scholarly journals Caspase‐8 regulates the expression of pro‐ and anti‐inflammatory cytokines in human bone marrow‐derived mesenchymal stromal cells

2016 ◽  
Vol 4 (3) ◽  
pp. 327-337 ◽  
Author(s):  
Siv H. Moen ◽  
Marita Westhrin ◽  
Muhammad Zahoor ◽  
Nikolai N. Nørgaard ◽  
Hanne Hella ◽  
...  
2015 ◽  
Vol 230 (6) ◽  
pp. 1258-1269 ◽  
Author(s):  
Sergio B. Sesia ◽  
Ralph Duhr ◽  
Carolina Medeiros da Cunha ◽  
Atanas Todorov ◽  
Stefan Schaeren ◽  
...  

Author(s):  
Tayebeh Mahmoudi ◽  
Ali Jalili ◽  
Kamal Abdolmohammadi ◽  
Shohreh Fakhari ◽  
Fatemeh Pahlavan ◽  
...  

Purpose: Acute pancreatitis (AP) which is distinguished by local pancreatic necrosis, following by systemic organ failure is known as an inflammatory disease. Up to now, there are only a few treatment options accessible for patients suffering from AP. In this study, we aimed to examine the anti-inflammatory capacities of human bone marrow-derived mesenchymal stromal cells (hBM-MSC) in a detailed AP model experiment. Methods: AP was induced in C57BL/6 mice by intraperitoneal administration of cerulein (100 µg/kg/h × 7 doses) at intervals of 1 hour (h). Then, 2×105 MSCs were infused in the AP mice by tail vein 6 h after the last cerulein injection. Mice were sacrificed 12 h following the injection of hBM-MSC, and blood samples and pancreas tissues were obtained. Results: We first determined the presence of transplanted hBM-MSC in the pancreas of mice with AP, but not the control mice. Our data indicate that administration of hBM-MSCs to mice with AP lead to (i) decreased serum levels of amylase, lipase and myeloperoxidase activities, (ii) downregulation of proinflammatory cytokine, macrophage inflammatory protein 2 (MIP-2), and (iii) upregulation of the anti-inflammatory cytokine, interleukin 10 (IL-10). Moreover, hBM-MSC administration results in notably attenuated cerulein-induced histopathological alternations and edema. Conclusion: we demonstrate that hBM-MSC attenuates AP signs and indicating that hMB-MSC therapy could be a suitable approach for the treatment of inflammatory disease such as AP.


Heliyon ◽  
2021 ◽  
Vol 7 (3) ◽  
pp. e06517
Author(s):  
Lyudmila M. Mezhevikina ◽  
Dmitriy A. Reshetnikov ◽  
Maria G. Fomkina ◽  
Nurbol O. Appazov ◽  
Saltanat Zh. Ibadullayeva ◽  
...  

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Shun Takao ◽  
Taku Nakashima ◽  
Takeshi Masuda ◽  
Masashi Namba ◽  
Shinjiro Sakamoto ◽  
...  

Abstract Background Mesenchymal stromal cells (MSCs) are a potential therapeutic tool for pulmonary fibrosis. However, ex vivo MSC expansion using serum poses risks of harmful immune responses or unknown pathogen infections in the recipients. Therefore, MSCs cultured in serum-free media (SF-MSCs) are ideal for clinical settings; however, their efficacy in pulmonary fibrosis is unknown. Here, we investigated the effects of SF-MSCs on bleomycin-induced pulmonary inflammation and fibrosis compared to those of MSCs cultured in serum-containing media (S-MSCs). Methods SF-MSCs and S-MSCs were characterized in vitro using RNA sequence analysis. The in vivo kinetics and efficacy of SF-MSC therapy were investigated using a murine model of bleomycin-induced pulmonary fibrosis. For normally distributed data, Student’s t test and one-way repeated measures analysis of variance followed by post hoc Tukey’s test were used for comparison between two groups and multiple groups, respectively. For non-normally distributed data, Kruskal–Wallis and Mann–Whitney U tests were used for comparison between groups, using e Bonferroni’s correction for multiple comparisons. All tests were two-sided, and P < 0.05 was considered statistically significant. Results Serum-free media promoted human bone marrow-derived MSC expansion and improved lung engraftment of intravenously administered MSCs in recipient mice. SF-MSCs inhibited the reduction in serum transforming growth factor-β1 and the increase of interleukin-6 in both the serum and the bronchoalveolar lavage fluid during bleomycin-induced pulmonary fibrosis. SF-MSC administration increased the numbers of regulatory T cells (Tregs) in the blood and lungs more strongly than in S-MSC administration. Furthermore, SF-MSCs demonstrated enhanced antifibrotic effects on bleomycin-induced pulmonary fibrosis, which were diminished by antibody-mediated Treg depletion. Conclusions SF-MSCs significantly suppressed BLM-induced pulmonary inflammation and fibrosis through enhanced induction of Tregs into the lungs and corrected the dysregulated cytokine balance. Therefore, SF-MSCs could be a useful tool for preventing pulmonary fibrosis progression without the demerits of serum use.


Sign in / Sign up

Export Citation Format

Share Document