scholarly journals The Key Role of PML in IFN-α Induced Cellular Senescence of Human Mesenchymal Stromal Cells

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3611-3611
Author(s):  
Shan Fu ◽  
Jieping Wei ◽  
Binsheng Wang ◽  
He Huang

Abstract As immunomodulatory cytokines, Type 1 interferons (IFNs) have a long history of efficacy in treating chronic myeloid leukemia (CML). Recently, many research reported the combination of IFN-α and imatinib significantly increased the rates of molecular responses, comparing to single imatinib treatment. Related molecular mechanism may be the direct effect of IFN-alpha on stem cells. Therefore, IFN-α was renewed to be a vital candidate for CML treatment. Bone marrow mesenchymal stem cells (MSCs), which also be defined as mesenchymal stromal cells, are important to hematopoiesis. IFN-α was indicated as a potential inhibitor of MSCs; however the exact mechanism remains unclear. PML is known as a tumor suppressor, which locates at the downstream of IFN-α pathway. In our previous research, we have proved that PML stably expressed in human MSCs (hMSCs), which was important in maintaining the normal function of hMSCs. To our knowledge, although PML has been extensively studied in tumor cells, little is known about PML gene regulation in MSCs. In this study, we investigated the effect of IFN-α on hMSCs and the role of PML involved in this process. After approval by institutional review board, hMSCs were isolated from the bone marrow of volunteers and confirmed by flow cytometry. Cells were treated with different concentration of IFN-α up to 14 days. We found that IFN-α treated cells were growing slowly, and had a dramatically decreased number of colone in a dose dependent manner (Fig A). However, IFN-α did not induce significant cell apoptosis. Then a variety of senescence-associated detection was measured. hMSCs senescence induced by IFN-α had a dose and time dependent manner (Fig B). After treated with IFN-α at 1000 U/ml for 7 or 14 days, we found that up to 18% ± 1.1 or 27.56% ± 1.33 of hMSCs became SA-b-gal-positive as compared with 7.53% ± 0.55 or 6.47% ± 2.5 of untreated cells (P<0.05 for both). Real time PCR analysis proved this process by an increase in production of the senescence marker p53 and p21. Expression of PML was detected by real-time PCR and immunofluorescence in hMSCs treated with IFN-α. Consistent with other studies, mRNA expression of PML can be up-regulated by IFN-α in hMSCs. When cells were treated with IFN-α at 1000 U/ml for 7 or 14 days, PML gene expression in hMSCs was increased by more than 2 fold. At the same time, both the number and size of PML-NBs were increased markedly and had a concentration dependent manner. These results indicate that PML protein can be up regulated by IFN-α in hMSCs. Then, PML expression was inhibited using an RNAi-mediated PML knockdown system. After treated with IFN-α at 1000 U/ml for 7 days, hMSCs senescence can be rescued by the knocking down of PML. The percentage of SA-b-gal positive cells in PML knocking down hMSCs has a significant decrease as compared with cell transfected with control-RNAi (4.49% ±1.27 vs. 17.26% ± 1.44, P < 0.05) (Fig C). To further characterize the effect of PML on cellular senescence in hMSCs, PML-overexpressed hMSCs were used. 7 days post-transfection, PML overexpressing hMSCs were strongly positive for SA-b-gal activity (47.43%±3.8), as compared with normal and empty vector transfected cells (4.9%±0.7, 5.97%±0.75) (P< 0.001) (Fig D). mRNA levels of P53 and P21 were also enhanced in PML-overexpressed hMSCs. P53 pathway contributes to cell senescence and the role of PML has been proved in the regulation of P53 activity, we wondered whether upregulation of PML induced by IFN-α has relationship with P53 pathway in hMSCs. In the process of IFN-α induced hMSCs senescence, an increasing co-localization of PML and P53 was observed in IFN-α treated cells (1000U/ml, 7 days) as compared with untreated cells (Fig E). To further confirm whether or not the change of P53 location was mediated through the upregulation of PML, we knocked-down the expression of PML in hMSCs. Treated with IFN-α (1000U/ml, 7 days), we did not found significant location of P53 in PML-knocking down cells as compared with control. Taken together, our results suggested that hMSCs incurred senescence upon IFN-α stimulation, while PML levels were observed significant increase. By knocking-down and overexpressing PML, we demonstrated that PML was indispensable to IFN-α mediated hMSCs senescence. The molecular mechanisms underlying this process may be an increased co-localization of PML and p53 induced by IFN-α. These findings provided a novel insight into the role of IFN-α on hMSCs. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.

2015 ◽  
Vol 39 (10) ◽  
pp. 1099-1110 ◽  
Author(s):  
Iordanis Pelagiadis ◽  
Eftichia Stiakaki ◽  
Christianna Choulaki ◽  
Maria Kalmanti ◽  
Helen Dimitriou

2018 ◽  
Vol 119 (7) ◽  
pp. 748-758
Author(s):  
Shihua Luo ◽  
Yinghai Chen ◽  
Lifen Zhao ◽  
Xia Qi ◽  
Xiaoyan Miao ◽  
...  

AbstractAplastic anaemia (AA) is characterised by pancytopenia resulting from a marked reduction in haemopoietic stem cells (HSC). The regulation of haemopoiesis depends on the interaction between HSC and various cells of the bone marrow (BM) microenvironment, including BM-derived mesenchymal stromal cells (BMSC). The purpose of this study was to analyse the biological effect of nutritional supplement (NS), a dietary supplement consisting of thirty-six compounds: amino acids, nucleotides, vitamins and micronutrients on the BMSC of AA rats. The AA rat model was established by irradiating X-ray (2·5 Gy) and intraperitoneal injections of cyclophosphamide (35 mg/kg; Sigma) and chloramphenicol (35 mg/kg; Sigma). Then AA rats were fed with NS in a dose-dependent manner (2266·95, 1511·3, 1057·91 mg/kg d) by intragastric administration. The effect of NS on the BMSC of AA rats was analysed. As compared with AA rats, NS treatment significantly improved these peripheral blood parameters and stimulated the proliferation of total femoral nucleated cells. NS treatment affected proliferative behaviour of BMSC and suppressed BMSC differentiation to adipocytes. Furthermore, NS treatment of AA rats accelerated osteogenic differentiation of BMSC and enhanced bone mineral density. Co-incubation of HSC with mesenchymal stromal cells and serum from AA rats subjected to high-dose NS markedly improved the yield of CD34+cells. Protein microarray analysis revealed that there were eleven differentially expressed proteins in the NS group compared with the AA rat group. The identified specific NS might be implicated in rehabilitation of BMSC in AA rats, suggesting their potential of nutritional support in AA treatment.


Cells ◽  
2020 ◽  
Vol 9 (11) ◽  
pp. 2396
Author(s):  
Katarzyna Zielniok ◽  
Anna Burdzinska ◽  
Beata Kaleta ◽  
Radoslaw Zagozdzon ◽  
Leszek Paczek

The therapeutic potential of mesenchymal stromal cells (MSCs) is largely attributed to their immunomodulatory properties, which can be further improved by hypoxia priming. In this study, we investigated the immunomodulatory properties of MSCs preconditioned with hypoxia-mimetic Vadadustat (AKB-6548, Akebia). Gene expression analysis of immunomodulatory factors was performed by real-time polymerase chain reaction (real-time PCR) on RNA isolated from six human bone-marrow derived MSCs populations preconditioned for 6 h with 40 μM Vadadustat compared to control MSCs. The effect of Vadadustat preconditioning on MSCs secretome was determined using Proteome Profiler and Luminex, while their immunomodulatory activity was assessed by mixed lymphocyte reaction (MLR) and Culturex transwell migration assays. Real-time PCR revealed that Vadadustat downregulated genes related to immune system: IL24, IL1B, CXCL8, PDCD1LG1, PDCD1LG2, HIF1A, CCL2 and IL6, and upregulated IL17RD, CCL28 and LEP. Vadadustat caused a marked decrease in the secretion of IL6 (by 51%), HGF (by 47%), CCL7 (MCP3) (by 42%) and CXCL8 (by 40%). Vadadustat potentiated the inhibitory effect of MSCs on the proliferation of alloactivated human peripheral blood mononuclear cells (PBMCs), and reduced monocytes-enriched PBMCs chemotaxis towards the MSCs secretome. Preconditioning with Vadadustat may constitute a valuable approach to improve the therapeutic properties of MSCs.


Biomolecules ◽  
2020 ◽  
Vol 10 (4) ◽  
pp. 557
Author(s):  
Elena Sánchez-Luis ◽  
Andrea Joaquín-García ◽  
Francisco J. Campos-Laborie ◽  
Fermín Sánchez-Guijo ◽  
Javier De las Rivas

Mesenchymal Stromal Cells (MSC) are multipotent cells characterized by self-renewal, multilineage differentiation, and immunomodulatory properties. To obtain a gene regulatory profile of human MSCs, we generated a compendium of more than two hundred cell samples with genome-wide expression data, including a homogeneous set of 93 samples of five related primary cell types: bone marrow mesenchymal stem cells (BM-MSC), hematopoietic stem cells (HSC), lymphocytes (LYM), fibroblasts (FIB), and osteoblasts (OSTB). All these samples were integrated to generate a regulatory gene network using the algorithm ARACNe (Algorithm for the Reconstruction of Accurate Cellular Networks; based on mutual information), that finds regulons (groups of target genes regulated by transcription factors) and regulators (i.e., transcription factors, TFs). Furtherly, the algorithm VIPER (Algorithm for Virtual Inference of Protein-activity by Enriched Regulon analysis) was used to inference protein activity and to identify the most significant TF regulators, which control the expression profile of the studied cells. Applying these algorithms, a footprint of candidate master regulators of BM-MSCs was defined, including the genes EPAS1, NFE2L1, SNAI2, STAB2, TEAD1, and TULP3, that presented consistent upregulation and hypomethylation in BM-MSCs. These TFs regulate the activation of the genes in the bone marrow MSC lineage and are involved in development, morphogenesis, cell differentiation, regulation of cell adhesion, and cell structure.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 688-688
Author(s):  
Sherine F. Elsawa ◽  
Anne J. Novak ◽  
Steven C. Ziesmer ◽  
Thomas E. Witzig ◽  
Steven P. Treon ◽  
...  

Abstract Waldenström macroglobulinemia (WM) is a B-cell malignancy that is characterized by the production of a monoclonal IgM protein, a lymphoplasmacytic infiltrate in the bone marrow, and associated symptoms including anemia, lymphadenopathy and hyperviscosity. The aberrant production of a monoclonal IgM in the serum is a major factor causing significant morbidity in patients with this disease, yet little is known about the mechanisms that regulate monoclonal protein synthesis. While recent gene array studies and serum analysis have shown that IL-6 is elevated in WM patients suggesting an important role for this cytokine in this disease, the precise role played by IL-6 in WM is unknown. Using a multiplex ELISA approach to screen sera from WM patients, we confirmed that IL-6 was significantly elevated (p<0.0019) in patients (n=20) compared to controls (n=20). Serum levels of IL-6 in WM patients correlated with elevated levels of β2-microglobulin (p<0.0019). Additionally, we also found that serum levels of CCL5 (Rantes) were significantly elevated in WM patients (p<0.0001). CCL5 has been shown to regulate IL-6 secretion, and we therefore wanted to determine if CCL5 influenced IL-6 expression in WM and what the subsequent consequence of IL-6 stimulation was on WM cells. To define the source of IL-6 in the tumor microenvironment, we used stromal cells from the bone marrow of healthy donors, malignant cells from patients with WM, and the BCWM.1 WM cell line, and tested their ability to secrete IL-6 by ELISA. All cell types secreted IL-6, with stromal cells secreting the most. We then tested the ability of CCL5 to induce IL-6 secretion by WM and stromal cells. CCL5 significantly increased IL-6 secretion by stromal cells (p<0.03) and also increased IL-6 secretion by fresh CD19+ CD138+ cells from WM patients (p<0.02). Using fresh patient WM cells and the BCWM.1 WM cell line as a model, we then determined the effect of IL-6 on growth of WM cells. We found that IL-6 had a modest effect (mean=20% increase, range=5–41%) on cell proliferation (p<0.0039) but had no effect on cell viability. In contrast, when we addressed the role of IL-6 on IgM secretion, we found that IL-6 increased IgM secretion by BCWM.1 cells in a dose-dependent manner. The IL-6 mediated increase in IgM secretion was abolished in the presence of neutralizing antibodies to IL-6. When we analyzed the downstream signaling events activated by IL-6 in WM cells we found that stimulation of BCWM.1 cells, which express the IL-6R, resulted in phosphorylation of Stat1, Stat3 and Erk1/2, but not Akt. Using a mitogen activated protein kinase (MAPK) inhibitor, we could inhibit the IL-6-mediated phosphorylation of Erk1/2. Similarly, using a JAK1 Inhibitor, we could inhibit IL-6 mediated signaling through Stat1 and Stat3. In summary, we have clearly shown that IL-6 significantly upregulates IgM secretion by WM cells and increases their proliferation. We have also demonstrated the ability of both the malignant cells and the stromal cells to secrete IL-6, and that this secretion is regulated in part by CCL5. We have found that WM cells express IL-6R, and that IL-6 induced signaling is through both the MAPK and Jak/Stat pathways. Therapies targeting IL-6 secretion or the IL-6 signaling pathways may therefore provide clinical benefit to patients with WM; not just by inhibiting the malignant cells but by down regulating the production of the monoclonal protein.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1367-1367 ◽  
Author(s):  
Patrick Wuchter ◽  
Rainer Saffrich ◽  
Wolfgang Wagner ◽  
Frederik Wein ◽  
Mario Stephan Schubert ◽  
...  

Abstract The interaction between human hematopoietic stem cells (HSC) and their niche plays a key role in regulating maintenance of “stemness” and differentiation. We have demonstrated that a feeder layer of human mesenchymal stromal cells (MSC) can serve as a surrogate model for the niche for human HSC. We could also show, MSC are intimately connected to one another by a novel kind of adhering junction, consisting of villiformto-vermiform cell projections (processus adhaerentes). With this background, we have analyzed the intercellular junctional complexes between HSC and MSC. In comparison, we also studied the cell-cell contacts between leukemia cells (LC) and MSC. MSC were derived from bone marrow aspirates from healthy voluntary donors. HSC were isolated from umbilical cord blood. Leukemia cells that were CD34+ were obtained from bone marrow aspirates from patients suffering from acute myeloid leukemia at the time point of initial diagnosis. After 24–48 hours of co-cultivation, we stained the cellular contacts with a panel of antibodies specific for various components of tight, gap and adherens junctions. Using advanced confocal laser scanning microscopy in combination with deconvolution and volume rendering software, we were able to produce 3D-images of intercellular junctions between HSC/MSC as well as between LC/MSC. To examine the specific function of N-cadherin, we analyzed the effect of siRNA knock down of N-cadherin in MSC upon co-cultures of HSC and MSC. Intercellular connections between HSC and MSC are mainly characterized by podia formation of the HSC linking to the adjacent MSC. At the intimate contact zone to the MSC, we have identified the cytoplasmic plaque proteins alpha- and beta-catenin, co-localized with the transmembrane glycoprotein N-cadherin. Additionally, we compared these findings with a similar setting consisting of human LC co-cultured with feeder-layer of MSC. Our results demonstrated that in comparison to HSC, the proportion of leukemia cells adherent to the feeder-layer is significantly lower and podia formation is less frequent (ratio 1:3). However, the mechanism of adhesion through cadherin-catenin-complex has remained the same. At a functional level, we found that siRNA knock down of N-cadherin in MSC resulted in decreased adhesion of HSC to MSC and in a reduction of cell divisions of HSC. These results confirm that direct cellular contact via N-cadherin-based junctions is essential for homing and adhesion of HSC to the cellular niche and subsequently for the regulation of self-renewal versus differentiation in HSC.


Sign in / Sign up

Export Citation Format

Share Document