Extracellular Vesicles isolated from mesenchymal stem cell (MSCs) negatively regulates human T cell survival by inducing caspase cascade and inhibiting anti-apoptotic proteins

Document Type : Original Research Article

Authors

1 Resident of oral and Maxillofacial Surgery, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran

2 Student Research Committee, Qom university of Medical Sciences, Qom, Iran

Abstract

Objective(s): Extracellular Vesicles (EVs) transports active biomolecules between human many cells. As natural nanoparticles (NP), exosomes (Exo), plays important roles in the diagnosis and also treatment, of human disorders. Recently, it has been exhibited that mesenchymal stem cell (MSC)-derived Exo could regulate immune cells' biological processes. We evaluated the influence of the MSCs-derived Exo on T cell survival.
Methods: Exo were procured from the supernatant of bone marrow (BM)-MSCs. The structure and form of MSC-Exo were investigated using transmission electron microscopy (TEM). Exo markers, CD9 and CD63, were detected by Western blotting. In co-culture condition with MSC-derived Exo, the survivals of T cells were assessed by flow cytometry. Also, the expression levels of the Caspase-3, Caspase-8, Bcl-2 and Mcl-1 were measured by Real-time PCR.
Results: MSCs-derived Exo induced apoptosis in human T cells more strongly within 72 hours but not 24 hours of co-culture. Real-time PCR results also showed that Exo co-culture led to down-regulation of Mcl-1, and Bcl-2 while inducing Caspase 3 and 8 expressions at mRNA levels. 
Conclusions: MSCs-derived Exo could induce apoptosis in T cells and lead to immunomodulation by triggering caspase cascade and also down-regulating anti-apoptotic protein levels.

Keywords


INTRODUCTION
Mesenchymal stem cell (MSC) therapy provides an encouraging therapeutic modality for treating autoimmune diseases, sepsis as well as transplant surgery [1, 2]. They are positive for MHC-I, CD90, CD105, and CD73, while lacking the CD45, CD34, CD14, or CD11b [3]. MSCs could be isolated from various sources, such as bone marrow (BM), adipose tissue (AT), umbilical cord (UC), skin, muscle, liver, etc. They could be differentiated into the various cells like connective tissue, bone, cartilage, and etc [4, 5]. MSCs mainly elicit strong immunosuppressive influences, largely achieved by targeting T cell proliferation, deterring the activation of B cell activation, influencing the DCs activation [6, 7]. However, recent studies have shown that the risk of tumor formation and cell aging might bypass their widespread utility in clinic.
Recent examinations indicated that the pleiotropic influence of MSCs is mainly due to the producing soluble paracrine factors ranging from cytokines to genetic material [8, 9]. Exosomes (Exo), nanoscale extracellular vesicles (EVs), largely involves in the MSCs-mediated paracrine effect. They are 30-150 nm in diameter and are categorized as one of the 3 main sub-populations of the EVs. They convey efficient cargos such as miRNA and mRNA, proteins, and lipids resulting from stem cell to the neighboring cells. Exo implicate in intercellular interactions and facilitate healing of damaged and destructed tissues and organs [10, 11]. Other sub-populations consist of microvesicles (MVs) and apoptotic bodies (ApoB) (both larger than 100 nm) [12]. Exo, as natural nanoparticles (NPs), are shaped by budding as intraluminal vesicles (ILVs) from the late endosomes luminal space [13, 14]. Many reports have evidenced that the Exo play central role in MSCs-mediated therapeutic effect on many experimental models. Importantly, they provoke similar effects to MSCs, more importantly, improving tissue repair, enhancing angiogenesis, immunmodulation, as well as neuroprotection [15, 16]. Indeed, Exo convey biological information between human cells and adjust in the physiological and pathological circumstances [17]. They express tetraspanin and CD63, CD9, CD81, and CD82. They are integrin proteins situated on the surface of Exo and participate in cell adhesion. Besides, Rab GTPases, Annexins, and HSP70 and HSP90 are other Exo-related proteins contributed to the membrane fusion [18, 19]. In vivo, MSC-Exo can inhibit the potential of peripheral blood mononuclear cells (PBMCs) to secret inflammatory biomolecules. Aside that, Exo can promotes anti-inflammatory TGF-β and IL-10 from PBMCs, thus triggering the growth and immunomodulatory capability of Tregs [20]. Furthermore, MSC-Exo can diminish the neuroinflammation in neurological diseases animal models by up-regulating Tregs activation and promoting M2/M1 macrophage ration [21]. However, the underlying cellular and molecular mechanisms behind the MSC-induced immunosuppression have not been completely explained. 
In the current study, we examine the potent effects of the MSCs-Exo on immune T cells survival in vitro and also study the effect of Exo on caspase cascade and expression of the anti-apoptotic proteins in these T cells.

MATERIAL AND METHODS 
Cell culture 
Bone marrow (BM)-MSCs were purchased from Royesh Stem Cell Biotechnology (Tehran, Iran). Cells were cultivated in DMEM with 10% fetal bovine serum (FBS) (Sigma-Aldrich, Germany) and 1% pen/strep. BM-MSCs then were maintained at a 37°C humidified atmosphere with CO2 5%.
To procure T cell isolation, human blood samples were prepared from healthy human donor. Human PBMCs were separated using ficoll density gradient centrifugation. Finally, human T cells were procured using MACS with respect to the producer recommendations. ​Isolated T cells were cultured in RPMI-1640 media (Sigma-Aldrich, Germany) enriched with 10% FBS, 1% pen/strep accompanied with 2 μg/mL phytohemagglutinin (PHA).

Exo isolation
BM-MSCs derived Exo were procured from the serum-free conditioned media by a MagCapture™ Exosome Isolation Kit PS (FUJIFILM Wako). The serum-free conditioned media was centrifuged at 10000 × g for 40 min to remove other types of the EVs. Then, supernatants were filtered by 0.2 mm filter membranes and ultimately were concentrated.

Transmission electron microscopy (TEM)
To evaluate the morphology of isolated Exo, these exosomes were examined by transmission electron microscope.

Western blotting
To determine the expression of Exo markers, including CD9 and CD63, the BM-MSCs-derived Exo and parental MSCs were firstly lysed in RIPA buffer containg the protease inhibitor (Thermo Fisher Scientific, U.S.A). Following the running of the lysate proteins on 10% sodium dodecyl sulfate-polyacrylamide gels, they were transported to polyvinylidene difluoride (PVDF) membranes. Then, primary and secondary anti-bodies (Abcam, UK) were used to detect proteins CD9 and CD63. Ultimately, the detection phase was conducted exploiting the Pierce ECL Western Blotting Substrate (Millipore).

Fluorescence-activated cell sorting (FACS) 
Firstly, BM-MSCs-derived Exo (50ng/ml) were added to the culture media including human T-cells. An Annexin V-based kit (Dako, Denmark) was applied to determine the apoptosis percentages of the T cells within 24 and 72 hours of co-culture. After that, 10 µL of propidium iodide (PI) and 10 µL of FITC-conjugated Annexin-V were added into T cells and BM-MSCs-derived Exo including wells. Finally, the apoptosis levels were measured by a FACSCalibur, and the results were examined by FlowJo software.

RNA extraction and cDNA synthesis
The RNX Plus solution kit (Sinaclon, Iran) was utilized to extract total RNA from T cells.  Firstly, cells were lysed and the yields were mainatined at -70°C and thawed when RNA extraction was prerequisite. A high-capacity kit (Bioneer, USA) was exploited to establish complementary DNA (cDNA) from the yielded RNA.    

Real-Time PCR
To examine the expression levels of Caspase-3, Caspase-8, Bcl-2 and Mcl-1 expression at mRNA levels, Real Time-PCR was conducted using cDNA, forward and reverse primers, distilled water, and PrimeTime™ Master Mix (Idtdna, USA) according to kit instructions. These genes expression ratio was assessed using Step One Plus Real-time PCR (Applied Biosystems, USA) in triplicate. This process was applied using pre-set cycling parameters. The GAPDH genes were employed to normalize the mRNAs expression. The primer pairs exploited in Real-Time PCR have been listed in Table 1.

Statistical analysis
Consequences are achieved from 3 or 6 independent experiments. Data are exposed in Mean ± SEM. Student T-test was conducted in order to determine the statistical differences. All analyses were performed by Graph Pad Prism software.  Finally, the P value < 0.05 was considered as statistically significant.

RESULTS AND DISCUSSION
The characterizing of Exo 
The western blotting results validated the presence of CD63 and CD9 in BM-MSCs-derived Exo (Fig. 1A). Also, based on TEM images, the procured Exo showed a spherical shape (Fig. 1A). 

MSCs-derived non-scale Exo induce T cell apoptosis
The apoptosis percentages of the human T cells were estimated in co-culture condition with Exo. The apoptosis percentage of T cells was measured at 24 as well as 72 hours of treatment with Exo using Annexin-V/PI staining and flow cytometry analysis. Consequences demonstrated that Exo did not induce apoptosis significantly in T cells within 24 hours of co-culture. Nonetheless, an enhancement in T cell apoptosis was found within 72 hours of co-culture (P<0.05) (Fig. 2A, B). The apoptosis levels in T cells (control) and T cells plus Exo within 24 hours of co-culture, and T cells plus Exo within 72 hours of co-culture were 4.78±1.41, 8.28±2.45, and 27.38±3.08, respectively (Figs. 2A, B).
Improved activity of Tregs modulates immune response and provide suitable environment to tissue regeneration. Other investigations also showed the inhibitory effects of the Exo derived from the adipose-MSCs on T cell [22]. Our findings indicate that MSCs-Exo could inhibit immune response by negative regulation of T cell survival and activity. Other studies also verified the positive effect of the MSCs-Exo on Tregs activity [23]. Meanwhile, Blazquez et al. (2014) found that Exo could diminish T cell proliferation and also bargained IFN-γ release [24]. Aside that, it has been proposed that BM-MSCs-Exo adjust B cells activation by targeting the expression of the miRNA-related genes [25]. These immunmodulatory properties make MSCs-Exo an authentically strategy to ameliorate immunological diseases [26].
MSCs-derived Exo augmented Caspases expression in T cell
Real-time PCR test was accomplished to deterimne the expression levels of the Caspase-3 and Caspase-9 in T cell following co-culture with Exo during 24- 72 hours of treatment. Accordingly, Exo co-culture resulted in an increase in the Csspase-3 expression in T cells at 24- 72 hours of treatment (P<0.05) (Fig. 3A). Besides, Caspase-9 expression at mRNA levels was up-regulated at 48 and 72 hours but not 24 hours of co-culture with Exo (P<0.05) (Fig. 3B).
These Caspases play key roles in Caspase cascade activation [27, 28]. The up-regulation of the Caspases expression could be a putative mechanisms by which Exo exert its suppressive effects on human T cells. Caspase cascade activation eventually results in cleavage of a many proteins in the cell and facilities cell apoptosis [29]. Similarly, other reports exhibited that MSCs inhibited leukemia T cell proliferation by two main mechanismS: 1) up-regulation of the expression of Caspase-3 and -8 and 2) improving Bax/Bcl-2 ratio [30]. 

MSCs-derived Exo reduced the expression of anti-apoptotic protein levels in T cell 
Real-time PCR test was managed to examine Mcl-1 and Bcl-2 expression in T cell upon co-culture with Exo within 24- 72 hours of exposure. According to analysis, Exo co-culture caused an attenuation in the Mcl-1 expression in T cells at 48 and 72 hours, while did not any effect at 24 hours of exposure (P<0.05) (Fig. 4A). Also, Bcl-2 expression at mRNA levels was reduced only within 72 hours but not 24 and 48 hours of  exposure (P<0.05) (Fig. 4B).
Bcl-2 and Mcl-1 serve important roles in down-regulation of Caspase cascade activation [31]. They mainly overexpressed in leukemia and lymphoma and play imperative roles in their pathogenesis [32]. They inhibit cytochrome C release and consequently deter Caspase-9 activation [33]. 

CONCLUSION
It has beforehand been demonstrated that MSCs-Exo is capable of alleviating T cell viability, thus introdings Exo as a putative strategy to stimulate immunomodulation. In the present study, MSCs-Exo induced apoptosis in human T cells mainly by two mechanis: 1) inducing caspase expression and 2) down-regulating anti-apoptotic protein levels. The current challenge of this intervention is the low yield of exosomse and lack of the universally acceptable isolation methods. Also, the contamination with other vesicles has been observed.

CONFLICT OF INTEREST
The authors declare no conflict of interest. 

1. Jiang W, Xu J. Immune modulation by mesenchymal stem cells. Cell proliferation, 2020;53(1):e12712.
https://doi.org/10.1111/cpr.12712
2. Nuschke A. Activity of mesenchymal stem cells in therapies for chronic skin wound healing. Organogenesis, 2014;10(1):29-37.
https://doi.org/10.4161/org.27405
3. Martinello T, Gomiero C, Perazzi A, Iacopetti I, Gemignani F, DeBenedictis GM, et al. Allogeneic mesenchymal stem cells improve the wound healing process of sheep skin. BMC Veterinary Research, 2018;14(1):202.
https://doi.org/10.1186/s12917-018-1527-8
4. Shariati A, Nemati R, Sadeghipour Y, Yaghoubi Y, Baghbani R, Javidi K, et al. Mesenchymal stromal cells (MSCs) for neurodegenerative disease: A promising frontier. European journal of cell biology, 2020;99(6):151097.
https://doi.org/10.1016/j.ejcb.2020.151097
5. Saleh M, Taher M, Sohrabpour AA, Vaezi AA, Nasiri Toosi M, Kavianpour M, et al. Perspective of placenta derived mesenchymal stem cells in acute liver failure. Cell & bioscience, 2020;10:71.
https://doi.org/10.1186/s13578-020-00433-z
6. Song N, Wakimoto H, Rossignoli F, Bhere D, Ciccocioppo R, Chen K-S, et al. Mesenchymal stem cell immunomodulation: In pursuit of controlling COVID-19 related cytokine storm. Stem Cells, 2021;39(6):707-22.
https://doi.org/10.1002/stem.3354
7. Fierabracci A, Del Fattore A, Luciano R, Muraca M, Teti A, Muraca M. Recent advances in mesenchymal stem cell immunomodulation: the role of microvesicles. Cell transplantation, 2015;24(2):133-49.
https://doi.org/10.3727/096368913X675728
8. Chen W, Huang Y, Han J, Yu L, Li Y, Lu Z, et al. Immunomodulatory effects of mesenchymal stromal cells-derived exosome. Immunologic research, 2016;64(4):831-40.
https://doi.org/10.1007/s12026-016-8798-6
9. Willis GR, Fernandez-Gonzalez A, Anastas J, Vitali SH, Liu X, Ericsson M, et al. Mesenchymal stromal cell exosomes ameliorate experimental bronchopulmonary dysplasia and restore lung function through macrophage immunomodulation. American journal of respiratory and critical care medicine, 2018;197(1):104-16.
https://doi.org/10.1164/rccm.201705-0925OC
10. Tan S, Wong J, Sim S, Tjio C, Wong K, Chew J, et al. Mesenchymal stem cell exosomes in bone regenerative strategies-a systematic review of preclinical studies. Materials Today Bio, 2020;7:100067.
https://doi.org/10.1016/j.mtbio.2020.100067
11. Wei H, Chen J, Wang S, Fu F, Zhu X, Wu C, et al. A nanodrug consisting of doxorubicin and exosome derived from mesenchymal stem cells for osteosarcoma treatment in vitro. International journal of nanomedicine, 2019;14:8603.
https://doi.org/10.2147/IJN.S218988
12. Nojehdehi S, Soudi S, Hesampour A, Rasouli S, Soleimani M, Hashemi SM. Immunomodulatory effects of mesenchymal stem cell-derived exosomes on experimental type‐1 autoimmune diabetes. Journal of cellular biochemistry, 2018;119(11):9433-43.
https://doi.org/10.1002/jcb.27260
13. Zhu W, Huang L, Li Y, Zhang X, Gu J, Yan Y, et al. Exosomes derived from human bone marrow mesenchymal stem cells promote tumor growth in vivo. Cancer letters, 2012;315(1):28-37.
https://doi.org/10.1016/j.canlet.2011.10.002
14. Xu X, Liang Y, Li X, Ouyang K, Wang M, Cao T, et al. Exosome-mediated delivery of kartogenin for chondrogenesis of synovial fluid-derived mesenchymal stem cells and cartilage regeneration. Biomaterials, 2021;269:120539.
https://doi.org/10.1016/j.biomaterials.2020.120539
15. Liu W, Rong Y, Wang J, Zhou Z, Ge X, Ji C, et al. Exosome-shuttled miR-216a-5p from hypoxic preconditioned mesenchymal stem cells repair traumatic spinal cord injury by shifting microglial M1/M2 polarization. Journal of neuroinflammation, 2020;17(1):1-22.
https://doi.org/10.1186/s12974-020-1726-7
16. Villamizar O, Waters SA, Scott T, Grepo N, Jaffe A, Morris KV. Mesenchymal Stem Cell exosome delivered Zinc Finger Protein activation of cystic fibrosis transmembrane conductance regulator. Journal of extracellular vesicles, 2021;10(3):e12053.
https://doi.org/10.1002/jev2.12053
17. Cho BS, Kim JO, Ha DH, Yi YW. Exosomes derived from human adipose tissue-derived mesenchymal stem cells alleviate atopic dermatitis. Stem cell research & therapy, 2018;9(1):1-5.
https://doi.org/10.1186/s13287-018-0939-5
18. Sengupta V, Sengupta S, Lazo A, Woods P, Nolan A, Bremer N. Exosomes derived from bone marrow mesenchymal stem cells as treatment for severe COVID-19. Stem cells and development, 2020;29(12):747-54.
https://doi.org/10.1089/scd.2020.0080
19. Wang R, Ji Q, Meng C, Liu H, Fan C, Lipkind S, et al. Role of gingival mesenchymal stem cell exosomes in macrophage polarization under inflammatory conditions. International immunopharmacology, 2020;81:106030.
https://doi.org/10.1016/j.intimp.2019.106030
20. Kalimuthu S, Gangadaran P, Rajendran RL, Zhu L, Oh JM, Lee HW, et al. A new approach for loading anticancer drugs into mesenchymal stem cell-derived exosome mimetics for cancer therapy. Frontiers in pharmacology, 2018;9:1116.
https://doi.org/10.3389/fphar.2018.01116
21. Liu L, Guo S, Shi W, Liu Q, Huo F, Wu Y, et al. Bone marrow mesenchymal stem cell-derived small extracellular vesicles promote periodontal regeneration. Tissue Engineering Part A, 2021;27(13-14):962-76.
https://doi.org/10.1089/ten.tea.2020.0141
22. Tian J, Zhu Q, Zhang Y, Bian Q, Hong Y, Shen Z, et al. Olfactory ecto-mesenchymal stem cell-derived exosomes ameliorate experimental colitis via modulating Th1/Th17 and Treg cell responses. Frontiers in immunology, 2020;11:598322.
https://doi.org/10.3389/fimmu.2020.598322
23. Zhang B, Yeo RWY, Lai RC, Sim EWK, Chin KC, Lim SK. Mesenchymal stromal cell exosome-enhanced regulatory T-cell production through an antigen-presenting cell-mediated pathway. Cytotherapy, 2018;20(5):687-96.
https://doi.org/10.1016/j.jcyt.2018.02.372
24. Blazquez R, Sanchez-Margallo FM, de la Rosa O, Dalemans W, Álvarez V, Tarazona R, et al. Immunomodulatory Potential of Human Adipose Mesenchymal Stem Cells Derived Exosomes on in vitro Stimulated T Cells. Frontiers in immunology, 2014;5.
https://doi.org/10.3389/fimmu.2014.00556
25. Khare D, Or R, Resnick I, Barkatz C, Almogi-Hazan O, Avni B. Mesenchymal Stromal Cell-Derived Exosomes Affect mRNA Expression and Function of B-Lymphocytes. Frontiers in immunology, 2018;9.
https://doi.org/10.3389/fimmu.2018.03053
26. Lai P, Chen X, Guo L, Wang Y, Liu X, Liu Y, et al. A potent immunomodulatory role of exosomes derived from mesenchymal stromal cells in preventing cGVHD. Journal of hematology & oncology, 2018;11(1):1-15.
https://doi.org/10.1186/s13045-018-0680-7
27. Fan T-J, Han L-H, Cong R-S, Liang J. Caspase family proteases and apoptosis. Acta biochimica et biophysica Sinica, 2005;37(11):719-27.
https://doi.org/10.1111/j.1745-7270.2005.00108.x
28. Yuan Y, Zhang C-J, Kwok RT, Mao D, Tang BZ, Liu B. Light-up probe based on AIEgens: dual signal turn-on for caspase cascade activation monitoring. Chemical science, 2017;8(4):2723-8.
https://doi.org/10.1039/C6SC04322D
29. Hayashi Y, Arakaki R, Ishimaru N. The role of caspase cascade on the development of primary Sjogren’s syndrome. Journal of Medical Investigation, 2003;50(1/2):32-8.
https://doi.org/10.1016/S1348-8643(04)80007-7
30. Rahbaran M, Baghini SS, Mardasi M, Razeghian E. MSCs modifies the proliferation of leukemia MOLT-4 cells and induces their apoptosis through up-regulating Bax, caspase-3, and-8, and down-regulating Bcl-2 expression. Ann Cancer Res Ther, 2021;29(1):79-84.
https://doi.org/10.4993/acrt.29.79
31. Zhang Q, Riley-Gillis B, Han L, Jia Y, Lodi A, Zhang H, et al. Activation of RAS/MAPK pathway confers MCL-1 mediated acquired resistance to BCL-2 inhibitor venetoclax in acute myeloid leukemia. Signal transduction and targeted therapy, 2022;7(1):1-13.
https://doi.org/10.1038/s41392-021-00870-3
32. He W, Li X, Morsch M, Ismail M, Liu Y, Rehman FU, et al. Brain-targeted codelivery of Bcl-2/Bcl-xl and Mcl-1 inhibitors by biomimetic nanoparticles for orthotopic glioblastoma therapy. ACS nano, 2022;16(4):6293-308.
https://doi.org/10.1021/acsnano.2c00320
33. Erlacher M, Labi V. MCL-1 and BCL-XL: blood brothers. Blood, The Journal of the American Society of Hematology, 2021;137(14):1850-1.
https://doi.org/10.1182/blood.2020010569