi.org/10.1016/j.celrep.2021.109160.

  9. Ledet RJ, Ruff SE, Wang Y, Nayak S, Schneider JA, Ueberheide B, Logan SK, Garabedian MJ (2021) Identification of PIM1 substrates reveals a role for NDRG1 phosphorylation in prostate cancer cellular migration and invasion. Communications Biology 4: https://doi.org/10.1038/s42003-020-01528-6.

  10. Li Y, Kong D, Wang Z, Ahmad A, Bao B, Padhye S, Sarkar FH (2011) Inactivation of AR/TMPRSS2-ERG/Wnt signaling networks attenuates the aggressive behavior of prostate cancer cells. Cancer Prev Res 4:1495–1506. https://doi.org/10.1158/1940-6207.capr-11-0077.

    Article  Google Scholar 

  11. Sacconi A, Donzelli S, Pulito C, Ferrero S, Spinella F, Morrone A, Rigoni M, Pimpinelli F, Ensoli F, Sanguineti G, Pellini R, Agrawal N, Izumchenko E, Ciliberto G, Giannì A, Muti P, Strano S, Blandino G (2020) TMPRSS2, a SARS-CoV-2 internalization protease is downregulated in head and neck cancer patients. Journal of Experimental & Clinical Cancer Research 39: https://doi.org/10.1186/s13046-020-01708-6.

  12. Rizzo P, Vieceli Dalla Sega F, Fortini F, Marracino L, Rapezzi C, Ferrari R (2020) COVID-19 in the heart and the lungs: could we “Notch” the inflammatory storm? Basic Research in Cardiology 115: https://doi.org/10.1007/s00395-020-0791-5.

  13. Breikaa RM, Lilly B (2021) The Notch pathway: a link between COVID-19 pathophysiology and its cardiovascular complications. Frontiers in Cardiovascular Medicine 8: https://doi.org/10.3389/fcvm.2021.681948.

  14. Santio NM, Landor SK-J, Vahtera L, Ylä-Pelto J, Paloniemi E, Imanishi SY, Corthals G, Varjosalo M, Manoharan GB, Uri A, Lendahl U, Sahlgren C, Koskinen PJ (2016) Phosphorylation of Notch1 by Pim kinases promotes oncogenic signaling in breast and prostate cancer cells. Oncotarget 7:43220–43238. https://doi.org/10.18632/oncotarget.9215.

  15. Ismail MMF, Farrag AM, Abou-El-Ela D (2020) Synthesis, anticancer screening, and in silico ADME prediction of novel2-pyridonesas Pim inhibitors. J Heterocycl Chem. https://doi.org/10.1002/jhet.4064.

    Article  Google Scholar 

  16. Kandeil A, Mostafa A, Hegazy RR, El-Shesheny R, El Taweel A, Gomaa MR, Shehata M, Elbaset MA, Kayed AE, Mahmoud SH, Moatasim Y, Kutkat O, Yassen NN, Shabana ME, GabAllah M, Kamel MN, Abo Shama NM, El Sayes M, Ahmed AN, Elalfy ZS, Mohamed BM, Abd El-Fattah SN, El Hariri HM, Abdel Kader M, Azmy O, Kayali G, Ali MA (2021) Immunogenicity and safety of an inactivated SARS-CoV-2 vaccine: preclinical studies. Vaccines 9:214. https://doi.org/10.3390/vaccines9030214.

    Article  Google Scholar 

  17. Mostafa A, Kandeil A, A. M. M. Elshaier Y, Kutkat O, Moatasim Y, Rashad AA, Shehata M, Gomaa MR, Mahrous N, Mahmoud SH, GabAllah M, Abbas H, Taweel AE, Kayed AE, Kamel MN, Sayes ME, Mahmoud DB, El-Shesheny R, Kayali G, Ali MA (2020) FDA-approved drugs with potent in vitro antiviral activity against severe acute respiratory syndrome coronavirus 2. Pharmaceuticals 13:443 https://doi.org/10.3390/ph13120443.

  18. Feoktistova M, Geserick P, Leverkus M (2016) Crystal violet assay for determining viability of cultured cells. Cold Spring Harbor Protocols 2016:pdb.prot087379. https://doi.org/10.1101/pdb.prot087379.

  19. Murakami Y, Mizuguchi K (2014) Homology-based prediction of interactions between proteins using averaged one-dependence estimators. BMC Bioinformatics 15. https://doi.org/10.1186/1471-2105-15-213.

  20. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, Jensen LJ, von Mering C (2019) STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res 47:D607–D613. https://doi.org/10.1093/nar/gky1131.

    Article  Google Scholar 

  21. Yang Y, Cui X, Wei H, Guo C, Zhang Y (2021) Potential anti-coronavirus agents and the pharmacologic mechanisms. Drug Des Dev Ther 15:1213–1223. https://doi.org/10.2147/dddt.s293216.

    Article  Google Scholar 

  22. Argyropoulos KV, Serrano A, Hu J, Black M, Feng X, Shen G, Call M, Kim MJ, Lytle A, Belovarac B, Vougiouklakis T, Lin LH, Moran U, Heguy A, Troxel A, Snuderl M, Osman I, Cotzia P, Jour G (2020) Association of initial viral load in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) patients with outcome and symptoms. Am J Pathol 190:1881–1887. https://doi.org/10.1016/j.ajpath.2020.07.001.

    Article  Google Scholar 

  23. Vandelli A, Monti M, Milanetti E, Armaos A, Rupert J, Zacco E, Bechara E, Delli Ponti R, Tartaglia GG (2020) Structural analysis of SARS-CoV-2 genome and predictions of the human interactome. Nucleic Acids Res 48:11270–11283. https://doi.org/10.1093/nar/gkaa864.

    Article  Google Scholar 

  24. Baindara P, Sarker MB, Earhart AP, Mandal SM, Schrum AG (2022) NOTCH signaling in COVID-19: a central hub controlling genes, proteins, and cells that mediate SARS-CoV-2 entry, the inflammatory response, and lung regeneration. Frontiers in Cellular and Infection Microbiology 12. https://doi.org/10.3389/fcimb.2022.928704.

  25. Smith JL, Jeng S, McWeeney SK, Hirsch AJ (2017) A microRNA screen identifies the Wnt signaling pathway as a regulator of the interferon response during Flavivirus infection. Journal of Virology 91: . https://doi.org/10.1128/jvi.02388-16.

  26. More S, Yang X, Zhu Z, Bamunuarachchi G, Guo Y, Huang C, Bailey K, Metcalf JP, Liu L (2018) Regulation of influenza virus replication by Wnt/β-catenin signaling. PLOS ONE 13:e0191010. https://doi.org/10.1371/journal.pone.0191010.

  27. Koujah L, Madavaraju K, Agelidis AM, Patil CD, Shukla D (2021) Heparanase-induced activation of AKT stabilizes β-catenin and modulates Wnt/β-Catenin signaling during herpes simplex virus 1 infection. mBio 12: . https://doi.org/10.1128/mbio.02792-21.

  28. Vallée A, Lecarpentier Y, Vallée J-N (2021) Interplay of opposing effects of the WNT/β-Catenin pathway and PPARγ and implications for SARS-CoV2 treatment. Frontiers in Immunology 12: . https://doi.org/10.3389/fimmu.2021.666693.

  29. Chatterjee S, Keshry SS, Ghosh S, Ray A, Chattopadhyay S (2022) Versatile β-Catenin is crucial for SARS-CoV-2 infection. Microbiology Spectrum 10. https://doi.org/10.1128/spectrum.01670-22.

  30. Swaine T, Dittmar M (2015) CDC42 use in viral cell entry processes by RNA viruses. Viruses 7:6526–6536. https://doi.org/10.3390/v7122955.

    Article  Google Scholar 

  31. Kolyvushko O, Kelch MA, Osterrieder N, Azab W (2020) Equine alphaherpesviruses require activation of the small GTPases Rac1 and Cdc42 for intracellular transport. Microorganisms 8:1013. https://doi.org/10.3390/microorganisms8071013.

    Article  Google Scholar 

  32. Segatori VI, Garona J, Caligiuri LG, Bizzotto J, Lavignolle R, Toro A, Sanchis P, Spitzer E, Krolewiecki A, Gueron G, Alonso DF (2021) Effect of ivermectin and atorvastatin on nuclear localization of importin alpha and drug target expression profiling in host cells from nasopharyngeal swabs of SARS-CoV-2- positive patients. Viruses 13:2084. https://doi.org/10.3390/v13102084.

    Article  Google Scholar 

  33. Zhang Y-Y, Liang R, Wang S-J, Ye Z-W, Wang T-Y, Chen M, Liu J, Na L, Yang Y-L, Yang Y-B, Yuan S, Yin X, Cai X-H, Tang Y-D (2022) SARS-CoV-2 hijacks macropinocytosis to facilitate its entry and promote viral spike–mediated cell-to-cell fusion. Journal of Biological Chemistry 298:102511. https://doi.org/10.1016/j.jbc.2022.102511.

  34. Vafaizadeh V, Buechel D, Rubinstein N, Kalathur RKR, Bazzani L, Saxena M, Valenta T, Hausmann G, Cantù C, Basler K, Christofori G (2021) The interactions of Bcl9/Bcl9L with β-catenin and Pygopus promote breast cancer growth, invasion, and metastasis. Oncogene 40:6195–6209. https://doi.org/10.1038/s41388-021-02016-9.

    Article  Google Scholar 

  35. Wang X, Feng M, Xiao T, Guo B, Liu D, Liu C, Pei J, Liu Q, Xiao Y, Rosin-Arbesfeld R, Shi Y, Zhou Y, Yang M, Feng Y-X, Jiang Y, Shao Z, Yu K, Zhu D (2021) BCL9/BCL9L promotes tumorigenicity through immune-dependent and independent mechanisms in triple negative breast cancer. Oncogene 40:2982–2997. https://doi.org/10.1038/s41388-021-01756-y.

    Article  Google Scholar 

  36. Mehedi M, McCarty T, Martin SE, Le Nouën C, Buehler E, Chen Y-C, Smelkinson M, Ganesan S, Fischer ER, Brock LG, Liang B, Munir S, Collins PL, Buchholz UJ (2016) Actin-related protein 2 (ARP2) and virus-induced filopodia facilitate human respiratory syncytial virus spread. PLOS Pathogens 12:e1006062. https://doi.org/10.1371/journal.ppat.1006062.

  37. Schmidt N, Lareau CA, Keshishian H, Ganskih S, Schneider C, Hennig T, Melanson R, Werner S, Wei Y, Zimmer M, Ade J, Kirschner L, Zielinski S, Dölken L, Lander ES, Caliskan N, Fischer U, Vogel J, Carr SA, Bodem J, Munschauer M (2021) The SARS-CoV-2 RNA–protein interactome in infected human cells. Nat Microbiol 6:339–353. https://doi.org/10.1038/s41564-020-00846-z.

    Article  Google Scholar 

  38. Ryu H-Y (2021) SUMO: a novel target for anti-coronavirus therapy. Pathog Glob Health 115:292–299. https://doi.org/10.1080/20477724.2021.1906562.

    Article  Google Scholar 

  39. Imbert F, Langford D (2021) Viruses, SUMO, and immunity: the interplay between viruses and the host SUMOylation system. J Neurovirol 27:531–541. https://doi.org/10.1007/s13365-021-00995-9.

    Article  Google Scholar 

  40. Imbert F, Leavitt G, Langford D (2022) SUMOylation and viral infections of the brain. Pathogens 11:818. https://doi.org/10.3390/pathogens11070818.

    Article  Google Scholar 

  41. Coey CT, Drohat AC (2018) Defining the impact of sumoylation on substrate binding and catalysis by thymine DNA glycosylase. Nucleic Acids Res 46:5159–5170. https://doi.org/10.1093/nar/gky278.

    Article  Google Scholar 

  42. Smet-Nocca C, Wieruszeski J-M, Léger H, Eilebrecht S, Benecke A (2011) SUMO-1 regulates the conformational dynamics of thymine-DNA glycosylase regulatory domain and competes with its DNA binding activity. BMC Biochemistry 12. https://doi.org/10.1186/1471-2091-12-4.

  43. Zolfaghari Emameh R, Eftekhari M, Nosrati H, Heshmatnia J, Falak R (2021) Identification and characterization of a silent mutation in RNA binding domain of N protein coding gene from SARS-CoV-2. BMC Research Notes 14. https://doi.org/10.1186/s13104-020-05439-x.

Download references

Acknowledgements

Not applicable

Funding

This research was partially funded by the NRC under contract number MP120801 (to M. A. A.) and by the Egyptian Academy of Scientific Research & Technology (ASRT) within the “Ideation Fund” program under contract number 7303 (to A. M.). This research was also funded by the National Institute of Allergy and Infectious Diseases, National Institutes of Health, and US Department of Health and Human Services (under contract HHSN272201400006C). The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Author information

Authors and Affiliations

Authors

Contributions

MMFI and AMF were responsible for design, preparation, and investigation of 2-pyridone PIM1 inhibitor potential effects. SHM and NMAS contributed to investigations. AM contributed to methodology, investigation, review and editing of the manuscript, and visualization. MAA was responsible for resources. MHR contributed to methodology and resources. RREA and IHI contributed to resources, methodology, investigations, bioinformatics study, and visualization, drafting, and reviewing the manuscript. The authors read and approved the final manuscript.

Corresponding authors

Correspondence to Rehab R. El-Awady or Iman H. Ibrahim.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1:

 PPI prediction between PIM1 and the studied genes.

Additional file 2:

 Functional PPI enrichments of the studied genes.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ismail, M.M.F., El-Awady, R.R., Farrag, A.M. et al. Potential role of PIM1 inhibition in the treatment of SARS-CoV-2 infection. J Genet Eng Biotechnol 21, 65 (2023). https://doi.org/10.1186/s43141-023-00520-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43141-023-00520-x

Keywords