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:

Targeting p97–Npl4 interaction inhibits tumor Treg cell development to enhance tumor immunity

Abstract

Targeting tumor-infiltrating regulatory T (TI-Treg) cells is a potential strategy for cancer therapy. The ATPase p97 in complex with cofactors (such as Npl4) has been investigated as an antitumor drug target; however, it is unclear whether p97 has a function in immune cells or immunotherapy. Here we show that thonzonium bromide is an inhibitor of the interaction of p97 and Npl4 and that this p97–Npl4 complex has a critical function in TI-Treg cells. Thonzonium bromide boosts antitumor immunity without affecting peripheral Treg cell homeostasis. The p97–Npl4 complex bridges Stat3 with E3 ligases PDLIM2 and PDLIM5, thereby promoting Stat3 degradation and enabling TI-Treg cell development. Collectively, this work shows an important role for the p97–Npl4 complex in controlling Treg–TH17 cell balance in tumors and identifies possible targets for immunotherapy.

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: TB disrupts p97–Npl4 interaction and suppresses tumor growth.
Fig. 2: TB boosts CD8+ T cell antitumor immunity.
Fig. 3: The antitumor effect of TB is dependent on T cells.
Fig. 4: Blocking the p97–Npl4 interaction skews the Treg–TH17 cell balance in the TME.
Fig. 5: Targeting p97–Npl4 interface does not disrupt Treg–TH17 balance outside the TME.
Fig. 6: Blocking the p97–Npl4 interaction inhibits infiltration and differentiation of TI-Treg cells.
Fig. 7: The p97–Npl4 complex promotes PDLIM2/PDLIM5-mediated protein degradation of Stat3.

Similar content being viewed by others

Data availability

All high-throughput sequencing data have been deposited in the Gene Expression Omnibus database under the accession code GSE240681. Source data are provided with this paper.

References

  1. Jarosch, E. et al. Protein dislocation from the ER requires polyubiquitination and the AAA-ATPase Cdc48. Nat. Cell Biol. 4, 134–139 (2002).

    Article  CAS  PubMed  Google Scholar 

  2. Ye, Y., Meyer, H. H. & Rapoport, T. A. The AAA ATPase Cdc48/p97 and its partners transport proteins from the ER into the cytosol. Nature 414, 652–656 (2001).

    Article  CAS  PubMed  Google Scholar 

  3. Heo, J. M. et al. A stress-responsive system for mitochondrial protein degradation. Mol. Cell 40, 465–480 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Kondo, H. et al. p47 is a cofactor for p97-mediated membrane fusion. Nature 388, 75–78 (1997).

    Article  CAS  PubMed  Google Scholar 

  5. Krastev, D. B. et al. The ubiquitin-dependent ATPase p97 removes cytotoxic trapped PARP1 from chromatin. Nat. Cell Biol. 24, 62–73 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Mouysset, J. et al. Cell cycle progression requires the CDC-48UFD-1/NPL-4 complex for efficient DNA replication. Proc. Natl Acad. Sci. USA 105, 12879–12884 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Ju, J. S. et al. Valosin-containing protein (VCP) is required for autophagy and is disrupted in VCP disease. J. Cell Biol. 187, 875–888 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Higashiyama, H. et al. Identification of ter94, Drosophila VCP, as a modulator of polyglutamine-induced neurodegeneration. Cell Death Differ. 9, 264–273 (2002).

    Article  CAS  PubMed  Google Scholar 

  9. Meyer, H., Bug, M. & Bremer, S. Emerging functions of the VCP/p97 AAA-ATPase in the ubiquitin system. Nat. Cell Biol. 14, 117–123 (2012).

    Article  CAS  PubMed  Google Scholar 

  10. Richly, H. et al. A series of ubiquitin binding factors connects CDC48/p97 to substrate multiubiquitylation and proteasomal targeting. Cell 120, 73–84 (2005).

    Article  CAS  PubMed  Google Scholar 

  11. Ciechanover, A. The ubiquitin-proteasome proteolytic pathway. Cell 79, 13–21 (1994).

    Article  CAS  PubMed  Google Scholar 

  12. Kilgas, S. & Ramadan, K. Inhibitors of the ATPase p97/VCP: from basic research to clinical applications. Cell Chem. Biol. 30, 3–21 (2023).

    Article  CAS  PubMed  Google Scholar 

  13. Anderson, D. J. et al. Targeting the AAA ATPase p97 as an approach to treat cancer through disruption of protein homeostasis. Cancer Cell 28, 653–665 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Roux, B. et al. Targeting acute myeloid leukemia dependency on VCP-mediated DNA repair through a selective second-generation small-molecule inhibitor. Sci. Transl. Med. 13, eabg1168 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Tay, C., Tanaka, A. & Sakaguchi, S. Tumor-infiltrating regulatory T cells as targets of cancer immunotherapy. Cancer Cell 41, 450–465 (2023).

    Article  CAS  PubMed  Google Scholar 

  16. Kamada, T. et al. PD-1+ regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer. Proc. Natl Acad. Sci. USA 116, 9999–10008 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Saito, T. et al. Two FOXP3+CD4+ T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat. Med. 22, 679–684 (2016).

    Article  CAS  PubMed  Google Scholar 

  18. Hao, Q. et al. A non-canonical role of the p97 complex in RIG-I antiviral signaling. EMBO J. 34, 2903–2920 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Rubtsov, Y. P. et al. Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 28, 546–558 (2008).

    Article  CAS  PubMed  Google Scholar 

  20. Sharma, P., Hu-Lieskovan, S., Wargo, J. A. & Ribas, A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 168, 707–723 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Geng, J. et al. The transcriptional coactivator TAZ regulates reciprocal differentiation of TH17 cells and Treg cells. Nat. Immunol. 18, 800–812 (2017).

    Article  CAS  PubMed  Google Scholar 

  23. Laurence, A. et al. STAT3 transcription factor promotes instability of nTreg cells and limits generation of iTreg cells during acute murine graft-versus-host disease. Immunity 37, 209–222 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Withers, D. R. et al. Transient inhibition of ROR-γt therapeutically limits intestinal inflammation by reducing TH17 cells and preserving group 3 innate lymphoid cells. Nat. Med. 22, 319–323 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Solt, L. A. & Burris, T. P. Action of RORs and their ligands in (patho)physiology. Trends Endocrinol. Metab. 23, 619–627 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Poria, D. K., Sheshadri, N., Balamurugan, K., Sharan, S. & Sterneck, E. The STAT3 inhibitor Stattic acts independently of STAT3 to decrease histone acetylation and modulate gene expression. J. Biol. Chem. 296, 100220 (2021).

    Article  CAS  PubMed  Google Scholar 

  27. Roux, K. J., Kim, D. I., Raida, M. & Burke, B. A promiscuous biotin ligase fusion protein identifies proximal and interacting proteins in mammalian cells. J. Cell Biol. 196, 801–810 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Harhouri, K. et al. MG132-induced progerin clearance is mediated by autophagy activation and splicing regulation. EMBO Mol. Med. 9, 1294–1313 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Tanaka, T. et al. PDLIM2 inhibits T helper 17 cell development and granulomatous inflammation through degradation of STAT3. Sci. Signal. 4, ra85 (2011).

    Article  PubMed  Google Scholar 

  30. Shi, Y. et al. PDLIM5 inhibits STUB1-mediated degradation of SMAD3 and promotes the migration and invasion of lung cancer cells. J. Biol. Chem. 295, 13798–13811 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhou, H. J. et al. Discovery of a first-in-class, potent, selective, and orally bioavailable inhibitor of the p97 AAA ATPase (CB-5083). J. Med. Chem. 58, 9480–9497 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Sharma, A. et al. Anti-CTLA-4 immunotherapy does not deplete FOXP3+ regulatory T cells (Tregs) in human cancers. Clin. Cancer Res. 25, 1233–1238 (2019).

    Article  CAS  PubMed  Google Scholar 

  33. Kryczek, I. et al. Phenotype, distribution, generation, and functional and clinical relevance of Th17 cells in the human tumor environments. Blood 114, 1141–1149 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Sfanos, K. S. et al. Phenotypic analysis of prostate-infiltrating lymphocytes reveals TH17 and Treg skewing. Clin. Cancer Res. 14, 3254–3261 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Massara, M. et al. ACKR2 in hematopoietic precursors as a checkpoint of neutrophil release and anti-metastatic activity. Nat. Commun. 9, 676 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Oguma, K. et al. Activated macrophages promote Wnt signalling through tumour necrosis factor-alpha in gastric tumour cells. EMBO J. 27, 1671–1681 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kryczek, I., Wei, S., Szeliga, W., Vatan, L. & Zou, W. Endogenous IL-17 contributes to reduced tumor growth and metastasis. Blood 114, 357–359 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Kryczek, I. et al. Human TH17 cells are long-lived effector memory cells. Sci. Transl. Med. 3, 104ra100 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Muranski, P. et al. Th17 cells are long lived and retain a stem cell-like molecular signature. Immunity 35, 972–985 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Paulos, C. M. et al. The inducible costimulator (ICOS) is critical for the development of human TH17 cells. Sci. Transl. Med. 2, 55ra78 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Martin-Orozco, N. et al. T helper 17 cells promote cytotoxic T cell activation in tumor immunity. Immunity 31, 787–798 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Tuovinen, H., Kekäläinen, E., Rossi, L. H., Puntila, J. & Arstila, T. P. Cutting edge: human CD4CD8 thymocytes express FOXP3 in the absence of a TCR. J. Immunol. 180, 3651–3654 (2008).

    Article  CAS  PubMed  Google Scholar 

  43. Liu, G. et al. Direct detection of FoxP3 expression in thymic double-negative CD4CD8 cells by flow cytometry. Sci. Rep. 4, 5781 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Barra, M. M., Richards, D. M., Hofer, A. C., Delacher, M. & Feuerer, M. Premature expression of Foxp3 in double-negative thymocytes. PLoS ONE 10, e0127038 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Mays, L. E. & Chen, Y. H. Maintaining immunological tolerance with Foxp3. Cell Res. 17, 904–918 (2007).

    Article  CAS  PubMed  Google Scholar 

  46. Lin, L. et al. Oleic acid availability impacts thymocyte preprogramming and subsequent peripheral Treg cell differentiation. Nat. Immunol. 25, 54–65 (2024).

    Article  CAS  PubMed  Google Scholar 

  47. Sun, Y. et al. The mediator subunit Med23 contributes to controlling T-cell activation and prevents autoimmunity. Nat. Commun. 5, 5225 (2014).

  48. Tang, Y. et al. Selective inhibition of STRN3-containing PP2A phosphatase restores hippo tumor-suppressor activity in gastric cancer. Cancer Cell 38, 115–128 (2020).

Download references

Acknowledgements

We thank Y. Shi from Soochow University, B. Li from Shanghai Jiao Tong University, and Y. Wang and X. Liu from University of Chinese Academy of Sciences for their helpful discussion. This work was supported by the National Key R&D Program of China (2020YFA0803200), National Natural Science Foundation of China grants (82150112, 82303148, 92168116, 82360502, 82060542, 31930026, 82361168638, 82372613, 82373251 and 82222052), China Postdoctoral Science Foundation (2023M742656), Yunnan Province’s Ten Thousand Talents Plan for Top Young Talents (YNWR-QNBJ-2019-275), Xingdian Talents and Famous Doctors Special Project (XDYC-YLWS-2023-0029) and Shanghai Super Postdoctoral Incentive Program.

Author information

Authors and Affiliations

Authors

Contributions

P.N. and R.Y. performed functional experiments. Z.C. performed biochemical experiments. Y.M. conducted scRNA-seq analysis. X.J. and S.W. performed genotyping of mice. B.L., C.D., L.T., C.L., Y.X., L.A., Y.P., Y.Z., W.Z., H.Z., Z.T., M.Z., Y.H. and W.W. discussed and analyzed the results. P.N., S.J. and Z.Z. designed the experiments and wrote the manuscript. S.J. and Z.Z. supervised the project.

Corresponding authors

Correspondence to Shi Jiao or Zhaocai Zhou.

Ethics declarations

Competing interests

Z.Z. (CN201510362643.5, CN201510359075.3, CN201910753411.0, CN2024108876845), S.J. (CN201510362643.5, CN201510359075.3, CN201910753411.0, CN2024108876845), P.N. (CN201910753411.0) and W.W. (CN201910753411.0) have filed patents regarding the potential application of TB in cancer therapy. The other authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Nick Bernard, in collaboration with the Nature Immunology team. Peer reviewer reports are available.

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 TB displayed a broader antitumor therapeutic window than CB-5083.

a, Summary of p97-related inhibitors. b, Strep pulldown assay to assess the interaction between p97 and Npl4. Data shown are representative results of two independent experiments. c, Co-IP assay to detect the interaction of p97 with Npl4. Data shown are representative results of two independent experiments. d, Inhibition of cell viability in HGC-27 cells. Data shown are representative results of two independent experiments. e, Inhibition of cell viability in GES-1 cells. Data shown are representative results of two independent experiments. f, mRNA levels of CHOP in the indicated cells that had been treated with TB (upper) or CB-5083 (lower). Data shown are representative results of two independent experiments. g, Schematic illustration of the tumor graft model and treatment with compounds. h, Experimental flow chart of a mouse model for MNNG/H. pylori-induced GC and the treatment with TB, CB-5083 or 5-FU. i, Therapeutic windows and tumor growth inhibition abilities of TB and CB-5083. IC50 values of the compounds are shown. j, Tumor weights of tumor graft models after treatment with TB (intragastrically administered). n = 6 biologically independent samples examined over three independent experiments. k, Tumor weights for MC38-bearing mice that had been treated with TB (intravenously injected, i.v.). n = 4 biologically independent samples examined over two independent experiments. l, Body weights of TB-treated (intraperitoneal administration) mice and control group. n = 4 biologically independent samples examined over two independent experiments. m, Body weights of TB-treated (i.v.) mice and control group. n = 4 biologically independent samples examined over two independent experiments. n, Body weights of TB-treated (25mg/kg, i.v.) mice and control group. n = 6 biologically independent samples examined over two independent experiments. o, Whole blood cell routine test of TB-treated mice and control group from panel n. n = 6 biologically independent samples examined over two independent experiments. Data shown as means ± s.d. (d-f) or means ± s.e.m. (j,k,l,m and n). Data were analyzed for significant differences by performing a one-way ANOVA with Dunnett’s post hoc analysis (f, j and k), or two-way ANOVA with Dunnett’s post hoc analysis (l,m), or two-tailed, unpaired Student’s t-tests (n).

Source data

Extended Data Fig. 2 TB treatment promoted the effector function of CD8+ T cells.

a, Apoptosis scores of CD8+ (left) and CD4+ T cells (right) based on the expression of T cell apoptosis-correlated genes from the indicated group. b, Violin plot showing the expression of genes correlated to CD8+ T cell effector function in the indicated group. c, Heatmap showing the expression of genes correlated to CD4+ T cell effector function in the indicated group. d, Flow cytometry analysis showing the proportions of CD4+ and CD8+ T cells among the tumor-infiltrated CD45+ cells from Ctrl and TB-treated mice. e, Proportions (left) and absolute numbers (right) of tumor-infiltrated CD4+ and CD8+ T cells per milligram tumor from Ctrl and TB-treated mice. n = 4 biologically independent samples examined over two independent experiments. f, Gating strategy for tumor-infiltrated CD8+ T cells and representative flow cytometry plots of unstimulated samples of the indicated cytokine-producing CD8+ T cells and samples of these cells stimulated with PMA and ionomycin. g, Proportions of tumor-infiltrated IFNγ+ CD8+ T cells per milligram tumor from Ctrl and TB-treated mice. n = 6 biologically independent samples examined over two independent experiments. Data shown as means ± s.e.m. (e,g). Data were analyzed for significant differences by performing two-tailed, unpaired Student’s t-tests (a), or one-way ANOVA with Dunnett’s post hoc analysis (b,e,g).

Source data

Extended Data Fig. 3 Mutational analysis of the p97-Npl4 complex in T cells.

a, Flow cytometry analysis of CD4+ and CD8+ T cell subpopulations in thymuses and spleens from Vcp3A/3A and Vcp3A/3A CD4Cre mice. b, Flow cytometry analysis showing the expression levels of GzmB and IFNγ in CD8+ T cells isolated from spleens of Vcp3A/3A or Vcp3A/3A CD4Cre mice. CD8+ T cells were sorted from spleens stimulated with anti-CD3 (2 μg/mL) and anti-CD28 (1 μg/mL) for the indicated times, and cytokine expression was determined by performing flow cytometry. c, Tumor growth curves of MC38-derived subcutaneous tumors from, respectively, Vcp3A/3A and Vcp3A/3A CD4Cre mice. n = 6 biologically independent samples examined over two independent experiments. d, Percentages of CD4+ and CD8+ T cells among the tumor-infiltrated CD45+ cells for the indicated mice. n = 4 biologically independent samples examined over two independent experiments. e, Frequencies of CD8+GzmB+, CD8+IFNγ+ and CD8+TNF+ cells among tumor-infiltrated CD8+ T cells. n = 4 biologically independent samples examined over two independent experiments. f, Number of tumor-infiltrated CD45.2+ Treg, Th17, Th1 and Th2 cells per milligram tumor from CD45.1 mice that were adoptively transferred with vector (Ctrl) or Flag-Vcp 3A lentivirus-infected (Vcp3A) CD45.2+ CD3+ T cells. n = 4 biologically independent samples examined over two independent experiments. Data shown as means ± s.e.m. (c-f). Data were analyzed for significant differences by performing two-way ANOVA with Dunnett’s post hoc analysis (c), or two-tailed, unpaired Student’s t-test (d-f).

Source data

Extended Data Fig. 4 Targeting the p97-Npl4 interaction skewed Treg-Th17 cell balance.

a, Gating strategy for TI-Treg and Th17 cells gated from tumor-infiltrated CD4+ T cells, with a fluorescence minus one (FMO) sample used as negative control. b, Absolute numbers of intratumoral Treg (TI-Treg, Foxp3+) and Th17 cells (IL17a+) per milligram tumor from tumor-bearing control (Ctrl) and TB-intraperitoneally (i.p.)-injected mice. n = 4 biologically independent samples examined over two independent experiments. c, Absolute numbers of TI-Treg and Th17 cells per milligram tumor from tumor-bearing Ctrl and TB-intravenously (i.v.)-injected mice. n = 4 biologically independent samples examined over two independent experiments. d, Representative flow cytometry plots and percentages of TI-Treg cells from tumor-bearing Ctrl or TB-intratumorally (i.t.)-injected mice. n = 4 biologically independent samples examined over two independent experiments. e, Absolute number of TI-Treg cells per milligram tumor in each of the indicated mice. n = 4 biologically independent samples examined over two independent experiments. f, Representative plots and percentages of tumor-infiltrated Th17 cells from tumor-bearing Ctrl or TB-intratumorally (i.t.)-injected mice. n = 4 biologically independent samples examined over two independent experiments. g, Absolute number of tumor-infiltrated Th17 cells per milligram tumor in each of the indicated mice. n = 4 biologically independent samples examined over two independent experiments. h, Schematic illustration of the iTreg-cell-transfer assay procedure. CD45.1 tumor-bearing mice were intravenously injected with Vcp3A/3A iTreg (WT) or Vcp3A/3A Foxp3Cre (3A) iTreg cells. i, Flow cytometry analysis of the purity of iTreg cells derived from WT and 3A mice for adoptive transfer assay. j, Absolute numbers of TI-Treg cells per milligram tumor from tumor-bearing Vcp3A/3A, Foxp3Cre and Vcp3A/3A Foxp3Cre mice. n = 10 biologically independent samples examined over four independent experiments. k, Absolute numbers of intratumoral Th17 cells per milligram tumor from tumor-bearing Vcp3A/3A, Foxp3Cre and Vcp3A/3AFoxp3Cre mice. n = 10 biologically independent samples examined over four independent experiments. l, Absolute numbers of tumor-infiltrated IFNγ+CD8+ T cells per milligram tumor from tumor-bearing Vcp3A/3A, Foxp3Cre and Vcp3A/3AFoxp3Cre mice. n = 10 biologically independent samples examined over four independent experiments. Data shown as means ± s.e.m. (b-g,j-l). Data were analyzed for significant differences by performing one-way ANOVA with Dunnett’s post hoc analysis (b–g) or two-tailed, unpaired Student’s t-test (j–l).

Source data

Extended Data Fig. 5 Targeting p97-Npl4 interaction did not disrupt immune homeostasis.

a, Flow cytometry analysis showing the proportions of Treg cells (Foxp3+) and Th17 cells (IL17a+RORγt+) among CD4+ T cells in lungs from Vcp3A/3A, Foxp3Cre and Vcp3A/3A Foxp3Cre mice. b, Flow cytometry analysis showing the proportions of Treg cells (Foxp3+) and Th17 cells (IL17a+RORγt+) among CD4+ T cells in the colon from Vcp3A/3A, Foxp3Cre and Vcp3A/3A Foxp3Cre mice. c, The proportions of CD44hiCD62Llow subpopulation among the CD4+ or CD8+ T cells from spleens and lymph nodes of Vcp3A/3A, Foxp3Cre and Vcp3A/3A Foxp3Cre mice. n = 5 biologically independent samples examined over two independent experiments. d, Absolute numbers of Treg (Foxp3+) and Th17 cells (IL17a+) in draining lymph nodes (dLN) from Ctrl and TB-treated mice. n = 4 biologically independent samples examined over two independent experiments. e, Absolute numbers of Treg (Foxp3+) and Th17 cells (IL17a+) in lungs from Ctrl and TB-treated mice. n = 4 biologically independent samples examined over two independent experiments. f, Absolute numbers of Treg (Foxp3+) and Th17 cells (IL17a+) in colons from Ctrl and TB-treated mice. n = 4 biologically independent samples examined over two independent experiments. Data shown as means ± s.e.m. (c-f). Data were analyzed for significant differences by performing one-way ANOVA with Dunnett’s post hoc analysis (c–f).

Source data

Extended Data Fig. 6 Target the p97-Npl4 interaction did not affect Treg cell stability and suppressive function.

a, Apoptosis scores of TI-Treg cells from Ctrl (grey) and TB (red)-treated groups. In each case, the apoptosis score was calculated based on the expression profile of the apoptosis-correlated transcriptomic signature. b, CTV proliferation analysis of iTreg (upper) and Th17 (lower) cells upon TB treatment at the indicated concentrations. Data shown are representative results from two independent experiments. c, Representative plots (left) and percentages (right) of Foxp3+ cells, IL17a+ cells and Foxp3+ IL17a+ cells among total tumor-infiltrating CD4+ T cells from Vcp3A/3A and Vcp3A/3AFoxp3Cre mice. n = 4 biologically independent samples examined over two independent experiments. d, Ex vivo suppression of carboxyfluorescein succinimidyl ester (CFSE)-labeled WT naïve CD8+ T cell proliferation by iTreg cells derived from Vcp3A/3A and Vcp3A/3AFoxp3Cre mice with the indicated ratios of Treg to Teff cells. Teff, CD8+ T effector cells (n = 3 repeats/group). Data shown are representative results from two independent experiments. e, Heatmap of downregulated regulon activities in TI-Treg cells from Ctrl and TB-treated tumor bearing mice. f, Heatmap showing the transcription levels of Stat3 target genes in the TI-Treg cells from Ctrl and TB-treated tumor bearing mice. g, Flow cytometry analysis of induced Treg (iTreg, CD25+Foxp3+, upper) cells and Th17 (IL17a+, lower) cells in the CD4+ T cells after treatment with TB or a mixture of TB with the Stat3 inhibitor Stattic. Naïve CD4+ T cells were sorted from spleen tissues and then stimulated with anti-CD3 (10 μg/mL) and anti-CD28 (10 μg/mL/) under Treg or Th17 differentiation conditions together with TB (1 μM) or a mixture of TB (1 μM) with Stattic (1, 2 μM). Data shown as means ± s.e.m. (c). Data were analyzed for significant differences by performing two-tailed, unpaired Student’s t-test (a), or one-way ANOVA with Dunnett’s post hoc analysis (c).

Source data

Extended Data Fig. 7 Targeting the p97-Npl4 interaction inhibited ubiquitination of Stat3.

a, Western blot analysis of Stat3 in wildtype (WT) cells and that in p97 KO 293FT cells, each transfected with Flag-UB, Myc-Stat3, wildtype or its 3A mutant HA-p97. Data shown are representative results from two independent experiments. b, Immunoblotting of the indicated proteins in CD4+ T cells at different activation and differentiation stages, and in CD8+ T cells upon TCR stimulation. Data shown are representative results from two independent experiments. c, Schematic illustration of the knocking out of p97, Npl4 or PDLIM2/5 using the CRISPR-Cas9 system. d, Immunoblotting analysis showing the efficiency of the knockout of each of p97, Npl4 and PDLIM2/5 in CD4+ T cells. Data shown are representative results from two independent experiments. e, Proportion of the iTreg (Foxp3+) cells among the CD4+ T cells after p97, Npl4 or PDLIM2/5 deficiency (n = 3 repeats/group). Data shown are representative results from two independent experiments. f, Proportion of the Th17 (IL17a+RORγt+) cells among the CD4+ T cells after p97, Npl4 or PDLIM2/5 deficiency (n = 3 repeats/group). Data shown are representative results from two independent experiments. g, Co-IP analysis to detect the interaction between p97 and PDLIM5 in 293FT cells. Data shown are representative results from two independent experiments. h, Co-IP analysis to detect the interaction between p97 and PDLIM2 in 293FT cells. Data shown are representative results from two independent experiments. i, qPCR analysis to determine the knockdown efficiency of PDLIM2/5 siRNAs (n = 3 repeat/group). Data shown are representative results from two independent experiments. j, Flow cytometry analysis showing the proportions of iTreg cells (Foxp3+) and Th17 cells (IL17a+RORγt+) among the indicated CD4+ T cells that had been subjected to knockdown of PDLIM2 or PDLIM5 with their specific siRNAs (n = 3 repeats/group). Data shown are representative results from two independent experiments. Data shown as means ± s.d. (e,f,I and j). Data were analyzed for significant differences by performing two-tailed, unpaired Student’s t-test (e,f,I and j).

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1–3. Supplementary Table 1: List of primers for mouse genotyping. Supplementary Table 2: List of key resources. Supplementary Table 3: List of primers for qPCR.

Reporting Summary

Peer Review File

Source data

Source Data Fig. 1

Statistical source data and unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data and unprocessed western blots.

Source Data Fig. 4

Statistical source data and unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data and unprocessed western blots.

Source Data Extended Data Fig. 1

Statistical source data and unprocessed western blots.

Source Data Extended Data Fig. 2

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. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data and unprocessed western blots.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) 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

Nie, P., Cao, Z., Yu, R. et al. Targeting p97–Npl4 interaction inhibits tumor Treg cell development to enhance tumor immunity. Nat Immunol 25, 1623–1636 (2024). https://doi.org/10.1038/s41590-024-01912-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41590-024-01912-y

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer