Int J Stem Cells.  2019 Nov;12(3):419-429. 10.15283/ijsc19115.

Perivascular Stem Cells Suppress Inflammasome Activation during Inflammatory Responses in Macrophages

Affiliations
  • 1Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Korea. shhong@kangwon.ac.kr
  • 2Scripps Korea Antibody Institute and Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea.
  • 3Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Korea.
  • 4Department of Physiology, School of Medicine, Kangwon National University, Chuncheon, Korea.
  • 5Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon, Korea.

Abstract

BACKGROUND AND OBJECTIVES
Perivascular stem cells (PVCs) have been identified as precursors of mesenchymal stem cells (MSCs) that offer promising prospects for application in the development of cellular therapies. Although PVCs have been demonstrated to have greater therapeutic potential compared to bone marrow and adipose tissue-derived MSCs in various diseases, the regulatory role of PVCs on inflammasome activation during macrophage-mediated inflammatory responses has not been investigated.
METHODS AND RESULTS
In this study, we found that the PVC secretome effectively alleviates secretion of both caspase-1 and interleukin-1β in lipopolysaccharide-primed and activated human and murine macrophages by blocking inflammasome activation and attenuating the production of mitochondrial reactive oxygen species (ROS). We further showed that the PVC secretome significantly reduces inflammatory responses and endoplasmic reticulum stress in peritoneal macrophages in a mouse model of monosodium urate-induced peritonitis. A cytokine antibody array analysis revealed that the PVC secretome contains high levels of serpin E1 and angiogenin, which may be responsible for the inhibitory effects on mitochondrial ROS generation as well as on inflammasome activation.
CONCLUSIONS
Our results suggest that PVCs may be therapeutically useful for the treatment of macrophage- and inflammation-mediated diseases by paracrine action via the secretion of various biological factors.

Keyword

Perivascular stem cells; Inflammation; Inflammasome; Macrophage

MeSH Terms

Animals
Biological Factors
Bone Marrow
Endoplasmic Reticulum Stress
Humans
Inflammasomes*
Inflammation
Macrophages*
Macrophages, Peritoneal
Mesenchymal Stromal Cells
Mice
Peritonitis
Plasminogen Activator Inhibitor 1
Reactive Oxygen Species
Stem Cells*
Biological Factors
Inflammasomes
Plasminogen Activator Inhibitor 1
Reactive Oxygen Species

Figure

  • Fig. 1 The inhibitory effect of PVC-CM on inflammasome activation in human macrophage. PMA-differentiated THP-1 cells were primed with LPS (1 μg/ml) in RPMI medium containing 10% FBS and antibiotics for 3 h, followed by incubation with inflammasome activators for 1 h. (A) Schematic diagram of steps involved in the process of inflammasome activation. (B) Cells were cultured in RPMI media containing the indicated concentration of PVC-CM or ATP (2 mM) as a positive control for 1 h. IL-1β secretion was analyzed by immunoblotting. (C) PMA-differentiated THP-1 cells were treated with ATP (2 mM), flagellin (0.5 μg/ml), or dsDNA (2 μg/ml) for 1 h in the presence of PVC-CM. Cellular supernatant (Sup), lysate (Lys) and cross-linked pellets (Pellet) from whole-cell lysates were analyzed with the indicated anti-sera in an immunoblot assay. (D, E) Secreted caspase-1 and IL-1β were quantitated by an ELISA-based assay kit and the data are presented as bar graphs. Error bars indicate SD (*p<0.05, **p<0.01, ***p<0.001).

  • Fig. 2 PVC-CM inhibits the inflammasomes activation and caspase-1 activity in mouse peritoneal macrophage. Peritoneal macrophages were isolated, primed with LPS for 3 h, and subjected to the indicated inflammasome triggers in the present of PVC-CM. (A) Sup, Lys, and Pellet from whole-cell lysates were analyzed for caspase-1, IL-1β, or ASC pyroptosome by immunoblot assay. (B) Supernatant mouse IL-1β levels were measured by ELISA. (C) rhCasp1 was incubated with its substrate (YVAD-pNA) in the presence of PVC-CM as indicated. Relative Casp1 activity in the reaction without the rhCasp1 was set as 0% and that in the reaction with Casp1 and YVAD-pNA without PVC-CM was set as 100%. Z-VAD indicates Z-VAD-FMK, a pan-caspase inhibitor. (D) The cytotoxicity of PVC-CM was measured after applying the indicated dosage of PVC-CM to PMA-differentiated THP-1 for 1 h, identical with the inflammasome-activating step. The survival rate of the Triton-treated group was set as 0% and that of the non-treated group was set as 100%. Error bars indicate SD (*p<0.05, **p<0.01, ***p<0.001).

  • Fig. 3 PVC-CM reduces inflammation and ER stress in in macrophages of MSU-induced peritonitis. (A) Schematic diagram of experiments using the peritonitis mouse model (n=8 per group). (B) PECs were determined by counting exudate cells with a cell counter. (C) Cytospins of isolated PECs were stained with Wright-Giemsa stain. (D, E) Relative mRNA levels of inflammatory (IL-1α, IL-1β, NLRP3, and Casp1) and ER stress-related genes (ATF6, PERK, elk2α, and IRE-1) were analyzed using SYBR green-based quantitative real-time PCR. Error bars indicate SD (*p<0.05, **p<0.01).

  • Fig. 4 Secretome analysis. Proteome profiler arrays were evaluated as described in Methods. (A) The heat map represents the subsequent image analysis and quantification of pixel intensity for each spot. (B, C) LPS-primed THP-1 cells treated with inflammasome triggers in the absence or presence of rhSerpin E1 or rhAngiogenin for 1 h. IL-1β levels in the supernatants were measured by ELISA. (D) Caspase-1 activity was determined by colorimetric assay. Error bars indicate SD (*p<0.05, **p<0.01, ***p<0.001).

  • Fig. 5 The Mechanism underlying the inhibitory effect of PVC-CM on NLRP3 inflammasome via mitochondrial ROS generation. (A) LPS-primed THP-1 cells treated with ATP (2 mM) and the indicated DPI (100 to 10 μM) for 1 h. Sup and Lys were analyzed by immunoblot using the indicated anti-sera. (B) LPS-primed THP-1 cells were treated with rotenone (20 μM) and the indicated dosages of PVC-CM for 6 h, after which mitochondrial ROS generation was analyzed by immunoblot. (C) Mitochondrial ROS (MitoSOX) were quantified using a fluorometric microplate reader. (D) Relative fluorescence intensities of Mito Tracker (mitochondria) and MitoSOX (mitochondrial ROS) were analyzed using confocal microscopy. Hoechst was used for nuclear staining. Error bars indicate SD (*p<0.05, **p<0.01, ***p<0.001).


Reference

References

1. Martinon F, Mayor A, Tschopp J. The inflammasomes: guardians of the body. Annu Rev Immunol. 2009; 27:229–265. DOI: 10.1146/annurev.immunol.021908.132715. PMID: 19302040.
Article
2. Chen G, Shaw MH, Kim YG, Nuñez G. NOD-like receptors: role in innate immunity and inflammatory disease. Annu Rev Pathol. 2009; 4:365–398. DOI: 10.1146/annurev.pathol.4.110807.092239. PMID: 18928408.
Article
3. Yang CA, Chiang BL. Inflammasomes and human autoimmunity: a comprehensive review. J Autoimmun. 2015; 61:1–8. DOI: 10.1016/j.jaut.2015.05.001. PMID: 26005048.
Article
4. Broderick L, De Nardo D, Franklin BS, Hoffman HM, Latz E. The inflammasomes and autoinflammatory syndromes. Annu Rev Pathol. 2015; 10:395–424. DOI: 10.1146/annurev-pathol-012414-040431. PMID: 25423351.
Article
5. Di Stefano A, Caramori G, Barczyk A, Vicari C, Brun P, Zanini A, Cappello F, Garofano E, Padovani A, Contoli M, Casolari P, Durham AL, Chung KF, Barnes PJ, Papi A, Adcock I, Balbi B. Innate immunity but not NLRP3 inflammasome activation correlates with severity of stable COPD. Thorax. 2014; 69:516–524. DOI: 10.1136/thoraxjnl-2012-203062. PMID: 24430176. PMCID: PMC4219154.
Article
6. Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol. 2010; 11:136–140. DOI: 10.1038/ni.1831. PMID: 20023662.
Article
7. Si YL, Zhao YL, Hao HJ, Fu XB, Han WD. MSCs: Biological characteristics, clinical applications and their outstanding concerns. Ageing Res Rev. 2011; 10:93–103. DOI: 10.1016/j.arr.2010.08.005. PMID: 20727988.
Article
8. Yi TG, Cho YK, Lee HJ, Kim J, Jeon MS, Ham DS, Kim WC, Song SU. A novel immunomodulatory mechanism dependent on acetylcholine secreted by human bone marrow-derived mesenchymal stem cells. Int J Stem Cells. 2019; 12:315–330. DOI: 10.15283/ijsc18098. PMID: 31242717. PMCID: PMC6657938.
Article
9. English K. Mechanisms of mesenchymal stromal cell immunomodulation. Immunol Cell Biol. 2013; 91:19–26. DOI: 10.1038/icb.2012.56. PMID: 23090487.
Article
10. Park HJ, Kim J, Saima FT, Rhee KJ, Hwang S, Kim MY, Baik SK, Eom YW, Kim HS. Adipose-derived stem cells ameliorate colitis by suppression of inflammasome formation and regulation of M1-macrophage population through prostaglandin E2. Biochem Biophys Res Commun. 2018; 498:988–995. DOI: 10.1016/j.bbrc.2018.03.096. PMID: 29550474.
Article
11. Mohamadi Y, Noori Moghahi SMH, Mousavi M, Borhani-Haghighi M, Abolhassani F, Kashani IR, Hassanzadeh G. Intrathecal transplantation of Wharton’s jelly mesenchymal stem cells suppresses the NLRP1 inflammasome in the rat model of spinal cord injury. J Chem Neuroanat. 2019; 97:1–8. DOI: 10.1016/j.jchemneu.2019.01.011. PMID: 30703433.
Article
12. Sun X, Hao H, Han Q, Song X, Liu J, Dong L, Han W, Mu Y. Human umbilical cord-derived mesenchymal stem cells ameliorate insulin resistance by suppressing NLRP3 inflammasome-mediated inflammation in type 2 diabetes rats. Stem Cell Res Ther. 2017; 8:241. DOI: 10.1186/s13287-017-0668-1. PMID: 29096724. PMCID: PMC5667486.
Article
13. Crisan M, Yap S, Casteilla L, Chen CW, Corselli M, Park TS, Andriolo G, Sun B, Zheng B, Zhang L, Norotte C, Teng PN, Traas J, Schugar R, Deasy BM, Badylak S, Buhring HJ, Giacobino JP, Lazzari L, Huard J, Péault B. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 2008; 3:301–313. DOI: 10.1016/j.stem.2008.07.003. PMID: 18786417.
Article
14. Kim JY, Lee JY, Ha KS, Han ET, Park WS, Min CK, Hong SH. Perivascular cells and NADPH oxidase inhibition partially restore hyperglycemia-induced alterations in hematopoietic stem cell and myeloid-derived suppressor cell populations in the bone marrow. Int J Stem Cells. 2018; 12:63–72. DOI: 10.15283/ijsc18097. PMID: 30595009. PMCID: PMC6457702.
Article
15. An B, Kim E, Song H, Ha KS, Han ET, Park WS, Ahn TG, Yang SR, Na S, Hong SH. Gestational diabetes affects the growth and functions of perivascular stem cells. Mol Cells. 2017; 40:434–439. DOI: 10.14348/molcells.2017.0053. PMID: 28614916. PMCID: PMC5523020.
Article
16. Baksh D, Yao R, Tuan RS. Comparison of proliferative and multilineage differentiation potential of human mesenchymal stem cells derived from umbilical cord and bone marrow. Stem Cells. 2007; 25:1384–1392. DOI: 10.1634/stemcells.2006-0709. PMID: 17332507.
Article
17. Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature. 2011; 469:221–225. DOI: 10.1038/nature09663. PMID: 21124315.
Article
18. Ogura S, Kurata K, Hattori Y, Takase H, Ishiguro-Oonuma T, Hwang Y, Ahn S, Park I, Ikeda W, Kusuhara S, Fukushima Y, Nara H, Sakai H, Fujiwara T, Matsushita J, Ema M, Hirashima M, Minami T, Shibuya M, Takakura N, Kim P, Miyata T, Ogura Y, Uemura A. Sustained inflammation after pericyte depletion induces irreversible blood-retina barrier breakdown. JCI Insight. 2017; 2:e90905. DOI: 10.1172/jci.insight.90905. PMID: 28194443. PMCID: PMC5291729.
Article
19. Leaf IA, Nakagawa S, Johnson BG, Cha JJ, Mittelsteadt K, Guckian KM, Gomez IG, Altemeier WA, Duffield JS. Pericyte MyD88 and IRAK4 control inflammatory and fibrotic responses to tissue injury. J Clin Invest. 2017; 127:321–334. DOI: 10.1172/JCI87532. PMID: 27869651. PMCID: PMC5199713.
Article
20. Oh JY, Ko JH, Lee HJ, Yu JM, Choi H, Kim MK, Wee WR, Prockop DJ. Mesenchymal stem/stromal cells inhibit the NLRP3 inflammasome by decreasing mitochondrial reactive oxygen species. Stem Cells. 2014; 32:1553–1563. DOI: 10.1002/stem.1608. PMID: 24307525.
Article
21. Kishimoto K, Liu S, Tsuji T, Olson KA, Hu GF. Endogenous angiogenin in endothelial cells is a general requirement for cell proliferation and angiogenesis. Oncogene. 2005; 24:445–456. DOI: 10.1038/sj.onc.1208223. PMID: 15558023.
Article
22. Mihai MC, Popa MA, Suica VI, Antohe F, Jackson EK, Simionescu M, Dubey RK. Mechanism of 17β-estradiol stimulated integration of human mesenchymal stem cells in heart tissue. J Mol Cell Cardiol. 2019; 133:115–124. DOI: 10.1016/j.yjmcc.2019.06.007. PMID: 31201797.
Article
23. Kim HK, Lee SG, Lee SW, Oh BJ, Kim JH, Kim JA, Lee G, Jang JD, Joe YA. A subset of paracrine factors as efficient biomarkers for predicting vascular regenerative efficacy of mesenchymal stromal/stem cells. Stem Cells. 2019; 37:77–88. DOI: 10.1002/stem.2920. PMID: 30281870.
Article
24. Chang PY, Zhang BY, Cui S, Qu C, Shao LH, Xu TK, Qu YQ, Dong LH, Wang J. MSC-derived cytokines repair radiation-induced intra-villi microvascular injury. Oncotarget. 2017; 8:87821–87836. DOI: 10.18632/oncotarget.21236. PMID: 29152123. PMCID: PMC5675675.
Article
25. Zhang Y, Xia X, Yan J, Yan L, Lu C, Zhu X, Wang T, Yin T, Li R, Chang HM, Qiao J. Mesenchymal stem cell-derived angiogenin promotes primodial follicle survival and angiogenesis in transplanted human ovarian tissue. Reprod Biol Endocrinol. 2017; 15:18. DOI: 10.1186/s12958-017-0235-8. PMID: 28274269. PMCID: PMC5343383.
Article
26. Lee SH, Kim KW, Min KM, Kim KW, Chang SI, Kim JC. Angiogenin reduces immune inflammation via inhibition of TANK-binding kinase 1 expression in human corneal fibroblast cells. Mediators Inflamm. 2014; 2014:861435. DOI: 10.1155/2014/861435. PMID: 24860242. PMCID: PMC4016892.
Article
27. Oikonomou KA, Kapsoritakis AN, Kapsoritaki AI, Manolakis AC, Tiaka EK, Tsiopoulos FD, Tsiompanidis IA, Potamianos SP. Angiogenin, angiopoietin-1, angiopoietin-2, and endostatin serum levels in inflammatory bowel disease. Inflamm Bowel Dis. 2011; 17:963–970. DOI: 10.1002/ibd.21410. PMID: 20629092.
Article
28. Etoh T, Shibuta K, Barnard GF, Kitano S, Mori M. Angiogenin expression in human colorectal cancer: the role of focal macrophage infiltration. Clin Cancer Res. 2000; 6:3545–3551. PMID: 10999742.
29. Yin Y, Hao H, Cheng Y, Zang L, Liu J, Gao J, Xue J, Xie Z, Zhang Q, Han W, Mu Y. Human umbilical cord-derived mesenchymal stem cells direct macrophage polarization to alleviate pancreatic islets dysfunction in type 2 diabetic mice. Cell Death Dis. 2018; 9:760. DOI: 10.1038/s41419-018-0801-9. PMID: 29988034. PMCID: PMC6037817.
Article
30. Ahn SY, Park WS, Kim YE, Sung DK, Sung SI, Ahn JY, Chang YS. Vascular endothelial growth factor mediates the therapeutic efficacy of mesenchymal stem cell-derived extracellular vesicles against neonatal hyperoxic lung injury. Exp Mol Med. 2018; 50:26. DOI: 10.1038/s12276-018-0055-8. PMID: 29650962. PMCID: PMC5938045.
Article
31. Lee SE, Jang JE, Kim HS, Jung MK, Ko MS, Kim MO, Park HS, Oh WI, Choi SJ, Jin HJ, Kim SY, Kim YJ, Kim SW, Kim MK, Sung CO, Pack CG, Lee KU, Koh EH. Mesenchymal stem cells prevent the progression of diabetic nephropathy by improving mitochondrial function in tubular epithelial cells. Exp Mol Med. 2019; 51:1–14. DOI: 10.1038/s12276-019-0268-5. PMID: 31285429. PMCID: PMC6802630.
Article
Full Text Links
  • IJSC
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr