Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Chemical genomics with pyrvinium identifies C1orf115 as a regulator of drug efflux

An Author Correction to this article was published on 24 August 2022

This article has been updated

Abstract

Pyrvinium is a quinoline-derived cyanine dye and an approved anti-helminthic drug reported to inhibit WNT signaling and have anti-proliferative effects in various cancer cell lines. To further understand the mechanism by which pyrvinium is cytotoxic, we conducted a pooled genome-wide CRISPR loss-of-function screen in the human HAP1 cell model. The top drug–gene sensitizer interactions implicated the malate–aspartate and glycerol-3-phosphate shuttles as mediators of cytotoxicity to mitochondrial complex I inhibition including pyrvinium. By contrast, perturbation of the poorly characterized gene C1orf115/RDD1 resulted in strong resistance to the cytotoxic effects of pyrvinium through dysregulation of the major drug efflux pump ABCB1/MDR1. Interestingly, C1orf115/RDD1 was found to physically associate with ABCB1/MDR1 through proximity-labeling experiments and perturbation of C1orf115 led to mis-localization of ABCB1/MDR1. Our results are consistent with a model whereby C1orf115 modulates drug efflux through regulation of the major drug exporter ABCB1/MDR1.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Pyrvinium inhibits mitochondrial respiration.
Fig. 2: Pooled CRISPR screen identifies chemical–genetic interactions with pyrvinium.
Fig. 3: Loss of GOT1, PGP, or pyrvinium treatment rewires central metabolic pathways.
Fig. 4: Loss of an uncharacterized gene suppresses the growth defects induced by pyrvinium.
Fig. 5: BioID reveals a proximity interaction between C1orf115 and ABCB1.
Fig. 6: C1orf115 regulates ABCB1 localization and drug efflux.

Similar content being viewed by others

Data availability

The datasets generated and analyzed during the current study are available in figures included in this article. Source data are provided with this paper.

Code availability

No advanced custom code was used for analyses in this study, and all tools and algorithms used are specified in the Methods.

Change history

References

  1. Costanzo, M. et al. Global genetic networks and the genotype-to-phenotype relationship. Cell 177, 85–100 (2019).

    CAS  PubMed  Google Scholar 

  2. Beck, J. W., Saavedra, D., Antell, G. J. & Tejeiro, B. The treatment of pinworm infections in humans (enterobiasis) with pyrvinium chloride and pyrvinium pamoate. Am. J. Trop. Med Hyg. 8, 349–352 (1959).

    CAS  PubMed  Google Scholar 

  3. Harada, Y., Ishii, I., Hatake, K. & Kasahara, T. Pyrvinium pamoate inhibits proliferation of myeloma/erythroleukemia cells by suppressing mitochondrial respiratory complex I and STAT3. Cancer Lett. 319, 83–88 (2012).

    CAS  PubMed  Google Scholar 

  4. Li, B. et al. Repurposing the FDA-approved pinworm drug pyrvinium as a novel chemotherapeutic agent for intestinal polyposis. PLoS ONE 9, e101969 (2014).

    PubMed  PubMed Central  Google Scholar 

  5. Venugopal, C. et al. Pyrvinium targets CD133 in human glioblastoma brain tumor-initiating cells. Clin. Cancer Res. 21, 5324–5337 (2015).

    CAS  PubMed  Google Scholar 

  6. Xiang, W. et al. Pyrvinium selectively targets blast phase-chronic myeloid leukemia through inhibition of mitochondrial respiration. Oncotarget 6, 33769–33780 (2015).

    PubMed  PubMed Central  Google Scholar 

  7. Thorne, C. A. et al. Small-molecule inhibition of Wnt signaling through activation of casein kinase 1α. Nat. Chem. Biol. 6, 829–836 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Steinhart, Z. et al. Genome-wide CRISPR screens reveal a Wnt-FZD5 signaling circuit as a druggable vulnerability of RNF43-mutant pancreatic tumors. Nat. Med. 23, 60–68 (2017).

    CAS  PubMed  Google Scholar 

  9. Cui, L., Zhao, J. & Liu, J. Pyrvinium sensitizes clear cell renal cell carcinoma response to chemotherapy via casein kinase 1α-dependent inhibition of Wnt/β-catenin. Am. J. Med Sci. 355, 274–280 (2018).

    PubMed  Google Scholar 

  10. Zhang, C., Zhang, Z., Zhang, S., Wang, W. & Hu, P. Targeting of Wnt/β-catenin by anthelmintic drug pyrvinium enhances sensitivity of ovarian cancer cells to chemotherapy. Med Sci. Monit. 23, 266–275 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Zheng, L., Liu, Y. & Pan, J. Inhibitory effect of pyrvinium pamoate on uveal melanoma cells involves blocking of Wnt/β-catenin pathway. Acta Biochim. Biophys. Sin. 49, 890–898 (2017).

    CAS  PubMed  Google Scholar 

  12. Hart, T. et al. High-resolution CRISPR screens reveal fitness genes and genotype-specific cancer liabilities. Cell 163, 1515–1526 (2015).

    CAS  PubMed  Google Scholar 

  13. Lau, M. T. et al. Systematic functional identification of cancer multi-drug resistance genes. Genome Biol. 21, 27 (2020).

    PubMed  PubMed Central  Google Scholar 

  14. Pal, S. K. et al. Mechanistic investigation of the androgen receptor DNA-binding domain inhibitor pyrvinium. ACS Omega 4, 2472–2481 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Chen, B. et al. Small molecule-mediated disruption of Wnt-dependent signaling in tissue regeneration and cancer. Nat. Chem. Biol. 5, 100–107 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Polosukhina, D. et al. Pharmacologic Inhibition of β-catenin with pyrvinium inhibits murine and human models of Wilms tumor. Oncol. Res 25, 1653–1664 (2017).

    PubMed  PubMed Central  Google Scholar 

  17. Birsoy, K. et al. Metabolic determinants of cancer cell sensitivity to glucose limitation and biguanides. Nature 508, 108–112 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Drahota, Z. et al. Biguanides inhibit complex I, II and IV of rat liver mitochondria and modify their functional properties. Physiol. Res 63, 1–11 (2014).

    CAS  PubMed  Google Scholar 

  19. Fu, J. et al. Biguanide MC001, a dual inhibitor of OXPHOS and glycolysis, shows enhanced anti-tumor activity without increasing lactate production. ChemMedChem 17, e202100674 (2022).

    CAS  PubMed  Google Scholar 

  20. Shitara, Y. et al. Role of organic cation/carnitine transporter 1 in uptake of phenformin and inhibitory effect on complex I respiration in mitochondria. Toxicol. Sci. 132, 32–42 (2013).

    CAS  PubMed  Google Scholar 

  21. Arroyo, J. D. et al. A genome-wide CRISPR death screen identifies genes essential for oxidative phosphorylation. Cell Metab. 24, 875–885 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Andrzejewski, S., Gravel, S. P., Pollak, M. & St-Pierre, J. Metformin directly acts on mitochondria to alter cellular bioenergetics. Cancer Metab. 2, 12 (2014).

    PubMed  PubMed Central  Google Scholar 

  23. Hart, T. et al. Evaluation and design of genome-wide CRISPR/SpCas9 knockout screens. G3 (Bethesda) 7, 2719–2727 (2017).

    CAS  PubMed  Google Scholar 

  24. Wang, T. et al. Gene essentiality profiling reveals gene networks and synthetic lethal interactions with oncogenic Ras. Cell 168, 890–903 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Hodges, L. M. et al. Very important pharmacogene summary: ABCB1 (MDR1, P-glycoprotein). Pharmacogenet. Genomics 21, 152–161 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. To, T. L. et al. A compendium of genetic modifiers of mitochondrial dysfunction reveals intra-organelle buffering. Cell 179, 1222–1238 e17 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Birsoy, K. et al. An essential role of the mitochondrial electron transport chain in cell proliferation is to enable aspartate synthesis. Cell 162, 540–551 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Garcia-Bermudez, J. et al. Aspartate is a limiting metabolite for cancer cell proliferation under hypoxia and in tumours. Nat. Cell Biol. 20, 775–781 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Garcia-Bermudez, J. et al. Publisher correction: aspartate is a limiting metabolite for cancer cell proliferation under hypoxia and in tumours. Nat. Cell Biol. 20, 1228 (2018).

    CAS  PubMed  Google Scholar 

  30. Sullivan, L. B. et al. Supporting aspartate biosynthesis is an essential function of respiration in proliferating cells. Cell 162, 552–563 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Sullivan, L. B. et al. Aspartate is an endogenous metabolic limitation for tumour growth. Nat. Cell Biol. 20, 782–788 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Mugabo, Y. et al. Identification of a mammalian glycerol-3-phosphate phosphatase: role in metabolism and signaling in pancreatic β-cells and hepatocytes. Proc. Natl Acad. Sci. USA 113, E430–E439 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Seifried, A. et al. Evolutionary and structural analyses of mammalian haloacid dehalogenase-type phosphatases AUM and chronophin provide insight into the basis of their different substrate specificities. J. Biol. Chem. 289, 3416–3431 (2014).

    CAS  PubMed  Google Scholar 

  34. Possik, E., Madiraju, S. R. M. & Prentki, M. Glycerol-3-phosphate phosphatase/PGP: role in intermediary metabolism and target for cardiometabolic diseases. Biochimie 143, 18–28 (2017).

    CAS  PubMed  Google Scholar 

  35. Prentki, M. & Madiraju, S. R. Glycerolipid/free fatty acid cycle and islet β-cell function in health, obesity and diabetes. Mol. Cell. Endocrinol. 353, 88–100 (2012).

    CAS  PubMed  Google Scholar 

  36. Collard, F. et al. A conserved phosphatase destroys toxic glycolytic side products in mammals and yeast. Nat. Chem. Biol. 12, 601–607 (2016).

    CAS  PubMed  Google Scholar 

  37. Bensaad, K. et al. TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell 126, 107–120 (2006).

    CAS  PubMed  Google Scholar 

  38. Ash, D. E., Goodhart, P. J. & Reed, G. H. ATP-dependent phosphorylation of α-substituted carboxylic acids catalyzed by pyruvate kinase. Arch. Biochem. Biophys. 228, 31–40 (1984).

    CAS  PubMed  Google Scholar 

  39. DeBerardinis, R. J. & Chandel, N. S. Fundamentals of cancer metabolism. Sci. Adv. 2, e1600200 (2016).

    PubMed  PubMed Central  Google Scholar 

  40. Seo, B. B., Matsuno-Yagi, A. & Yagi, T. Modulation of oxidative phosphorylation of human kidney 293 cells by transfection with the internal rotenone-insensitive NADH-quinone oxidoreductase (NDI1) gene of Saccharomyces cerevisiae. Biochim. Biophys. Acta 1412, 56–65 (1999).

    CAS  PubMed  Google Scholar 

  41. Wheaton, W. W. et al. Metformin inhibits mitochondrial complex I of cancer cells to reduce tumorigenesis. Elife 3, e02242 (2014).

    PubMed  PubMed Central  Google Scholar 

  42. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    PubMed  Google Scholar 

  43. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal 6, pl1 (2013).

    PubMed  PubMed Central  Google Scholar 

  44. Nagy, A., Lanczky, A., Menyhart, O. & Gyorffy, B. Validation of miRNA prognostic power in hepatocellular carcinoma using expression data of independent datasets. Sci. Rep. 8, 9227 (2018).

    PubMed  PubMed Central  Google Scholar 

  45. Nagy, A., Lanczky, A., Menyhart, O. & Gyorffy, B. Author correction: validation of miRNA prognostic power in hepatocellular carcinoma using expression data of independent datasets. Sci. Rep. 8, 11515 (2018).

    PubMed  PubMed Central  Google Scholar 

  46. Gerin, I. et al. Phosphoglycolate has profound metabolic effects but most likely no role in a metabolic DNA response in cancer cell lines. Biochem. J. 476, 629–643 (2019).

    CAS  PubMed  Google Scholar 

  47. Colic, M. et al. Identifying chemogenetic interactions from CRISPR screens with drugZ. Genome Med 11, 52 (2019).

    PubMed  PubMed Central  Google Scholar 

  48. Hart, T., Brown, K. R., Sircoulomb, F., Rottapel, R. & Moffat, J. Measuring error rates in genomic perturbation screens: gold standards for human functional genomics. Mol. Syst. Biol. 10, 733 (2014).

    PubMed  PubMed Central  Google Scholar 

  49. Hart, T. & Moffat, J. BAGEL: a computational framework for identifying essential genes from pooled library screens. BMC Bioinf. 17, 164 (2016).

    Google Scholar 

  50. Sanjana, N. E., Shalem, O. & Zhang, F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods 11, 783–784 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Wan, L. C. et al. Reconstitution and characterization of eukaryotic N6-threonylcarbamoylation of tRNA using a minimal enzyme system. Nucleic Acids Res. 41, 6332–6346 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Hesketh, G. G., Youn, J. Y., Samavarchi-Tehrani, P., Raught, B. & Gingras, A. C. Parallel exploration of interaction space by BioID and affinity purification coupled to mass spectrometry. Methods Mol. Biol. 1550, 115–136 (2017).

    CAS  PubMed  Google Scholar 

  53. Branon, T. C. et al. Efficient proximity labeling in living cells and organisms with TurboID. Nat. Biotechnol. 36, 880–887 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Aregger, M. et al. Systematic mapping of genetic interactions for de novo fatty acid synthesis identifies C12orf49 as a regulator of lipid metabolism. Nat. Metab. 2, 499–513 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Burke, D., Dawson, D. & Stearns, T. Methods in Yeast Genetics: a Cold Spring Harbor Laboratory Course Manual (Cold Spring Harbor Laboratory Press, 2000).

  56. van Leeuwen, J., Andrews, B., Boone, C. & Tan, G. Rapid and efficient plasmid construction by homologous recombination in yeast. Cold Spring Harb. Protoc. 2015, pdb.prot085100 (2015).

    PubMed  Google Scholar 

Download references

Acknowledgements

We thank the Donnelly sequencing center for assistance with sequencing. We thank the flow cytometry facility at the Temerty Faculty of Medicine for providing the BD LSR-Fortessa cell analyzer to assess mitochondrial membrane potential and drug efflux. We thank the Microscopy Imaging Laboratory at the Temerty faculty of medicine for providing the Zeiss LSM 880 for imaging experiments. We thank the SPARC BioCentre at Sickkids for assistance with mitochondrial stress test experiments. We thank Agios for assistance with metabolite profiling experiments. We thank the NBCC Proteomics Facility for assistance with mass spectrometry. Funds for this study were provided by CIHR to J.M. (PJT-463531) and the Ontario Research Fund RE9 Program to J.M. and C.B. J.M was a Canada Research Chair in Functional Genomics of Cancer and the Anne and Max Tanenbaum Chair in Molecular Medicine in the Temerty Faculty of Medicine at the University of Toronto.

Author information

Authors and Affiliations

Authors

Contributions

S.N.M., M.C., M.A., X.Z., P.M., and D.A.P. performed experiments and analyzed data. S.N.M., M.C., D.A.P., O.Z., J.R.M.-B., and G.A.S. helped perform and analyze the metabolomics experiments. G.T. and C.B. performed yeast experiments. M.A., Z.-Y.L., C.J.W., S.N.M., J.M. and A.-C.G. helped perform and analyze BioID experiments. J.M. supervised the project. S.N.M., M.C., P.M. and J.M. made the figures and wrote the manuscript with input from all co-authors.

Corresponding author

Correspondence to Jason Moffat.

Ethics declarations

Competing interests

J.M. is an advisor and shareholder of Century Therapeutics and Aelian Biotechnology.

Peer review

Peer review information

Nature Chemical Biology thanks Navdeep Chandel and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Fitness assays reveal a role for mitochondrial respiration following pyrvinium treatment.

(a) The effect of pyrvinium or LGK974 on cell viability at the indicated concentrations after a 5-day treatment in HPAF-II cells cultured in standard cell growth medium. (b) The effect of phenformin on cell viability at the indicated concentrations after a 5-day treatment in HAP1 cells cultured in either glucose or galactose containing medium. The effect of pyrvinium on cell viability at the indicated concentrations after a 5-day treatment in (c) HCT116 or (d) A375 cells cultured in either glucose or galactose containing medium. Viability was measured using Alamar blue staining at the end point. All measurements are relative to untreated cells. All data are represented as means ± SD (n = 3 replicates).

Extended Data Fig. 2 Validation of GOT1 as a sensitizer upon pyrvinium treatment in Cas9-expressing cells.

Cas9 expressing clonal (a) HAP1, (b) HCT116, or (c) A375 cell lines infected with lentiviruses carrying expression cassettes for non-targeting LacZ or different GOT1 gRNAs, as indicated. After puromycin selection, cells were seeded for a proliferation assay and treated with pyrvinium at the indicated doses. Cells were manually counted after 5 days of drug treatment. (d, e, f) cell viability of clonal KO’s of GOT1, PGP, and parental HAP1 cells assessed under pyrvinium treatment in limiting growth conditions. Cells were seeded in (d) minimal medium (MM), (e) MM supplemented with 10 mM aspartate, or (f) MM supplemented with 1 mM alpha-ketobutyrate (AKB). Cell viability was measured using Alamar blue after 5 days of treatement. All measurements are relative to untreated cells and data are represented as means ± SD (n = 3 replicates). (g) HAP1, (h) HCT116, or (i) A375 cell lines infected with lentiviruses expressing both Cas9 and a gRNAs targeting LacZ, the targeting control locus AAVS1, or PGP. After puromycin selection, cells were seeded for a proliferation assay and treated with pyrvinium at the indicated doses. Viability was measured using Alamar blue staining at the end point. All data are represented as means ± SD (n = 3 replicates). (j) Schematic depicting the function of PGP in glycerophospholipid/free fatty (GL/FFA) cycle (left) and as a metabolite repair enzyme in glycolysis (middle), and the pentose phosphate pathway (PPP) (right). Cell viability assay showing the effect of indicated doses of pyrvinium on (k) HAP1 parental, (l) PGPΔ-C1 and (m) PGPΔ-C2 cells after a 4-day pyrvinium treatment in different media conditions supplemented with indicated concentrations of glycerol. Viability was measured using Alamar blue staining. All measurements are relative to untreated cells. All data are represented as means ± SD (n = 3 replicates). (n) Levels of F-1,6-BP measured by LC-MS metabolite profiling of HAP1, PGPΔ, and GOT1Δ cells in the presence or absence of pyrvinium. Two-way ANOVA, **p < 0.001, ***p < 0.0001. Measurements consist of 3 biological replicates. Data represented as relative to untreated HAP1 parental cells as means ± SD. G-6-P, glucose-6-phosphate; F-6-P, frcutose-6-phosphate; F-1,6-BP, fructose-1,6-bisphosphate; F-2,6-BP, fructose-2,6-bisphosphate; G-3-P, glyceraldehyde-3-phosphate; DHAP, dihydroxyacetonephosphate; 1,3-BP-glycerate, 1,3-biphosphoglycerate; 3-PG, 3-phosphoglycerate; 2-PG, 2-phosphoglycerate; PEP, phosphoenolpyruvate; Glycerol-3-P, glycerol-3-phosphate; 2-P-lactate, 2-phospho-L-lactate; FA-CoA, fatty acyl coenzyme A; GL/FFA, glycerolipid/free fatty acid; G6PD, glucose 6-phosphate dehydrogenase; PFK2, phosphofructokinase 2; 6PGL, 6-phosphoglucolactonase; 6PGDH, 6-phosphogluconate dehydrogenase; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; PK, pyruvate kinase; GK, glycerokinase; Acyl-pase, acylphosphatase.

Extended Data Fig. 3 Validation of PGP as a sensitizer upon pyrvinium treatment in Cas9-expressing cells.

(a) Schematic of different metabolic pathways required for cancer cell growth. Relative abundance of indicated metabolites measured by LC/MS in glycolysis, (b) Essential amino acids, (c) Non-essential amino acids in parental HAP1 cells untreated (black) or treated (gray) with 100 nM pyrvinium for 5 hours. All measurements are relative to untreated parental HAP1 cells. All data are represented as means ± SD (n = 3 replicates). *p < 0.05; two-tailed unpaired t-test. (d) schematic showing the effect of different mitochondrial respiratory complex inhibitors. (e-j) Cell viability assays of parental, PGPΔ and GOT1Δ cells treated with indicated doses of different mitochondrial inhibitors including (e) rotenone, (f) phenformin, (g) metformin, (h) antimycin, (i) sodium azide and (j) oligomycin. Rotenone, metformin and phenformin are complex I inhibitors, antimycin inhibits complex III, sodium azide inhibits complex IV and oligomycin inhibits complex V or ATP synthase. Viability was measured using Alamar Blue staining. All measurements are relative to untreated cells. All data are represented as means ± SD (n = 3 replicates). (k, l) Cell viability assays in HCT116 parental and HCT116 NDI1 overexpressing cells (top panel) across indicated doses of either phenformin or pyrvinium. Cell viability assays in PGPΔ cells, as well as PGPΔ cells expressing NDI1 across indicated doses of phenformin or pyrvinium. All measurements presented as relative to DMSO control assessed by alamar blue (n = 4 biological replicates times 3 technical replicates).

Source data

Extended Data Fig. 4 C1orf115 mediates resistance to pyrvinium.

(a) Schematic of C1orf115 protein. (b) Top alteration frequencies of the C1orf115 gene across cancer types retrieved from cBioportal using the TCGA PanCan data. (c) and (d) show Kaplan Meier plots for clear cell renal carcinoma (ccRCC) and ovarian cancer patient outcomes predicted by C1orf115 expression. (e) HAP1-Cas9 cells were transduced with lentiviruses expressing the indicated gRNAs targeting C1orf115. After puromycin selection, cells were seeded for a viability assay and treated with pyrvinium at the indicated doses. Relative abundance of (f) lactate and (g) pyruvate in parental and C1orf115 knockout cells in the presence or absence of 100 nM pyrvinium treatment for 5-hours. (h, i) Clonal C1orf115- KO cell lines were subjected to different doses of (h) phenformin, (i) metformin, and (j) antimycin for 5 days. C1orf115-V5 overexpression did not rescue the effects of antimycin on cell viability. (k) Cell viability of C1orf115-KO cell lines assessed in either glucose or galactose media conditions with pyrvinium treatment. Cell viability was measured using Alamar Blue staining at the end point. All measurements are relative to untreated cells. All data are represented as means ± SD (n = 3 replicates). p < 0.05; two-tailed unpaired t test.

Source data

Extended Data Fig. 5 Functional enrichment of BioID data.

(a) Bar plot illustrating the cellular compartments for preys captured by either BioID2 (gray) or mini-Turbo (black) C-terminal tagged C1orf115 HAP1 cells.

Source data

Extended Data Fig. 6 ABCC1 localization in relation to C1orf115 expression.

(a) ABCC1 localization (green) and DAP1 visualizing nuclei (blue) across genetic backgrounds (HAP1 parental, C1orf115/RDD1 KO clones C10, C3, C1orf115-V5 overexpression, and ABCC1 KO cells). In contrast to ABCB1 localization, no apparent changes in localization are evident for ABCC1 across C1orf115 genetic backgrounds. (b) FACs results of drug efflux of rhodamine-123, data representative of two independent biological replicates. Light gray bars depict drug load at 4 °C, green bars depict drug efflux at 37 °C. Results illustrate that C1orf115 attenuates drug efflux.

Extended Data Fig. 7 Quantitative measurements of drug efflux.

(a) Gating strategy applied to percentage efflux measurements shown in Fig. 6d. Data is representative of 3 independent biological replicates measuring DiOC(2)3 drug efflux. (b) Barplot representing percent drug efflux of DiOC(2) 3 in either 4 °C or 37 °C conditions across cell lines. (c) Gating strategy for quantifying drug efflux.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Masud, S.N., Chandrashekhar, M., Aregger, M. et al. Chemical genomics with pyrvinium identifies C1orf115 as a regulator of drug efflux. Nat Chem Biol 18, 1370–1379 (2022). https://doi.org/10.1038/s41589-022-01109-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-022-01109-0

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research