CC BY-NC-ND 4.0 · AJP Rep 2024; 14(01): e69-e73
DOI: 10.1055/a-2216-9194
Original Article

Mesenchymal Stem Cells Suppress Inflammatory Cytokines in Lipopolysaccharide Exposed Preterm and Term Human Pregnant Myometrial Cells

Arunmani Mani
1   Department of Obstetrics Gynecology and Reproductive Sciences, University of Texas Health Science Center at Houston, Houston, Texas
,
John Hotra
1   Department of Obstetrics Gynecology and Reproductive Sciences, University of Texas Health Science Center at Houston, Houston, Texas
,
Sean C. Blackwell
2   Department of Obstetrics and Gynecology, University of Texas Health Science Center at Houston, Houston, Texas
,
Laura Goetzl
1   Department of Obstetrics Gynecology and Reproductive Sciences, University of Texas Health Science Center at Houston, Houston, Texas
,
Jerrie S. Refuerzo
1   Department of Obstetrics Gynecology and Reproductive Sciences, University of Texas Health Science Center at Houston, Houston, Texas
› Author Affiliations

Abstract

Objective The objective of this study was to determine the cytokine response in human pregnant preterm and term myometrial cells exposed to lipopolysaccharide (LPS) and cocultured with mesenchymal stem cells (MSCs).

Study Design Myometrium was obtained at cesarean delivery in term and preterm patients. Human myometrial cells were exposed to 5 μg/mL LPS for 4 hours followed by 1 μg/mL LPS for 24 hours and were cocultured with MSCs for 24 hours. Culture supernatants were collected at 24 hours and expression of cytokines, including interleukin-1β (IL-1β), IL-6, IL-8, tumor necrosis factor-α (TNF-α), transforming growth factor-β (TGF-β), and IL-10, was quantified by enzyme-linked immunosorbent assay.

Results There was significantly increased expression of the proinflammatory cytokines IL-1β, IL-6, IL-8, and TNF-α in preterm myometrial cells treated with LPS compared with untreated preterm myometrial cells. Coculture with MSCs significantly suppressed the proinflammatory cytokine levels in LPS-treated preterm versus treated term myometrial cells. Moreover, MSC cocultured preterm myometrial cells expressed increased levels of the anti-inflammatory cytokines TGF-β and IL-10 compared with treated term myometrial cells.

Conclusion MSCs ameliorate LPS-mediated inflammation in preterm human myometrial cells compared with term myometrial cells. Immunomodulatory effects of MSCs mediated through anti-inflammatory cytokine regulation suggest a potential cell-based therapy for preterm birth.



Publication History

Received: 27 June 2023

Accepted: 19 October 2023

Accepted Manuscript online:
23 November 2023

Article published online:
18 February 2024

© 2024. 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, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

 
  • References

  • 1 Keelan JA, Blumenstein M, Helliwell RJ, Sato TA, Marvin KW, Mitchell MD. Cytokines, prostaglandins and parturition–a review. Placenta 2003; 24 (Suppl A): S33-S46
  • 2 Nadeau HC, Subramaniam A, Andrews WW. Infection and preterm birth. Semin Fetal Neonatal Med 2016; 21 (02) 100-105
  • 3 Sivarajasingam SP, Imami N, Johnson MR. Myometrial cytokines and their role in the onset of labour. J Endocrinol 2016; 231 (03) R101-R119
  • 4 Törnblom SA, Klimaviciute A, Byström B, Chromek M, Brauner A, Ekman-Ordeberg G. Non-infected preterm parturition is related to increased concentrations of IL-6, IL-8 and MCP-1 in human cervix. Reprod Biol Endocrinol 2005; 3: 39
  • 5 Raba G, Tabarkiewicz J. Cytokines in preterm delivery: proposal of a new diagnostic algorithm. J Immunol Res 2018; 8073476
  • 6 Pandey M, Chauhan M, Awasthi S. Interplay of cytokines in preterm birth. Indian J Med Res 2017; 146 (03) 316-327
  • 7 Singh N, Herbert B, Sooranna GR. et al. Is myometrial inflammation a cause or a consequence of term human labour?. J Endocrinol 2017; 235 (01) 69-83
  • 8 Xu S, Liu C, Ji HL. Concise review: therapeutic potential of the mesenchymal stem cell derived secretome and extracellular vesicles for radiation-induced lung injury: progress and hypotheses. Stem Cells Transl Med 2019; 8 (04) 344-354
  • 9 Saldaña L, Bensiamar F, Vallés G, Mancebo FJ, García-Rey E, Vilaboa N. Immunoregulatory potential of mesenchymal stem cells following activation by macrophage-derived soluble factors. Stem Cell Res Ther 2019; 10 (01) 58
  • 10 Kyurkchiev D, Bochev I, Ivanova-Todorova E. et al. Secretion of immunoregulatory cytokines by mesenchymal stem cells. World J Stem Cells 2014; 6 (05) 552-570
  • 11 Putra A, Ridwan FB, Putridewi AI. et al. The role of TNF-α induced MSCs on suppressive inflammation by increasing TGF-β and IL-10. Open Access Maced J Med Sci 2018; 6 (10) 1779-1783
  • 12 Nuzzo AM, Moretti L, Mele P, Todros T, Eva C, Rolfo A. Effect of placenta-derived mesenchymal stromal cells conditioned media on an LPS-induced mouse model of preeclampsia. Int J Mol Sci 2022; 23 (03) 1674
  • 13 Li YH, Zhang D, Du MR. Advances and challenges of mesenchymal stem cells for pregnancy-related diseases. Cell Mol Immunol 2021; 18 (08) 2075-2077
  • 14 Suvakov S, Richards C, Nikolic V. et al. Emerging therapeutic potential of mesenchymal stem/stromal cells in preeclampsia. Curr Hypertens Rep 2020; 22 (05) 37
  • 15 Kusuma GD, Georgiou HM, Perkins AV, Abumaree MH, Brennecke SP, Kalionis B. Mesenchymal stem/stromal cells and their role in oxidative stress associated with preeclampsia. Yale J Biol Med 2022; 95 (01) 115-127
  • 16 Jin S, Wu C, Chen M, Sun D, Zhang H. The pathological and therapeutic roles of mesenchymal stem cells in preeclampsia. Front Med (Lausanne) 2022; 9: 923334
  • 17 Mani A, Hotra JW, Blackwell SC, Goetzl L, Refuerzo JS. Mesenchymal stem cells attenuate lipopolysaccharide-induced inflammatory response in human uterine smooth muscle cells. AJP Rep 2020; 10 (03) e335-e341
  • 18 Mosher AA, Rainey KJ, Bolstad SS. et al. Development and validation of primary human myometrial cell culture models to study pregnancy and labour. BMC Pregnancy Childbirth 2013; 13 (Suppl 1): S7
  • 19 Li H, Shen S, Fu H. et al. Immunomodulatory functions of mesenchymal stem cells in tissue engineering. Stem Cells Int 2019; 9671206
  • 20 Schäfer R. Advanced cell therapeutics are changing the clinical landscape: will mesenchymal stromal cells be a part of it?. BMC Med 2019; 17 (01) 53
  • 21 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
  • 22 Rajagopal SP, Hutchinson JL, Dorward DA, Rossi AG, Norman JE. Crosstalk between monocytes and myometrial smooth muscle in culture generates synergistic pro-inflammatory cytokine production and enhances myocyte contraction, with effects opposed by progesterone. Mol Hum Reprod 2015; 21 (08) 672-686
  • 23 Cappelletti M, Della Bella S, Ferrazzi E, Mavilio D, Divanovic S. Inflammation and preterm birth. J Leukoc Biol 2016; 99 (01) 67-78
  • 24 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; 2631439
  • 25 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
  • 26 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 (01) 65
  • 27 Kim H, Darwish I, Monroy MF, Prockop DJ, Liles WC, Kain KC. Mesenchymal stromal (stem) cells suppress pro-inflammatory cytokine production but fail to improve survival in experimental staphylococcal toxic shock syndrome. BMC Immunol 2014; 15: 1
  • 28 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
  • 29 Wang J, Huang C, Jiang R. et al. Decreased endometrial IL-10 impairs endometrial receptivity by downregulating HOXA10 expression in women with adenomyosis. BioMed Res Int 2018; 2549789
  • 30 Gotsch F, Romero R, Kusanovic JP. et al. The anti-inflammatory limb of the immune response in preterm labor, intra-amniotic infection/inflammation, and spontaneous parturition at term: a role for interleukin-10. J Matern Fetal Neonatal Med 2008; 21 (08) 529-547
  • 31 Pereira TB, Thomaz EB, Nascimento FR. et al. Regulatory cytokine expression and preterm birth: case-control study nested in a cohort. PLoS One 2016; 11 (08) e0158380
  • 32 Costa EM, de Araujo Figueiredo CS, Martins RFM. et al. Periodontopathogenic microbiota, infectious mechanisms and preterm birth: analysis with structural equations (cohort-BRISA). Arch Gynecol Obstet 2019; 300 (06) 1521-1530
  • 33 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 (01) e0190358
  • 34 Nakazaki M, Morita T, Lankford KL, Askenase PW, Kocsis JD. Small extracellular vesicles released by infused mesenchymal stromal cells target M2 macrophages and promote TGF-β upregulation, microvascular stabilization and functional recovery in a rodent model of severe spinal cord injury. J Extracell Vesicles 2021; 10 (11) e12137