About the Authors:
Su-Min Park
Roles Conceptualization, Investigation, Writing – original draft, Writing – review & editing
Affiliation: Department of Clinical Veterinary Science, Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
Ju-Hyun An
Roles Resources
Affiliation: Department of Clinical Veterinary Science, Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
Jeong-Hwa Lee
Roles Resources
Affiliation: Department of Clinical Veterinary Science, Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
ORCID logo https://orcid.org/0000-0003-1716-8152
Kyung-Bo Kim
Roles Resources
Affiliation: Department of Clinical Veterinary Science, Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
Hyung-Kyu Chae
Roles Validation
Affiliation: Department of Clinical Veterinary Science, Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
Ye-In Oh
Roles Validation
Affiliation: Department of Clinical Veterinary Science, Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
Woo-Jin Song
Roles Supervision, Validation
* E-mail: [email protected] (WJS); [email protected] (HYY)
Affiliation: Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Jeju National University, Jeju, Republic of Korea
ORCID logo https://orcid.org/0000-0002-9195-551X
Hwa-Young Youn
Roles Supervision
* E-mail: [email protected] (WJS); [email protected] (HYY)
Affiliation: Department of Clinical Veterinary Science, Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
Introduction
Among the secretomes of mesenchymal stem/stromal cells (MSCs), extracellular vesicles (EVs) have been studied frequently for playing an important role in transmitting signals across cells [1]. EVs are small membrane vesicles that are 40–100 nm in diameter. They are released from cell membrane after fusion with multivesicular endosome and cell membrane, and contain proteins, metabolites, and nucleic acids [2]. Studies have shown the cells to be modified via transmission of mRNA, RNA, and protein via EVs [3,4].
Further studies are being conducted on how to increase the productivity and efficacy of EVs [5]. Some studies have been conducted using hypoxia-preconditioned methods to improve anti-inflammatory effect of MSC-derived EVs [6]. Several studies have shown deferoxamine (DFO), a hypoxia mimetic agent, to be usable in hypoxia preconditioning [7] and improve angiogenesis effect of MSC-derived EVs [8]. DFO inhibits hydroxylation of hypoxia-inducible factor-1 alpha (HIF-1α) and chelates the iron necessary for prolyl-4 hydroxylase, owing to which, HIF-1α is accumulated in the cell nucleus similar to that in hypoxic condition [9]. Although some previous papers had reported the modification of stem cells in hypoxic culture [10,11], there have not been many studies reporting the changes in MSC-derived EVs when treated with DFO.
Hypoxic stimulation induces HIF-1α accumulation in the nucleus, and control various signal pathways, such as inflammation, energy deprivation, or proliferation [12]. We focused on the activation of cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE 2) synthase axis [13] by HIF-1α, since the former has anti-inflammatory function in stem cells [14]. Moreover, COX-2 and signal transducer and activator of transcription 3 (STAT3) are linked to macrophages polarization [15], which is the main signal molecule in M2 polarization [16,17].
Therefore, in this study, we aimed to investigate a hypoxic culture method using DFO in order to increase the anti-inflammatory efficacy of MSC-derived EVs. Moreover, we investigated the association of HIF-1α/COX-2 anti-inflammatory pathway in DFO preconditioned canine adipose tissue derived MSC (cAT-MSC). Further, we revealed the molecular changes of MSCs to be reflected on the derived EVs, the latter then transferring the molecules to macrophages and reprogramming them into M2 phase.
Materials and methods
Cell preparation and culture
Canine adipose tissues (cAT) were obtained using a protocol approved by the Institutional Animal Care and Use Committee of Seoul National University (SNU; protocol no. SNU-180621-27). Briefly, Canine adipose tissue was obtained from a healthy dog < 1 year old during routine spay surgery. The tissue was washed three times with phosphate-buffered saline (PBS; PAN Biotech, Aidenbach, Germany) which was contained 100 U/ml penicillin and 100 g/ml streptomycin. Then the tissues were cut into small pieces and digested for 1 h at 37°C by collagenase type IA (1 mg/ml; Sigma-Aldrich, St. Louis, MO, USA). After 1 hour, the collagenase was inhibited with Dulbecco’s Modified Eagle’s Medium (DMEM; PAN Biotech) with 10% fetal bovine serum (FBS; PAN Biotech). To remove debris, the cell pellet was obtained after centrifugation at 1200 × g for 5 min and filtered through a 70-μm Falcon cell strainer (Fisher Scientific, Waltham, MA, USA). Then cells were incubated in DMEM containing 10% FBS at 37°C in a humidified atmosphere of 5% CO2.
cAT-MSCs were characterized as described in Supporting information. Cells were differentiated into adipocytes, chondrocytes, and osteocytes to confirm their multilineage features. They were also characterized by detecting stem cell markers with flow cytometry. After characterization, cAT-MSCs at passages 3–4 were used for subsequent experiments.
Cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM; PAN Biotech, Aidenbach, Germany) with 10% fetal bovine serum (FBS; PAN Biotech) and 1% penicillin-streptomycin (PS; PAN Biotech) at 37°C in 5% CO2 atmosphere. The culture medium was changed every 2–3 days, and cells were sub-cultured at 70–80% confluency. When cAT-MSCs were approximately 70% confluent, 100 μM DFO was added for 48 h in DMEM with 10% exosome-depleted FBS (Thermo Fischer Scientific, Massachusetts, USA) and 1% PS (PAN Biotech).
The canine macrophage cell line DH82 was purchased from the Korean Cell Line Bank (Seoul, Korea) and cultured in DMEM with 15% FBS at 37°C in 5% CO2 atmosphere until they reached 70–80% confluency.
Transfection of cAT-MSCs with siRNA
When the confluency of cAT-MSC was approximately 40%, they were transfected with COX-2 siRNA or control siRNA (sc-29279 and sc-37007, respectively; Santa Cruz Biotechnology, Santa Cruz, CA, USA) for 48 h using Lipofectamine RNAiMAX (Invitrogen, Carlsbad, CA, USA), following the manufacturers’ instructions. COX-2 knockdown was confirmed by qRT-PCR before further experiments.
Isolation and characterization of EVs derived from canine adipose tissue-derived (cAT)-MSCs
cAT-MSCs were cultured for 48 h in DMEM with 10% exosome-depleted FBS (Thermo Fischer Scientific) and 1% PS (PAN Biotech). The medium from each cultured cAT-MSC sample was collected and centrifuged at 2600 × g for 20 min to remove cells and cell debris. Each supernatant was transferred to a fresh tube and appropriate volume of ExoQuick-CG (System Biosciences, USA) added. EVs were isolated according to the manufacturer’s instructions.
Protein markers of isolated EVs were identified by western blotting using antibodies against CD81 (Aviva system biology, CA, USA) and CD9 (GeneTex, Irvine, CA, USA). Morphology of the EVs was characterized using transmission electron microscopy. Briefly, 10 μl of EV suspension was placed on a 300-mesh formvar/carbon-coated electron microscopy grid with the coated side facing the suspension. Distilled water was placed on the mesh for washing and a 10-μl drop of uranyl acetate was placed on the mesh for negative staining for 1 min, followed by observation under a transmission electron microscope (TEM; LIBRA 120, Carl Zeiss, Germany) at 120 kV. Size distribution of the particles was determined using a zeta-potential and particle size analyzer (ELSZ-1000ZS, Otsuka Electronics, Osaka, Japan).
RNA extraction, cDNA synthesis, and quantitative real-time polymerase chain reaction (qRT-PCR)
Total RNA was extracted from cAT-MSCs preconditioned with DFO or from control group, and from DH82 cells using the Easy-Blue total RNA extraction kit (Intron Biotechnology, Sungnam, Korea). cDNA was synthesized using the CellScript All-in-One 5× first strand cDNA synthesis master mix (CellSafe, Seoul, Korea). Samples were analyzed using AMPIGENE qPCR green mix Hi-ROX with SYBR Green dye (Enzo Life Sciences, Farmingdale, NY, USA) and 400 nM forward and reverse primers (Table 1) in the qRT-PCR thermal cycler (Bionics, Seoul, Korea). The expression level of each gene was normalized to that of glyceraldehyde 3-phosphate dehydrogenase (GAPDH), and relative expression calculated against the contrasting control group.
[Figure omitted. See PDF.]
Table 1. Sequences of PCR primers used in this study.
https://doi.org/10.1371/journal.pone.0254657.t001
Protein extraction, cell fractionation, and western blotting
Protein was extracted from preconditioned cAT-MSC-derived exosomes and DH82 using the Pro-Prep protein extraction solution (Intron Biotechnology). Concentration of the protein samples was analyzed using the DC Protein Assay Kit (Bio-Rad, Hercules, CA, USA). Nuclear proteins were isolated using the Cell Fractionation Kit-Standard (Abcam, Cambridge, MA, USA). For western blot assays, 25 μg of proteins were loaded and separated by SDS-PAGE. Bands from SDS-PAGE were transferred to polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA), which were then blocked with 5% non-fat dry milk and Tris-buffered saline. Membranes were incubated with primary antibodies against HIF-1α (1:500; LifeSpan BioSciences, Seattle, WA, USA), COX-2 (1:500, Santa Cruz Biotechnology, Dallas, TX, USA), STAT3 (1:500, LifeSpan BioSciences), phosphorylated (Tyr705) STAT3 (1:500, LifeSpan BioSciences), lamin A (1:500, Santa Cruz Biotechnology) and β-actin (1:1000, Santa Cruz Biotechnology) at 4°C overnight. The membranes were subsequently incubated with the appropriate secondary antibody for 1 h. Using an enhanced chemiluminescence detection kit (Advansta, Menlo Park, CA, USA), immunoreactive bands were detected and normalized to the housekeeping protein (β-actin).
IF analyses
DH82 cells were cultured at 2 × 105 cells in cell-culture slide (SPL, Korea), and 200 ng/ml lipopolysaccharides (LPS; Sigma-Aldrich) were stimulated for 24 h. After LPS stimulation, DH82 cells were treated with EVs at concentrations of 50 μg/well for 48 h. The slide was fixed with 4% paraformaldehyde and blocked with a blocking buffer containing 5% bovine serum albumin and 0.3% Triton X-100 (both from Sigma-Aldrich) for 1 h. They were then incubated overnight at 4°C with antibodies against FITC-conjugated CD206 (1:200; Santa Cruz Biotechnology) and phycoerythrin-conjugated CD11b (1:100; Abcam). After three washes, the slides were mounted in a VECTASHIELD mounting medium containing 4’,6-diamidino-2-phenylindole (Vector Laboratories, Burlingame, CA, USA). The samples were observed under a EVOS FL microscope (Life Technologies, Darmstadt, Germany). Immunoreactive cells were calculated with 20 random fields per group as per the ratio of DAPI/CD206-positive cells.
Statistical analyses
Data are shown as mean ± standard deviation. Mean values from the different groups were compared by Student’s t-test and one-way analysis of variance using GraphPad Prism v.6.01 software (GraphPad Software, La Jolla, CA, USA). P value < 0.05 was considered statistically significant.
Results
Characterization of cAT-MSC EVs
cAT-MSCs were characterized by flow cytometry and differentiation (Fig 1A–1G). EVs were separated from stem cell culture media by Exo-quickTM. They were round in shape, with diameter ranging from 50 nm to 100 nm, as per electron microscopic analysis (Fig 1H). Using a particle-size analyzer, the diameter of EVs was confirmed to be approximately 100 nm (Fig 1I). To identify the surface markers of exosomes, CD81 and CD 9 were confirmed as positive while β-actin was negative in western blotting (Fig 1J).
[Figure omitted. See PDF.]
Fig 1. cAT-MSC characterization and cAT-MSC derived EV characterization.
To identify multilineage differentiation, cAT-MSC were differentiated into (a) adipocyte, (b) osteocyte and (c) chondrocyte. Differentiated cells were stained each specific dye. (d-g) To immunotype, the markers of cAT-MCS were analyzed with CD29, CD44 as positive and CD45, CD 34 as negative. (h) In electron microscopic analysis, EVs were measured as 50-100nm. (i) Through a particles-size analyzer, the diameter of EVs was confirmed as around 100nm. (j) CD81 and CD 9, surface marker of EV, were confirmed as positive and β-actin was confirmed as negative in western blotting.
https://doi.org/10.1371/journal.pone.0254657.g001
Elevation of HIF-1α/COX-2 expression in DFO-preconditioned MSCs
In CCK assay, the viability of cAT-MSCs was found to be decreased upon treatment with 1 mM DFO; therefore, we decided to use DFO < 1 mM in further experiments (Fig 2A). We confirmed that HIF-1α accumulated in DFO-preconditioned cAT-MSCs (cAT-MSCDFO) at both 100 μM and 500 μM of DFO concentration (Fig 2B). Considering the previous report, which found no significant difference between 100 μM and 500 μM DFO [18], 100 μM DFO was chosen for the current treatment.
[Figure omitted. See PDF.]
Fig 2. The expression levels of COX-2 in cAT-MSCDFO.
(a) The mRNA level of COX-2 is increased in DFO preconditioned and treated with siCTL group. However, COX-2 is not increased in transfected with siCOX-2 group. (b) The protein level of COX-2 is increased in DFO preconditioned and decreased in transfected with siCOX-2 group, same as mRNA result. Results are shown as means ± standard deviation. ***P < 0.001. ns, not significant.
https://doi.org/10.1371/journal.pone.0254657.g002
To evaluate the role of COX-2 in cAT-MSCs, siCOX-2 was transfected into the cells before DFO conditioning. RNA levels of COX-2 were significantly increased in cAT-MSCDFO and DFO preconditioning plus control siRNA (cAT-MSCsiRNA) groups; the expression not being significantly different between the two. In DFO-preconditioned group, with COX-2 siRNA (cAT-MSCsiCOX-2), COX-2 expression was not increased (Fig 2C). At protein level, COX-2 was increased in cAT-MSCDFO and cAT-MSCsiRNA, but not in cAT-MSCsiCOX-2 (Fig 2D).
cAT-MSC-derived EVs transport COX-2 to DH82 and activate the phosphorylation of STAT3
When expression of COX-2 protein was evaluated in EVs, it was found increased in cAT-MSCDFO-derived EV (EVDFO) than in non-preconditioned cAT-MSC-derived EV (EVnon).
In cAT-MSCsiCOX-2-derived EV (EVsiCOX-2), COX-2 was not increased, similar to that in cAT-MSCs (Fig 3A).
[Figure omitted. See PDF.]
Fig 3. Protein levels of COX-2 in cAT-MSC derived EVs and the effect of DFO preconditioned EV in DH82.
(a) COX-2 level was increased in EVs derived from cAT-MSCDFO (EVDFO). Otherwise, COX-2 was decreased in EVsiCOX-2. The result of COX-2 protein level in EVs was similar with cytosol COV-2 protein level in cAT-MSC. (b) Protein levels of STAT3 and p-STAT3 in DH82 treated with EVs was measured by western blotting to evaluate the effect of DFO preconditioned EVs. In the all EV treated groups, p-STAT3 (Tyr 705) was increased. Compared to EVnon treated group, p-STAT3 is much more increased in EVDFO treated group, but similar in EVsiCOX-2 treated group. Results are shown as means ± standard deviation. *P < 0.05, ***P < 0.001. ns, not significant.
https://doi.org/10.1371/journal.pone.0254657.g003
DH82 cells were treated with EVs to verify their anti-inflammatory effect on macrophages. They were treated with LPS before EV treatment. In the LPS-treated group, expression of p-STAT3 was increased, whereas that of STAT3 decreased compared to that in naïve group. The expression of both STAT3 and p-STAT3 increased in EV-treated groups. In the groups treated with EVDFO, the expression of p-STAT3 was significantly increased than in non-preconditioned group. However, in the group treated with EVsiCOX-2, p-STAT3 was not increased compared to that in the group treated with EVnon (Fig 3B).
Change of polarization of DH82 when treated with preconditioned EVs
When DH82 cells were treated with LPS, the markers of M1 proinflammatory phase, namely IL-1b and IL-6, were significantly increased. However, expression of both cytokines was decreased in the groups with EV treatment. Expression of pro-inflammatory cytokines was decreased in the groups treated with EVDFO, than in the groups treated with EVnon group. The group with EVsiCOX-2 showed no significant difference from that with EVnon (Fig 4A and 4B).
[Figure omitted. See PDF.]
Fig 4. The polarization phase of DH82 is directed into M2 phase when treated with EVs.
(a, b) The mRNA level of IL-1b and IL-6, which is the marker of M1 macrophage phase, was decreased in EV treated groups and significantly decreased in EVDFO treated group compared to EVnon treated group. Conversely, the mRNA levels were similar in EVsiCOX-2 treated group and EVnon treated group. (c-h) CD11b, which is the surface marker of macrophage, was analyzed by immunofluorescence. CD206, which is the surface marker of M2 macrophage phase, was analyzed by immunofluorescence. (e-i) In the EV treated groups, CD 206 with green fluorescence is increased and (f, g) especially more increased in EVDFO group. (h) In the EVsiCOX-2 treated group, CD206 is also increased, but not much compared to EVDFO group. (i) The ratio of CD206/DAPI. Results are shown as means ± standard deviation. ***P < 0.001. ns, not significant.
https://doi.org/10.1371/journal.pone.0254657.g004
Using immunofluorescence of CD206, which is a marker of M2 anti-inflammatory phase, we evaluated the polarization phase of DH82 cells. Red staining was against CD11b, which is a marker of macrophage. In naïve DH82 group, red staining was visibly detected and when LPS inducing in DH82, the marker was not changed (Fig 4C and 4D). In EVs treated groups, green stain of CD206 was significantly increased and the red stain was difficult to recognize (Fig 4E–4I). Especially, CD206 was significantly increased in the groups treated with EVDFO (Fig 4F). The group with EVsiCOX-2 showed slight enhancement of CD206, which was not significant compared to that in the group with EVDFO (Fig 4H and 4I).
Discussion
Mesenchymal stem cells (MSCs) have been studied for their anti-inflammatory therapeutic functions due to paracrine effect [19,20]. Since they interact with immune cells, including macrophages and T cells, MSC secretome is considered to be immunomodulatory and can regulate the anti-inflammatory phase [21,22]. We have recently demonstrated that paracrine effect of cAT-MSCs can significantly reprogram macrophages from M1 pro-inflammatory phase to M2 anti-inflammatory phase [18]. The study proved that secretory function of MSCs plays an important role in cell interaction, especially with inflammatory cells like macrophages. Recently, EVs were reported to play an important role among secretomes, owing to their ability to communicate and deliver substances [23–26]. Therefore, we focused on the EVs derived from cAT-MSCs. In the current study, we investigated whether DFO-preconditioned EVs have the potential to direct macrophages to M2 phase. To prove this hypothesis, HIF-1α/COX-2 axis was analyzed in cAT-MSCDFO and EVDFO. After confirming that DFO could effectively increase COX-2 expression in EVs, Canine macrophage cells, DH82, were treated with EVs to confirm the delivery function of EVs and evaluate their effect on macrophage polarization.
First, accumulation of HIF-1α in the nucleus, which is an important indicator of hypoxia, was confirmed by western blot in cAT-MSCDFO (S1B Fig). Some reports had earlier suggested that COX-2 increases significantly with hypoxic culture under HIF-1α signal pathway [27] and is associated with both anti-inflammatory effect and macrophage polarization [28,29]. In cAT-MSCDFO, both RNA and protein expression of COX-2 was increased, implying that DFO preconditioning could increase HIF-1α accumulation, and transcription of COX-2 was activated under HIF-1α pathway (Fig 2A and 2B).
Some reports have revealed that the same proteins may be detected either in MSC cytoplasm or in MSC secretomes and EVs [30–32]. In the current study as well, expression change of COX-2 protein in the cytosol of cAT-MSCs were also reflected in the EVs. EVs from DFO-preconditioned cAT-MSC were enriched with COX-2 molecules (Fig 3A).
HIF-1α and COX-2/PGE2 pathway are known to be associated with STAT3 activation [33,34], which is an important factor in macrophage M2 anti-inflammatory phase [35–37]. Previously published reports had suggested that macrophages are polarized into M2 phase when various molecules, such as miRNA, are delivered through EVs and stimulate STAT3 pathway in the macrophages [38–40]. Another study had revealed that not only miRNA, but proteins also can be transmitted into macrophage through EVs, and play their role [37].
The delivery system of EVs has been reported to play an important role in the anti-inflammatory effect of stem cells, especially in relation to macrophages [41]. Reprogramming macrophages from pro-inflammatory M1 phase to anti-inflammatory M2 phase is the most important mechanism in controlling immune homeostasis [42], and EVs regulate this polarization phase by transferring substances from stem cells to macrophages [43,44]. Therefore, we focused on the effect of COX-2 transferred by EVs to STAT3 signaling in macrophages. When the expression of pSTAT3 and STAT3 in LPS-induced DH82 were evaluated, these factors in LPS-induced DH82 treated with EVDFO were increased over than with EVnon (Fig 3B).
The expression change of pSTAT3 in LPS-induced DH82 could be associated with macrophage phase. Thus, we evaluated macrophage phase of LPS-induced DH82 with EVs treating. In LPS-induced DH82 cells treated with EVs, which were increased with pSTAT, the marker of M1 was reduced while that of M2 was increased (Fig 4A–4I). These changes were greater with EVDFO. Considering all the results, EVs could deliver COX-2 to LPS-induced DH82, and might lead to M2 polarization by activating the phosphorylation of STAT3. Moreover, DFO preconditioning in cAT-MSCs enhance the macrophage directing effect through activating HIF-1α/COX-2 axis.
In this study, a limitation was that it was not clear whether substances such as miRNA in EVs could be stimulated to activate STAT3. Therefore, further studies would be required to analyze the changes in miRNA levels in cAT-MSCDFO and EVDFO. However, this study found that preconditioning with DFO could affect COX-2 in cAT-MSCs and acted as anti-inflammatory molecules. EVDFO contained COX-2 protein and could effectively reprogram macrophage polarization into M2 phase via protein delivery system. The findings presented the therapeutic possibility of EVDFO, which could be used in treating inflammatory diseases through macrophage reprogramming.
Conclusions
To the best of our knowledge, this report is the first to reveal that DFO preconditioning affects EVs and macrophage polarization via EVs. DFO preconditioning increased anti-inflammatory factors, such as COX-2 in cAT-MSCs, and also increased COX-2 molecules in EVs. EVDFO delivered COX-2 to macrophages, which then led to M2 anti-inflammatory phase by activating STAT3 phosphorylation.
Supporting information
S1 Fig.
(a) Depending on DFO concentration, cell viability was not affected under 500 μΜ and was decreased in 1mM. (b) HIF-1α was accumulated in the nuclear of cAT-MSCDFO, which showed that DFO treatment could accumulate HIF-1α in cAT-MSC nucleus. Results are shown as means ± standard deviation. *P < 0.05. ns, not significant.
https://doi.org/10.1371/journal.pone.0254657.s001
(TIF)
S1 Raw images.
https://doi.org/10.1371/journal.pone.0254657.s002
(PDF)
Citation: Park S-M, An J-H, Lee J-H, Kim K-B, Chae H-K, Oh Y-I, et al. (2021) Extracellular vesicles derived from DFO-preconditioned canine AT-MSCs reprogram macrophages into M2 phase. PLoS ONE 16(7): e0254657. https://doi.org/10.1371/journal.pone.0254657
1. Lai RC, Yeo RWY, Lim SK, editors. Mesenchymal stem cell exosomes. Seminars in cell & developmental biology; 2015: Elsevier.
2. Hessvik NP, Llorente A. Current knowledge on exosome biogenesis and release. Cellular and Molecular Life Sciences. 2018;75(2):193–208. pmid:28733901
3. Phinney DG, Pittenger MF. Concise review: MSC‐derived exosomes for cell‐free therapy. Stem cells. 2017;35(4):851–8. pmid:28294454
4. Sterzenbach U, Putz U, Low L-H, Silke J, Tan S-S, Howitt J. Engineered exosomes as vehicles for biologically active proteins. Molecular Therapy. 2017;25(6):1269–78. pmid:28412169
5. Hong P, Yang H, Wu Y, Li K, Tang Z. The functions and clinical application potential of exosomes derived from adipose mesenchymal stem cells: a comprehensive review. Stem Cell Research & Therapy. 2019;10(1):242. pmid:31391108
6. Cui GH, Wu J, Mou FF, Xie WH, Wang FB, Wang QL, et al. Exosomes derived from hypoxia‐preconditioned mesenchymal stromal cells ameliorate cognitive decline by rescuing synaptic dysfunction and regulating inflammatory responses in APP/PS1 mice. The FASEB Journal. 2018;32(2):654–68. pmid:28970251
7. Fujisawa K, Takami T, Okada S, Hara K, Matsumoto T, Yamamoto N, et al. Analysis of metabolomic changes in mesenchymal stem cells on treatment with desferrioxamine as a hypoxia mimetic compared with hypoxic conditions. Stem Cells. 2018;36(8):1226–36. pmid:29577517
8. Ding J, Wang X, Chen B, Zhang J, Xu J. Exosomes derived from human bone marrow mesenchymal stem cells stimulated by deferoxamine accelerate cutaneous wound healing by promoting angiogenesis. BioMed research international. 2019;2019. pmid:31192260
9. Wang GL, Semenza GL. Desferrioxamine induces erythropoietin gene expression and hypoxia-inducible factor 1 DNA-binding activity: implications for models of hypoxia signal transduction. 1993.
10. Basciano L, Nemos C, Foliguet B, de Isla N, de Carvalho M, Tran N, et al. Long term culture of mesenchymal stem cells in hypoxia promotes a genetic program maintaining their undifferentiated and multipotent status. BMC cell biology. 2011;12(1):12. pmid:21450070
11. Pezzi A, Amorin B, Laureano Á, Valim V, Dahmer A, Zambonato B, et al. Effects of hypoxia in long‐term in vitro expansion of human bone marrow derived mesenchymal stem cells. Journal of cellular biochemistry. 2017;118(10):3072–9. pmid:28240374
12. Zagórska A, Dulak J. HIF-1: the knowns and unknowns of hypoxia sensing. Acta Biochimica Polonica. 2004;51(3). pmid:15448722
13. Lee JJ, Natsuizaka M, Ohashi S, Wong GS, Takaoka M, Michaylira CZ, et al. Hypoxia activates the cyclooxygenase-2–prostaglandin E synthase axis. Carcinogenesis. 2010;31(3):427–34. pmid:20042640
14. Yang H-M, Song W-J, Li Q, Kim S-Y, Kim H-J, Ryu M-O, et al. Canine mesenchymal stem cells treated with TNF-α and IFN-γ enhance anti-inflammatory effects through the COX-2/PGE2 pathway. Research in veterinary science. 2018;119:19–26. pmid:29783120
15. Han Y, Guo W, Ren T, Huang Y, Wang S, Liu K, et al. Tumor-associated macrophages promote lung metastasis and induce epithelial-mesenchymal transition in osteosarcoma by activating the COX-2/STAT3 axis. Cancer letters. 2019;440:116–25. pmid:30343113
16. Nakamura R, Sene A, Santeford A, Gdoura A, Kubota S, Zapata N, et al. IL10-driven STAT3 signalling in senescent macrophages promotes pathological eye angiogenesis. Nature communications. 2015;6(1):1–14. pmid:26260587
17. Roberts J, Fallon PG, Hams E. The Pivotal Role of Macrophages in Metabolic Distress. Macrophage Activation-Biology and Disease: IntechOpen; 2019.
18. Park S-M, Li Q, Ryu M-O, Nam A, An J-H, Yang J-I, et al. Preconditioning of canine adipose tissue-derived mesenchymal stem cells with deferoxamine potentiates anti-inflammatory effects by directing/reprogramming M2 macrophage polarization. Veterinary immunology and immunopathology. 2020;219:109973. pmid:31765882
19. Samadi P, Saki S, Khoshinani HM, Sheykhhasan M. Therapeutic applications of mesenchymal stem cells: A comprehensive review. Current Stem Cell Research & Therapy. 2020.
20. Huang P, Gebhart N, Richelson E, Brott TG, Meschia JF, Zubair AC. Mechanism of mesenchymal stem cell–induced neuron recovery and anti-inflammation. Cytotherapy. 2014;16(10):1336–44. pmid:24927715
21. Sargent A, Miller RH. MSC therapeutics in chronic inflammation. Current stem cell reports. 2016;2(2):168–73. pmid:28133600
22. Kim J, Hematti P. Mesenchymal stem cell–educated macrophages: A novel type of alternatively activated macrophages. Experimental hematology. 2009;37(12):1445–53. pmid:19772890
23. Riau A, Ong HS, Yam GH, Mehta J. Sustained delivery system for stem cell-derived exosomes. Frontiers in pharmacology. 2019;10:1368. pmid:31798457
24. Baek G, Choi H, Kim Y, Lee HC, Choi C. Mesenchymal stem cell‐derived extracellular vesicles as therapeutics and as a drug delivery platform. Stem cells translational medicine. 2019;8(9):880–6. pmid:31045328
25. Bruno S, Grange C, Deregibus MC, Calogero RA, Saviozzi S, Collino F, et al. Mesenchymal stem cell-derived microvesicles protect against acute tubular injury. Journal of the American Society of Nephrology. 2009;20(5):1053–67. pmid:19389847
26. Kusuma GD, Carthew J, Lim R, Frith JE. Effect of the microenvironment on mesenchymal stem cell paracrine signaling: opportunities to engineer the therapeutic effect. Stem cells and development. 2017;26(9):617–31. pmid:28186467
27. Han KH, Kim AK, Kim MH, Kim DH, Go HN, Kim DI. Enhancement of angiogenic effects by hypoxia‐preconditioned human umbilical cord‐derived mesenchymal stem cells in a mouse model of hindlimb ischemia. Cell biology international. 2016;40(1):27–35. pmid:26222206
28. Luo C, Urgard E, Vooder T, Metspalu A. The role of COX-2 and Nrf2/ARE in anti-inflammation and antioxidative stress: Aging and anti-aging. Medical hypotheses. 2011;77(2):174–8. pmid:21530094
29. Jin L, Deng Z, Zhang J, Yang C, Liu J, Han W, et al. Mesenchymal stem cells promote type 2 macrophage polarization to ameliorate the myocardial injury caused by diabetic cardiomyopathy. Journal of translational medicine. 2019;17(1):251. pmid:31382970
30. Selmani Z, Naji A, Zidi I, Favier B, Gaiffe E, Obert L, et al. Human leukocyte antigen‐G5 secretion by human mesenchymal stem cells is required to suppress T lymphocyte and natural killer function and to induce CD4+ CD25highFOXP3+ regulatory T cells. Stem cells. 2008;26(1):212–22. pmid:17932417
31. Schaeffer D, Tsanova B, Barbas A, Reis FP, Dastidar EG, Sanchez-Rotunno M, et al. The exosome contains domains with specific endoribonuclease, exoribonuclease and cytoplasmic mRNA decay activities. Nature structural & molecular biology. 2009;16(1):56–62. pmid:19060898
32. Jansen FH, Krijgsveld J, van Rijswijk A, van den Bemd G-J, van den Berg MS, van Weerden WM, et al. Exosomal secretion of cytoplasmic prostate cancer xenograft-derived proteins. Molecular & Cellular Proteomics. 2009;8(6):1192–205. pmid:19204029
33. Yu X, Wan Q, Cheng G, Cheng X, Zhang J, Pathak JL, et al. CoCl2, a mimic of hypoxia, enhances bone marrow mesenchymal stem cells migration and osteogenic differentiation via STAT3 signaling pathway. Cell biology international. 2018;42(10):1321–9. pmid:29908007
34. Liu Q, Ji S, Xia T, Liu J, Liu Z, Chen X, et al. MCP-1 Priming Enhanced the Therapeutic Effects of Mesenchymal Stromal Cells on Contact Hypersensitivity by Activating the COX2-PGE2/STAT3 Pathway. Stem cells and development. 2020. pmid:32552417
35. Hollander L, Guo X, Velazquez H, Chang J, Safirstein R, Kluger H, et al. Renalase expression by melanoma and tumor-associated macrophages promotes tumor growth through a STAT3-mediated mechanism. Cancer research. 2016;76(13):3884–94. pmid:27197188
36. Shiraishi D, Fujiwara Y, Komohara Y, Mizuta H, Takeya M. Glucagon-like peptide-1 (GLP-1) induces M2 polarization of human macrophages via STAT3 activation. Biochemical and biophysical research communications. 2012;425(2):304–8. pmid:22842565
37. Zhao H, Shang Q, Pan Z, Bai Y, Li Z, Zhang H, et al. Exosomes from adipose-derived stem cells attenuate adipose inflammation and obesity through polarizing M2 macrophages and beiging in white adipose tissue. Diabetes. 2018;67(2):235–47. pmid:29133512
38. Heo JS, Choi Y, Kim HO. Adipose-derived mesenchymal stem cells promote M2 macrophage phenotype through exosomes. Stem cells international. 2019;2019. pmid:31781246
39. Sun Y, Li Q, Gui H, Xu D-P, Yang Y-L, Su D-F, et al. MicroRNA-124 mediates the cholinergic anti-inflammatory action through inhibiting the production of pro-inflammatory cytokines. Cell research. 2013;23(11):1270–83. pmid:23979021
40. Haneklaus M, Gerlic M, O’Neill LA, Masters S. miR‐223: infection, inflammation and cancer. Journal of internal medicine. 2013;274(3):215–26. pmid:23772809
41. Sicco CL, Reverberi D, Balbi C, Ulivi V, Principi E, Pascucci L, et al. Mesenchymal stem cell‐derived extracellular vesicles as mediators of anti‐inflammatory effects: Endorsement of macrophage polarization. Stem cells translational medicine. 2017;6(3):1018–28. pmid:28186708
42. Genua M, Ingangi V, Fonteyne P, Piontini A, Yousif AM, Merlino F, et al. Treatment with a urokinase receptor-derived cyclized peptide improves experimental colitis by preventing monocyte recruitment and macrophage polarization. Inflammatory Bowel Diseases. 2016;22(10):2390–401. pmid:27537052
43. Lankford KL, Arroyo EJ, Nazimek K, Bryniarski K, Askenase PW, Kocsis JD. Intravenously delivered mesenchymal stem cell-derived exosomes target M2-type macrophages in the injured spinal cord. PloS one. 2018;13(1):e0190358. pmid:29293592
44. Willis GR, Fernandez-Gonzalez A, Reis M, Mitsialis SA, Kourembanas S. Macrophage immunomodulation: the gatekeeper for mesenchymal stem cell derived-exosomes in pulmonary arterial hypertension? International journal of molecular sciences. 2018;19(9):2534. pmid:30150544
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2021 Park et al. This is an open access article distributed under the terms of the Creative Commons Attribution License: http://creativecommons.org/licenses/by/4.0/ (the “License”), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Background
Mesenchymal stem/stromal cells (MSCs) are effective therapeutic agents that ameliorate inflammation through paracrine effect; in this regard, extracellular vesicles (EVs) have been frequently studied. To improve the secretion of anti-inflammatory factors from MSCs, preconditioning with hypoxia or hypoxia-mimetic agents has been attempted and the molecular changes in preconditioned MSC-derived EVs explored. In this study, we aimed to investigate the increase of hypoxia-inducible factor 1-alpha (HIF-1α)/cyclooxygenase-2 (COX-2) in deferoxamine (DFO)-preconditioned canine MSC (MSCDFO) and whether these molecular changes were reflected on EVs. Furthermore, we focused on MSCDFO derived EVs (EVDFO) could affect macrophage polarization via the transfer function of EVs.
Results
In MSCDFO, accumulation of HIF-1α were increased and production of COX-2 were activated. Also, Inside of EVDFO were enriched with COX-2 protein. To evaluate the transferring effect of EVs to macrophage, the canine macrophage cell line, DH82, was treated with EVs after lipopolysaccharide (LPS) stimulation. Polarization changes of DH82 were evaluated with quantitative real-time PCR and immunofluorescence analyses. When LPS-induced DH82 was treated with EVDFO, phosphorylation of signal transducer and transcription3 (p-STAT3), which is one of key factor of inducing M2 phase, expression was increased in DH82. Furthermore, treated with EVDFO in LPS-induced DH82, the expression of M1 markers were reduced, otherwise, M2 surface markers were enhanced. Comparing with EVDFO and EVnon.
Conclusion
DFO preconditioning in MSCs activated the HIF-1α/COX-2 signaling pathway; Transferring COX-2 through EVDFO could effectively reprogram macrophage into M2 phase by promoting the phosphorylation of STAT3.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer