Article Text

Download PDFPDF
Extended report
PPARβ/δ directs the therapeutic potential of mesenchymal stem cells in arthritis
  1. P Luz-Crawford1,2,
  2. N Ipseiz3,
  3. G Espinosa-Carrasco1,2,
  4. A Caicedo1,2,4,
  5. G Tejedor1,2,
  6. K Toupet1,2,
  7. J Loriau1,2,
  8. C Scholtysek3,
  9. C Stoll4,
  10. M Khoury5,
  11. D Noël1,2,6,
  12. C Jorgensen1,2,6,
  13. G Krönke3,
  14. F Djouad1,2
  1. 1Inserm U1183, Montpellier, France
  2. 2Université Montpellier, Montpellier, France
  3. 3Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany
  4. 4Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud, Escuela de Medicina, Hospital de los Valles, Quito Ecuador
  5. 5Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
  6. 6Service d'Immuno-Rhumatologie Thérapeutique, Hôpital Lapeyronie, Montpellier, France
  1. Correspondence to Dr Farida Djouad, Inserm U1183, Hôpital Saint-Eloi, IRMB, 80 avenue Augustin Fliche, Montpellier 34295, cedex 5, France; farida.djouad{at}inserm.fr

Abstract

Objectives To define how peroxisome proliferator-activated receptor (PPAR) β/δ expression level in mesenchymal stem cells (MSCs) could predict and direct both their immunosuppressive and therapeutic properties. PPARβ/δ interacts with factors such as nuclear factor-kappa B (NF-κB) and regulates the expression of molecules including vascular cell adhesion molecule (VCAM)-1 and intercellular adhesion molecule (ICAM)-1. Since these molecules are critical for MSC function, we investigated the role of PPARβ/δ on MSC immunosuppressive properties.

Methods We either treated human MSCs (hMSCs) with the irreversible PPARβ/δ antagonist (GSK3787) or derived MSCs from mice deficient for PPARβ/δ (PPARβ/δ−/− MSCs). We used the collagen-induced arthritis (CIA) as model of immune-mediated disorder and the MSC-immune cell coculture assays.

Results Modulation of PPARβ/δ expression in hMSCs either using GSK3787 or hMSCs from different origin reveals that MSC immunosuppressive potential is inversely correlated with Ppard expression. This was consistent with the higher capacity of PPARβ/δ−/− MSCs to inhibit both the proliferation of T lymphocytes, in vitro, and arthritic development and progression in CIA compared with PPARβ/δ+/+ MSCs. When primed with proinflammatory cytokines to exhibit an immunoregulatory phenotype, PPARβ/δ−/− MSCs expressed a higher level of mediators of MSC immunosuppression including VCAM-1, ICAM-1 and nitric oxide (NO) than PPARβ/δ+/+ MSCs. The enhanced NO2 production by PPARβ/δ−/− MSCs was due to the increased retention of NF-κB p65 subunit on the κB elements of the inducible nitric oxide synthase promoter resulting from PPARβ/δ silencing.

Conclusions Our study is the first to show that the inhibition or knockdown of PPARβ/δ in MSCs primes their immunoregulatory functions. Thus, the regulation of PPARβ/δ expression provides a new strategy to generate therapeutic MSCs with a stable regulatory phenotype.

  • Arthritis
  • Autoimmune Diseases
  • Treatment

Statistics from Altmetric.com

Request Permissions

If you wish to reuse any or all of this article please use the link below which will take you to the Copyright Clearance Center’s RightsLink service. You will be able to get a quick price and instant permission to reuse the content in many different ways.

Footnotes

  • Handling editor Tore K Kvien

  • PL-C, NI contributed equally.

  • Twitter Follow Andres Caicedo at @AveCaicedo

  • Contributors Design of the study: PL-C and FD. Acquisition of data: PL-C, NI, AC, GT, KT, JL, CSc, CSt and MK. Interpretation of data: PL-C, NI, GEC, AC, GT, KT, JL, CSc, CSt, MK, DN, CJ, GK and FD. Manuscript preparation: PL-C, NI, DN, CJ, GK and FD. All authors reviewed the manuscript and gave final approval for the work.

  • Competing interests None declared.

  • Provenance and peer review Not commissioned; externally peer reviewed.