Abstract

BackgroundMesenchymal stromal cells (MSCs) have been extensively used in the clinic due to their exquisite tissue repair capacity. However, they also hold promise in the field of cellular vaccination as they can behave as conditional antigen presenting cells in response to interferon (IFN)-gamma treatment under a specific treatment regimen. This suggests that the immune function of MSCs can be pharmacologically modulated. Given the capacity of the agonist pyrimido-indole derivative UM171a to trigger the expression of various antigen presentation-related genes in human hematopoietic progenitor cells, we explored the potential use of UM171a as a means to pharmacologically instill and/or promote antigen presentation by MSCs.MethodsBesides completing a series of flow-cytometry-based phenotypic analyses, several functional antigen presentation assays were conducted using the SIINFEKL-specific T-cell clone B3Z. Anti-oxidants and electron transport chain inhibitors were also used to decipher UM171a’s mode of action in MSCs. Finally, the potency of UM171a-treated MSCs was evaluated in the context of therapeutic vaccination using immunocompetent C57BL/6 mice with pre-established syngeneic EG.7T-cell lymphoma.ResultsTreatment of MSCs with UM171a triggered potent increase in H2-Kb cell surface levels along with the acquisition of antigen cross-presentation abilities. Mechanistically, such effects occurred in response to UM171a-mediated production of mitochondrial-derived reactive oxygen species as their neutralization using anti-oxidants or Antimycin-A mitigated MSCs’ ability to cross-present antigens. Processing and presentation of the immunogenic ovalbumin-derived SIINFEKL peptide was caused by de novo expression of the Psmb8 gene in response to UM171a-triggered oxidative stress. When evaluated for their anti-tumoral properties in the context of therapeutic vaccination, UM171a-treated MSC administration to immunocompetent mice with pre-established T-cell lymphoma controlled tumor growth resulting in 40% survival without the need of additional supportive therapy and/or standard-of-care.ConclusionsAltogether, our findings reveal a new immune-related function for UM171a and clearly allude to a direct link between UM171a-mediated ROS induction and antigen cross-presentation by MSCs. The fact that UM171a treatment modulates MSCs to become antigen-presenting cells without the use of IFN-gamma opens-up a new line of investigation to search for additional agents capable of converting immune-suppressive MSCs to a cellular tool easily adaptable to vaccination.Graphical abstract

Highlights

  • Mesenchymal stromal cells (MSCs) have been extensively used in the clinic due to their exquisite tissue repair capacity

  • UM171a is well tolerated by primary MSCs and triggers Major histocompatibility complex I (MHCI) up‐regulation The parent UM171a compound was initially discovered by a high-throughput screening assay designed for the identification of compounds capable of triggering leukemic stem cell proliferation [19]

  • When further studied to decipher its potential mode of action on human C­ D34+ hematopoietic stem cells (HSCs), UM171a was found to trigger a marked increase in the expression of several immune-related genes including human leukocyte antigens (HLA)-A and B—ortholog of the murine MHCI, beta 2-microglobulin (β2M) as well as the co-stimulatory molecule CD86 [21]

Read more

Summary

Introduction

Mesenchymal stromal cells (MSCs) have been extensively used in the clinic due to their exquisite tissue repair capacity They hold promise in the field of cellular vaccination as they can behave as conditional antigen presenting cells in response to interferon (IFN)-gamma treatment under a specific treatment regimen. Several characteristics support the extensive use of culture-adapted mesenchymal stromal cells (MSCs) as cellular biopharmaceuticals [1, 2] These include: (1) simple isolation protocols from small volumes of bone marrow (BM) aspirates, (2) ease of in vitro proliferation/expansion, (3) standard and defined culture medium, (4) low senescence through multiple passages, (5) ability to be gene modified, and (6) distinct in vivo migration capabilities toward damaged or inflamed tissues [1,2,3,4,5,6,7]. The design of novel pharmacological strategies capable of triggering consistent pro-inflammatory functions in both murine and human MSCs while bypassing the above-mentioned limitations remains a central goal for the development of immunestimulatory MSC-based therapeutics

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call