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:

Regulating the expression of therapeutic transgenes by controlled intake of dietary essential amino acids

Abstract

Widespread application of gene therapy will depend on the development of simple methods to regulate the expression of therapeutic genes. Here we harness an endogenous signaling pathway to regulate therapeutic gene expression through diet. The GCN2-eIF2α signaling pathway is specifically activated by deficiencies in any essential amino acid (EAA); EAA deficiency leads to rapid expression of genes regulated by ATF4-binding cis elements. We found that therapeutic genes under the control of optimized amino acid response elements (AAREs) had low basal expression and high induced expression. We applied our system to regulate the expression of TNFSF10 (TRAIL) in the context of glioma therapy and found that intermittent activation of this gene by EEA-deficient meals retained its therapeutic efficacy while abrogating its toxic effects on normal tissue. The GCN2-eIF2α pathway is expressed in many tissues, including the brain, and is highly specific to EAA deficiency. Our system may be particularly well suited for intermittent regulation of therapeutic transgenes over short or long time periods.

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

Figure 1: Generation and in vivo validation of the AARE-driven expression system.
Figure 2: Determination of the optimal nutritional induction of the AARE-driven expression system.
Figure 3: Effective transgene regulation by the AARE-driven expression system after gene transfer methods in several organs.
Figure 4: TRAIL expression driven by the nutrition-based AARE gene system prevents growth of xenografted glioblastoma cells.

Similar content being viewed by others

References

  1. Kaufmann, K.B., Büning, H., Galy, A., Schambach, A. & Grez, M. Gene therapy on the move. EMBO Mol. Med. 5, 1642–1661 (2013).

    Article  CAS  Google Scholar 

  2. Benítez, J.A., Domínguez-Monzón, G. & Segovia, J. Conventional and gene therapy strategies for the treatment of brain tumors. Curr. Med. Chem. 15, 729–742 (2008).

    Article  Google Scholar 

  3. Sheridan, C. Gene therapy finds its niche. Nat. Biotechnol. 29, 121–128 (2011).

    Article  CAS  Google Scholar 

  4. Misra, S. Human gene therapy: a brief overview of the genetic revolution. J. Assoc. Physicians India 61, 127–133 (2013).

    PubMed  Google Scholar 

  5. Maurin, A.C. et al. The GCN2 kinase biases feeding behavior to maintain amino acid homeostasis in omnivores. Cell Metab. 1, 273–277 (2005).

    Article  CAS  Google Scholar 

  6. Chaveroux, C. et al. Molecular mechanisms involved in the adaptation to amino acid limitation in mammals. Biochimie 92, 736–745 (2010).

    Article  CAS  Google Scholar 

  7. Kilberg, M.S., Balasubramanian, M., Fu, L. & Shan, J. The transcription factor network associated with the amino acid response in mammalian cells. Adv. Nutr. 3, 295–306 (2012).

    Article  CAS  Google Scholar 

  8. Dever, T.E. et al. Phosphorylation of initiation factor 2 alpha by protein kinase GCN2 mediates gene-specific translational control of GCN4 in yeast. Cell 68, 585–596 (1992).

    Article  CAS  Google Scholar 

  9. Lu, P.D., Harding, H.P. & Ron, D. Translation reinitiation at alternative open reading frames regulates gene expression in an integrated stress response. J. Cell Biol. 167, 27–33 (2004).

    Article  CAS  Google Scholar 

  10. Vattem, K.M. & Wek, R.C. Reinitiation involving upstream ORFs regulates ATF4 mRNA translation in mammalian cells. Proc. Natl. Acad. Sci. USA 101, 11269–11274 (2004).

    Article  CAS  Google Scholar 

  11. Bruhat, A. et al. Amino acids control mammalian gene transcription: activating transcription factor 2 is essential for the amino acid responsiveness of the CHOP promoter. Mol. Cell. Biol. 20, 7192–7204 (2000).

    Article  CAS  Google Scholar 

  12. Harding, H.P. et al. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol. Cell 11, 619–633 (2003).

    Article  CAS  Google Scholar 

  13. Carraro, V. et al. Amino acid availability controls TRB3 transcription in liver through the GCN2/eIF2α/ATF4 pathway. PLoS One 5, e15716 (2010).

    Article  CAS  Google Scholar 

  14. Donnelly, N., Gorman, A.M., Gupta, S. & Samali, A. The eIF2α kinases: their structures and functions. Cell. Mol. Life Sci. 70, 3493–3511 (2013).

    Article  CAS  Google Scholar 

  15. Crosby, J.S., Lee, K., London, I.M. & Chen, J.J. Erythroid expression of the heme-regulated eIF-2 alpha kinase. Mol. Cell. Biol. 14, 3906–3914 (1994).

    Article  CAS  Google Scholar 

  16. Chaveroux, C. et al. In vivo imaging of the spatiotemporal activity of the eIF2α-ATF4 signaling pathway: Insights into stress and related disorders. Sci. Signal. 8, rs5 (2015).

    Article  Google Scholar 

  17. Ezzine, S. et al. RILES, a novel method for temporal analysis of the in vivo regulation of miRNA expression. Nucleic Acids Res. 41, e192 (2013).

    Article  CAS  Google Scholar 

  18. Keller, T.L. et al. Halofuginone and other febrifugine derivatives inhibit prolyl-tRNA synthetase. Nat. Chem. Biol. 8, 311–317 (2012).

    Article  CAS  Google Scholar 

  19. Pines, M. & Spector, I. Halofuginone - the multifaceted molecule. Molecules 20, 573–594 (2015).

    Article  Google Scholar 

  20. Seol, J.Y. et al. Adenovirus-TRAIL can overcome TRAIL resistance and induce a bystander effect. Cancer Gene Ther. 10, 540–548 (2003).

    Article  CAS  Google Scholar 

  21. Griffith, T.S. et al. TRAIL gene therapy: from preclinical development to clinical application. Curr. Gene Ther. 9, 9–19 (2009).

    Article  CAS  Google Scholar 

  22. Bellail, A.C., Qi, L., Mulligan, P., Chhabra, V. & Hao, C. TRAIL agonists on clinical trials for cancer therapy: the promises and the challenges. Rev. Recent Clin. Trials 4, 34–41 (2009).

    Article  CAS  Google Scholar 

  23. Kim, C.Y. et al. Preclinical studies for pharmacokinetics and biodistribution of Ad-stTRAIL, an adenovirus delivering secretable trimeric TRAIL for gene therapy. Exp. Mol. Med. 43, 580–586 (2011).

    Article  CAS  Google Scholar 

  24. Nagane, M., Cavenee, W.K. & Shiokawa, Y. Synergistic cytotoxicity through the activation of multiple apoptosis pathways in human glioma cells induced by combined treatment with ionizing radiation and tumor necrosis factor-related apoptosis-inducing ligand. J. Neurosurg. 106, 407–416 (2007).

    Article  Google Scholar 

  25. Walczak, H. et al. Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo. Nat. Med. 5, 157–163 (1999).

    Article  CAS  Google Scholar 

  26. Nitsch, R. et al. Human brain-cell death induced by tumour-necrosis-factor-related apoptosis-inducing ligand (TRAIL). Lancet 356, 827–828 (2000).

    Article  CAS  Google Scholar 

  27. Nesterov, A., Ivashchenko, Y. & Kraft, A.S. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) triggers apoptosis in normal prostate epithelial cells. Oncogene 21, 1135–1140 (2002).

    Article  CAS  Google Scholar 

  28. Volkmann, X. et al. Increased hepatotoxicity of tumor necrosis factor-related apoptosis-inducing ligand in diseased human liver. Hepatology 46, 1498–1508 (2007).

    Article  CAS  Google Scholar 

  29. Hingtgen, S. et al. Targeting multiple pathways in gliomas with stem cell and viral delivered S-TRAIL and Temozolomide. Mol. Cancer Ther. 7, 3575–3585 (2008).

    Article  CAS  Google Scholar 

  30. Agha-Mohammadi, S. & Lotze, M.T. Regulatable systems: applications in gene therapy and replicating viruses. J. Clin. Invest. 105, 1177–1183 (2000).

    Article  CAS  Google Scholar 

  31. Mortimore, G.E. & Pösö, A.R. Intracellular protein catabolism and its control during nutrient deprivation and supply. Annu. Rev. Nutr. 7, 539–568 (1987).

    Article  CAS  Google Scholar 

  32. Goverdhana, S. et al. Regulatable gene expression systems for gene therapy applications: progress and future challenges. Mol. Ther. 12, 189–211 (2005).

    Article  CAS  Google Scholar 

  33. Naidoo, J. & Young, D. Gene regulation systems for gene therapy applications in the central nervous system. Neurol. Res. Int. 2012, 595410 (2012).

    Article  Google Scholar 

  34. Bartus, R.T., Weinberg, M.S. & Samulski, R.J. Parkinson's disease gene therapy: success by design meets failure by efficacy. Mol. Ther. 22, 487–497 (2014).

    Article  CAS  Google Scholar 

  35. Di Stasi, A. et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N. Engl. J. Med. 365, 1673–1683 (2011).

    Article  CAS  Google Scholar 

  36. Ron, D. & Walter, P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat. Rev. Mol. Cell Biol. 8, 519–529 (2007).

    Article  CAS  Google Scholar 

  37. O'Connor, T. et al. Phosphorylation of the translation initiation factor eIF2alpha increases BACE1 levels and promotes amyloidogenesis. Neuron 60, 988–1009 (2008).

    Article  CAS  Google Scholar 

  38. Hoozemans, J.J. et al. Activation of the unfolded protein response in Parkinson's disease. Biochem. Biophys. Res. Commun. 354, 707–711 (2007).

    Article  CAS  Google Scholar 

  39. Shah, K., Tang, Y., Breakefield, X. & Weissleder, R. Real-time imaging of TRAIL-induced apoptosis of glioma tumors in vivo. Oncogene 22, 6865–6872 (2003).

    Article  CAS  Google Scholar 

  40. Harding, H.P., Zhang, Y., Bertolotti, A., Zeng, H. & Ron, D. Perk is essential for translational regulation and cell survival during the unfolded protein response. Mol. Cell 5, 897–904 (2000).

    Article  CAS  Google Scholar 

  41. Harding, H.P. et al. Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol. Cell 6, 1099–1108 (2000).

    Article  CAS  Google Scholar 

  42. Abraham, N. et al. Characterization of transgenic mice with targeted disruption of the catalytic domain of the double-stranded RNA-dependent protein kinase, PKR. J. Biol. Chem. 274, 5953–5962 (1999).

    Article  CAS  Google Scholar 

  43. Maurin, A.C. et al. Hypothalamic eIF2α signaling regulates food intake. Cell Rep. 6, 438–444 (2014).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank K. Shah (Harvard Medical School) for Gli36-Luc cells, D. Ron and H. Harding (Institute of Metabolic Science, Cambridge, UK) for GCN2−/− and PERK−/− MEFs, J.C. Bell (Ottawa health research institute, Canada) for PKR−/− MEFs, M. Petera for statistical analysis assistance, and A. Terrisse and A Teygnié for animal care and technical assistance. This study was supported by 'Programmes Emergence' (ANR-12-EMMA-0024) and (ANR-12-BSV2-0025-03) of 'Agence Nationale de la Recherche'.

Author information

Authors and Affiliations

Authors

Contributions

C.C., A.B., J.M. and P.F. designed the experiment; C.C., A.B., V.C., A.M., P.B., A.D.T. and P.R. performed the experiments; P.C. supervised A.M.'s work; J.A., A-C.M., L.P., C.J., Y.M. and F.M. provided reagents, materials, or analysis tools; C.C., P.F., J.M., P.R. and A.B. analyzed the data; J.M., A.B. and P.F. wrote the paper.

Corresponding author

Correspondence to Pierre Fafournoux.

Ethics declarations

Competing interests

P.F., A.B., C.J., A.-C.M. and J.A. are authors of patent application WO/2013/068096 (also US14357160A1 and EP2776568), “Inducible Expression Cassette, And Uses Thereof.”

Integrated supplementary information

Supplementary Figure 1 Generation of the AARE-Gene system construct.

(a) Position and sequence alignment of AAREs extracted from human Trb3, Chop or Atf3 promoters. AARE core sequences are boxed in grey. (b) Comparison of strength of AAREs from Trb3, Chop or Atf3 promoters in response to Leucine starvation. MEFs were transiently transfected with constructs containing two copies of each AARE. Twenty-four hours after transfection, cells were incubated for 16 h in control or medium lacking leucine and assayed for luciferase activity (Student's t-test: ***p <0.001, n=9; error bars: means ± s.e.m.). (c) The TK minimal promoter provides a lower baseline level compared to the β- globin one. MEFs were transiently transfected with Luc constructs containing two copies of AAREs from Trb3 promoter and a minimal promoter coming from either the TK or β-globin gene. Twenty-four hours after transfection, cells were incubated for 16 h in control or medium lacking leucine and assayed for luciferase activity (Student's t-test: ***p <0.001, n=9; error bars: means ± s.e.m.). (d) Mouse Embryonic Fibroblasts (MEF) or HeLa cells stably transduced with LV-AARE-Luc lentiviral vector were incubated for 16 h either in control (Ctrl) or leucine-free medium (-Leu) and assayed for luciferase activity (Student's t-test: ***p <0.001, n=9; error bars: means ± s.e.m.).

Supplementary Figure 2 GCN2 requirement for the AARE-Driven Expression System inducibility by leucine starvation.

(a) Wild type (WT), GCN2 -/-, PERK -/- or PKR -/- MEFs were transiently transfected with the construction described above. Twenty-four hours after transfection, cells were leucine-starved for 16 h and assayed for luciferase activity (Student's t-test: ***p <0.001, n=9; error bars: means ± s.e.m.). (b) Leucine concentration response of the AARE-Gene system inducibility. MEFs were transiently transfected with the AARE-Luc construction. Twenty-four hours after, cells were incubated for 16 h in media containing decreasing concentrations of leucine and assayed for luciferase activity (Student's t-test: ***p <0.001 in comparison to the control condition (420 μM), n=9; error bars: means ± s.e.m.).

Supplementary Figure 3 Consumption of a diet devoid of one essential amino acid causes a drop of the corresponding EAA in the plasma.

Following an overnight fasting period, AARE-Luc transgenic mice were fed for 6 hours, a control diet (Ctrl) or a diet devoid of alanine (-Ala), or histidine (-His), or isoleucine (-Ile), or methionine (-Met), or leucine (-Leu), or lysine (-Lys), or phenylalanine (-Phe), or threonine (-Thr), or tryptophan (-Trp), or valine (-Val). At the end of the experiments, blood was collected and amino acid measurement was performed for the different nutritional conditions (Student's t-test: ***p <0.001, n=6 male mice; error bars: means ± s.e.m.).

Supplementary Figure 4 Identification of the target tissues of the eIF2α-ATF4 pathway induction in transgenic mice, in which the AARE-gene expressing luciferase was stably integrated.

Mice were fed on a complete or a leucine-deficient diet for 6 h. Mice were sacrificed by cervical dislocation and organs were rapidly excised followed by snap-freezing for subsequent luciferase activity measurement. Results are given as fold induction relative to the control condition arbitrary set at 1 for each analyzed organ (Student's t-test: *p <0.05, **p <0.01, ***p <0.001 in comparison to the control condition, n=6 male mice; error bars: means ± s.e.m.).

Supplementary Figure 5 Assessment by bioluminescence imaging of the lentivirus dissemination in the mouse body following pancreatic transduction with LV-AARE-Luc.

(a) Administration of lentiviral particles containing the AARE-TK-Luc was performed into the pancreas of wild type mice. Ten days after injection, mice were challenged with the nutritional protocol (-Ile diet) for 6h and bioluminescence was measured. After pancreas and the adjacent spleen have been collected, organs and the body were imaged for bioluminescence. (b) PCR amplification for genotyping of the spleen and pancreas collected in Figure Supplementary 5a (primers used for PCR: forward primer, 5′-TCTGGTGGCGCTCCCCTCTC-3′; reverse primer, 5′-AACAACAACGGCGGCGGGAA-3′).

Supplementary Figure 6 Induction of the AARE-Driven Expression system by halofuginone in liver and pancreas.

Halofuginone (Hf) exerts its effects by acting as a high affinity inhibitor of the enzyme Glutamyl-Prolyl tRNA synthetase. Inhibition of prolyl tRNA charging leads to the accumulation of uncharged prolyl tRNAs, which serves as a signal to initiate the amino acid starvation response. Hf has anti-metastatic and anti-proliferative effects. It has already been used to treat patients with solid tumor, unfortunatly with low antitumor effects and few side effects (nosea, vomiting, fatigue…) (De Jonge et al 2006; European J of Cancer 42; 1768-1774). (a) Bioluminescence imaging of mice following hydrodynamics-based DNA injection of the pGL3-AARE-Luc. Wild type mice received 25 micrograms of plasmid according to the hydrodynamic injection method. Twenty-four hours later, mice were injected with Hf (0.05μg/g of body weight) or PBS (Ctrl). Bioluminescence imaging was performed 6 hours after injection and light emission quantified using ROIs covering the abdominal area (red shape) (Student's t-test: ***p <0.001 versus Control (Ctrl), n=6 male mice; error bars: means ± s.e.m.). Signal intensity as a result of photon detection is graded from red (highest number of photons) to blue (lowest intensity). Next the livers have been collected and imaged for bioluminescence and then the corresponding protein homogenates were assayed for luciferase activity determination (Student's test: ***p <0.001 versus Control diet, n=6 male mice; error bars: means ± s.e.m.). (b) Intra-pancreatic delivery and induction of the AARE-Luc by halofuginone following lentiviral transduction. Administration of lentiviral particles containing the AARE-TK-Luc (LV-AARE-Luc) was performed into the pancreas of wild type mice. Ten days after injection, mice were injected with halofuginone (Hf) or PBS (Ctrl). Light emission was quantified 6 h after injection using ROIs covering the pancreatic area (red shape). Signal intensity as a result of photon detection is graded from red (highest number of photons) to blue (lowest intensity). (Student's t-test: **p <0.01 versus Control (Ctrl), n=6 male mice; error bars: means ± s.e.m.). Then the pancreas have been collected and imaged for bioluminescence and the corresponding protein homogenates were assayed for luciferase activity determination (Student's t-test: ***p <0.001 versus Control (Ctrl), n=6 male mice; error bars: means ± s.e.m.).

Supplementary Figure 7 Apoptosis induction through the AARE-Gene mediated expression of TRAIL in human glioblastoma cells.

(a) Scheme for the AARE-Gene expression system driving either eGFP (LV-AARE-eGFP) or TRAIL (LV-AARE-TRAIL) expression. The corresponding lentiviral particles were then used for the following experiments. (b) Time course analysis of cell death induction in human glioblastoma cells expressing TRAIL under the control of the AARE-Gene system. Gli36-luc cells were transduced with the lentiviral particles described above. Forty-eight hours post-transduction, cells were incubated in control medium (Ctrl) or medium without leucine (-Leu) for the indicated time periods. TRAIL, cleaved-PARP (marker of apoptosis, short and long exposure), and ATF4 levels were analyzed by western blot. Actin levels are shown as a loading control. (c) Monitoring of apoptosis induction in glioblastoma cells following TRAIL expression in response to an EAA starvation. LV-AAREeGFP or LV-AARE-TRAIL Gli36-luc cells were incubated in control (Ctrl) or leucine-deprived (-Leu) medium for 4-24 h and then stained with 7-AAD and Annexin V for subsequent FACS analysis. (d) Quantification of TRAIL release by Gli36-luc cells following TRAIL expression in response to a leucine starvation. LV-AARE-eGFP or LV-AARE-TRAIL Gli36-luc cells were incubated in control medium (white bars) or starved for leucine (black bars) for 16 h. Quantification of TRAIL concentrations in the media was performed by ELISA (Student's t-test: ***p <0.001, n=9; error bars: means ± s.e.m.). (e) Paracrine effect of secreted TRAIL. Parental cells were incubated for 16 h with media collected in Supplementary Figure 7d. Cleaved-PARP and ATF4 levels were analyzed by western blot. Actin levels are shown as a loading control.

Supplementary Figure 8 Growth of xenografted glioblastoma cells is not impaired in mice fed diets devoid of one EAA.

(a) 2x106 Gli36-luc cells were implanted subcutaneously in nude mice. A week after, mice were divided into two groups: one fed a control diet and a second group fed with an alternation of EAA-deficient diets as described in figure 2C. For tumor growth monitoring, mice bearing tumors were imaged on the first day after Gli36-luc implantation (T0) and subsequently every week. Signal intensity within the region of interest was quantified using ROIs covering the tumors. Photon detection is graded from red (highest number of photons) to blue (lowest intensity). No significant difference was observed for radiance, between the 2 groups along the time. (b) Tumors weight analysis. At the end of the experiment, excised tumors were photographed and weighted. No significant difference was observed for tumor weight, between the 2 groups along the time. (c) Nutritional control of the GCN2-eIF2α-ATF4 pathway. At the end of the experiment, tumors from the Ctrl group fed a complete diet (Ctrl/Ctrl) were excised at day 21. Within the -EAA group, half of the lot was sacrificed at day 21 whereas the other half was killed a day later. For more details see legend of Figure 4. Proteins homogenates from the excised tumors were analyzed by western blot for detection of P-eIF2α and ATF4 levels. Actin levels are shown as a loading control.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 (PDF 1470 kb)

Supplementary Table 1

Composition of the experimental diets (PDF 159 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chaveroux, C., Bruhat, A., Carraro, V. et al. Regulating the expression of therapeutic transgenes by controlled intake of dietary essential amino acids. Nat Biotechnol 34, 746–751 (2016). https://doi.org/10.1038/nbt.3582

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3582

This article is cited by

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