Spontaneous apoptosis of cells in therapeutic stem cell preparation exert immunomodulatory effects through release of phosphatidylserine

Xuemei He, Weiqi Hong, Jingyun Yang, Hong Lei, Tianqi Lu, Cai He, Zhenfei Bi, Xiangyu Pan, Yu Liu, Lunzhi Dai, Wei Wang, Canhua Huang, Hongxin Deng, Xiawei Wei, Xuemei He, Weiqi Hong, Jingyun Yang, Hong Lei, Tianqi Lu, Cai He, Zhenfei Bi, Xiangyu Pan, Yu Liu, Lunzhi Dai, Wei Wang, Canhua Huang, Hongxin Deng, Xiawei Wei

Abstract

Mesenchymal stem cell (MSC)-mediated immunomodulation has been harnessed for the treatment of human diseases, but its underlying mechanism has not been fully understood. Dead cells, including apoptotic cells have immunomodulatory properties. It has been repeatedly reported that the proportion of nonviable MSCs in a MSC therapeutic preparation varied from 5~50% in the ongoing clinical trials. It is conceivable that the nonviable cells in a MSC therapeutic preparation may play a role in the therapeutic effects of MSCs. We found that the MSC therapeutic preparation in the present study had about 5% dead MSCs (DMSCs), characterized by apoptotic cells. Namely, 1 × 106 MSCs in the preparation contained about 5 × 104 DMSCs. We found that the treatment with even 5 × 104 DMSCs alone had the equal therapeutic effects as with 1 × 106 MSCs. This protective effect of the dead MSCs alone was confirmed in four mouse models, including concanavalin A (ConA)- and carbon tetrachloride (CCl4)-induced acute liver injury, LPS-induced lung injury and spinal cord injury. We also found that the infused MSCs died by apoptosis in vivo. Furthermore, the therapeutic effect was attributed to the elevated level of phosphatidylserine (PS) upon the injection of MSCs or DMSCs. The direct administration of PS liposomes (PSLs) mimic apoptotic cell fragments also exerted the protective effects as MSCs and DMSCs. The Mer tyrosine kinase (MerTK) deficiency or the knockout of chemokine receptor C-C motif chemokine receptor 2 (CCR2) reversed these protective effects of MSCs or DMSCs. These results revealed that DMSCs alone in the therapeutic stem cell preparation or the apoptotic cells induced in vivo may exert the same immunomodulatory property as the "living MSCs preparation" through releasing PS, which was further recognized by MerTK and participated in modulating immune cells.

Conflict of interest statement

All authors declare no competing interests. C.H. is the editorial board member of Signal Transduction and Targeted Therapy, but he has not been involved in the process of the manuscript handling.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Culture and identification of MSCs and DMSCs. a The morphology of MSCs. Scale bar represents 100 μm. b MSCs were identified as CD45−CD11b−CD31−CD86−CD29+CD44+Sca-1+. c Morphology of living MSCs and DMSCs. Scale bar represents 100 μm. d Identification of living and dead MSCs by Trypan blue. Scale bar represents 100 μm. e Living and dead MSCs were stained using LIVE/DEADTM Near-IR Dead Cell Stain Kit and were analyzed by flow cytometry. f The apoptosis of MSCs and DMSCs detected by flow cytometry. g The PI staining of MSCs and DMSCs. Scale bar represents 50 μm. h, i The expression of cleaved caspase 3 (h) and Cathepsin B (i) in MSCs and DMSCs. Scale bar represents 10 μm. C-Cas 3, cleaved caspase 3
Fig. 2
Fig. 2
MSCs and DMSCs attenuate ConA-induced liver injury in mice. a Mice were intravenously injected with 12 mg/kg ConA, followed by intravenous injection with PBS, 1 × 106 MSCs (containing 5 × 104 DMSCs) or 5 × 104 to 5 × 106 DMSCs. Twelve hours after administration of ConA, mice were killed and representative macroscopic images of livers were shown. n = 6~8. b Representative images of liver histopathology with H&E staining. Scale bar represents 200 μm. ce Quantitative analysis of necrotic area (c), serum ALT and AST levels (d), and TUNEL-positive cells (e, scale bar represents 50 μm) in mice with PBS, MSCs, or DMSCs treatment after ConA injection. n = 6~8 in c, d and n = 5 in e. f Survival of 25 mg/kg ConA-injected mice treated with PBS, MSCs, and DMSCs. n = 10. g, h The levels of IL-6, IFN-γ, and TNF-α in serum (g), and IL-10 and HGF in hepatic tissues (h) were determined by ELISA in each group. n = 3~5 in each group. Data are represented as mean ± SEM. ANOVA with Dunnett’s multiple comparison test was performed. Statistical significance is indicated by *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001, compared with PBS group
Fig. 3
Fig. 3
MSCs and DMSCs attenuate tissue injury in mice. a Mice were intraperitoneally injected with 0.5 ml/kg CCl4, followed by intravenous injection with PBS, MSCs, or DMSCs. Representative macroscopic images of livers 24 h after CCl4 injection. b Representative images of liver histopathology with H&E staining 24 h after CCl4 administration. Scale bar represents 200 μm. c Quantification of serum AST and ALT after CCl4 injection in each group. n = 7 in each group. d Survival 5 ml/kg CCl4-injected mice treated with PBS, MSCs, and DMSCs. n = 10. e, f The levels of IL-6, IFN-γ, TNF-α in serum (e) and HGF in hepatic tissues (f) were determined by ELISA in each group. n = 3~5 in each group. g Mice were intratracheally instilled with 5 mg/kg LPS, followed by intravenous injection with PBS, MSCs, or DMSCs. The representative macroscopic images of the lungs 72 h after LPS administration were shown. h, i Representative images of lung histopathology with H&E staining (i) and histopathology score (h) 72 h after LPS administration. Scale bar represents 200 μm. j MSCs and DMSCs improved the behavior of mice in spinal cord injury model. Data are represented as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001, compared with PBS group in cf, h. **p < 0.01, ***p < 0.001, MSC compared with PBS group; #p < 0.05, ##p < 0.01, DMSC compared with PBS group in j
Fig. 4. Transplanted MSCs undergo apoptosis within…
Fig. 4. Transplanted MSCs undergo apoptosis within few hours and release phospholipid in vivo.
a Here, 1 × 106 GFP-MSCs were injected into mice by i.v. and the lung and liver tissues were taken for frozen section and stained for C-Cas 3 (red) and DAPI (blue) at 0.5, 2, and 4 h. Scale bar represents 50 μm (upper row) or 10 μm (lower panel). b Quantification of living MSC number in the lung and liver after GFP-MSCs injection. **p < 0.01, ***p < 0.001, compared with the 0.5 h group. c Mice were injected with 1 × 106 MSCs (containing 5 × 104 DMSCs), or 5 × 104 DMSCs through tail vein. Plasma lipid was isolated at 0.5 or 4 h, and the lipid levels were detected by Nano high-resolution liquid mass analyzer. PCA plot on all samples using the normalized lipid levels of total lipids. d, e Fold change of total PS (d), PS (18:0/16:1)-H, PS (18:0/18:1)-H, PS (18:0/22:6)-H, PS (18:0/22:5)-H, PS (37:2)-H, and PS (39:1)-H (e) abundance after injection of MSCs and DMSCs at 0.5 and 4 h. Data are represented relative to the Control group. *p < 0.05, **p < 0.01, ***p < 0.001, compared with the Control group. ce Every dot represents one individual animal. Data are represented as mean ± SEM. n = 4 in each group in be. ANOVA with Dunnett’s multiple comparison test was performed. C-Cas 3, cleaved Caspase 3
Fig. 5. PSLs ameliorate ConA-induced ALI and…
Fig. 5. PSLs ameliorate ConA-induced ALI and LPS-induced lung injury.
a Mice were intravenously injected with 12 mg/kg ConA, followed by treatment with PBS, PCLs, or PSLs. Representative images of liver histopathology with H&E staining in each group. Scale bar represents 200 μm. bd Quantitative analysis of necrosis area (b), TUNEL-positive cells (c), and serum ALT and AST levels (d) in mice with PBS, PCLs, or PSLs treatment. n = 5. e Survival of 25 mg/kg ConA-injected mice treated with PBS, PCLs, and PSLs. n = 10. f, g The levels of IL-6, IFN-γ, and TNF-α in serum (f) and IL-10, HGF in hepatic tissues (g) were determined by ELISA. n = 3~5 in each group. h Representative images of lung histopathology 72 h after LPS administration. Scale bar represents 200 μm. i Histopathology score of lung sections in each group. n = 5 in each group. Data are represented as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001
Fig. 6
Fig. 6
MSCs, DMSCs, and PSLs rescued ConA-induced liver injury via MerTK signaling. a Representative images of liver histopathology with H&E staining in ConA-injected mice treated with Vehicle and TAM receptor inhibitor LDC1267. Scale bar represents 200 μm. b Quantitative analysis of necrotic area in the liver of PBS-, MSCs-, DMSCs-, or PSLs-treated mice. c Representative H&E staining analysis of liver sections for necrosis area, inflammatory cell infiltration in WT or MerTK−/− mice after PBS, MSCs, DMSCs, or PSLs treatment. Scale bar represents 200 μm. df Quantitative analysis of necrosis area (d), serum ALT and AST levels (e), and TUNEL-positive cells (f) in WT or MerTK−/− mice with PBS, MSCs, DMSCs, or PSLs treatment. n = 4 in each group in af. g Survival of ConA-injected MerTK−/− mice treated with PBS, MSCs, DMSCs, or PSLs. n = 4 in each group. h Protein levels of p-p38-MAPK, t-p38-MAPK, p-NF-κB p65, t-NF-κB p65, and IL-10 in each group were determined using western blotting. Data are represented as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001
Fig. 7
Fig. 7
MSCs, DMSCs, and PSLs recruit Ly-6Chigh MoMF and reduce inflammatory cell infiltration partly through CCR2. a Flow cytometric analyses of active NK cells (CD45+CD3−NK1.1+CD69+), neutrophils (CD45+CD11b+Ly-6G+), Ly-6Chigh MoMF (CD45+CD11b+Ly-6G−F4/80lowLy-6Chigh), and total number of Ly-6Chigh IL-10 producing MoMF in the livers of WT and MerTK−/− mice 12 h after ConA injection followed by PBS, MSCs, DMSCs, and PSLs treatment. n = 3 in each group. b Representative H&E staining analysis of liver sections in WT or CCR2−/− mice after PBS, MSCs, DMSCs, or PSLs treatment. Scale bar represents 200 μm. c, d Quantitative analysis of necrotic area (c) and TUNEL-positive cells (d) in liver sections of WT or CCR2−/− mice with PBS, MSCs, DMSCs, or PSLs treatment. n = 4 in each group. e Quantitative analysis of the proportion of active NK cells, neutrophils, and Ly-6Chigh MoMF, and the total number of Ly-6Chigh IL-10 producing MoMF in the livers of CCR2−/− mice by flow cytometry. n = 3 in each group. Data are represented as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, compared with PBS group in WT mice. #p < 0.05, ##p < 0.01, compared with each other

References

    1. Hofmann M, et al. Monitoring of bone marrow cell homing into the infarcted human myocardium. Circulation. 2005;111:2198–2202.
    1. Lee KC, Lin HC, Huang YH, Hung SC. Allo-transplantation of mesenchymal stem cells attenuates hepatic injury through IL1Ra dependent macrophage switch in a mouse model of liver disease. J. Hepatol. 2015;63:1405–1412.
    1. Lee JS, et al. A long-term follow-up study of intravenous autologous mesenchymal stem cell transplantation in patients with ischemic stroke. Stem Cells. 2010;28:1099–1106.
    1. Banas A, et al. IFATS collection: in vivo therapeutic potential of human adipose tissue mesenchymal stem cells after transplantation into mice with liver injury. Stem Cells. 2008;26:2705–2712.
    1. Islam D, et al. Identification and modulation of microenvironment is crucial for effective mesenchymal stromal cell therapy in acute lung injury. Am. J. Respir. Crit. Care Med. 2019;199:1214–1224.
    1. Duan X, et al. The long-term fate of mesenchymal stem cells labeled with magnetic resonance imaging-visible polymersomes in cerebral ischemia. Int. J. Nanomed. 2017;12:6705–6719.
    1. Bruder, S. P. et al. Mesenchymal stem cells in osteobiology and applied bone regeneration. Clin. Orthop. Relat. Res. S247–S256 (1998).
    1. Zhou, Y., Yamamoto, Y., Xiao, Z. & Ochiya, T. The immunomodulatory functions of mesenchymal stromal/stem cells mediated via paracrine activity. J. Clin. Med. 8, 1025 (2019).
    1. Song N, Scholtemeijer M, Shah K. Mesenchymal stem cell immunomodulation: mechanisms and therapeutic potential. Trends Pharmacol. Sci. 2020;41:653–664.
    1. Fan XL, Zhang Y, Li X, Fu QL. Mechanisms underlying the protective effects of mesenchymal stem cell-based therapy. Cell Mol. Life Sci. 2020;77:2771–2794.
    1. Murphy MB, Moncivais K, Caplan AI. Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine. Exp. Mol. Med. 2013;45:e54.
    1. Li L, Chen X, Wang WE, Zeng C. How to improve the survival of transplanted mesenchymal stem cell in ischemic heart? Stem Cells Int. 2016;2016:9682757.
    1. Schrepfer S, et al. Stem cell transplantation: the lung barrier. Transplant. Proc. 2007;39:573–576.
    1. Sun T, et al. A combination of ultrasound-targeted microbubble destruction with transplantation of bone marrow mesenchymal stem cells promotes recovery of acute liver injury. Stem Cell Res. Ther. 2018;9:356.
    1. Ramalho BDS, et al. Injection of bone marrow mesenchymal stem cells by intravenous or intraperitoneal routes is a viable alternative to spinal cord injury treatment in mice. Neural Regen. Res. 2018;13:1046–1053.
    1. Fadok VA, et al. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J. Clin. Invest. 1998;101:890–898.
    1. Ipseiz N, et al. The nuclear receptor Nr4a1 mediates anti-inflammatory effects of apoptotic cells. J. Immunol. 2014;192:4852–4858.
    1. Morimoto K, et al. Alveolar macrophages that phagocytose apoptotic neutrophils produce hepatocyte growth factor during bacterial pneumonia in mice. Am. J. Respir. Cell Mol. Biol. 2001;24:608–615.
    1. Gonzalez NA, et al. Apoptotic cells promote their own clearance and immune tolerance through activation of the nuclear receptor LXR. Immunity. 2009;31:245–258.
    1. Ramos GC, et al. Apoptotic mimicry: phosphatidylserine liposomes reduce inflammation through activation of peroxisome proliferator-activated receptors (PPARs) in vivo. Br. J. Pharmacol. 2007;151:844–850.
    1. Fadok VA, et al. The role of phosphatidylserine in recognition of apoptotic cells by phagocytes. Cell Death Differ. 1998;5:551–562.
    1. Birge RB, et al. Phosphatidylserine is a global immunosuppressive signal in efferocytosis, infectious disease, and cancer. Cell Death Differ. 2016;23:962–978.
    1. Zhu H, et al. A protocol for isolation and culture of mesenchymal stem cells from mouse compact bone. Nat. Protoc. 2010;5:550–560.
    1. Sun S, et al. Isolation of mouse marrow mesenchymal progenitors by a novel and reliable method. Stem Cells. 2003;21:527–535.
    1. Qian H, et al. Molecular characterization of prospectively isolated multipotent mesenchymal progenitors provides new insight into the cellular identity of mesenchymal stem cells in mouse bone marrow. Mol. Cell Biol. 2013;33:661–677.
    1. Gnecchi M, Melo LG. Bone marrow-derived mesenchymal stem cells: isolation, expansion, characterization, viral transduction, and production of conditioned medium. Methods Mol. Biol. 2009;482:281–294.
    1. Mossanen JC, et al. Chemokine (C-C motif) receptor 2-positive monocytes aggravate the early phase of acetaminophen-induced acute liver injury. Hepatology. 2016;64:1667–1682.
    1. Yang W, et al. Neutrophils promote the development of reparative macrophages mediated by ROS to orchestrate liver repair. Nat. Commun. 2019;10:1076.
    1. Krenkel O, et al. Therapeutic inhibition of inflammatory monocyte recruitment reduces steatohepatitis and liver fibrosis. Hepatology. 2018;67:1270–1283.
    1. Zigmond E, et al. Infiltrating monocyte-derived macrophages and resident kupffer cells display different ontogeny and functions in acute liver injury. J. Immunol. 2014;193:344–353.
    1. Pan GZ, et al. Bone marrow mesenchymal stem cells ameliorate hepatic ischemia/reperfusion injuries via inactivation of the MEK/ERK signaling pathway in rats. J. Surg. Res. 2012;178:935–948.
    1. Li S, et al. Mesenchymal stem cells ameliorate hepatic ischemia/reperfusion injury via inhibition of neutrophil recruitment. J. Immunol. Res. 2018;2018:7283703.
    1. Bishop CJ, Donald KJ. Non-immunological cell death of intravenously injected murine tumour cells. Br. J. Exp. Pathol. 1979;60:29–37.
    1. Eggenhofer E, et al. Mesenchymal stem cells are short-lived and do not migrate beyond the lungs after intravenous infusion. Front. Immunol. 2012;3:297.
    1. Panes J, et al. Expanded allogeneic adipose-derived mesenchymal stem cells (Cx601) for complex perianal fistulas in Crohn’s disease: a phase 3 randomised, double-blind controlled trial. Lancet. 2016;388:1281–1290.
    1. Hare JM, et al. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J. Am. Coll. Cardiol. 2009;54:2277–2286.
    1. Wilson JG, et al. Mesenchymal stem (stromal) cells for treatment of ARDS: a phase 1 clinical trial. Lancet Respir. Med. 2015;3:24–32.
    1. Gupta N, et al. Intrapulmonary delivery of bone marrow-derived mesenchymal stem cells improves survival and attenuates endotoxin-induced acute lung injury in mice. J. Immunol. 2007;179:1855–1863.
    1. Kim JY, et al. Pretreatment with lycopene attenuates oxidative stress-induced apoptosis in human mesenchymal stem cells. Biomol. Ther. (Seoul) 2015;23:517–524.
    1. Leventis PA, Grinstein S. The distribution and function of phosphatidylserine in cellular membranes. Annu. Rev. Biophys. 2010;39:407–427.
    1. Triantafyllou E, et al. MerTK expressing hepatic macrophages promote the resolution of inflammation in acute liver failure. Gut. 2018;67:333–347.
    1. Cai B, et al. Macrophage MerTK promotes liver fibrosis in nonalcoholic steatohepatitis. Cell Metab. 2020;31:406–421.
    1. Cai B, et al. MerTK cleavage limits proresolving mediator biosynthesis and exacerbates tissue inflammation. Proc. Natl Acad. Sci. USA. 2016;113:6526–6531.
    1. Cai, B. et al. MerTK signaling in macrophages promotes the synthesis of inflammation resolution mediators by suppressing CaMKII activity. Sci. Signal. 11, eaar3721 (2018).
    1. Luk F, et al. Inflammatory conditions dictate the effect of mesenchymal stem or stromal cells on B cell function. Front. Immunol. 2017;8:1042.
    1. Luz-Crawford P, et al. Mesenchymal stem cell-derived interleukin 1 receptor antagonist promotes macrophage polarization and inhibits B cell differentiation. Stem Cells. 2016;34:483–492.
    1. Nemeth K, et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat. Med. 2009;15:42–49.
    1. Deng Y, et al. Umbilical cord-derived mesenchymal stem cells instruct monocytes towards an IL10-producing phenotype by secreting IL6 and HGF. Sci. Rep. 2016;6:37566.
    1. Vagnozzi RJ, et al. An acute immune response underlies the benefit of cardiac stem cell therapy. Nature. 2020;577:405–409.
    1. Volarevic V, et al. Protective role of IL-33/ST2 axis in Con A-induced hepatitis. J. Hepatol. 2012;56:26–33.
    1. Mei SH, et al. Prevention of LPS-induced acute lung injury in mice by mesenchymal stem cells overexpressing angiopoietin 1. PLoS Med. 2007;4:e269.
    1. Scheff SW, et al. Experimental modeling of spinal cord injury: characterization of a force-defined injury device. J. Neurotrauma. 2003;20:179–193.
    1. Basso DM, et al. Basso Mouse Scale for locomotion detects differences in recovery after spinal cord injury in five common mouse strains. J. Neurotrauma. 2006;23:635–659.
    1. Liu, L. et al. Anti-spike IgG causes severe acute lung injury by skewing macrophage responses during acute SARS-CoV infection. JCI Insight. 4, e123158 (2019).
    1. Nguyen-Lefebvre AT, et al. The innate immune receptor TREM-1 promotes liver injury and fibrosis. J. Clin. Invest. 2018;128:4870–4883.
    1. Jordan S, et al. Dietary intake regulates the circulating inflammatory monocyte pool. Cell. 2019;178:1102–1114.
    1. Harel-Adar T, et al. Modulation of cardiac macrophages by phosphatidylserine-presenting liposomes improves infarct repair. Proc. Natl Acad. Sci. USA. 2011;108:1827–1832.

Source: PubMed

3
Subskrybuj