Mesenchymal stromal cell-dependent reprogramming of Kupffer cells is mediated by TNF-α and PGE2 and is crucial for liver transplant tolerance

2015 ◽  
Vol 62 (3) ◽  
pp. 292-305 ◽  
Author(s):  
Yu You ◽  
Jiqin Zhang ◽  
Jianping Gong ◽  
Yupei Chen ◽  
Yue Li ◽  
...  
2020 ◽  
Vol 9 (4) ◽  
pp. 427-432 ◽  
Author(s):  
Federica Casiraghi ◽  
Norberto Perico ◽  
Eliana Gotti ◽  
Marta Todeschini ◽  
Marilena Mister ◽  
...  

Nano LIFE ◽  
2015 ◽  
Vol 05 (02) ◽  
pp. 1550001 ◽  
Author(s):  
Andrea Gray ◽  
Ileana Marrero-Berrios ◽  
Mehdi Ghodbane ◽  
Timothy Maguire ◽  
Jonathan Weinberg ◽  
...  

Anti-fibrotic and tissue regenerative mesenchymal stromal cell (MSC) properties are largely mediated by secreted cytokines and growth factors. MSCs are implanted to augment joint cartilage replacement and to treat diabetic ulcers and burn injuries simultaneously with local anesthetics, which reduce pain. However, the effect of anesthetics on therapeutic human MSC secretory function has not been evaluated. In order to assess the effect of local anesthetics on the MSC secretome, a panel of four anesthetics with different potencies — lidocaine, procaine, ropivacaine and bupivacaine — was evaluated. Since injured tissues secrete inflammatory cytokines, the effects of anesthetics on MSCs stimulated with tumor necrosis factor (TNF)-α and interferon (IFN)-γ were also measured. Dose dependent and anesthesia specific effects on cell viability, post exposure proliferation and secretory function were quantified using alamar blue reduction and immunoassays, respectively. Computational pathway analysis was performed to identify upstream regulators and molecular pathways likely associated with the effects of these chemicals on the MSC secretome. Our results indicated while neither lidocaine nor procaine greatly reduced unstimulated cell viability, ropivacaine and bupivacaine induced dose dependent viability decreases. This pattern was exaggerated in the simulated inflammatory environment. The reversibility of these effects after withdrawal of the anesthetics was attenuated for TNF-α/IFN-γ-stimulated MSCs exposed to ropivacaine and bupivacaine. In addition, secretome analysis indicated that constitutive secretion changes were clearly affected by both anesthetic alone and anesthetic plus TNFα/IFNγ cell stimulation, but the secretory pattern was drug specific and did not necessarily coincide with viability changes. Pathway analysis identified different intracellular regulators for stimulated and unstimulated MSCs. Within these groups, ropivacaine and bupivacaine appeared to act on MSCs similarly via the same regulatory mechanisms. Given the variable effect of local anesthetics on MSC viability and function, these studies underscore the need to evaluate MSC in the presence of medications, such as anesthetics, that are likely to accompany cell implantation.


Sign in / Sign up

Export Citation Format

Share Document