CC BY-NC-ND 4.0 · AJP Rep 2020; 10(03): e335-e341
DOI: 10.1055/s-0040-1715166
Case Report

Mesenchymal Stem Cells Attenuate Lipopolysaccharide-Induced Inflammatory Response in Human Uterine Smooth Muscle Cells

Arunmani Mani
1   Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, UT Health-McGovern Medical School, Houston, Texas
,
John W. Hotra
1   Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, UT Health-McGovern Medical School, Houston, Texas
,
Sean C. Blackwell
1   Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, UT Health-McGovern Medical School, Houston, Texas
,
Laura Goetzl
1   Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, UT Health-McGovern Medical School, Houston, Texas
,
Jerrie S. Refuerzo
1   Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology and Reproductive Sciences, UT Health-McGovern Medical School, Houston, Texas
› Institutsangaben
Funding The study was supported from the Department of Obstetrics, Gynecology, and Reproductive Sciences, UTHealth-McGovern Medical School, Houston TX and The Larry C. Gilstrap, MD, Center for Perinatal and Women's Health Research, University of Texas Health Science Center at Houston.

Abstract

Objective The aim of this study was to determine if mesenchymal stem cells (MSCs) would suppress the inflammatory response in human uterine cells in an in vitro lipopolysaccharide (LPS)-based preterm birth (PTB) model.

Study Design Cocultures of human uterine smooth muscle cells (HUtSMCs) and MSCs were exposed to 5 μg/mL LPS for 4 hours and further challenged with 1 μg/mL LPS for a subsequent 24 hours. Key elements of the parturition cascade regulated by toll-like receptors (TLRs) through activation of mitogen-activated protein kinases (MAPKs) were quantified in culture supernatant as biomarkers of MSC modulation.

Results Coculture with MSCs significantly attenuated TLR-4, p-JNK, and p- extracellular signal-regulated kinase 1/2 (ERK1/2) protein levels compared with HUtSMCs monoculture (p = 0.05). In addition, coculture was associated with significant inhibition of proinflammatory cytokines interleukin (IL)-6 and IL-8 (p = 0.0001) and increased production of anti-inflammatory cytokines IL-10 and transforming growth factor (TGF)-β1 (p = 0.0001).

Conclusion MSCs appear to play a role in significantly attenuating LPS-mediated inflammation via alteration of down-stream MAPKs. MSCs may represent a novel, cell-based therapy in women with increased risk of inflammatory-mediated preterm birth.

Note

This study was presented as Posters at the 38th Annual Meeting of the Society for Maternal-Fetal Medicine; January 29 to February 3, 2018; Dallas, TX, and at the 65th Annual Scientific Meeting of the Society for Reproductive Investigation; March 6 to 10, 2018 in San Diego, CA.




Publikationsverlauf

Eingereicht: 27. März 2020

Angenommen: 24. April 2020

Artikel online veröffentlicht:
23. September 2020

© 2020. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial-License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/).

Thieme Medical Publishers
333 Seventh Avenue, New York, NY 10001, USA.

 
  • References

  • 1 Cappelletti M, Della Bella S, Ferrazzi E, Mavilio D, Divanovic S. Inflammation and preterm birth. J Leukoc Biol 2016; 99 (01) 67-78
  • 2 Frey HA, Klebanoff MA. The epidemiology, etiology, and costs of preterm birth. Semin Fetal Neonatal Med 2016; 21 (02) 68-73
  • 3 Nadeau HC, Subramaniam A, Andrews WW. Infection and preterm birth. Semin Fetal Neonatal Med 2016; 21 (02) 100-105
  • 4 Martin JA, Hamilton BE, Osterman MJK, Driscoll AK, Drake P. Births: Final Data for 2016. Natl Vital Stat Rep 2018; 67 (01) 1-55
  • 5 Boyle AK, Rinaldi SF, Norman JE, Stock SJ. Preterm birth: Inflammation, fetal injury and treatment strategies. J Reprod Immunol 2017; 119: 62-66
  • 6 Bollapragada S, Youssef R, Jordan F, Greer I, Norman J, Nelson S. Term labor is associated with a core inflammatory response in human fetal membranes, myometrium, and cervix. Am J Obstet Gynecol 2009; 200 (01) 104.e1-104.e11
  • 7 Peltier MR. Immunology of term and preterm labor. Reprod Biol Endocrinol 2003; 1: 122
  • 8 Yuan M, Jordan F, McInnes IB, Harnett MM, Norman JE. Leukocytes are primed in peripheral blood for activation during term and preterm labour. Mol Hum Reprod 2009; 15 (11) 713-724
  • 9 Goldenberg RL, Hauth JC, Andrews WW. Intrauterine infection and preterm delivery. N Engl J Med 2000; 342 (20) 1500-1507
  • 10 Agrawal V, Hirsch E. Intrauterine infection and preterm labor. Semin Fetal Neonatal Med 2012; 17 (01) 12-19
  • 11 Kemp MW. Preterm birth, intrauterine infection, and fetal inflammation. Front Immunol 2014; 5: 574
  • 12 Mogensen TH. Pathogen recognition and inflammatory signaling in innate immune defenses. Clin Microbiol Rev 2009; 22 (02) 240-273 Table of Contents.
  • 13 Newton K, Dixit VM. Signaling in innate immunity and inflammation. Cold Spring Harb Perspect Biol 2012; 4 (03) a006049
  • 14 Amirchaghmaghi E, Taghavi SA, Shapouri F, Saeidi S, Rezaei A, Aflatoonian R. The role of toll like receptors in pregnancy. Int J Fertil Steril 2013; 7 (03) 147-154
  • 15 Elovitz MA, Wang Z, Chien EK, Rychlik DF, Phillippe M. A new model for inflammation-induced preterm birth: the role of platelet-activating factor and Toll-like receptor-4. Am J Pathol 2003; 163 (05) 2103-2111
  • 16 Noguchi T, Sado T, Naruse K. , et al. Evidence for activation of Toll-like receptor and receptor for advanced glycation end products in preterm birth. Mediators Inflamm 2010; 2010: 490406
  • 17 Thaxton JE, Nevers TA, Sharma S. TLR-mediated preterm birth in response to pathogenic agents. Infect Dis Obstet Gynecol 2010; 2010: 378472
  • 18 Koga K, Izumi G, Mor G, Fujii T, Osuga Y. Toll-like receptors at the maternal-fetal interface in normal pregnancy and pregnancy complications. Am J Reprod Immunol 2014; 72 (02) 192-205
  • 19 Kawai T, Akira S. The roles of TLRs, RLRs and NLRs in pathogen recognition. Int Immunol 2009; 21 (04) 317-337
  • 20 Guo J, Zheng L, Chen L. , et al. Lipopolysaccharide activated TLR4/NF-κB signaling pathway of fibroblasts from uterine fibroids. Int J Clin Exp Pathol 2015; 8 (09) 10014-10025
  • 21 Lyon D, Cheng CY, Howland L. , et al. Integrated review of cytokines in maternal, cord, and newborn blood: part I--associations with preterm birth. Biol Res Nurs 2010; 11 (04) 371-376
  • 22 Sivarajasingam SP, Imami N, Johnson MR. Myometrial cytokines and their role in the onset of labour. J Endocrinol 2016; 231 (03) R101-R119
  • 23 Lei J, Firdaus W, Rosenzweig JM. , et al. Murine model: maternal administration of stem cells for prevention of prematurity. Am J Obstet Gynecol 2015; 212 (05) 639.e1-639.e10
  • 24 Lindström TM, Bennett PR. The role of nuclear factor kappa B in human labour. Reproduction 2005; 130 (05) 569-581
  • 25 Otun HA, MacDougall MW, Bailey J, Europe-Finner GN, Robson SC. Spatial and temporal expression of the myometrial mitogen-activated protein kinases p38 and ERK1/2 in the human uterus during pregnancy and labor. J Soc Gynecol Investig 2005; 12 (03) 185-190
  • 26 Robertson SA, Wahid HH, Chin PY, Hutchinson MR, Moldenhauer LM, Keelan JA. Toll-like receptor-4: a new target for preterm labour pharmacotherapies?. Curr Pharm Des 2018; 24 (09) 960-973
  • 27 Bhaskar B, Mekala NK, Baadhe RR, Rao PS. Role of signaling pathways in mesenchymal stem cell differentiation. Curr Stem Cell Res Ther 2014; 9 (06) 508-512
  • 28 Oh JY, Ko JH, Lee HJ. , et al. Mesenchymal stem/stromal cells inhibit the NLRP3 inflammasome by decreasing mitochondrial reactive oxygen species. Stem Cells 2014; 32 (06) 1553-1563
  • 29 Sangiorgi B, Panepucci RA. Modulation of immunoregulatory properties of mesenchymal stromal cells by toll-like receptors: potential applications on GVHD. Stem Cells Int 2016; 2016: 9434250
  • 30 Zhang ZH, Zhu W, Ren HZ. , et al. Mesenchymal stem cells increase expression of heme oxygenase-1 leading to anti-inflammatory activity in treatment of acute liver failure. Stem Cell Res Ther 2017; 8 (01) 70
  • 31 Zhu H, Yang F, Tang B. , et al. Mesenchymal stem cells attenuated PLGA-induced inflammatory responses by inhibiting host DC maturation and function. Biomaterials 2015; 53: 688-698
  • 32 Pedrazza L, Cubillos-Rojas M, de Mesquita FC. , et al. Mesenchymal stem cells decrease lung inflammation during sepsis, acting through inhibition of the MAPK pathway. Stem Cell Res Ther 2017; 8 (01) 289
  • 33 Song IH, Jung KJ, Lee TJ. , et al. Mesenchymal stem cells attenuate adriamycin-induced nephropathy by diminishing oxidative stress and inflammation via downregulation of the NF-kB. Nephrology (Carlton) 2018; 23 (05) 483-492
  • 34 Krikun G, Trezza J, Shaw J. , et al. Lipopolysaccharide appears to activate human endometrial endothelial cells through TLR-4-dependent and TLR-4-independent mechanisms. Am J Reprod Immunol 2012; 68 (03) 233-237
  • 35 Faure E, Equils O, Sieling PA. , et al. Bacterial lipopolysaccharide activates NF-kappaB through toll-like receptor 4 (TLR-4) in cultured human dermal endothelial cells. Differential expression of TLR-4 and TLR-2 in endothelial cells. J Biol Chem 2000; 275 (15) 11058-11063
  • 36 Arima K, Nasu K, Narahara H, Fujisawa K, Matsui N, Miyakawa I. Effects of lipopolysaccharide and cytokines on production of RANTES by cultured human endometrial stromal cells. Mol Hum Reprod 2000; 6 (03) 246-251
  • 37 Oude Nijhuis CS, Vellenga E, Daenen SM, Kamps WA, De Bont ES. Endothelial cells are main producers of interleukin 8 through Toll-like receptor 2 and 4 signaling during bacterial infection in leukopenic cancer patients. Clin Diagn Lab Immunol 2003; 10 (04) 558-563
  • 38 Li H, Shen S, Fu H. , et al. Immunomodulatory functions of mesenchymal stem cells in tissue engineering. Stem Cells Int 2019; 2019: 9671206
  • 39 Li D, Wang C, Chi C. , et al. Bone marrow mesenchymal stem cells inhibit lipopolysaccharide-induced inflammatory reactions in macrophages and endothelial cells. Mediators Inflamm 2016; 2016: 2631439
  • 40 Rahmat Z, Jose S, Ramasamy R, Vidyadaran S. Reciprocal interactions of mouse bone marrow-derived mesenchymal stem cells and BV2 microglia after lipopolysaccharide stimulation. Stem Cell Res Ther 2013; 4 (01) 12
  • 41 Cai SX, Liu AR, Chen S. , et al. Activation of Wnt/β-catenin signalling promotes mesenchymal stem cells to repair injured alveolar epithelium induced by lipopolysaccharide in mice. Stem Cell Res Ther 2015; 6: 65
  • 42 Kubosch EJ, Heidt E, Bernstein A, Böttiger K, Schmal H. The trans-well coculture of human synovial mesenchymal stem cells with chondrocytes leads to self-organization, chondrogenic differentiation, and secretion of TGFβ. Stem Cell Res Ther 2016; 7 (01) 64
  • 43 Zhu H, Xiong Y, Xia Y. , et al. Therapeutic effects of human umbilical cord-derived mesenchymal stem cells in acute lung injury mice. Sci Rep 2017; 7: 39889
  • 44 Németh K, Leelahavanichkul A, Yuen PS. , 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 (01) 42-49
  • 45 Gu W, Song L, Li XM, Wang D, Guo XJ, Xu WG. Mesenchymal stem cells alleviate airway inflammation and emphysema in COPD through down-regulation of cyclooxygenase-2 via p38 and ERK MAPK pathways. Sci Rep 2015; 5: 8733
  • 46 Martire A, Bedada FB, Uchida S. , et al. Mesenchymal stem cells attenuate inflammatory processes in the heart and lung via inhibition of TNF signaling. Basic Res Cardiol 2016; 111 (05) 54