Mesenchymal stem cells alleviate oxidative stress–induced mitochondrial dysfunction in the airways

2018 ◽  
Vol 141 (5) ◽  
pp. 1634-1645.e5 ◽  
Author(s):  
Xiang Li ◽  
Charalambos Michaeloudes ◽  
Yuelin Zhang ◽  
Coen H. Wiegman ◽  
Ian M. Adcock ◽  
...  
2019 ◽  
Vol 20 (18) ◽  
pp. 4545 ◽  
Author(s):  
Yeo Min Yoon ◽  
Hyung Joo Kim ◽  
Jun Hee Lee ◽  
Sang Hun Lee

Human mesenchymal stem cells (hMSCs) are a potent source of cell-based regenerative therapeutics used to treat patients with ischemic disease. However, disease-induced oxidative stress disrupts mitochondrial homeostasis in transplanted hMSCs, resulting in hMSC apoptosis and reducing their efficacy post-transplantation. To address this issue, we evaluated the effects of melatonin on cellular defense mechanisms and mitophagy in hMSCs subjected to oxidative stress. H2O2-induced oxidative stress increases the levels of reactive oxygen species and reduces membrane potential in hMSCs, leading to mitochondrial dysfunction and cell death. Oxidative stress also decreases the expression of 70-kDa heat shock protein 1L (HSPA1L), a molecular chaperone that assists in the recruitment of parkin to the autophagosomal mitochondrial membrane. Decreased expression of HSPA1L destabilizes parkin, thereby impairing mitophagy. Our results indicate that treating hMSCs with melatonin significantly inhibited mitochondrial dysfunction induced by oxidative stress, which decreased hMSCs apoptosis. In damaged hMSCs, treatment with melatonin increased the levels of HSPA1L, which bound to parkin. The interaction between HSPA1L and parkin increased membrane potential and levels of oxidative phosphorylation, resulting in enhanced mitophagy. Our results indicate that melatonin increased the expression of HSPA1L, thereby upregulating mitophagy and prolonging cell survival under conditions of oxidative stress. In this study, we have shown that melatonin, a readily available compound, can be used to improve hMSC-based therapies for patients with pathologic conditions involving oxidative stress.


Author(s):  
Elham Shiri ◽  
Parichehr Pasbakhsh ◽  
Maryam Borhani‑Haghighi ◽  
Zohreh Alizadeh ◽  
Saied Nekoonam ◽  
...  

2020 ◽  
Vol 79 (Suppl 1) ◽  
pp. 483.2-484
Author(s):  
L. Zaripova ◽  
A. Midgley ◽  
S. Christmas ◽  
E. Baildam ◽  
R. Oldershaw

Background:Juvenile idiopathic arthritis (JIA) is a well-known chronic rheumatic disease of childhood characterised by progressive joint destruction and severe systemic complications.Immune cells are known to trigger the pathophysiological cascade in JIA, but there is little information regarding the contribution made by Mesenchymal stem cells (MSCs). These cells are able to modulate the immune response and decrease the level of pro-inflammatory cytokines. With addition of regenerative property it makes MSCs potential candidates for clinical application as immunosuppressants in treatment of autoimmune diseases.Objectives:To investigate MSCs proliferation, viability and immunomodulatory function in JIA and healthy children.Methods:MSCs were separated from peripheral blood (PB) and synovial fluid (SF) of JIA patients and healthy controls. Cell proliferation rate was counted by Population doublings per day (PDD) during 9 days, in the last of which alamarBlue™ assays were performed to assess cell viability. Due to measure senescence MSCs were stained with SA-β-galactosidase. Immunofluorescence was used to examine the expression of p16, p21, p53. Oxidative stress was measured with DCFH-DA. Cell cycle analysis was evaluated with Propidium Iodide and analysed by Accuri® C6 Flow Cytometer.Commercially-available bone marrow mesenchymal stem cells (BM-MSCs) were treated with graded concentrations of pro-inflammatory cytokines (0.1-100 ng/ml) with following examination of cell viability. Mixed lymphocyte reactions (MLR) were performed to measure MSC immunomodulatory abilityin vitro.Results:The growth kinetics of JIA-MSCs were different from healthy controls. JIA-MSCs divided slowly and appeared disorganised with large cytoplasm and loads of outgrowth. They demonstrated a decrease in cell proliferation (negative PDD) and metabolic activity. Difference in growth kinetics and metabolic activity were found inside the JIA PB group with some evidence of response following biological treatment. Thus, PB-MSCs from patients treated with TNFi and anti-IL6 medications had notably higher cell proliferation and metabolic activity against JIA patients received other therapy. Considering this difference, it was hypothesised that cytokines obtained in a high amount in PB and SF of JIA patients may influence MSCs viability. To prove this BM-MSCs were treated with cytokines and demonstrated a dose-dependent decrease in metabolic activity significantly after TNFα and IL1, no significantly after treatment with IL6. Both BM-MSCs treated with cytokines and JIA-MSCs displayed high level of reactive oxygen species.Cell cycle analysis revealed that JIA-MSCs were arrested in G0/G1 phase with low number of mitotic cells. In addition, the number of senescence-associated SA-β-gal-positive cells was notably higher in JIA-MSCs. Furthermore, JIA-MSCs expressed high level of immunofluorescence for p16, p21 and p53 which played an important role in regulating the senescence progress of MSCs.Results of MLR showed the ability of BM-MSCs to decrease the percentage of activated T-helpers, T-suppressors, B-cells and natural killers proliferation, while JIA-MSCs lost this property.Conclusion:Taken together current research has demonstrated that under the influence of proinflammatory cytokines JIA-MSCs suffered from oxidative stress and disruption of metabolic activity acquire senescent morphology, shorten of telomere length, arrest in G0 phase of cell cycle and finally loss of immune regulation. We are continuing our research to determine the mechanisms that are responsible for the impaired phenotype with the aim of identifying new therapeutic strategies for the treatment of JIA.Disclosure of Interests: :None declared


Stem Cells ◽  
2009 ◽  
pp. N/A-N/A ◽  
Author(s):  
Xiaohong Wang ◽  
Tiemin Zhao ◽  
Wei Huang ◽  
Tao Wang ◽  
Jiang Qian ◽  
...  

Biomolecules ◽  
2018 ◽  
Vol 8 (4) ◽  
pp. 181 ◽  
Author(s):  
Wang Liao ◽  
Yuqiu Zheng ◽  
Wenli Fang ◽  
Shaowei Liao ◽  
Ying Xiong ◽  
...  

Alzheimer’s disease (AD) is a devastating neurodegenerative disease with limited treatment options and no cure. Beta-amyloid (Aβ) is a hallmark of AD that has potent neurotoxicity in neural stem cells (NSCs). Dual specificity phosphatase 6 (DUSP6) is a member of the mitogen-activated protein kinases (MAPKs), which is involved in regulating various physiological and pathological processes. Whether DUSP6 has a protective effect on Aβ-induced NSC injury remains to be explored. C17.2 neural stem cells were transfected with DUSP6-overexpressed plasmid. NSCs with or without DUSP6 overexpression were administrated with Aβ25–35 at various concentrations (i.e., 0, 2.5, 5 μM). DUSP6 expression after Aβ treatment was detected by Real-Time Polymerase Chain Reaction (RT-PCR) and Western blot and cell vitality was examined by the CCK8 assay. The oxidative stress (intracellular reactive oxygen species (ROS) and malondialdehyde (MDA)), endoplasmic reticulum stress (ER calcium level) and mitochondrial dysfunction (cytochrome c homeostasis) were tested. The expression of p-ERK1/2 and ERK1/2 were assayed by Western blot. Our results showed that Aβ decreased the expression of DUSP6 in a dose-dependent manner. The overexpression of DUSP6 increased the cell vitality of NSCs after Aβ treatment. Oxidative stress, ER stress, and mitochondrial dysfunction induced by Aβ could be restored by DUSP6 overexpression. Additionally, the Aβ-induced ERK1/2 activation was reversed. In summary, DUSP6 might have a neuroprotective effect on Aβ-induced cytotoxicity, probably via ERK1/2 activation.


2014 ◽  
Vol 19 (5) ◽  
pp. 685-693 ◽  
Author(s):  
Bahareh Bahmani ◽  
Mehryar Habibi Roudkenar ◽  
Raheleh Halabian ◽  
Ali Jahanian-Najafabadi ◽  
Fatemeh Amiri ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document