Skip to main content

Advertisement

Log in

In vitro–Generated MDSCs Reduce the Pregnancy Complications in an Abortion-Prone Murine Model

  • Reproductive Biology: Original Article
  • Published:
Reproductive Sciences Aims and scope Submit manuscript

Abstract  

Recurrent spontaneous abortion (RSA) is one of the major pregnancy-related complications. The roles of different immune cells have been studied in pregnancy complications. The current study aimed to investigate myeloid-derived suppressor cells (MDSCs) in a murine abortion model and introduce a therapeutic approach by using in vitro–generated MDSCs in this model. CBA/J × DBA/2 (abortion prone) and CBA/J × Balb/C (normal pregnancy) mice were used. The frequency of granulocytic MDSCs, monocytic MDSCs, and Tregs was checked in the bone marrow and uteroplacental tissue of mice on three gestational days (gd9.5, gd13.5, and gd17.5) using the flow cytometry approach. MDSCs were generated in vitro from bone marrow-isolated cells using GM-CSF and IL-6 cytokines. Abortion-prone mice were injected intravenously with in vitro–generated MDSCs at gd0.5, and pregnancy outcomes were recorded in treated mice. The frequency of G-MDSCs and M-MDSCs in the bone marrow of abortion-prone mice was decreased at gd9.5 (p = 0.026 and p = 0.05, respectively). In uteroplacental tissue, the frequency of G-MDSCs was significantly lower at gd9.5 and gd13.5 (p = 0.001, p = 0.029, respectively), while M-MDSCs only showed decreased number at gd9.5 (p = 0.05) in abortion-prone mice. Injection of in vitro–generated MDSCs resulted in the increased fetus and placenta weights (p = 0.049 and p = 0.012, respectively) but showed no effect on the number of live fetuses and abortion rate. The reduced frequency of both G-MDSCs and M-MDSCs in the bone marrow and at the feto-maternal interface is associated with pregnancy complications. In vitro–generated MDSCs could be considered as a potential approach to reduce these complications.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10

Similar content being viewed by others

Data Availability

Data and material will be available if demanded.

Code Availability

Not applicable.

Abbreviations

MDSC:

Myeloid-derived suppressor cell

G-MDSC:

Granulocytic myeloid-derived suppressor cell

M-MDSC:

Monocytic myeloid-derived suppressor cell

RSA:

Recurrent spontaneous abortion

TGF-β:

Transforming growth factor beta

Arg-1:

Arginase-1

IDO:

Indoleamine 2,3-dioxygenase

INOS:

Inducible nitric oxide synthase

GAPDH:

Glyceraldehyde 3-phosphate dehydrogenase

References

  1. Rpl EGGO, Bender Atik R, Christiansen OB et al. ESHRE guideline: recurrent pregnancy loss. Hum Reprod Open 2018(2), hoy004 (2018).

  2. Ford HB, Schust DJ. Recurrent pregnancy loss: etiology, diagnosis, and therapy. Rev Obstet Gynecol. 2009;2(2):76.

    PubMed  PubMed Central  Google Scholar 

  3. Mor G, Cardenas I. The immune system in pregnancy: a unique complexity. Am J Reprod Immunol. 2010;63(6):425–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Orefice R. Immunology and the immunological response in pregnancy. Best Pract Res Clin Obstet Gynaecol. 2021;76:3–12.

    Article  CAS  PubMed  Google Scholar 

  5. Saito S, Nakashima A, Shima T, Ito M. Th1/Th2/Th17 and regulatory T-cell paradigm in pregnancy. Am J Reprod Immunol. 2010;63(6):601–10.

    Article  CAS  PubMed  Google Scholar 

  6. Carlino C, Stabile H, Morrone S, et al. Recruitment of circulating NK cells through decidual tissues: a possible mechanism controlling NK cell accumulation in the uterus during early pregnancy. Blood. 2008;111(6):3108–15.

    Article  CAS  PubMed  Google Scholar 

  7. Saito S, Sasaki Y, Sakai M. CD4+ CD25high regulatory T cells in human pregnancy. J Reprod Immuno. 2005;65(2):111–20.

    Article  CAS  Google Scholar 

  8. Jena MK, Nayak N, Chen K, Nayak NR. Role of macrophages in pregnancy and related complications. Arch Immunol Ther Exp. 2019;67(5):295–309.

    Article  CAS  Google Scholar 

  9. Ali-Hassanzadeh M, Hosseini MS, Ahmadi M, et al. Analysis of the frequency of type 2 innate lymphoid cells and regulatory T cells in abortion-prone mice. Immunol Lett. 2020;220:1–10.

    Article  CAS  PubMed  Google Scholar 

  10. Ahmadi M, Mohammadi M, Ali-Hassanzadeh M, Zare M, Gharesi-Fard B. MDSCs in pregnancy: critical players for a balanced immune system at the feto-maternal interface. Cell Immunol. 2019;346: 103990.

    Article  CAS  PubMed  Google Scholar 

  11. Yang F, Zheng Q, Jin L. Dynamic function and composition changes of immune cells during normal and pathological pregnancy at the maternal-fetal interface. Front immunol. 2019;10:2317.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Han X, Ghaemi MS, Ando K, et al. Differential dynamics of the maternal immune system in healthy pregnancy and preeclampsia. Front immunol. 2019;10:1305.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Young MR, Newby M, Wepsic HT. Hematopoiesis and suppressor bone marrow cells in mice bearing large metastatic Lewis lung carcinoma tumors. Cancer Res. 1987;47(1):100–5.

    CAS  PubMed  Google Scholar 

  14. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9(3):162–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Sanchez-Pino MD, Dean MJ, Ochoa AC. Myeloid-derived suppressor cells (MDSC): when good intentions go awry. Cell Immunol. 2021;362: 104302.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Dumitru CA, Moses K, Trellakis S, Lang S, Brandau S. Neutrophils and granulocytic myeloid-derived suppressor cells: immunophenotyping, cell biology and clinical relevance in human oncology. Cancer Immunol Immunother. 2012;61(8):1155–67.

    Article  CAS  PubMed  Google Scholar 

  17. Mandruzzato S, Brandau S, Britten CM, et al. Toward harmonized phenotyping of human myeloid-derived suppressor cells by flow cytometry: results from an interim study. Cancer Immunol Immunother. 2016;65(2):161–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Rodriguez PC, Hernandez CP, Quiceno D, et al. Arginase I in myeloid suppressor cells is induced by COX-2 in lung carcinoma. J Exp Med. 2005;202(7):931–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Donkor MK, Lahue E, Hoke TA, et al. Mammary tumor heterogeneity in the expansion of myeloid-derived suppressor cells. Int Immunopharmacol. 2009;9(7–8):937–48.

    Article  CAS  PubMed  Google Scholar 

  20. Mao Y, Poschke I, Wennerberg E, et al. Melanoma-educated CD14+ cells acquire a myeloid-derived suppressor cell phenotype through COX-2–dependent mechanisms. Cancer Res. 2013;73(13):3877–87.

    Article  CAS  PubMed  Google Scholar 

  21. Kang X, Zhang X, Liu Z et al. Granulocytic myeloid-derived suppressor cells maintain feto-maternal tolerance by inducing Foxp3 expression in CD4+ CD25− T cells by activation of the TGF-β/β-catenin pathway. Mhr: Mol Hum Reprod 22(7), 499–511 (2016).

  22. Ren J, Zeng W, Tian F, et al. Myeloid-derived suppressor cells depletion may cause pregnancy loss via upregulating the cytotoxicity of decidual natural killer cells. Am J Reprod Immunol. 2019;81(4): e13099.

    Article  PubMed  Google Scholar 

  23. Pan T, Liu Y, Zhong LM, et al. Myeloid-derived suppressor cells are essential for maintaining feto-maternal immunotolerance via STAT3 signaling in mice. J Leukoc Biol. 2016;100(3):499–511.

    Article  CAS  PubMed  Google Scholar 

  24. Foks AC, Van Puijvelde GH, Wolbert J, et al. CD11b+ Gr-1+ myeloid-derived suppressor cells reduce atherosclerotic lesion development in LDLr deficient mice. Cardiovasc Res. 2016;111(3):252–61.

    Article  CAS  PubMed  Google Scholar 

  25. Zhang Y, Bi Y, Yang H, et al. mTOR limits the recruitment of CD11b+ Gr1+ Ly6Chigh myeloid-derived suppressor cells in protecting against murine immunological hepatic injury. J Leukoc Biol. 2014;95(6):961–70.

    Article  PubMed  Google Scholar 

  26. Carson, Danielle. “Attaching and effacing bacterial pathogenesis: characterisation of a severe disease model and the role of effector protein Map.” PhD diss., Imperial College London, 2019.

  27. Gu J, Liu X, Wang Q-X, et al. Angiotensin II increases CTGF expression via MAPKs/TGF-β1/TRAF6 pathway in atrial fibroblasts. Exp Cell Res. 2012;318(16):2105–15.

    Article  CAS  PubMed  Google Scholar 

  28. Karimian P, Kavoosi G, Amirghofran Z. Anti-inflammatory effect of Mentha longifolia in lipopolysaccharide-stimulated macrophages: reduction of nitric oxide production through inhibition of inducible nitric oxide synthase. J Immunotoxicol. 2013;10(4):393–400.

    Article  CAS  PubMed  Google Scholar 

  29. Solito S, Pinton L, De Sanctis F, et al. Methods to measure MDSC immune suppressive activity in vitro and in vivo. Curr Protoc Immunol. 2019;124(1): e61.

    Article  PubMed  Google Scholar 

  30. Muzikova E, Clark DA. Is spontaneous resorption in the DBA/2-mated CBA/J mouse due to a defect in “seed” or in “soil”? Am J Reprod Immunol. 1995;33(1):81–5.

    Article  CAS  PubMed  Google Scholar 

  31. Flores RR, Clauson CL, Cho J, et al. Expansion of myeloid-derived suppressor cells with aging in the bone marrow of mice through a NF-κB-dependent mechanism. Aging Cell. 2017;16(3):480–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Porembka MR, Mitchem JB, Belt BA, et al. Pancreatic adenocarcinoma induces bone marrow mobilization of myeloid-derived suppressor cells which promote primary tumor growth. Cancer Immunol Immunother. 2012;61(9):1373–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kang X, Zhang X, Liu Z, et al. CXCR2-mediated granulocytic myeloid-derived suppressor cells’ functional characterization and their role in maternal fetal interface. DNA Cell Biol. 2016;35(7):358–65.

    Article  CAS  PubMed  Google Scholar 

  34. Wysoczynki M, Khan A, Bolli R. New paradigms in cell therapy: repeated dosing, intravenous delivery, immunomodulatory actions, and new cell types. Circ Res. 2018;123(2):138–58.

    Article  CAS  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors would like to thank the Comparative Medicine Center of Shiraz University of Medical Sciences for providing appropriate conditions for establishing the mouse model.

Funding

This paper was extracted from the PhD thesis done by Moslem Ahmadi and was financially supported by Shiraz University of Medical Sciences (grant number 16763).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Behrouz Gharesi-Fard.

Ethics declarations

Ethical Approval.

This study was performed in line with the principles of the Declaration of Helsinki. Approval was granted by the Ethics Committee of Shiraz University of Medical Sciences (IR.SUMS.REC.1398.404).

Consent to Participate

All authors declare their consent to participate in this work.

Consent for Publication

All authors declare their consent for publication of this paper.

Conflict of Interest

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ahmadi, M., Ali-Hassanzadeh, M., Hosseini, M.S. et al. In vitro–Generated MDSCs Reduce the Pregnancy Complications in an Abortion-Prone Murine Model. Reprod. Sci. 30, 1217–1228 (2023). https://doi.org/10.1007/s43032-022-00995-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s43032-022-00995-y

Keywords

Navigation