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:

A targeted antisense therapeutic approach for Hutchinson–Gilford progeria syndrome

Abstract

Hutchinson–Gilford progeria syndrome (HGPS) is a rare accelerated aging disorder characterized by premature death from myocardial infarction or stroke. It is caused by de novo single-nucleotide mutations in the LMNA gene that activate a cryptic splice donor site, resulting in the production of a toxic form of lamin A, which is termed progerin. Here we present a potential genetic therapeutic strategy that utilizes antisense peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs) to block pathogenic splicing of mutant transcripts. Of several candidates, PPMO SRP-2001 provided the most significant decrease in progerin transcripts in patient fibroblasts. Intravenous delivery of SRP-2001 to a transgenic mouse model of HGPS produced significant reduction of progerin transcripts in the aorta, a particularly critical target tissue in HGPS. Long-term continuous treatment with SRP-2001 yielded a 61.6% increase in lifespan and rescue of vascular smooth muscle cell loss in large arteries. These results provide a rationale for proceeding to human trials.

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: Design and testing of a PPMO-based strategy for blocking the production of progerin.
Fig. 2: Effect of SRP-2001 on cellular proliferation and B-type lamin protein expression in the fibroblasts of a patient with HGPS.
Fig. 3: Determination of SRP-2001 efficacy in culture.
Fig. 4: Efficient in vivo delivery of PPMO to murine vascular cells and tissues.
Fig. 5: Long-term treatment of HGPS mice with SRP-2001 significantly extends their lifespan.
Fig. 6: Partial rescue of vascular tissue degradation in mice treated with SRP-2001.

Similar content being viewed by others

Data availability

All requests for raw and analyzed data and materials will be promptly reviewed by the National Human Genome Research Institute and Sarepta Therapeutics to verify whether the request is subject to any intellectual property or confidentiality obligations. Any data and materials that can be shared will be released via a Data/Material Sharing Agreement. All requests should be made to the primary or corresponding author. All detailed PPMO sequence and assay primer data are presented in the extended and supplementary data.

References

  1. Burke, B. & Stewart, C. L. The nuclear lamins: flexibility in function. Nat. Rev. Mol. Cell Biol. 14, 13–24 (2013).

    Article  CAS  PubMed  Google Scholar 

  2. Capell, B. C. & Collins, F. S. Human laminopathies: nuclei gone genetically awry. Nat. Rev. Genet. 7, 940–952 (2006).

    Article  CAS  PubMed  Google Scholar 

  3. Schreiber, K. H. & Kennedy, B. K. When lamins go bad: nuclear structure and disease. Cell 152, 1365–1375 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hennekam, R. C. M. Hutchinson–Gilford progeria syndrome: review of the phenotype. Am. J. Med. Genet. A 140, 2603–2624 (2006).

    Article  PubMed  Google Scholar 

  5. Eriksson, M. et al. Recurrent de novo point mutations in lamin A cause Hutchinson–Gilford progeria syndrome. Nature 423, 293–298 (2003).

    Article  CAS  PubMed  Google Scholar 

  6. De Sandre-Giovannoli, A. et al. Lamin a truncation in Hutchinson–Gilford progeria. Science 300, 2055 (2003).

    Article  CAS  PubMed  Google Scholar 

  7. Dechat, T. et al. Nuclear lamins: major factors in the structural organization and function of the nucleus and chromatin. Genes Dev. 22, 832–853 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Gordon, L. B. et al. Impact of farnesylation inhibitors on survival in Hutchinson–Gilford progeria syndrome. Circulation 130, 27–34 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Gordon, L. B. et al. Association of lonafarnib treatment vs no treatment with mortality rate in patients with Hutchinson–Gilford progeria syndrome. JAMA 319, 1687–1695 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Gordon, L. B. et al. Clinical trial of a farnesyltransferase inhibitor in children with Hutchinson–Gilford progeria syndrome. Proc. Natl Acad. Sci. USA 109, 16666–16671 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. Cao, K. et al. Rapamycin reverses cellular phenotypes and enhances mutant protein clearance in Hutchinson–Gilford progeria syndrome cells. Sci. Transl. Med. 3, 89ra58 (2011).

    Article  CAS  PubMed  Google Scholar 

  12. Harhouri, K. et al. An overview of treatment strategies for Hutchinson–Gilford progeria syndrome. Nucleus 9, 246–257 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Lebleu, B. et al. Cell penetrating peptide conjugates of steric block oligonucleotides. Adv. Drug Deliv. Rev. 60, 517–529 (2008).

    Article  CAS  PubMed  Google Scholar 

  14. Syed, Y. Y. Eteplirsen: first global approval. Drugs 76, 1699–1704 (2016).

    Article  CAS  PubMed  Google Scholar 

  15. Cubria, M. B. et al. Evaluation of musculoskeletal phenotype of the G608G progeria mouse model with lonafarnib, pravastatin, and zoledronic acid as treatment groups. Proc. Natl Acad. Sci. USA 117, 12029–12040 (2020).

    Article  CAS  PubMed  Google Scholar 

  16. Hanson, G. J. Peptide oligonucleotide conjugates. US Patent US9161948 (2015); https://patents.google.com/patent/US9161948B2/en

  17. Tsai, M.-Y. et al. A mitotic lamin B matrix induced by RanGTP required for spindle assembly. Science 311, 1887–1893 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Freund, A., Laberge, R.-M., Demaria, M. & Campisi, J. Lamin B1 loss is a senescence-associated biomarker. Mol. Biol. Cell 23, 2066–2075 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sazani, P. et al. Systemically delivered antisense oligomers upregulate gene expression in mouse tissues. Nat. Biotechnol. 20, 1228–1233 (2002).

    Article  CAS  PubMed  Google Scholar 

  20. Echigoya, Y. et al. Effects of systemic multiexon skipping with peptide-conjugated morpholinos in the heart of a dog model of Duchenne muscular dystrophy. Proc. Natl Acad. Sci. USA 114, 4213–4218 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Stehbens, W. E., Delahunt, B., Shozawa, T. & Gilbert-Barness, E. Smooth muscle cell depletion and collagen types in progeric arteries. Cardiovasc. Pathol. 10, 133–136 (2001).

    Article  CAS  PubMed  Google Scholar 

  22. Gerhard-Herman, M. et al. Mechanisms of premature vascular aging in children with Hutchinson–Gilford progeria syndrome. Hypertension 59, 92–97 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Olive, M. et al. Cardiovascular pathology in Hutchinson–Gilford progeria: correlation with the vascular pathology of aging. Arterioscler. Thromb. Vasc. Biol. 30, 2301–2309 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Varga, R. et al. Progressive vascular smooth muscle cell defects in a mouse model of Hutchinson–Gilford progeria syndrome. Proc. Natl Acad. Sci. USA 103, 3250–3255 (2006).

    Article  CAS  PubMed  Google Scholar 

  25. Rusiñol, A. E. & Sinensky, M. S. Farnesylated lamins, progeroid syndromes and farnesyl transferase inhibitors. J. Cell Sci. 119, 3265–3272 (2006).

    Article  PubMed  Google Scholar 

  26. Young, S. G., Yang, S. H., Davies, B. S. J., Jung, H.-J. & Fong, L. G. Targeting protein prenylation in progeria. Sci. Transl. Med. 5, 171ps173 (2013).

    Article  Google Scholar 

  27. Evangelisti, C., Cenni, V. & Lattanzi, G. Potential therapeutic effects of the MTOR inhibitors for preventing ageing and progeria-related disorders. Br. J. Clin. Pharmacol. 82, 1229–1244 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wu, D., Yates, P. A., Zhang, H. & Cao, K. Comparing lamin proteins post-translational relative stability using a 2A peptide-based system reveals elevated resistance of progerin to cellular degradation. Nucleus 7, 585–596 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Scaffidi, P. & Misteli, T. Reversal of the cellular phenotype in the premature aging disease Hutchinson–Gilford progeria syndrome. Nat. Med. 11, 440–445 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Osorio, F. G. et al. Splicing-directed therapy in a new mouse model of human accelerated aging. Sci. Transl. Med. 3, 106ra107 (2011).

    Article  PubMed  Google Scholar 

  31. Shimi, T. et al. The role of nuclear lamin B1 in cell proliferation and senescence. Genes Dev. 25, 2579–2593 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Dreesen, O. et al. Lamin B1 fluctuations have differential effects on cellular proliferation and senescence. J. Cell Biol. 200, 605–617 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Moir, R. D., Montag-Lowy, M. & Goldman, R. D. Dynamic properties of nuclear lamins: lamin B is associated with sites of DNA replication. J. Cell Biol. 125, 1201–1212 (1994).

    Article  CAS  PubMed  Google Scholar 

  34. Van Berlo, J. H. et al. A-type lamins are essential for TGF-β1 induced PP2A to dephosphorylate transcription factors. Hum. Mol. Genet. 14, 2839–2849 (2005).

    Article  CAS  PubMed  Google Scholar 

  35. Skvortsov, S. et al. Proteomics profiling of microdissected low- and high-grade prostate tumors identifies lamin A as a discriminatory biomarker. J. Proteome Res. 10, 259–268 (2011).

    Article  CAS  PubMed  Google Scholar 

  36. Aljada, A. et al. Altered lamin A/C splice variant expression as a possible diagnostic marker in breast cancer. Cell. Oncol. (Dordr.) 39, 161–174 (2016).

    Article  CAS  Google Scholar 

  37. Fong, L. G. et al. Prelamin A and lamin A appear to be dispensable in the nuclear lamina. J. Clin. Invest. 116, 743–752 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Guo, Y., Kim, Y., Shimi, T., Goldman, R. D. & Zheng, Y. Concentration-dependent lamin assembly and its roles in the localization of other nuclear proteins. Mol. Biol. Cell 25, 1287–1297 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Kole, R. & Krieg, A. M. Exon skipping therapy for Duchenne muscular dystrophy. Adv. Drug Deliv. Rev. 87, 104–107 (2015).

    Article  CAS  PubMed  Google Scholar 

  40. Maharshi, V. & Hasan, S. Nusinersen: the first option beyond supportive care for spinal muscular atrophy. Clin. Drug Investig. 37, 807–817 (2017).

    Article  CAS  PubMed  Google Scholar 

  41. Jearawiriyapaisarn, N. et al. Sustained dystrophin expression induced by peptide-conjugated morpholino oligomers in the muscles of mdx mice. Mol. Ther. 16, 1624–1629 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sazani, P. et al. Nuclear antisense effects of neutral, anionic and cationic oligonucleotide analogs. Nucleic Acids Res. 29, 3965–3974 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the NIH Intramural Research Program. F.S.C., M.R.E., W.A.C., J.G.-J., Y. B. and U.L.T. were funded by the National Human Genome Research Institute (no. HG200305). K.C. was supported by NIHR01 HL126784. F.S.C. and J.G.-J. were funded by a Progeria Research Foundation grant (no. 2015-57). L.B.G. was funded by the Progeria Research Foundation. We thank T. Yan for assistance with data analysis.

Author information

Authors and Affiliations

Authors

Contributions

M.R.E, W.A.C., G.H., D.V.M., R.K., K.C., U.L.T. and F.S.C. designed the research. M.R.E., W.A.C., K.C., J.G.-J., S.C. and U.L.T. performed the experiments. M.R.E., W.A.C., K.C., N.N., P.M.Z., S.C., Y.B. and M.A.E. analyzed the data. M.R.E., L.B.G. and F.S.C. supervised the project. M.R.E., W.A.C. and F.S.C. wrote the manuscript with input from all other authors.

Corresponding author

Correspondence to Francis S. Collins.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Medicine thanks Thomas Glover and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Joao Monteiro was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 PPMO structure.

SRP-2001 is a peptide-conjugated phosphorodiamidate morpholino oligomer (PPMO) comprising a cell-penetrating peptide (CPP) covalently linked to a phosphorodiamidate morpholino oligomer (PMO). The PMO contains 25 morpholino subunits each bearing a nucleobase forming the sequence 5’-GAGGAGATGGGTCCACCCACCTGGG-3’. The conjugated peptide comprises the amino acid sequence R6G, where R is arginine and G is glycine. The glycine residue covalently links the CPP to the PMO by an amide bound to the 3’ end of the PMO. Mass spectrometric characterization was performed to confirm the structure and sequence of SRP-2001.

Extended Data Fig. 2 Quantitative PCR screening of candidate PPMOs for targeted reduction of progerin in proband fibroblasts.

Quantitation of LMNA (A), Progerin (P), and LMNC (C) expression by ddPCR analysis of Classic HGPS (LMNA c.1824C > T) fibroblasts treated with 6uM candidate PPMOs. a, Transcript levels relative to TFRC. b, Each isoforms’ fraction relative to all LMNA transcripts. c, Each isoforms’ transcript level relative to LMNC transcripts. The greatest reduction of progerin transcripts in culture was achieved with the centrally located morpholino (SRP-2001). A, LMNA; P, Progerin; C, LMNC; WT, oligo complementary to normal LMNA sequence; CTRL, scrambled control; Values and error bars represent mean ± SD determined from triplicate reactions of biological triplicates. * p < 0.05, ** p < 0.01, *** p < 0.001 versus transcripts or protein from cells receiving saline (NT).

Extended Data Fig. 3 Western immunoblot screening of candidate PPMOs for targeted reduction of progerin in proband fibroblasts.

Quantitation of LMNA (A), Progerin (P), and LMNC (C) protein from immunoblots of lysates from Classic HGPS (LMNA c.1824C > T) fibroblasts treated with 6uM candidate PPMOs. Data was derived from the analysis shown in Fig. 1d and is expressed as isoform levels relative to ACTB (a), the isoforms’ fraction relative to all LMNA gene products (b), and the isoforms’ level relative to LMNC protein (c). The greatest reduction of progerin protein in culture was achieved with the centrally located morpholino (SRP-2001). A, LMNA; P, progerin; C, LMNC; WT, oligo complementary to normal LMNA sequence; CTRL, scrambled control; Values and error bars represent mean ± SD determined from biological triplicates shown in Fig. 1d. * p < 0.05, ** p < 0.01, *** p < 0.001 versus transcripts or protein from cells receiving saline NT).

Extended Data Fig. 4 Quantitative PCR screening of SRP-2001 for targeted reduction of progerin in Classic and Non-classic HGPS cell lines.

a, Transcript copy number in normal control (NL Control), Classic HGPS (c.1824C>T), and Non-classic HGPS (c.1822G>A, c.1968+1G>A, c.1968+2T>C, c.1968+5G>C) fibroblasts following 2 weeks of treatment with SRP-2001. b, Individual LMNA transcripts expressed as a fraction of all total LMNA-derived transcripts in each cell line. c, Full-length LMNA and progerin transcript levels relative to LMNC transcripts. A, LMNA; P, progerin; C, LMNC; Values and error bars represent mean ± SD determined from triplicate reactions of biological triplicates. * p < 0.05; ** p < 0.01; *** p < 0.001 vs same transcript in cells receiving saline only (NT).

Extended Data Fig. 5 Reduction of progerin protein by SRP-2001 in Classic and Non-classic HGPS cell lines.

a, Western immunoblots of normal control (NL Control), Classic HGPS (c.1824C>T), and Non-classic HGPS (c.1822G>A, c.1968+1G>A, c.1968+2T>C, c.1968+5G>C) fibroblasts following 2 weeks of treatment with SRP-2001. b, Quantitation of LMNA gene products relative to ACTB. c, Quantitation of LMNA, progerin and LMNC isoforms expressed as a fraction of all total LMNA-derived gene products in each cell line. d, Full-length LMNA and progerin protein levels relative to LMNC. A, LMNA; P, progerin; C, LMNC; Graphs represent mean ± SD determined from biological triplicates shown in panel A. * p < 0.05; ** p < 0.01; *** p < 0.001 vs same protein isoform in cells receiving saline only (NT).

Extended Data Fig. 6 SRP-2001 reduces expression of transgene-derived progerin in murine aortas.

a, Quantitative PCR analysis of transgene transcript levels in aortas of LMNAG/G mice receiving long-term treatment with saline (vehicle) or 60 mg/kg SRP-2001. Transcripts are quantitated relative to Hprt expression, as a fraction of all transgene-derived LMNA transcripts or relative to transgene-derived LMNC transcripts. Data are presented as mean values ± SD determined from 12 independent samples per treatment group (n = 6 males, 6 females). b, Western immunoblots of A-type lamins and beta actin (ACTB) immunoprecipitated from aorta tissue. c, Quantitation of A-type lamins from immunoblots expressed relative to beta actin (ACTB), as a fraction of all A-type lamins and relative to LMNC. Data are presented as mean values ± SD determined from 12 independent samples per treatment group (n = 6 males, 6 females), shown in panel b. A, LMNA; P, progerin; C, LMNC; * p < 0.05; ** p < 0.01; *** p < 0.001 vs same transcript or protein in mice receiving saline only (vehicle).

Extended Data Fig. 7 SRP-2001 reduces expression of transgene-derived progerin in murine hearts.

a, Quantitative PCR analysis of transgene transcript levels in hearts of LMNAG/G mice receiving long-term treatment with saline (vehicle) or 60 mg/kg SRP-2001. Transcripts are quantitated relative to Hprt expression, as a fraction of all transgene-derived LMNA transcripts or relative to transgene-derived LMNC transcripts. Data are presented as mean values ± SD determined from 12 independent samples per treatment group (n = 6 males, 6 females). b, Western immunoblots of A-type lamins and smooth muscle actin (SMA) immunoprecipitated from heart tissue. c, Quantitation of A-type lamins from immunoblots expressed relative to smooth muscle actin (SMA), as a fraction of all A-type lamins and relative to LMNC. Data are presented as mean values ± SD determined from 12 independent samples per treatment group (n = 6 males, 6 females), shown in panel b. A, LMNA; P, progerin; C, LMNC; * p < 0.05; ** p < 0.01; *** p < 0.001 vs same transcript or protein in mice receiving saline only (vehicle).

Extended Data Fig. 8 SRP-2001 reduces expression of transgene-derived progerin in murine liver.

a, Quantitative PCR analysis of transgene transcripts in liver tissue of LMNAG/G mice receiving long-term treatment with saline (vehicle) or 60 mg/kg SRP-2001. Expression of LMNA (A), progerin (P) and LMNC (C) is quantitated relative to Hprt expression, as a fraction of all transgene-derived LMNA transcripts or relative to transgene-derived LMNC transcripts. Data are presented as mean values ± SD determined from 12 independent samples per treatment group (n = 6 males, 6 females). b, Western immunoblots of A-type lamins and beta actin (ACTB) immunoprecipitated from liver homogenates. c, Quantitation of A-type lamins from immunoblots expressed relative to beta actin (ACTB), as a fraction of all A-type lamins and relative to LMNC. Data are presented as mean values ± SD determined from 12 independent samples per treatment group (n = 6 males, 6 females), shown in panel b. * p < 0.05; ** p < 0.01; *** p < 0.001 vs same transcript or protein in mice receiving saline only (vehicle).

Extended Data Fig. 9 SRP-2001 reduces progerin transcripts in multiple mesenchyme-derived murine tissues.

Quantitative PCR analysis of transgene transcripts in a, quadriceps, b, femoral bone, and c, kidneys of LMNAG/G mice receiving long-term treatment with saline (vehicle) or 60 mg/kg SRP-2001. Expression of LMNA (A), progerin (P) and LMNC (C) is quantitated relative to Hprt expression, as a fraction of all transgene-derived LMNA transcripts or relative to transgene-derived LMNC transcripts. Data are presented as mean values ± SD determined from 12 independent samples per treatment group (n = 6 males, 6 females). * p < 0.05; ** p < 0.01; *** p < 0.001 vs same transcript or protein in mice receiving saline only (vehicle).

Extended Data Fig. 10 Histologic analysis of kidneys shows minimal toxicity due to long-term treatment with SRP-2001.

Representative images of haemotoxylin and eosin (HE) staining of sectioned kidneys from treated (n = 12) and untreated (n = 12) mice. Mice receiving long-term treatment with SRP-2001 developed irregular cortical surfaces (ic), degenerate tubules (dt) and basophilic granules (bg) in tubular epithelial cells. Two sections per tissue sample were analyzed.

Supplementary information

Supplementary Information

Supplementary Tables 1–3 and Figs. 1–3.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Erdos, M.R., Cabral, W.A., Tavarez, U.L. et al. A targeted antisense therapeutic approach for Hutchinson–Gilford progeria syndrome. Nat Med 27, 536–545 (2021). https://doi.org/10.1038/s41591-021-01274-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-021-01274-0

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