Open Access

Suppressive effect mediated by human adipose-derived stem cells on T cells involves the activation of JNK

  • Authors:
    • Yinmin Wang
    • Xiuxia Wang
    • Xianyu Zhou
    • Zhu Zhu
    • Jun Yang
    • Fei Liu
  • View Affiliations

  • Published online on: October 24, 2018     https://doi.org/10.3892/ijmm.2018.3953
  • Pages: 177-184
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Adipose‑derived stem cells (ADSCs) have an immunomodulatory role in vascularized composite tissue allo‑transplantation (VCA). However, the specific effects of ADSCs on lymphocytes remain to be fully elucidated. The present study examined the changes in T cells co‑cultured with ADSCs in terms of the proliferation by Cell Counting Kit‑8 assay, cell cycle profile and apoptosis by flow cytometry, inflammatory cytokine production by polymerase chain reaction and ELISA, in addition to the expression of survival proteins by western blotting. The ADSCs reduced the viability of Jurkat T cells and downregulated the transcription of tumor necrosis factor‑α and transforming growth factor‑β1. Co‑culture with ADSCs also induced apoptosis and increased the levels of phosphorylated c‑Jun N‑terminal kinase in the T cells. Taken together, these findings confirmed that ADSCs modulate the host immune response by suppressing T cells.

Introduction

The allo-transplantation of vascularized composite tissue (VCA) has been increasingly used in recent years to reconstruct tissues following amputation or severe injury (1,2). However, the immunosuppressive agents that are administered post-transplantation may cause life-threatening complications, including nephrotoxicity, opportunistic infections and tumorigenesis (3). The cumulative side effects of chronic immunosuppression outweigh the benefits of VCA. In the last decade, cellular therapy has attracted increased attention in the field of allo-transplantation. Mesenchymal stem cells (MSCs), particularly adipose-derived stem cells (ADSCs), can regulate the host immune response to allografts and, in addition to immunosuppressants, improve graft survival (4,5).

The combination therapy of MSCs and immunosuppres-sants induces immune tolerance to VCA, indicating that a cellular component can reduce the dependency on immuno-suppressants in maintaining allografts. However, cell-based therapy also has safety concerns, including the increased risk of infection during ex vivo expansion and cell aggregation during systematic infusion (6,7). Therefore, it is essential to understand the interactions between ADSCs and host immune cells in order to improve the outcomes of cellular therapy in allo-transplantation.

ADSCs secrete immunomodulatory cytokines, including prostaglandin E2 (PGE-2), which inhibit the proliferation of peripheral blood mononuclear cells (PBMCs) in a mixed lymphocyte reaction (8), and express higher levels of cyclooxygenase-2 (COX-2) and indoleamine-2,3- dioxygenase when co-cultured with lymphocytes or pro-inflammatory cytokines (9). In addition, ADSCs and other MSCs regulate the function of T cells, the major driver of allo-rejection, and dendritic cells and macrophages during allo- transplantation (10,11).

The studies performed so far on the mechanisms of ADSC-mediated immunosuppression have not analyzed the molecular changes induced by ADSCs in lymphocytes. The aim of the present study was to determine the effect of ADSCs on T cells; to this end, ADSCs were isolated from adipose tissues and their interaction with the human Jurkat T cell line was investigated.

Materials and methods

Isolation and expansion of ADSCs, and co-culture with Jurkat cells

The human ADSCs were cultured as described previously (12). Briefly, adipose tissue was obtained by liposuction of the abdominal wall from three different donors (samples 1, 2 and 3; females aged 36, 54 and 56 years; Shanghai 9th People's Hospital, Shanghai, China), who had provided informed consent. The tissues were digested in 0.01% collagenase IV (Roche Diagnostics GmbH, Mannheim, Germany) for 1 h, washed twice with PBS, and seeded in 10-cm culture dishes at the density of 1x105 cells/ml with low-glucose Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS; ScienCell Research Laboratories, Inc., San Diego, CA, USA), 100 U/ml penicillin and 100 mg/ml streptomycin (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA). The cells were cultured at 37˚C under 5% CO2 until they reached 80-90% confluence, following which they were dissociated with 0.05% Trypsin-EDTA and passaged. The cells of passages 2-5 were combined, and used for further characterization and in vitro differentiation. The ADSCs were identified by immune- detection of surface CD29 (1:100, cat. no. B195249), CD44 (1:100, cat. no. B162932), CD90 (1:100, cat. no. B205317), CD34 (1:100, cat. no. B203565) and CD45 (1:100, cat. no. B215193) (all BioLegend, Inc., San Diego, CA, USA). The cells were stained with the labeled antibodies for 15 min in the dark at 4˚C and analyzed using the BD FACSCalibur flow cytom-eter (BD Biosciences, San Jose, CA, USA). Adipogenesis, osteogenesis and chondrogenesis were induced by suitable differentiation media (human adipose-derived stem cell adipogenic differentiation medium, HUXMD-90031; human adipose-derived stem cell osteogenic differentiation medium, HUXMD-90021; human adipose-derived stem cell chondro-genic differentiation medium, HUXMD-9004; all Cyagen Bioscience, Inc., Guangzhou, China) at 37˚C under 5% CO2 for >28 days, and the ensuing differentiated cells were identified by staining with oil red, alizarin red and alcian blue, respectively. Images were captured using an inverted microscope (Leica Microsystems GmbH, Wetzlar, Germany).

The Jurkat cells (purchased from GENE, Inc., Shanghai, China) were suspended in RPMI 1640 medium (HyClone; GE Healthcare, Logan, UT, USA) with 10% FBS, 100 U/ml penicillin and 100 mg/ml streptomycin, and seeded in 100-mm dishes at the density of 1x106 cells each. The culture medium was replaced every second day. The ADSCs and Jurkat cells were co-cultured for subsequent experiments in the same media in a 0.4-µm Transwell system (Corning Incorporated, Corning, NY, USA), wherein the ADSCs were seeded in the upper chamber and Jurkat cells in the lower chamber at the ratio of 1:5. The Jurkat cells were treated with 40 µM of the JNK inhibitor SP600125 (Selleck Chemicals, Houston, TX, USA) or DMSO (1 µl/ml cell suspension) for 30 min at 37°C per the requirements of the experiment.

Proliferation, cell cycle and apoptosis assays

The effect of the ADSCs on Jurkat cell proliferation was measured using a CCK-8 (Doijndo Molecular Technologies, Inc., Kumamoto, Japan) assay according to the manufacturer's protocol. The Jurkat cells were seeded into the lower chamber of a 24-well Transwell plate at a density of 1×105 cells/ml per well in 600 µl medium. The upper chambers were filled with either ADSC suspension or sterile culture medium (control). The cells were cultured for 1, 3, or 5 days, and then incubated with 60 µl CCK-8 per well at 37°C for 3 h. The supernatants were collected and the absorbance was measured at 450 nm with a microplate reader (Molecular Devices LLC, Sunnyvale, CA, USA). Each test sample was assayed in triplicate and the experiment was repeated three times.

For cell cycle profiling, the Jurkat cells were harvested following 24 h of mono- or co-culture, and fixed overnight with 70% ethanol. The cells were stained with the reagents provided in a Cell Cycle kit (Qihai Biotechnology, Shanghai, China) as per the manufacturer's protocol. To detect apoptosis, the mono- and co-cultured Jurkat cells were harvested and washed twice with phosphate-buffered saline (PBS), and stained using the FITC-Annexin V/PI Apoptosis Detection kit (BD Biosciences, Franklin Lakes, NJ, USA) as per the manufacturer's protocol. All stained samples were analyzed by flow cytometry (BD Biosciences, San Jose, CA, USA), and the ModFit LT v2.0 program (BD Biosciences).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis

Total RNA was extracted from the Jurkat cells harvested following 48 h of mono- or co-culture using an RNA isolation kit (Takara Bio, Inc., Otsu, Japan). The purity of the RNA was evaluated by calculating the A260/A280 ratio, and samples with ratios between 1.8 and 2.0 were used for further analysis. The mRNA was reverse transcribed into cDNA using the PrimeScript™ II First Strand cDNA Synthesis kit (Takara Bio, Inc.). The RT-qPCR analysis was accomplished with a SYBR-Green PCR Master mix kit (Applied Biosystems; Thermo Fisher Scientific, Inc., MA, USA) containing 2 µl cDNA and 0.5 µl of each primer (10 µM), according to the manufacturer's protocol. The following primer pairs were used for gene amplification: TGF-β1, forward 5'-ACA CCA ACT ATT GCT TCA G-3' and reverse 5'-TGT CCA GGC TCC AAA TG-3'; TNF-a, forward 5'-CTC GAA CCC CGA GTG ACA AG-3' and reverse 5'-TGA GGT ACA GGC CCT CT G AT-3'; β-actin, forward 5'-AAG CAG GAG TAT GAC GAG TCC G-3' and reverse 5'-GCC TTC ATA CAT CTC AAG TTG G-3', with the following thermal cycling conditions for 40 cycles in total: 10 min at 95°C; 15 sec at 95°C; and 30 sec at 60°C. The relative gene expression levels were calculated using the 2-ΔΔCq method (13), and normalized against those of β-actin. Three independent experiments were performed.

ELISA

After 72 h, the supernatants were collected from the mono- and co-cultured Jurkat cells and stored at -80°C until use. The levels of TGF-β1 were measured using an ELISA kit (R&D Systems, Inc., Minneapolis, MN, USA) according to the manufacturer's protocol.

Western blotting

Total protein was extracted from the Jurkat cells harvested following 24, 48 or 72 h of co-culture with RIPA lysis buffer as described previously (14). The nuclear proteins were extracted with NE-PER nuclear and cytoplasmic extraction reagents (Pierce; Thermo Fisher Scientific, Inc.) from the 48 and 72 h co-cultured cells. Equal quantities of protein (40 µg) per sample were loaded on an 12% SDS-polyacrylamide gel, electrophoresed and transferred onto a PVDF membrane and blocked with TBS with Tween-20 containing 5% non-fat dry milk for 1 h at room temperature. The membranes were probed with primary antibodies against phosphorylated (p)-P65 (1:2,000, cat. no. AF2006, Affinity Biosciences, Cambridge, UK), P65 (1:2,000, cat. no. 10745-1-AP, ProteinTech Group, Inc., Wuhan, China), inhibitor of NF-κB (IκB) kinase (IKK)β (1:1,000, cat. no. Ab124957, Abcam, Cambridge, MA, USA), IκBα (1:1,000, cat. no. 4812, Cell Signaling Technology, Inc., Danvers, MA, USA), B-cell lymphoma 2 (Bcl-2; 1:2,000, cat. no. 12789-1-AP, ProteinTech Group, Inc.), Bcl-2-associated X protein (Bax; 1:5,000, cat. no. 50599-2-IG, Protein Tech Group, Inc), JNK1/2 (1:1,000, cat. no. 9252, Cell Signaling Technology, Inc.), p-JNK1/2 (1:1,000, cat. no. 4668, Cell Signaling Technology, Inc.), extracellular signal-regulated kinase (ERK)1/2 (1:1,000, cat. no. 9102, Cell Signaling Technology, Inc.), p-ERK1/2 (1:2,000, cat. no. 4370, Cell Signaling Technology, Inc.), P38 (1:1,000, cat. no. 9212, Cell Signaling, Technology, Inc.), p-P38 (1:1,000, cat. no. 9211, Cell Signaling Technology, Inc.), mothers against decapentaplegic (Smad)2/3 (1:1,000, cat. no. AF6367, Affinity Biosciences), p-Smad2/3 (1:200, cat. no. MAB8935, R&D Systems, Inc.) and the loading control β-actin (1:200, cat. no. BM0627, Boster Biological Technology, Wuhan, China). This step was followed by incubation with horseradish peroxidase-conjugated secondary antibodies (1:50,000, goat anti-mouse, cat no. BA1051, Boster Biological Technology) for 2 h at room temperature. The specific protein bands were visualized using an enhanced chemiluminescence detection kit (GE Healthcare Life Sciences, Chalfont, UK), and the ratio of the grayscale values of the target protein were calculated.

Statistical analysis

All data are presented as the mean ± standard deviation. Statistical significance was calculated with Student's t-test and one-way analysis of variance; Tukey's multiple comparison test was used for post hoc analysis. Statistical analyses were performed using Prism 6.0 software (GraphPad Software, Inc., La Jolla, CA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of ADSCs on Jurkat cell proliferation, cell cycle and apoptosis

The ADSCs isolated from human adipose tissue were characterized using MSC surface markers CD90, CD44, CD29, CD34 and CD45 (12). The multipotency of the ADSCs was determined by the adipogenic, osteogenic and chondrogenic differentiation assays (Fig. 1A-H).

The effect of ADSCs on Jurkat cell behavior was assessed using established proliferation, cell cycle and apoptosis assays. Although no significant differences were observed in the proliferation rates of the mono- and co-cultured Jurkat cells on days 1 and 3, the ADSC-co-cultured cells exhibited a significantly lower proliferation rate on day 5 compared with the control cells (P<0.05; Fig. 2A). Following 24 h of co-culture with ADSCs, the proportion of Jurkat cells in the G0/G1 and G2/M phases increased significantly and there was a concomitant decrease in the proportion of cells in the S phase compared with the control cells (Fig. 2B). Finally, the Jurkat cells co-cultured with ADSCs for 48 h exhibited significantly higher apoptotic rates than the control cells (Fig. 3A-D). The Jurkat cells co-cultured with ADSCs for 48 h also showed significantly higher levels of pro-apoptotic Bax and lower levels of anti-apoptotic Bcl-2 compared with the control cells (Fig. 3E). Taken together, the ADSCs suppressed Jurkat cell proliferation by arresting them at the G0/G1 and G2/M phases, and inducing apoptosis.

Effect of ADSCs on Jurkat cell cytokine secretion

The mRNA and protein levels of TNF-α and TGF-β1 were measured in the Jurkat cells co-cultured with ADSCs by RT-qPCR and ELISA analyses, respectively. Compared with the control cells, the mRNA levels of TNF-α and TGF-β1 were signifi-cantly decreased in the co-cultured cells at 48 h, whereas the secreted levels of TGF-β1 were significantly increased in the supernatants of the co-cultured cells at 72 h. (Fig. 4).

Regulation of Jurkat cell signaling pathways by ADSCs

In addition to the cells co-cultured with ADSCs for 48 h showing higher levels of pro-apoptotic Bax and lower levels of anti-apoptotic Bcl-2, the NF-κB p-P65 subunit levels in the cellular and nuclear fractions were marginally lower in the co-cultured cells compared with those in the control cells after 72 h. No apparent differences were observed in the levels of P65, IκBα or IKKβ between the two groups, as well as the level of Erk, P38 and Smad2/3 (Figs. 5A-D and 6). By contrast, the level of p-JNK1/2 was higher in the co-cultured cells after 48 h (Fig. 6A-C). These findings indicated that the ADSCs induced apoptosis of the Jurkat cells by suppressing NF-κB P65 subunit phosphorylation and activating the JNK signaling pathway.

ADSC induces apoptosis in Jurkat cells via the JNK pathway

In order to evaluate the role of the activation of JNK in ADSC-induced Jurkat cell apoptosis, the latter were treated with 40 µM of the specific JNK inhibitor SP600125 prior to the co-culture, and the percentage of apoptotic cells was assessed. As shown in Fig. 7A-E, the inhibition of JNK in Jurkat cells significantly inhibited ADSC-induced apoptosis.

Discussion

As the prognosis of allo-transplantation is dependent on the host immune response to the allografts, reducing T cell-mediated acute rejection is vital in order to protect the allograft. The immunomodulatory effects of ADSCs observed in pre-clinical studies make it a suitable addition to routine immunosuppressants for preventing allograft rejection (4,5). Therefore, it is important to determine the specific effects of ADSCs on immune cells, particularly CD4+ and CD8+ T cells, which are the major elicitors of acute and chronic rejection (15,16).

Gonzalez-Rey et al (17) reported suppressive effects of human ADSCs on the proliferation of activated PBMCs and production of TNF-α. A study on the miniature swine hind-limb model showed that the infusion of ADSCs prolonged allograft survival, increased regulatory T cells in allografts and blood, and decreased the levels of circulating pro-inflammatory cytokine TNF-α (4). In addition, Matula et al (18) reported a similar inhibitory effect of ADSCs on Jurkat T cells following 3 and 4 days of co-culture. However, Mousavi et al (19) did not report any differences in the proliferation rates of Jurkat cells cultured with or without ADSC for 2 and 3 days. In the present study, ADSC co-culture reduced Jurkat cell viability on day 5, with no significant effects observed until day 3, indicating a time-dependent effect of the ADSCs on Jurkat cells.

The NFAT and NF-κB signaling pathways synergistically activate T cells (20). NF-κB is a transcription factor that regulates the expression of inflammatory cytokines and genes essential for the proliferation and survival of T cells (21). MSCs have been shown to inhibit NF-κB signaling in responder cells (22-24). Consistent with this, the present study observed a reduction in the levels of p-p65 and its nuclear translocation in Jurkat cells co-cultured with ADSCs, thereby confirming the suppressive effect of ADSCs on T cells.

Consistent with the pro-apoptotic effects of MSCs on T cells reported in previous studies (25,26), the present study observed increased apoptosis in the Jurkat cells co-cultured with ADSCs. Previous studies have also reported an inhibitory effect of MSCs on T cell apoptosis (27-29). This ambiguity may be the result of differences in responder cells (28), different tissue sources of MSCs (e.g. bone marrow, placenta or adipose tissue) (27,29), and different T-cell stimulators (e.g., PHA or anti-Fas antibody) (27,29) used in these studies. Therefore, MSCs from different origins may affect T cell function via different mechanisms.

Apoptosis can be induced by either inhibiting survival signals, for example, BCL-2 as an inner mitochondrial membrane anti-apoptotic protein, or by increasing death signals, for example BAX as a pro-apoptotic effector of mitochondrial outer membrane permeabilization (30). Furthermore, Bcl-2-associated apoptosis in Jurkat cells is associated with the activation of JNK (31). The present study observed increased levels of p-JNK1/2 in the co-cultured Jurkat cells, indicating the ADSC-mediated activation of JNK. The involvement of the JNK pathway in ADSC-induced apoptosis was further verified by the JNK-specific inhibitor SP600125 inhibiting apoptosis in the co-cultured Jurkat cells. However, the inhibitory effect was not complete, indicating the involvement of other pathways in ADSC-induced apoptosis. Higher levels of secreted TGF-β1, an activator of JNK, were present in the superna-tants of the co-cultured Jurkat cells. However, the mRNA levels of TGF-β1 were significantly lower in the co-cultured Jurkat cells; it is likely that TGF-β1 was secreted by the ADSCs and not Jurkat cells. TGF-β1 is an important component of the ADSC secretome, and has immunosuppressive and apoptotic effects on T cells (32,33). Taken together, the ADSCs induced apoptosis in Jurkat cells by activating the JNK pathway.

MSCs have shown potential in improving transplantation outcomes (34-36). A previous study showed that the inclusion of bone marrow-derived MSCs (BMSCs) during renal transplantation improved renal function at the one-year follow-up (34). In addition, the BMSCs also reduced tubulitis, interstitial fibrosis and tubule atrophy post-transplantation (35). The immunomodulatory effects ADSCs, although evident in pre-clinical and experimental studies, have not been verified in clinical trials. In the present study, the mechanism underlying the effect of ADSC on T cells was elucidated, to further underscore the clinical potential of ADSCs in immunological diseases and transplantation, and to improve current understanding of stem cell-based therapy.

In conclusion, the present study showed that ADSCs suppressed Jurkat T cell viability by inducing apoptosis and cell cycle arrest by activating the JNK signaling and mitochondrial apoptotic pathways. These findings provide novel insights into the molecular mechanisms underlying the immunomodulatory effects of ADSCs, and provide an experimental basis for the clinical application of ADSCs.

Funding

The present study was supported by the National Natural Science Foundation for Young Scholars (grant no. 81401613).

Availability of data and materials

The data used during the present study are available from the corresponding authors on reasonable request.

Authors' contributions

FL conceived and designed the experiments, and JY guided the design of the experiments. YMW and XXW performed the experiments under the direction of JY. ZZ and XYZ were involved in the completion of the experiments. YMW and XXW analyzed the data. YMW wrote the manuscript. FL and JY revised the manuscript. All the authors read and approved the final manuscript.

Ethics approval and consent to participate

All experiments were approved by the Ethics Committee of Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine (no. 2017-452-T348). The donors provided written informed consent.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Abbreviations:

ADSCs

adipose-derived stem cells

VCA

vascularized composite allotransplantation

JNK

c-Jun N-terminal kinase

MSCs

mesenchymal stem cells

PBMCs

peripheral blood mononuclear cells

FBS

fetal bovine serum

FITC

fluorescein isothiocyanate

PI

propidium iodide

DMSO

dimethyl sulfoxide

Acknowledgments

The authors would like to thank the Laboratory of Tissue Engineering at Shanghai Ninth People's Hospital, Shanghai Jiao Tong University (Shanghai, China) for the provision of experimental equipment and technical guidance necessary to complete the study.

References

1 

Khalifian S, Brazio PS, Mohan R, Shaffer C, Brandacher G, Barth RN and Rodriguez ED: Facial transplantation: The first 9 years. Lancet. 384:2153–2163. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Zuo KJ and Olson JL: The evolution of functional hand replacement: From iron prostheses to hand transplantation. Plast Surg (Oakv). 22:44–51. 2014. View Article : Google Scholar :

3 

Bamoulid J, Staeck O, Halleck F, Khadzhynov D, Brakemeier S, Durr M and Budde K: The need for minimization strategies: Current problems of immunosuppression. Transpl Int. 28:891–900. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Kuo YR, Chen CC, Chen YC and Chien CM: Recipient Adipose-derived stem cells enhance recipient cell engraftment and prolong allotransplant survival in a miniature swine hind-limb model. Cell Transplant. 26:1418–1427. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Plock JA, Schnider JT, Zhang W, Schweizer R, Tsuji W, Kostereva N, Fanzio PM, Ravuri S, Solari MG, Cheng HY, et al: Adipose- and bone marrow-derived mesenchymal stem cells prolong graft survival in vascularized composite allotransplantation. Transplantation. 99:1765–1773. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Munir H and McGettrick HM: Mesenchymal stem cell therapy for autoimmune disease: Risks and rewards. Stem Cells Dev. 24:2091–2100. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Boltze J, Arnold A, Walczak P, Jolkkonen J, Cui L and Wagner DC: The dark side of the force-constraints and complications of cell therapies for stroke. Front Neurol. 6:1552015. View Article : Google Scholar

8 

Cui L, Yin S, Liu W, Li N, Zhang W and Cao Y: Expanded adipose-derived stem cells suppress mixed lymphocyte reaction by secretion of prostaglandin E2. Tissue Eng. 13:1185–1195. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Crop MJ, Baan CC, Korevaar SS, Ijzermans JN, Pescatori M, Stubbs AP, van Ijcken WF, Dahlke MH, Eggenhofer E, Weimar W and Hoogduijn MJ: Inflammatory conditions affect gene expression and function of human adipose tissue-derived mesenchymal stem cells. Clin Exp Immunol. 162:474–486. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Kuo YR, Chen CC, Goto S, Lin PY, Wei FC and Chen CL: Mesenchymal stem cells as immunomodulators in a vascularized composite allotransplantation. Clin Dev Immunol. 2012.854846:2012.

11 

Zhao Q, Ren H and Han Z: Mesenchymal stem cells: Immunomodulatory capability and clinical potential in immune diseases. J Cell Immunotherapy. 2:3–20. 2016. View Article : Google Scholar

12 

Bunnell BA, Flaat M, Gagliardi C, Patel B and Ripoll C: Adipose-derived stem cells: Isolation, expansion and differentiation. Methods. 45:115–120. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar

14 

Hwang GS, Hu S, Lin YH, Chen ST, Tang TK, Wang PS and Wang SW: Arecoline inhibits interleukin-2 secretion in Jurkat cells by decreasing the expression of alpha7-nicotinic acetylcho-line receptors and prostaglandin E2. J Physiol Pharmacol. 64:535–543. 2013.PubMed/NCBI

15 

Liu Z, Fan H and Jiang S: CD4(+) T-cell subsets in transplantation. Immunol Rev. 252:183–191. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Yap M, Brouard S, Pecqueur C and Degauque N: Targeting CD8 T-cell metabolism in transplantation. Front Immunol. 6:5472015. View Article : Google Scholar : PubMed/NCBI

17 

Gonzalez-Rey E, Gonzalez MA, Varela N, O'Valle F, Hernandez-Cortes P, Rico L, Büscher D and Delgado M: Human adipose-derived mesenchymal stem cells reduce inflammatory and T cell responses and induce regulatory T cells in vitro in rheumatoid arthritis. Ann Rheum Dis. 69:241–248. 2010. View Article : Google Scholar

18 

Matula Z, Németh A, Lőrincz P, Szepesi Á, Brózik A, Buzás EI, Lőw P, Német K, Uher F and Urbán VS: The role of extracellular vesicle and tunneling Nanotube-mediated intercellular cross-talk between mesenchymal stem cells and human peripheral T cells. Stem Cells Dev. 25:1818–1832. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Mousavi Niri N, Jaberipour M, Razmkhah M, Ghaderi A and Habibagahi M: Mesenchymal stem cells do not suppress lymphoblastic leukemic cell line proliferation. Iran J Immunol. 6:186–194. 2009.

20 

Bronk CC, Yoder S, Hopewell EL, Yang S, Celis E, Yu XZ and Beg AA: NF-κB is crucial in proximal T-cell signaling for calcium influx and NFAT activation. Eur J Immunol. 44:3741–3746. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Reale C, Zotti T, Scudiero I, Vito P and Stilo R: Chapter nine-the NF-κB family of transcription factors and its role in thyroid physiology. Vitamins and Hormones. Litwack G: Academic Press; pp. 195–210. 2018, View Article : Google Scholar

22 

Letourneau PA, Menge TD, Wataha KA, Wade CE, S Cox C Jr, Holcomb JB and Pati S: Human bone marrow derived mesenchymal stem cells regulate Leukocyte-endothelial interactions and activation of transcription factor NF-Kappa B. J Tissue Sci Eng. 3(Suppl): S0012011.

23 

Onishi R, Ohnishi S, Higashi R, Watari M, Yamahara K, Okubo N, Nakagawa K, Katsurada T, Suda G, Natsuizaka M, et al: Human amnion-derived mesenchymal stem cell transplantation ameliorates dextran sulfate sodium-induced severe colitis in rats. Cell Transplant. 24:2601–2614. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Wang PP, Xie DY, Liang XJ, Peng L, Zhang GL, Ye YN, Xie C and Gao ZL: HGF and direct mesenchymal stem cells contact synergize to inhibit hepatic stellate cells activation through TLR4/NF-kB pathway. PLoS One. 7:e434082012. View Article : Google Scholar : PubMed/NCBI

25 

Plumas J, Chaperot L, Richard MJ, Molens JP, Bensa JC and Favrot MC: Mesenchymal stem cells induce apoptosis of activated T cells. Leukemia. 19:1597–1604. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Akiyama K, Chen C, Wang D, Xu X, Qu C, Yamaza T, Cai T, Chen W, Sun L and Shi S: Mesenchymal-stem-cell-induced immunoregulation involves FAS-ligand-/FAS-mediated T cell apoptosis. Cell Stem Cell. 10:544–555. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Benvenuto F, Ferrari S, Gerdoni E, Gualandi F, Frassoni F, Pistoia V, Mancardi G and Uccelli A: Human mesenchymal stem cells promote survival of T cells in a quiescent state. Stem Cells. 25:1753–1760. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Ramasamy R, Lam EW, Soeiro I, Tisato V, Bonnet D and Dazzi F: Mesenchymal stem cells inhibit proliferation and apoptosis of tumor cells: Impact on in vivo tumor growth. Leukemia. 21:304–310. 2007. View Article : Google Scholar

29 

Gu YZ, Xue Q, Chen YJ, Yu GH, Qing MD, Shen Y, Wang MY, Shi Q and Zhang XG: Different roles of PD-L1 and FasL in immunomodulation mediated by human placenta-derived mesenchymal stem cells. Hum Immunol. 74:267–276. 2013. View Article : Google Scholar

30 

Zheng JH, Viacava Follis A, Kriwacki RW and Moldoveanu T: Discoveries and controversies in BCL-2 protein-mediated apoptosis. FEBS J. 283:2690–2700. 2016. View Article : Google Scholar

31 

Lee SH, Park SW, Pyo CW, Yoo NK, Kim J and Choi SY: Requirement of the JNK-associated Bcl-2 pathway for human lactoferrin-induced apoptosis in the Jurkat leukemia T cell line. Biochimie. 91:102–108. 2009. View Article : Google Scholar

32 

Kapur SK and Katz AJ: Review of the adipose derived stem cell secretome. Biochimie. 95:2222–2228. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Wang J, Guan E, Roderiquez G and Norcross MA: Synergistic induction of apoptosis in primary CD4(+) T cells by macrophage-tropic HIV-1 and TGF-beta1. J Immunol. 167:3360–3366. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Tan J, Wu W, Xu X, Liao L, Zheng F, Messinger S, Sun X, Chen J, Yang S, Cai J, Gao X, et al: Induction therapy with autologous mesenchymal stem cells in living-related kidney transplants: A randomized controlled trial. JAMA. 307:1169–1177. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Reinders ME, de Fijter JW, Roelofs H, Bajema IM, de Vries DK, Schaapherder AF, Claas FH, van Miert PP, Roelen DL, van Kooten C, et al: Autologous bone marrow-derived mesen-chymal stromal cells for the treatment of allograft rejection after renal transplantation: Results of a phase I study. Stem Cells Transl Med. 2:107–111. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Peng Y, Ke M, Xu L, Liu L, Chen X, Xia W, Li X, Chen Z, Ma J, Liao D, et al: Donor-derived mesenchymal stem cells combined with low-dose tacrolimus prevent acute rejection after renal transplantation: A clinical pilot study. Transplantation. 95:161–168. 2013. View Article : Google Scholar

Related Articles

Journal Cover

January-2019
Volume 43 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang Y, Wang X, Zhou X, Zhu Z, Yang J and Liu F: Suppressive effect mediated by human adipose-derived stem cells on T cells involves the activation of JNK. Int J Mol Med 43: 177-184, 2019
APA
Wang, Y., Wang, X., Zhou, X., Zhu, Z., Yang, J., & Liu, F. (2019). Suppressive effect mediated by human adipose-derived stem cells on T cells involves the activation of JNK. International Journal of Molecular Medicine, 43, 177-184. https://doi.org/10.3892/ijmm.2018.3953
MLA
Wang, Y., Wang, X., Zhou, X., Zhu, Z., Yang, J., Liu, F."Suppressive effect mediated by human adipose-derived stem cells on T cells involves the activation of JNK". International Journal of Molecular Medicine 43.1 (2019): 177-184.
Chicago
Wang, Y., Wang, X., Zhou, X., Zhu, Z., Yang, J., Liu, F."Suppressive effect mediated by human adipose-derived stem cells on T cells involves the activation of JNK". International Journal of Molecular Medicine 43, no. 1 (2019): 177-184. https://doi.org/10.3892/ijmm.2018.3953