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 Jul 10;16(1):6397.
doi: 10.1038/s41467-025-61759-1.

PIP4K2C inhibition reverses autophagic flux impairment induced by SARS-CoV-2

Affiliations

PIP4K2C inhibition reverses autophagic flux impairment induced by SARS-CoV-2

Marwah Karim et al. Nat Commun. .

Abstract

In search for broad-spectrum antivirals, we discover a small molecule inhibitor, RMC-113, that potently suppresses the replication of multiple RNA viruses including SARS-CoV-2 in human lung organoids. We demonstrate selective inhibition of the lipid kinases PIP4K2C and PIKfyve by RMC-113 and target engagement by its clickable analog. Lipidomics analysis reveals alteration of SARS-CoV-2-induced phosphoinositide signature by RMC-113 and links its antiviral effect with functional PIP4K2C and PIKfyve inhibition. We identify PIP4K2C's roles in SARS-CoV-2 entry, RNA replication, and assembly/egress, validating it as a druggable antiviral target. Integrating proteomics, single-cell transcriptomics, and functional assays, reveals that PIP4K2C binds SARS-CoV-2 nonstructural protein 6 and regulates virus-induced autophagic flux impairment. Promoting viral protein degradation by reversing autophagic flux impairment is a mechanism of antiviral action of RMC-113. These findings reveal virus-induced autophagy regulation via PIP4K2C, an understudied kinase, and propose dual PIP4K2C and PIKfyve inhibition as a candidate strategy to combat emerging viruses.

PubMed Disclaimer

Conflict of interest statement

Competing interests: The authors declare no competing interests.

Figures

Fig. 1
Fig. 1. RMC-113 inhibits SARS-CoV-2 infection in vitro and in human ALOs with a high genetic barrier to resistance.
a Chemical structure of RMC-113. b Rescue assay for virus-induced cell lethality. RMC-113 (10 μM) was incubated with Vero E6-eGFP cells for 20 h followed by SARS-CoV-2 infection. eGFP signal measured at 96 hpi indicates cell survival. c and d Fluorescence images (c) and corresponding graph (d) of Vero-eGFP cells rescued from SARS-CoV-2-induced lethality by RMC-113 (Belgium-GHB-03021 strain, MOI = 0.05). Original magnification, ×5 (c). e and j, Dose response to RMC-113 of SARS-CoV-2 infection [black, USA-WA1/2020 strain, MOI = 0.05 (e), 1 (j)] and cell viability (blue) in Calu-3 cells (e) or ALO-derived monolayer supernatants (j) via plaque and alamarBlue assays at 24 (e) or 48 (j) hpi, respectively. f and i, Schematics of the experiments shown in g (f) and j, k, l (i), respectively. Figure was created in BioRender. Karim, M. (2025) https://biorender.com/2yx5sx9. g, Vero E6-TMPRSS2 cells were infected with rSARS-CoV-2-nLuc virus (MOI = 0.05) and passaged daily under RMC-113 (0.1–0.3 μM) or DMSO over nine passages. Viral titers were measured by plaque assays. h, Dose response to RMC-113 of rSARS-CoV-2-nLuc virus harvested after nine passages under RMC-113 or DMSO via luciferase assays. k Dose response to RMC-113 of SARS-CoV-2 (MOI = 1) nucleocapsid copy number in ALO lysates measured by RT-qPCR assays at 48 hpi. l Confocal IF microscopy images of F-actin (violet), nucleocapsid (green), and DAPI (blue) in uninfected and SARS-CoV-2–infected ALOs, pretreated with DMSO or RMC-113 (5 μM) at 24 hpi. Representative merged images at x40 magnification are shown. Scale bars: 50 μm. Data shown are representative of independent experiments (c, d, j, l) or represent combined results from two (n = 2; e, g, h) or three (n = 3; k) independent experiments. Data in e, h, j, and k are relative to DMSO. Means ± SD are shown. Source data are provided as a Source Data file.
Fig. 2
Fig. 2. RMC-113 is a selective kinase inhibitor that stably binds PIKfyve and PIP4K2C.
a Kinase abundance ratio between RMC-113 (0.1, 1.0, and 10 µM)- and DMSO-treated SUM159 cell lysates measured by multiplexed inhibitor beads kinome profiling and mass spectrometry (MIB/MS). Shown are the Log2 fold change mean values ± SD of 2 replicates of a subset of the screen panel (see Supplementary Fig. 2a). b and c In vitro dose response of PIKfyve activity (b) and PIP4K2C binding (c) to RMC-113. d Biochemical parameters for RMC-113 and SRN2-002. ND = Not determined. e Chemical structure of SRN2-002. f Dose response of SARS-CoV-2 infection (MOI = 0.05, black) and cell viability (blue) to SRN2-002 in Calu-3 cells via plaque and alamarBlue assays at 24 hpi, respectively (n = 2 independent experiments). g Kinase expression in SARS-CoV-2-infected A549-ACE2 lysates following incubation with SRN2-002 individually or combined with RMC-113, UV irradiation, and streptavidin pull-down measured by Western blotting at 24 hpi. Representative membranes of n = 2 experiments are shown. Lanes: 1: SRN2-002, no UV; 2: SRN2-002, no pull-down; 3: SRN2-002 (1 µM); 4: SRN2-002 (5 µM); 5: SRN2-002 (1 µM) competitively inhibited by RMC-113 (10 µM); 6: SRN2-002 (5 µM) competitively inhibited by RMC-113 (50 µM). h, Putative binding mode of RMC-113 into the kinase ATP-binding pockets based on microsecond timescale MD simulations. A representative snapshot with key interactions is shown (see i–l for details). Binding pocket residues with >10% interaction frequencies to RMC-113 are shown. Positively charged residues are highlighted in blue; negative charge, red; hydrophobic, green; polar, cyan. RMC-113 is shown in yellow (carbon) stick model. H-bonds are illustrated with purple dashed lines, π–π interactions with green. i and k, Summary of main protein-ligand interactions ( > 10% frequency) in the MD simulations of PIKfyve (i) and PIP4K2C (k). j and l Aggregate of protein-ligand interactions (residues with >10%) in the simulations of PIKfyve (j) and PIP4K2C (l). Means ± SD are shown (f). Data in (i and j) consist of 72 μs, and (k and l) consist of 20 μs, both analyzed each 1 ns. Source data are provided as a Source Data file.
Fig. 3
Fig. 3. PIKfyve and PIP4K2C are essential for SARS-CoV-2 infection and mediate the antiviral effect of RMC-113.
a, g, k, Schematics of the experiments in b, c, d (a); h, i (g); and l, m, n (k). b and c Confirmation of siRNA-mediated knockdown of PIKfyve and PIP4K2C by RT-qPCR (b) and Western blot (c) in Calu-3 cells. Shown are gene expression levels normalized to GAPDH relative to respective gene levels in siNT control (b) and representative membranes and quantitative data (c) at 48 h post-transfection. d Viral titers (PFU/ml) and cell viability (blue) in Calu-3 cells transfected with the indicated siRNAs at 24 hpi with SARS-CoV-2 (MOI = 0.05) via plaque and alamarBlue assays, respectively. e Chemical structures of PIKfyve and/or PIP4K2C inhibitors. f Dose response of rSARS-CoV-2-nLuc (black, USA-WA1/2020 strain, MOI = 0.05) infection and cell viability (blue) in Calu-3 cells via luciferase and alamarBlue assays at 24 hpi, respectively. h and i Rescue of rVSV-SARSCoV-2-S infection under RMC-113 treatment upon ectopic expression of WT PIKfyve (h) and PIP4K2C (i) their kinase-dead mutants or empty control plasmids measured by luciferase assays at 24 hpi in Vero cells. j Schematic representation of phosphoinositides and associated kinases. Figure was created in BioRender. Karim, M. (2025) https://biorender.com/yynkvs0. l and m, Fold change (FC) of peak area ratio of the indicated phosphoinositides in SARS-CoV-2-infected (MOI = 0.5) vs. uninfected (l) and RMC-113- vs. DMSO-treated infected A549-ACE2 cells (m) as measured via PRMC-MS. n, Product-to-substrate ratios in RMC-113- vs. DMSO-treated cells infected with SARS-CoV-2 (l, m). Data shown are representative of independent experiments (c) or represent combined results from two (n = 2; f, h, i) or three (n = 3; b, d) independent experiments. Data is relative to siNT (b, d) or DMSO (f, h, i). Means ± SD are shown (b, d, f, h, i). l, m, n represent one of two independent experiments. See an associated experiment in Supplementary Fig. 6e–g and Supplementary Data 2. P values by 1-way ANOVA followed by Dunnett’s (b, d) multiple-comparison test are indicated. Source data are provided as a Source Data file.
Fig. 4
Fig. 4. PIP4K2C is required for temporally distinct stages of the SARS-CoV-2 life cycle, whereas PIKfyve is required for viral entry only.
a Schematic of the time-of-addition experiments shown in b, c. b and c Calu-3 cells were infected with WT SARS-CoV-2 (MOI = 1). At the indicated times, RMC-113 (10 μM) (b), apilimod (10 μM) (c), or DMSO were added. Supernatants were collected at 10 hpi, and viral titers measured by plaque assays. d, f and h, Schematics of the experiments shown in e, k (d); g, j, l (f) and i (h). e, Dose response to RMC-113 of WT SARS-CoV-2 entry (MOI = 1) in Calu-3 cell lysates measured by RT-qPCR assays at 2 hpi. g, Dose response to RMC-113 and ensitrelvir of viral RNA replication measured by luciferase assay in Vero E6 cells 24 h post-transfection of in vitro transcribed nano-luciferase reporter-based SARS-CoV-2 subgenomic non-infectious replicon. i Dose response to apilimod of rVSV-SARS-CoV-2-S infection (black) and cell viability (blue) in Vero cells via luciferase and alamarBlue assays at 24 hpi, respectively. j Dose response to apilimod of viral RNA replication measured by luciferase assay in Vero E6-TMPRSS2 cells 24 h post-transfection of in vitro transcribed nano-luciferase reporter-based SARS-CoV-2 subgenomic non-infectious replicon. k WT SARS-CoV-2 (MOI = 1) entry measured in Calu-3 cells depleted of the indicated kinases using corresponding siRNAs by RT-qPCR at 2 hpi. l, Viral RNA replication and cell viability (blue) measured by luciferase and alamarBlue assays, respectively, in Vero E6 cells depleted of the indicated kinases, 24 h post-transfection of in vitro transcribed nano-luciferase reporter-based SARS-CoV-2 subgenomic non-infectious replicon. Data shown are representative of independent experiments (i) or represent combined results from two (n = 2; e, g, j) or three (n = 3; b, c, k, l) independent experiments. Means ± SD are shown. Data is relative to DMSO (b, c, e, g, i, j) or siNT (k, l). P values by 1-way ANOVA followed by Tukey’s (b, c) or Dunnett’s (k, l) multiple-comparison test are indicated. ns= non-significant. Source data are provided as a Source Data file.
Fig. 5
Fig. 5. RMC-113 reverses SARS-CoV-2-induced impairment of autophagic flux and accelerates viral protein degradation.
a Schematic for c and d. b Autophagy flux: from autophagosomes (yellow, RFP + /GFP + ) to autolysosomes (red, RFP+/GFP-). Figure was created in BioRender. Karim, M. (2025) https://biorender.com/yynkvs0. c, Representative confocal images (x40) of A549-ACE2 cells expressing GFP-RFP-LC3, infected with SARS-CoV-2 (MOI = 0.5), treated with DMSO, RMC-113 (5 μM), apilimod (5 μM) or CQ (10 μM) for 24 h, and stained for nucleocapsid (violet). Scale bars: 10 μm. Zoomed-in images: autophagosomes (yellow); autolysosomes (red). d, Autolysosome-to-autophagosome ratio in 70 single cells (c). e–g Autophagy marker expression in uninfected and SARS-CoV-2-infected (MOI = 1) A549-ACE2 lysates treated with RMC-113 (5 µM) and/or CQ (10 µM) or DMSO at 24 hpi via immunoblotting (e). Quantification of p62/actin (f) and LC3II/actin (g) ratios normalized to DMSO (n = 4 membranes). h and i, Representative membranes (h) and quantification (i) (n = 3 membranes) showing ORF3A half-life in A549-ACE2 lysates after RMC-113 (5 µM) or DMSO treatment. j, Marker genes annotating cell populations. Color: expression (cpm); dot size: expressing cell fraction. k and l UMAP embedding of scRNA-seq dataset indicating distinct cell types (k) or SARS-CoV-2 transcripts (l). m and n Pathway enrichment in AT2 cells infected vs. uninfected (m) and RMC-113 vs. DMSO treated (n) ALOs. o, Heatmap of log2 fold-change in autophagy-related genes between infected vs. uninfected (DMSO), RMC-113 vs. DMSO (uninfected), and RMC-113 vs. DMSO (infected) AT2-like cells at 24 hpi. Black rectangles: significant changes (Wilcoxon test). See Supplementary Fig. 8b. p–r Box plots of gene expression in individual AT2 cells at 24 hpi. Horizontal lines indicate quartiles; whiskers extend to ±1.5 × interquartile range. Adjusted P values (two-sided Wilcoxon test, Benjamini-Hochberg correction). Data are representative of independent experiments (c, e, h) or combined results from three (n = 3; d, i–r) or four (n = 4; f, g) independent experiments. Means ± SD are shown (d, f, g, i). P values by 1-way (d) or 2-way (i) ANOVA with Tukey’s (d) or two-tailed unpaired t-test (f, g) or Šídák’s (i) multiple comparison test are indicated. ns= non-significant. Source data are provided as Source Data file.
Fig. 6
Fig. 6. PIP4K2C binds SARS-CoV-2 NSP6 and mediates virus-induced impairment of autophagic flux.
a Schematic of the experiment shown in b and c. b Representative confocal microscopic images of A549-ACE2 cells transfected with indicated siRNAs and GFP-RFP-LC3 tandem plasmid, and infected with SARS-CoV-2 (MOI = 0.5) for 24 h and stained for nucleocapsid (violet). Representative merged images at x40 magnification are shown. Scale bars: 10 μm. Zoomed-in images show autophagosomes (yellow) and autolysosomes (red). c Autolysosome-to-autophagosomes ratio (autophagy flux) in single cells (n = 27 cells per category) (b). d–i The expression levels of p62, LC3-I and LC3-II following transfection of siPIP4K2C (d–f) and siPIKfyve (g–i) in uninfected and SARS-CoV-2-infected A549-ACE2 cell lysates at 24 hpi. Bar graphs show quantitative analysis of p62/actin (e, h) and LC3II/actin ratios (f, i), from 2 membranes and normalized to corresponding siNT controls. j PIP4K2C and PIKfyve interactions with 15 SARS-CoV-2 nonstructural proteins, ORF3a, ORF7a, and empty plasmid as measured via protein-fragment complementation assay (PCAs) in HEK293T cells. Dots depict mean NLR values. The dotted line depicts the cutoff (NLR > 10) used to define PIP4K2C-interacting proteins (green), representing greater than two SDs above the mean NLR of a non-interacting reference set. k and l Confocal IF microscopy images of PIP4K2C (red) and NSP6 (green) in A549-ACE2 cells ectopically expressing FLAG-NSP6 24 h after transfection. Shown are images at 60× magnification with a 6-fold zoom in (right panel) (k) and Manders’ colocalization coefficients (M2: NSP6 overlapping PIP4K2C) (l) in 32 cells. Scale bar: 10 μm. m Proposed model for the roles of PIP4K2C and PIKfyve in SARS-CoV-2 infection and the mechanism of antiviral action of RMC-113. Figure was created in BioRender. Karim, M. (2025) https://biorender.com/glw3z17. Data are representative of independent experiments (b, d, g, k) or combined from two (n = 2; e, f, h, i, j) or three (n = 3; c, l) independent experiments. Data are relative to siNT (c, e, f, h, i). Means ± SD are shown (c, l). P values by 1-way ANOVA followed by Dunnett’s multiple comparison test are shown (c). Source data are provided as a Source Data file.

Update of

References

    1. Weaver, S. C., Ferro, C., Barrera, R., Boshell, J. & Navarro, J.-C. Venezuelan equine encephalitis. Annu. Rev. Entomol.49, 141–174 (2004). - PubMed
    1. Karim, M., Lo, C.-W. & Einav, S. Preparing for the next viral threat with broad-spectrum antivirals. J. Clin. Investig.133, e170236 (2023). - PMC - PubMed
    1. Llorente, A., Arora, G. K., Grenier, S. F. & Emerling, B. M. PIP kinases: a versatile family that demands further therapeutic attention. Adv. Biol. Regul.87, 100939 (2023). - PMC - PubMed
    1. Sbrissa, D., Ikonomov, O. C. & Shisheva, A. PIKfyve, a mammalian ortholog of yeast Fab1p lipid kinase, synthesizes 5-phosphoinositides: effect of insulin. J. Biol. Chem.274, 21589–21597 (1999). - PubMed
    1. Sharma, G. et al. A family of PIKFYVE inhibitors with therapeutic potential against autophagy-dependent cancer cells disrupt multiple events in lysosome homeostasis. Autophagy15, 1694–1718 (2019). - PMC - PubMed

MeSH terms

Substances

LinkOut - more resources