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:

HOXB13 suppresses de novo lipogenesis through HDAC3-mediated epigenetic reprogramming in prostate cancer

Abstract

HOXB13, a homeodomain transcription factor, critically regulates androgen receptor (AR) activities and androgen-dependent prostate cancer (PCa) growth. However, its functions in AR-independent contexts remain elusive. Here we report HOXB13 interaction with histone deacetylase HDAC3, which is disrupted by the HOXB13 G84E mutation that has been associated with early-onset PCa. Independently of AR, HOXB13 recruits HDAC3 to lipogenic enhancers to catalyze histone deacetylation and suppress lipogenic regulators such as fatty acid synthase. Analysis of human tissues reveals that the HOXB13 gene is hypermethylated and downregulated in approximately 30% of metastatic castration-resistant PCa. HOXB13 loss or G84E mutation leads to lipid accumulation in PCa cells, thereby promoting cell motility and xenograft tumor metastasis, which is mitigated by pharmaceutical inhibition of fatty acid synthase. In summary, we present evidence that HOXB13 recruits HDAC3 to suppress de novo lipogenesis and inhibit tumor metastasis and that lipogenic pathway inhibitors may be useful to treat HOXB13-low PCa.

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: HOXB13 WT, but not G84E mutant, inhibits lipogenic programs in PCa.
Fig. 2: HOXB13 interacts with HDAC3 protein through its MEIS domain.
Fig. 3: HOXB13 recruits HDAC3 to catalyze histone deacetylation.
Fig. 4: HDAC3 is required for HOXB13-mediated suppression of de novo lipogenesis.
Fig. 5: HOXB13 is hypermethylated and downregulated in CRPC.
Fig. 6: HOXB13 loss promotes cell motility and PCa metastasis.
Fig. 7: Therapeutic targeting of HOXB13-low tumors with an FASN inhibitor.

Similar content being viewed by others

Data availability

All sequencing data (RNA-seq and ChIP–seq) generated for the study have been deposited in the Gene Expression Omnibus (GSE153586) at https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE153586. The mass spectrometry proteomic data have been deposited to the ProteomeXchange Consortium via the PRIDE60 partner repository with the dataset identifier PXD030810. Source data are provided with this paper.

Code availability

The code for NGS analyses performed in this paper has been uploaded to https://github.com/JYULAB/HOXB13_project.

References

  1. Zadra, G. et al. Inhibition of de novo lipogenesis targets androgen receptor signaling in castration-resistant prostate cancer. Proc. Natl Acad. Sci. USA 116, 631–640 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Poulose, N. et al. Genetics of lipid metabolism in prostate cancer. Nat. Genet. 50, 169–171 (2018).

    Article  CAS  PubMed  Google Scholar 

  3. Butler, L. M., Centenera, M. M. & Swinnen, J. V. Androgen control of lipid metabolism in prostate cancer: novel insights and future applications. Endocr. Relat. Cancer 23, R219–R227 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Swinnen, J. V., Esquenet, M., Goossens, K., Heyns, W. & Verhoeven, G. Androgens stimulate fatty acid synthase in the human prostate cancer cell line LNCaP. Cancer Res. 57, 1086–1090 (1997).

    CAS  PubMed  Google Scholar 

  5. Swinnen, J. V., Ulrix, W., Heyns, W. & Verhoeven, G. Coordinate regulation of lipogenic gene expression by androgens: evidence for a cascade mechanism involving sterol regulatory element binding proteins. Proc. Natl Acad. Sci. USA 94, 12975–12980 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Han, W. et al. Reactivation of androgen receptor-regulated lipid biosynthesis drives the progression of castration-resistant prostate cancer. Oncogene 37, 710–721 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. Mitra, R., Chao, O., Urasaki, Y., Goodman, O. B. & Le, T. T. Detection of lipid-rich prostate circulating tumour cells with coherent anti-Stokes Raman scattering microscopy. BMC Cancer 12, 540 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Yue, S. et al. Cholesteryl ester accumulation induced by PTEN loss and PI3K/AKT activation underlies human prostate cancer aggressiveness. Cell Metab. 19, 393–406 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Chen, J. et al. Compartmentalized activities of the pyruvate dehydrogenase complex sustain lipogenesis in prostate cancer. Nat. Genet. 50, 219–228 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Chen, M. et al. An aberrant SREBP-dependent lipogenic program promotes metastatic prostate cancer. Nat. Genet. 50, 206–218 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Berger, M. F. et al. Variation in homeodomain DNA binding revealed by high-resolution analysis of sequence preferences. Cell 133, 1266–1276 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Huang, Q. et al. A prostate cancer susceptibility allele at 6q22 increases RFX6 expression by modulating HOXB13 chromatin binding. Nat. Genet. 46, 126–135 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Sreenath, T., Orosz, A., Fujita, K. & Bieberich, C. J. Androgen-independent expression of hoxb-13 in the mouse prostate. Prostate 41, 203–207 (1999).

    Article  CAS  PubMed  Google Scholar 

  14. Edwards, S. et al. Expression analysis onto microarrays of randomly selected cDNA clones highlights HOXB13 as a marker of human prostate cancer. Br. J. Cancer 92, 376–381 (2005).

    Article  CAS  PubMed  Google Scholar 

  15. Economides, K. D. & Capecchi, M. R. Hoxb13 is required for normal differentiation and secretory function of the ventral prostate. Development 130, 2061–2069 (2003).

    Article  CAS  PubMed  Google Scholar 

  16. Pomerantz, M. M. et al. The androgen receptor cistrome is extensively reprogrammed in human prostate tumorigenesis. Nat. Genet. 47, 1346–1351 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Norris, J. D. et al. The homeodomain protein HOXB13 regulates the cellular response to androgens. Mol. Cell 36, 405–416 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Chen, Z. et al. Diverse AR-V7 cistromes in castration-resistant prostate cancer are governed by HoxB13. Proc. Natl Acad. Sci. USA 115, 6810–6815 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Augello, M. A. et al. CHD1 loss alters AR binding at lineage-specific enhancers and modulates distinct transcriptional programs to drive prostate tumorigenesis. Cancer Cell 35, 603–617 e8 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Jung, C., Kim, R. S., Zhang, H. J., Lee, S. J. & Jeng, M. H. HOXB13 induces growth suppression of prostate cancer cells as a repressor of hormone-activated androgen receptor signaling. Cancer Res. 64, 9185–9192 (2004).

    Article  CAS  PubMed  Google Scholar 

  21. VanOpstall, C. et al. MEIS-mediated suppression of human prostate cancer growth and metastasis through HOXB13-dependent regulation of proteoglycans. eLife 9, e53600 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Whitington, T. et al. Gene regulatory mechanisms underpinning prostate cancer susceptibility. Nat. Genet. 48, 387–397 (2016).

    Article  CAS  PubMed  Google Scholar 

  23. Mazrooei, P. et al. Cistrome partitioning reveals convergence of somatic mutations and risk variants on master transcription regulators in primary prostate tumors. Cancer Cell 36, 674–689 e6 (2019).

    Article  CAS  PubMed  Google Scholar 

  24. Spisak, S. et al. CAUSEL: an epigenome- and genome-editing pipeline for establishing function of noncoding GWAS variants. Nat. Med. 21, 1357–1363 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ewing, C. M. et al. Germline mutations in HOXB13 and prostate-cancer risk. N. Engl. J. Med. 366, 141–149 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Johng, D. et al. HOXB13 interaction with MEIS1 modifies proliferation and gene expression in prostate cancer. Prostate 79, 414–424 (2019).

    Article  CAS  PubMed  Google Scholar 

  27. Heinzel, T. et al. A complex containing N-CoR, mSin3 and histone deacetylase mediates transcriptional repression. Nature 387, 43–48 (1997).

    Article  CAS  PubMed  Google Scholar 

  28. Abbas, A. & Gupta, S. The role of histone deacetylases in prostate cancer. Epigenetics 3, 300–309 (2008).

    Article  PubMed  Google Scholar 

  29. Horlein, A. J. et al. Ligand-independent repression by the thyroid hormone receptor mediated by a nuclear receptor co-repressor. Nature 377, 397–404 (1995).

    Article  CAS  PubMed  Google Scholar 

  30. Chen, J. D. & Evans, R. M. A transcriptional co-repressor that interacts with nuclear hormone receptors. Nature 377, 454–457 (1995).

    Article  CAS  PubMed  Google Scholar 

  31. Guenther, M. G., Barak, O. & Lazar, M. A. The SMRT and N-CoR corepressors are activating cofactors for histone deacetylase 3. Mol. Cell. Biol. 21, 6091–6101 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. You, S. H. et al. Nuclear receptor co-repressors are required for the histone-deacetylase activity of HDAC3 in vivo. Nat. Struct. Mol. Biol. 20, 182–187 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Knutson, S. K. et al. Liver-specific deletion of histone deacetylase 3 disrupts metabolic transcriptional networks. EMBO J. 27, 1017–1028 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Sun, Z. et al. Hepatic Hdac3 promotes gluconeogenesis by repressing lipid synthesis and sequestration. Nat. Med. 18, 934–942 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Feng, D. et al. A circadian rhythm orchestrated by histone deacetylase 3 controls hepatic lipid metabolism. Science 331, 1315–1319 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kent, L. N. & Leone, G. The broken cycle: E2F dysfunction in cancer. Nat. Rev. Cancer 19, 326–338 (2019).

    Article  CAS  PubMed  Google Scholar 

  37. Wang, S. et al. Target analysis by integration of transcriptome and ChIP-seq data with BETA. Nat. Protoc. 8, 2502–2515 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Pomerantz, M. M. et al. Prostate cancer reactivates developmental epigenomic programs during metastatic progression. Nat. Genet. 52, 790–799 (2020).

    Article  CAS  PubMed  Google Scholar 

  39. Sun, Z. et al. Deacetylase-independent function of HDAC3 in transcription and metabolism requires nuclear receptor corepressor. Mol. Cell 52, 769–782 (2013).

    Article  CAS  PubMed  Google Scholar 

  40. Guo, C. et al. Regulated clearance of histone deacetylase 3 protects independent formation of nuclear receptor corepressor complexes. J. Biol. Chem. 287, 12111–12120 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Li, J. et al. Both corepressor proteins SMRT and N-CoR exist in large protein complexes containing HDAC3. EMBO J. 19, 4342–4350 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Wang, Z. et al. Genome-wide mapping of HATs and HDACs reveals distinct functions in active and inactive genes. Cell 138, 1019–1031 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Nguyen, H. M. et al. LuCaP prostate cancer patient-derived xenografts reflect the molecular heterogeneity of advanced disease and serve as models for evaluating cancer therapeutics. Prostate 77, 654–671 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Takeshima, H., Yamashita, S., Shimazu, T. & Ushijima, T. Effects of genome architecture and epigenetic factors on susceptibility of promoter CpG islands to aberrant DNA methylation induction. Genomics 98, 182–188 (2011).

    Article  CAS  PubMed  Google Scholar 

  45. Zhao, S. G. et al. The DNA methylation landscape of advanced prostate cancer. Nat. Genet. 52, 778–789 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Yu, Y. P. et al. Whole-genome methylation sequencing reveals distinct impact of differential methylations on gene transcription in prostate cancer. Am. J. Pathol. 183, 1960–1970 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Vire, E. et al. The Polycomb group protein EZH2 directly controls DNA methylation. Nature 439, 871–874 (2006).

    Article  CAS  PubMed  Google Scholar 

  48. Huang, Y. H. et al. DNA epigenome editing using CRISPR-Cas SunTag-directed DNMT3A. Genome Biol. 18, 176 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Jin, H. J., Zhao, J. C., Ogden, I., Bergan, R. C. & Yu, J. Androgen receptor-independent function of FoxA1 in prostate cancer metastasis. Cancer Res. 73, 3725–3736 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Heuer, T. S. et al. FASN inhibition and taxane treatment combine to enhance anti-tumor efficacy in diverse xenograft tumor models through disruption of tubulin palmitoylation and microtubule organization and FASN inhibition-mediated effects on oncogenic signaling and gene expression. eBioMedicine 16, 51–62 (2017).

    Article  PubMed  Google Scholar 

  51. Kote-Jarai, Z. et al. Prevalence of the HOXB13 G84E germline mutation in British men and correlation with prostate cancer risk, tumour characteristics and clinical outcomes. Ann. Oncol. 26, 756–761 (2015).

    Article  CAS  PubMed  Google Scholar 

  52. Ventura, R. et al. Inhibition of de novo palmitate synthesis by fatty acid synthase induces apoptosis in tumor cells by remodeling cell membranes, inhibiting signaling pathways, and reprogramming gene expression. eBioMedicine 2, 808–824 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Xu, B. et al. Altered chromatin recruitment by FOXA1 mutations promotes androgen independence and prostate cancer progression. Cell Res. 29, 773–775 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Fong, K. W., Zhao, J. C., Song, B., Zheng, B. & Yu, J. TRIM28 protects TRIM24 from SPOP-mediated degradation and promotes prostate cancer progression. Nat. Commun. 9, 5007 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Corces, M. R. et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nat. Methods 14, 959–962 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Song, B. et al. Targeting FOXA1-mediated repression of TGF-β signaling suppresses castration-resistant prostate cancer progression. J. Clin. Invest. 129, 569–582 (2019).

    Article  PubMed  Google Scholar 

  57. Li, Y., Ge, D. & Lu, C. The SMART App: an interactive web application for comprehensive DNA methylation analysis and visualization. Epigenetics Chromatin 12, 71 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Diez-Villanueva, A., Mallona, I. & Peinado, M. A. Wanderer, an interactive viewer to explore DNA methylation and gene expression data in human cancer. Epigenetics Chromatin 8, 22 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Shen, S. Y., Burgener, J. M., Bratman, S. V. & De Carvalho, D. D. Preparation of cfMeDIP-seq libraries for methylome profiling of plasma cell-free DNA. Nat. Protoc. 14, 2749–2780 (2019).

    Article  CAS  PubMed  Google Scholar 

  60. Perez-Riverol, Y. et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 47, D442–D450 (2019).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Y. Jia and W. Xie for providing some technical support for in vivo experiments. We thank all members of the Yu laboratory for helpful discussions. This work was partially supported by the Northwestern University Pathology Core Facility, the Center for Advanced Microscopy/Nikon Imaging Center and the Robert H. Lurie Comprehensive Cancer Center Support Grant (grant no. NCI P30CA060553). NGS was done at the University of Chicago Genomics Facility and lipid profiling at the Mass Spectrometry Core, University of Illinois at Chicago. IP-MS was done at the Taplin Mass Spectrometry Facility of Harvard Medical School. We thank the patients and their families, C. Higano, E. Yu, E. Mostaghel, H. Cheng, P. Nelson, B. Montgomery, M. Schweizer, A. Hsieh, J. Wright, D. Lin, F. Vakar-Lopez, X. Zhang, M. Roudier, L. True and the rapid autopsy teams for their contributions to the University of Washington Medical Center Prostate Cancer Donor Rapid Autopsy Program and the Development of the LuCaP PDX models. Funding supports for the work include the NIH/NCI training grant no. T32CA09560 (to G.G., L.T.B.); prostate cancer SPORE grant no. P50CA180995 (to W.J.C., J.Y., X.Y.); grants no. R01CA257446 and no. R01CA227918 (to J.Y.); grant no. R50CA211271 (to J.C.Z.); NIH-R35 grant no. CA197532 (to N.S.C.); grant no. R01CA251555 (to M.L.F.); Prostate Cancer Foundation grant no. 2017CHAL2008 (to J.Y.); and the Department of Defense grants no. W81XWH-19-1-0565 and no. W81XWH-21-1-0234 (to M.L.F.) and no. W81XWH-17-1-0405 and no. W81XWH-17-1-0578 (to J.Y.). Generation and maintenance of the LuCaP PDX models were partially funded by NIH awards no. P50CA97186 and no. P01CA163227.

Author information

Authors and Affiliations

Authors

Contributions

J.Y. and X.L. conceived the project and designed the experiments. J.C.Z., S.C.B., M.L.F. and J.Y. conducted bioinformatic and statistical analyses. F.W. and G.G. assisted with in vivo mouse experiments. K.F. performed IP-MS experiments and tissue microarray acquisition. J.R., X.L. and X.Y. performed HOXB13 and FASN IHC scoring. L.T.B. validated key experiments. J.E.B. carried out MeDIP-seq experiments. S.S. generated G84E isogenic 22Rv1. E.C. provided LuCaP PDX tissues. C.M. generated CRPC TMAs. X.L., J.C.Z. and J.Y. wrote the original manuscript. N.S.C., W.J.C. and M.L.F. consulted on the project and edited the manuscript.

Corresponding authors

Correspondence to Jonathan C. Zhao or Jindan Yu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Andrea Alimonti 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 HOXB13 WT, but not G84E mutant, inhibits lipogenic programs in PCa.

a. Heatmap showing HOXB13-induced gene expression in PCa cells with HOXB13 KD and/or rescue. HOXB13-induced (n = 276) genes were derived by comparing shHOXB13 with pGIPZ using FDR < 0.05 and fold change > =2.5. b. GO analysis of HOXB13-induced genes identified in a. GO analyses was performed by DAVID, top enriched molecular concepts are shown. The X-axis indicates enrichment significance. One-sided Fisher’s Exact test was performed and -log10 (p-value) are shown. c. QRT-PCR validation of cell cycle gene regulation by HOXB13 in LNCaP cells. d. RT-qPCR validation of PSA and other key lipogenic gene regulation by HOXB13 in PC-3, C4-2B, and 22Rv1 cells. e. IGV view of the percentage of HOXB13 WT (C) and G84E alleles (T) (left) and genome browser view of mRNA expression of FASN (right, chr17:80,036,214-80,056,106, hg19, hg19) in WT and five isogenic G84E clones of 22Rv1. The isogenic G84E cells of HOXB13 were generated by CRISPR editing. f. WB validating the expression of FASN, PSA, AR, and HOXB13 in control and HOXB13-KD LNCaP cells. LNCaP cells were infected with control shRNA (pGIPZ) or shRNA targeting HOXB13 (shHOXB13) for four days, followed by hormone starvation (Ethl) or regular medium (10% FBS) for three days. g. Pie chart showing the genomic distribution of HOXB13 binding sites in LNCaP cells. h-i. ChIP-qPCR analyses of HOXB13 at lipogenic gene enhancers in LNCaP (h) cells with HOXB13 KD and/or rescue and PC-3 (i) cells with HOXB13 KD. Data in c,d and h,i were shown as technical replicates from one of three (n = 3) independent experiments. Data shown are mean ±s.e.m, P values by unpaired two-sided t-test.

Source data

Extended Data Fig. 2 HOXB13 interacts with HDAC3 protein through its MEIS domain.

a. Heatmap showing the number of peptides of AR-cofactors and HDAC3/NCoR complex enriched by HOXB13 WT or G84E mutant mass spectrometry. The complete lists are included in Supplemental Table 6. b-c. Co-IP of V5-NcoR1 full-length (FL) or deletion mutants (b) expressed in 293 T cells along with HOXB13, with or without HDAC3 co-expression (c). Co-IP using whole-cell lysates showed no interactions between HOXB13 and NCoR1 in cells without HDAC3 co-expression. d-e. Co-IP of HA-HDAC3 FL or deletion mutants (d) expressed in 293 T cells along with HOXB13 (e). Whole-cell lysates were subjected to co-IP using an anti-HA antibody. f. Fractionation assay showing cellular localization of HOXB13 WT and mutants in LNCaP cells. WT, G84E and ΔMEIS HOXB13 were detected on the chromatin, whereas ΔHOX and WFQ-3A have impaired ability to bind chromatin. Asterisk indicates endogenous HOXB13. g-h. Schematic illustration of a series of HOXB13 deletion mutants (g) and their interaction with HDAC3 (h). Whole-cell lysates from 293 T cells co-transfected with HA-HDAC3 along with Flag-tagged HOXB13 FL or its deletion mutants were subjected to co-IP using an anti-Flag antibody. Asterisk indicates the size of corresponding mutants. i. The interaction between HOXB13 and MEIS1 is not interrupted by HDAC3. Whole-cell lysates from 293 T cells co-transfected with Flag-HOXB13 along with HA-tagged MEIS1 and gradually increased amount of HA-tagged HDAC3 were subjected to co-IP using an anti-Flag antibody.

Source data

Extended Data Fig. 3 HOXB13 recruits HDAC3 to catalyze histone deacetylation.

a-b. MA plot showing differential HOXB13 (a) and HDAC3 ChIP-seq enrichment (b) in control (pGIPZ) and HOXB13 KD (shHOXB13) LNCaP cells. Color encodes the intensity of HOXB13 ChIP-seq in control cells. Dotted lines represent 2-fold differences. c. Venn diagram showing the overlap between HDAC3 and HOXB13 cistromes in PC-3 and LNCaP cells (top). Bottom: H3K27ac ChIP-seq was performed in PC-3 cells with control or HOXB13 KD, and their average intensity plots centered (±5 kb) on co-occupied sites are shown. d-e. ChIP-qPCR analyses of HDAC3 (d) and H3K27ac (e) at lipogenic gene enhancers in PC3 cells with control or HOXB13 KD. Data were normalized to 2% of input DNA. Shown are the mean ±s.e.m of technical replicates from one of three (n = 3) independent experiments. P values were calculated by unpaired two-sided t-test. f-h. ChIP-qPCR analysis of HOXB13 (f), HDAC3 (g), and H3K27ac (h) at PSA and FASN enhancer in 7 CRPC PDX tumors. Data were normalized to 5% of input DNA. Shown are the mean ±s.e.m of technical replicates from one of three (n = 3) independent experiments. Unpaired two-sided t-test was performed between indicated groups.

Extended Data Fig. 4 HDAC3 is required for HOXB13-mediated suppression of de novo lipogenesis.

a. Venn Diagram showing the overlap between HOXB13- or HDAC1/3-induced or –repressed genes in LNCaP cells. b. GO analysis of HDAC1/3-repressed (left) and -induced (right) genes in LNCaP cells. Top enriched molecular concepts are shown. p values were calculated by one-sided Fisher’s exact test. c. QRT-PCR analysis of SREBF2, FASN, and SCD expression in PC-3 cells with KD of HOXB13, HDAC3, or both. Data were normalized to GAPDH. Shown are mean ±s.e.m of technical replicates from one of three (n = 3) independent experiments. P values were calculated by unpaired two-sided t-test. d. WB analysis of HOXB13 and HDAC1/3 co-regulated genes in LNCaP cells with HOXB13 KD and rescue by HOXB13 WT or mutants. HOXB13 depletion expectedly led to up-regulation of FASN, SREBF1, and PSA, which were again repressed by re-introduction of WT HOXB13. HOXB13 ΔHOX/WFQ-3A mutants, although capable of interacting with HDAC3, are unable to bind DNA17 and consequently failed to repress lipogenic genes. Importantly, ΔMEIS and G84E mutants, which have impaired ability to interact with HDAC3, also failed to fully repress these genes. e-f. ORO staining and quantification of lipid accumulation in 22Rv1 (e) and PC-3 (f) with shHOXB13 and/or rescue. Scale bar: 30 µm. Quantification data are the mean ±s.d of technical replicates from one of two (n = 2) independent experiments. P values were calculated by unpaired two-sided t-test. h. LipidSpot staining and quantification of lipid droplets in HOXB13 WT and five clones of isogenic G84E 22Rv1. Scale bar:20 µm. LipidSpot staining intensity was quantified and calculated as the fold change of LipidSpot staining in G84E mutant cells compared with WT controls (right). Each data point represents an average of 6 (n = 6) fields per independent experiment by image J. Data are the mean ±s.d. Unpaired two-sided t-test was performed between indicated groups as show in figure.

Source data

Extended Data Fig. 5 HOXB13 KD induces de novo lipogenesis in PCa cells cultured in either lipid-free or regular medium.

a-e. LipidSpot staining and quantification of lipid droplets in control (sgNC) or HOXB13 KD (sgHOXB13) LNCaP (a-c) and PC-3M (d-f) cultured in lipid-free or regular medium (10% FBS). WB (a,d) validating FASN up-regulation in HOXB13-KD cells cultured in lipid-free medium, confirming de novo lipogenesis. Data (b,e) shown are representative images of LipidSpot staining with a scale bar of 20 µm. LipidSpot staining intensity was quantified and calculated as fold change of LipidSpot staining in each condition normalized to sgNC non-target control under lipid-free medium (c,f). Each data point represents an average of 6 (n = 6) fields per independent experiment by image J. Data are the mean ±s.d. Unpaired two-sided t-test were performed between indicated groups as show in figure. g. Quantification of lipid accumulation of PDXs in Fig.4i. Six (n = 6) representative areas per PDX were quantified using Image J. Data are shown as mean±s.d. Unpaired one-sided t-test was performed between indicated groups as show in figure.

Source data

Extended Data Fig. 6 HOXB13 is hypermethylated and down-regulated in CRPC.

a. HOXB13 gene methylation levels in a variety of cancer types and corresponding benign (normal) samples using TCGA methylation data. Blue arrow indicates PCa (PRAD). *marks tumor types of significant differences compared with matched benign samples by Wilcoxon rank sum test. The sample sizes were included in Supplemental Table 7. b. Average methylation levels of Illumina Infinium probes targeting different regions of the HOXB13 gene (chr17:46,802,127-46,806,111. hg19) based on TCGA methylation data. Probes in green fonts target CpG islands. *probes with significantly differential methylation between normal and prostate tumor samples (Wilcoxon rank sum test with adjusted p-value < 0.05). c-d. Genome browser view of RNA-seq data (c) of HOXB13 (chr17:48,724,763-48,728,750, hg38) and average methylation levels (d) of 3 CpG islands within the HOXB13 gene loci (chr17:44,157,125-44,161,110. hg18) in LNCaP, 22Rv1, PC-3, DU145, and RWPE-1 cells. e. Expression of HOXB13 in Log2(FKPM) in LuCaP PDXs based on RNA-seq data. Blue indicates high- and red for low-HOXB13 samples. f. IGV track showing methylation at HOXB13 gene (chr17:46,802,127-46,806,111. hg19) in LuCaP PDX tumors with high or low HOXB13 as indicated in e. HOXB13 gene methylation in LuCaP PDX tumors was analyzed using MeDIP-seq data. g-h. RT-qPCR and WB analyses of PC-3M cells subjected to EZH2 or GFP-DNMT3A overexpression. PCR data shown are mean ±s.e.m. of technical replicates from one of three (n = 3) independent experiments and analyzed by unpaired two-sided t-test. i. WB analyses of LNCaP cells treated with dCas9-SunTag-DNMT3A system with gRNAs targeting the 4 CpGs within the HOXB13 gene loci. The gRNAs were cloned into the pLKO5.sgRNA.EFS.tRFP657 vector and an sgRNA (NC) that had no cognate target in the human genome was used as a negative control. WB was performed 7 days after co-infection of indicated constructs.

Source data

Extended Data Fig. 7 HOXB13 loss induces PCa cell motility in vitro and tumor metastasis in vivo.

a. Colony formation assays were performed in LNCaP, C4-2B and PC-3 cells with HOXB13 de-regulation or indicated treatment. The data showed that androgen-dependent LNCaP cell growth was abolished by HOXB13 KD, which could be restored by re-expression of either WT or G84E HOXB13, suggesting an AR-dependent effect. By contrast, C4-2B is only partially sensitive to HOXB13 KD, whereas the growth of C4-2B cells pre-treated by enzalutamide (ENZ) and the AR-negative PC-3 cells is unaffected by HOXB13 KD. b. Cell invasion assays of control or HOXB13-KD PC-3M cells cultured in lipid-free or regular medium. Representative images are shown (left panels), and the number of invaded cells are quantified (right panel). Scale bar: 50 µm. Quantified data shown are mean ±s.d. of three representative fields from one of three (n = 3) independent experiments. P values were calculated by unpaired two-sided t-test. c. Cell migration assays of PC-3 cells with control or shHOXB13. Images shown were taken at 0 and 48 hours after a scratch was created on the cell monolayer. d. Tumor volume was measured by IVIS after two weeks of intra-prostate inoculation of PC-3M cells. Y-axis shows the normalized luciferase intensity. Statistical significance was evaluated by one-way ANOVA test (P = 0.836). e. HOXB13 de-regulation did not affect PC-3M xenograft tumor growth. Tumor volume was measured every week by IVIS live mice imaging. Y-axis shows the normalized luciferase intensity. Data shown in each time point are mean ± s.d. Statistical significance was evaluated by one-way ANOVA test (P = 0.365).

Extended Data Fig. 8 Effect of various lipogenic inhibitors on PCa cells.

a. FASN mRNA levels in publically available PCa gene expression profiling datasets. Data shown (Y-axis) are log2-transformed microarray expression values for GSE6919 and GSE35988 and FPKM values for TCGA-dbGaP dataset, which combines prostate samples from dbGaP datasets with accession#: phs000178(TCGA), phs000443, phs000915, and phs000909. P values between primary and metastatic PCa were calculated using unpaired two-sided t-test. Boxplots represent the median and bottom and upper quartiles; Whisker edges indicate minimum and maximum values. b. Cell invasion of control or HOXB13-KD cells treated with various lipid inhibitors. PC-3M cells were treated with 5 µM TVB-2640, 5 µM TOFA, 2 µM Fatostatin or 2 µM Simvastatin for 3 days. C4-2B cells were treated with 1 µM of indicated inhibitors for 3 days. Representative images are shown for C4-2B with a Scale bar of 50 µm and PC-3M with a Scale bar of 30 µm. c. Quantification of cell invasion shown in b. Data shown are the mean ±s.d of three representative fields from one of two (n = 2) independent experiments. P values were calculated by unpaired two-sided t-test comparing inhibitor groups with DMSO in control or shHOXB13 cells. d-e. Representative images of ORO staining (d) and quantification (e) of lipid accumulation in HOXB13-KD PC-3M cells treated with DMSO or 5 μM TVB-2640 for 3 days. Scale bar: 30 µm. Data shown are the mean ± s.d of triplicate wells from one of two (n = 2) independent experiments. Unpaired two-sided t test was performed between indicated groups as shown in figures.

Extended Data Fig. 9 Therapeutic targeting of HOXB13-low tumors with FASN inhibitors in an orthotopic xenograft model.

a. Graph showing tumor volumes after intraprostatic inoculation of PC-3M cells. Tumor formation was confirmed by IVIS one week after inoculation. Then the mice were randomized to receive vehicle (30% PEG400) or TVB-2640 (100 mg/Kg) every day for 30 days. Tumor volumes were measured twice per week by IVIS after two weeks of inoculation. Y-axis shows the normalized luciferase intensity. Data in each time point are mean ±s.d. Statistical significance was evaluated by one-way ANOVA (P = 0.0007) and comparisons between indicated groups by post hoc Tukey test. PC-3M xenograft tumor growth was not affected by HOXB13 KD (p = 0.3212). Importantly, primary tumor growth in both control (pGIPZ, p = 0.0171) and HOXB13-KD (shHOXB13, p = 0.0082) mice was significantly inhibited by TVB-2640. b-c. Representative ex vivo IVIS images (b) and quantifications (c) of PC-3M tumor metastasis to the liver and lung (n = 6 mice per group). Heatmap shows IVIS signal intensity color scale. Indicated p-values were shown by unpaired two-sided t-test. HOXB13 KD significantly promoted PCa metastases, which was abolished by TVB-2640. d. Validation of PC-3M tumor metastasis to the liver by H&E and IHC. Luciferase and Pan-keratin IHC were used to identify metastasized PC-3M cells in mouse liver. Representative images of H&E, Luciferase and Pan-keratin staining in indicated group (n = 3 mice in each group) are shown. Scale bar: 30 µm. “T” indicates tumor and “n” for normal liver. e. Kaplan-Meier analyses of overall survival of pGIPZ and HOXB13 KD mice treated with vehicle or TVB-2640 (n = 6 mice per group). P values were determined by the log-rank test. TVB-2640 treatment prolonged the overall survival of mice in both control (p = 0.017) and HOXB13-KD (p = 0.005) groups.

Extended Data Fig. 10 Therapeutic targeting of HOXB13-low tumors with FASN inhibitors in an intravenous xenograft model.

a. Body weight analysis in mice inoculated by control or HOXB13-KD PC-3M cells and treated with vehicle or TVB-2640. Data in each time point are mean ±s.d. Y-axis shows the percentage of body weight change. b-c. Representative ex vivo IVIS images (b) of PC-3M tumor metastasis to lung, hind leg, liver, and rib and quantification (c) of metastasis to liver, hind leg, and rib (n = 4 mice for pGIPZ vehicle group; n = 5 mice for the rest groups). At the endpoint, lung, hind leg, liver, and rib were collected and analyzed using ex vivo IVIS. Heatmap in b shows IVIS signal intensity color scale. Y-axis in c shows the normalized luciferase intensity. Indicated p values were by unpaired two-sided t test.

Supplementary information

Supplementary Information

Supplementary Figs. 1–7.

Reporting Summary

Supplementary Tables

Supplementary Table 1: Antibodies that were used in this study. Supplementary Table 2: Oligonucleotides that were used in this study. Supplementary Table 3: Genes not rescued by G84E and rescued by G84E in Fig. 1a. Supplementary Table 4: Genes involved in fatty acid uptake related to Supplementary Fig. 4. Supplementary Table 5: Key enzymes involved in lipogenesis related to Supplementary Fig. 7. Supplementary Table 6: Interacting proteins in HOXB13 WT and G84E mass spec related to Extended Data Fig. 2a. Supplementary Table 7: The numbers of normal and tumor in different cancer types related to Extended Data Fig. 6a.

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lu, X., Fong, Kw., Gritsina, G. et al. HOXB13 suppresses de novo lipogenesis through HDAC3-mediated epigenetic reprogramming in prostate cancer. Nat Genet 54, 670–683 (2022). https://doi.org/10.1038/s41588-022-01045-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-022-01045-8

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing