4.8 Article

Induction of muscle-regenerative multipotent stem cells from human adipocytes by PDGF-AB and 5-azacytidine

Journal

SCIENCE ADVANCES
Volume 7, Issue 3, Pages -

Publisher

AMER ASSOC ADVANCEMENT SCIENCE
DOI: 10.1126/sciadv.abd1929

Keywords

-

Funding

  1. Endeavour International Postgraduate Research scholarship from the Australian Government
  2. International Postgraduate Student scholarship from UNSW
  3. Prince of Wales Clinical School
  4. UNSW
  5. St. Vincent's Clinic Foundation
  6. Arrow BMT Foundation
  7. Australian Research Council [FT180100417]
  8. Olivia Lambert Foundation
  9. NHMRC [APP1091261, APP1119152]
  10. MTPConnect MedTech and Pharma Growth Centre as part of the Australian Government [PRJ2017-55, BMTH06]
  11. Peter Doherty Fellowship from the National Health and Medical Research Council of Australia
  12. Cancer Institute NSW Early Career Fellowship
  13. Anthony Rothe Memorial Trust
  14. Jasper Medical Innovations (Sydney, Australia)
  15. National Health and Medical Research Council of Australia [APP1139811]
  16. Gilead Sciences

Ask authors/readers for more resources

By optimizing culture conditions, human adipocyte-derived induced multipotent stem cells have been successfully generated and can regenerate tissues in a context-dependent manner without ectopic or neoplastic growth.
Terminally differentiated murine osteocytes and adipocytes can be reprogrammed using platelet-derived growth factor-AB and 5-azacytidine into multipotent stem cells with stromal cell characteristics. We have now optimized culture conditions to reprogram human adipocytes into induced multipotent stem (iMS) cells and characterized their molecular and functional properties. Although the basal transcriptomes of adipocyte-derived iMS cells and adipose tissue-derived mesenchymal stem cells were similar, there were changes in histone modifications and CpG methylation at cis-regulatory regions consistent with an epigenetic landscape that was primed for tissue development and differentiation. In a non-specific tissue injury xenograft model, iMS cells contributed directly to muscle, bone, cartilage, and blood vessels, with no evidence of teratogenic potential. In a cardiotoxin muscle injury model, iMS cells contributed specifically to satellite cells and myofibers without ectopic tissue formation. Together, human adipocyte-derived iMS cells regenerate tissues in a context-dependent manner without ectopic or neoplastic growth.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available