Next Article in Journal
Finding a Direct Method for a Dynamic Process: The DD (Direct and Dynamic) Cell-Tox Method
Previous Article in Journal
Recent Advances in Genome Maintenance Processes
Previous Article in Special Issue
Uterine Deletion of Bmal1 Impairs Placental Vascularization and Induces Intrauterine Fetal Death in Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Reproductive Immunology and Pregnancy 2.0

by
Dariusz Szukiewicz
Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
Int. J. Mol. Sci. 2024, 25(10), 5132; https://doi.org/10.3390/ijms25105132
Submission received: 25 April 2024 / Accepted: 6 May 2024 / Published: 9 May 2024
(This article belongs to the Special Issue Reproductive Immunology and Pregnancy 2.0)
This Special Issue comprises original articles in the field of clinical studies whose major topics concern the genetic and immunological aspects of miscarriage and pre-eclampsia, the isolation of decidua macrophages and Hofbauer cells in the placenta for diagnostic purposes, and epigenetic mechanisms that trigger labor. Similar topics will be covered in the third edition of this series of Special Issues (“Reproductive Immunology and Pregnancy 3.0”) [1].
The appropriate level of activity for many genes in the uterine spiral arteries, as well as decidual changes in the endometrium and placenta determine the optimal development of vessels at the site of implantation and fetal growth and development [2]. Genes responsible for proper vascularization at the implantation site are mainly related to the synthesis of representatives of two families of growth factors, i.e., the vascular endothelial growth factor (VEGF) family and the angiopoietin/tyrosine kinase receptor (TEK) family [3]. Low levels of VEGF and ANGPT2 transcripts were reported in cases of pregnancies complicated by fetal growth restriction (FGR). Chronic placental hypoxia, the features of which were histopathologically confirmed, was accompanied by increased placental growth factor (PGF) expression, which may indicate a compensatory mechanism that was ineffective in this patient [4].
An increasing number of studies indicate that human endogenous retrovirus (HERV) genes play a significant role in placental pathology. HERVs constitute up to 8% of human genomic DNA and have been termed “fossil viruses” because they represent footprints of previous retroviral infections [5]. The placenta actively expresses a number of HERV genes [6]. For example, HERV-W env (syncytin-1), a captive retroviral envelope protein, is a membrane glycoprotein that is crucial for placental morphogenesis. Syncytin-1 is involved in fusogenic processes, during which cytotrophoblast cells continuously fuse with the overlying syncytiotrophoblast layers to establish barrier functions and transport activities [7]. In addition, the nonfusogenic effects of syncytin may be responsible for pathological placental morphogenesis based on increased apoptosis and proliferation [8]. Disturbed syncytin-1 expression is associated with increased risk of infertility, pre-eclampsia and FGR, but it has also been observed in patients with tumors such as neuroblastomas, endometrial cancer, and endometriosis [8,9]. Poor placental angiogenesis is a typical histopathological feature of syncytin-1 functional disruption and is often found in patients with pre-eclampsia [10]. Therefore, there are many arguments that syncytin-1 may be a new biological marker and a potential therapeutic target.
Clinically significant abnormal vascularization of placental tissue may be caused by reduced expression/deletion of the basic helix–loop–helix ARNT-like 1 (BMAL1) gene [11]. As the core circadian locomotor output cycle kaput (CLOCK) gene, BMAL1 is rhythmically expressed in many tissues, including uterine and placental tissues, and is responsible for controlling the circadian expression of numerous target genes involved in many physiological processes [11,12]. It has been suggested that reduced endometrial BMAL1 expression is associated with an increased risk of spontaneous recurrent miscarriages in humans [13,14]. Decidual cells deficient in BMAL1 expression showed inhibitory effects on trophoblast invasion into uterine spiral arteries [15]. These findings at the molecular level should prompt a reassessment of the systemic consequences of disruption of the biological clock, especially with regard to fertility [16]. Moreover, the insufficient invasion of trophoblast cells, which is the main factor involved in the development of pre-eclampsia, is caused by the impaired activity of natural killer (NK) cells [14,15]. Therefore, if the cytolytic activity in NK cells required for trophoblast invasion follows a daily rhythm, it may be disrupted as a result of decreased BMAL1 expression [17].
Because NK cells are the most abundant immune cells in the uterus, some researchers suggest that that the immune communication between the fetus and mother is moderated primarily by natural killer (NK) cells rather than T cells [18]. Notably, based on cytokine production, the T helper type 1 (Th1)/Th2 balance in PE gradually shifts to an NK1/NK2 balance [18,19]. The central role of NK cells in PE may be due to the fact that decidual NK (dNK) cells have a cytokine profile that is favored by the presence of HLA-E and HLA-C, and contributes to vascular remodeling and trophoblast invasion into the uterine spiral artery with the subsequent development of the placenta, whereas the cytotoxic phenotypes of peripheral or circulating NK (cNK) cells may be crucial for successful immune escape from fetal to maternal immunity at the maternal–fetal interface [20]. Vascular remodeling in PE is accompanied by an increased number of both dNK and cNK cells, exhibiting increased cytolytic activity, which is measured according to the intracellular production of interferon γ (IFN-γ), perforin, and granzyme B [21]. The increased number of NK cells may also be a manifestation of a compensatory mechanism in the event of the insufficient activation of dNK cells, as is the case in an improper combination of killer cell immunoglobulin-like receptors (KIRs) and human leukocyte antigens (HLA)-C expressed by extravillous trophoblast cells (EVTs) [19,22]. The involvement of NK cells in the pathomechanism of PE is shown in Figure 1.
The results from the latest research on the molecular mechanisms of miscarriage have sparked interest in regulatory T cells (Tregs) as central modulators of the intensity of the immune response in the maternal–fetal system. This specific role of Tregs is to maintain homeostasis during pregnancy by suppressing the activation of autoreactive or alloreactive effector T cells through a diverse repertoire of molecular mechanisms [24,25]. The application of Treg epitopes capable of inducing tolerance seems to be a promising approach for the treatment of immunological causes of pregnancy loss [26,27]. Moreover, the lack of immunogenicity of Treg epitopes in vivo means that they should prove safe for future clinical applications in humans [28,29]. However, the selection/creation of such epitopes is challenging because predicting the action of a given epitope is difficult due to the high degree of MHC polymorphism and disparity in the volume of data on various steps encountered in the generation and presentation of T-cell epitopes in living systems [30].
Among the immune system cells in the human placenta, the dominant type in terms of number and importance are macrophages. The macrophages on the maternal side (decidual) and the macrophages on the fetal side (Hofbauer cells) of the placenta show phenotypic differences associated with changes in activity (polarization). The precise determination of the polarization of placental macrophages in physiological and complicated pregnancies may provide valuable information, as the important role of these immune cells in implantation, placentation and throughout the course of pregnancy is unquestionable [31,32]. The development of a representative isolation method for the direct comparison of maternal and fetal macrophages may shed new light on both the physiology and immunopathology of the placenta, possibly leading to the development of new therapeutic approaches [33].

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Szukiewicz, D. Reproductive Immunology and Pregnancy 3.0. Int. J. Mol. Sci. 2023, 24, 16606. [Google Scholar] [CrossRef]
  2. Shibata, S.; Endo, S.; Nagai, L.A.E.; HKobayashi, E.; Oike, A.; Kobayashi, N.; Kitamura, A.; Hori, T.; Nashimoto, Y.; Nakato, R.; et al. Modeling embryo-endometrial interface recapitulating human embryo implantation. Sci. Adv. 2024, 10, eadi4819. [Google Scholar] [CrossRef] [PubMed]
  3. Umapathy, A.; Chamley, L.W.; James, J.L. Reconciling the distinct roles of angiogenic/anti-angiogenic factors in the placenta and maternal circulation of normal and pathological pregnancies. Angiogenesis 2020, 23, 105–117. [Google Scholar] [CrossRef] [PubMed]
  4. Ravikumar, G.; Mukhopadhyay, A.; Mani, C.; Kocchar, P.; Crasta, J.; Thomas, T.; Dwarkanath, P.; Thomas, A.; Kurpad, A.V.; Sridhar, T.S. Placental expression of angiogenesis-related genes and their receptors in IUGR pregnancies: Correlation with fetoplacental and maternal parameters. J. Matern. Fetal Neonatal. Med. 2020, 33, 3954–3961. [Google Scholar] [CrossRef]
  5. Gröger, V.; Cynis, H. Human Endogenous Retroviruses and Their Putative Role in the Development of Autoimmune Disorders Such as Multiple Sclerosis. Front. Microbiol. 2018, 9, 265. [Google Scholar] [CrossRef] [PubMed]
  6. Bergallo, M.; Marozio, L.; Botta, G.; Tancredi, A.; Daprà, V.; Galliano, I.; Montanari, P.; Coscia, A.; Benedetto, C.; Tovo, P.A. Human Endogenous Retroviruses Are Preferentially Expressed in Mononuclear Cells From Cord Blood Than From Maternal Blood and in the Fetal Part of Placenta. Front. Pediatr. 2020, 8, 244. [Google Scholar] [CrossRef]
  7. Huang, Q.; Chen, H.; Li, J.; Oliver, M.; Ma, X.; Byck, D.; Gao, Y.; Jiang, S.W. Epigenetic and non-epigenetic regulation of syncytin-1 expression in human placenta and cancer tissues. Cell Signal. 2014, 26, 648–656. [Google Scholar] [CrossRef] [PubMed]
  8. Wang, Y.N.; Ye, Y.; Zhou, D.; Guo, Z.W.; Xiong, Z.; Gong, X.X.; Jiang, S.W.; Chen, H. The Role of Syncytin in Placental Angiogenesis and Fetal Growth. Front. Cell Dev. Biol. 2022, 10, 852561. [Google Scholar] [CrossRef] [PubMed]
  9. Wang, Q.; Shi, Y.; Bian, Q.; Zhang, N.; Wang, M.; Wang, J.; Li, X.; Lai, L.; Zhao, Z.; Yu, H. Molecular mechanisms of syncytin-1 in tumors and placental development related diseases. Discov. Oncol. 2023, 14, 104. [Google Scholar] [CrossRef]
  10. Qiao, S.; Wang, F.; Chen, H.; Jiang, S.W. Inducible knockout of Syncytin-A gene leads to an extensive placental vasculature deficiency, implications for preeclampsia. Clin. Chim. Acta 2017, 474, 137–146. [Google Scholar] [CrossRef]
  11. Waddell, B.J.; Wharfe, M.D.; Crew, R.C.; Mark, P.J. A rhythmic placenta? Circadian variation, clock genes and placental function. Placenta 2012, 33, 533–539. [Google Scholar] [CrossRef]
  12. Yang, G.; Chen, L.; Grant, G.R.; Paschos, G.; Song, W.L.; Musiek, E.S.; Lee, V.; McLoughlin, S.C.; Grosser, T.; Cotsarelis, G.; et al. Timing of expression of the core clock gene Bmal1 influences its effects on aging and survival. Sci. Transl. Med. 2016, 8, 324ra16. [Google Scholar] [CrossRef]
  13. Lv, S.; Wang, N.; Ma, J.; Li, W.P.; Chen, Z.J.; Zhang, C. Impaired decidualization caused by downregulation of circadian clock gene BMAL1 contributes to human recurrent miscarriage. Biol. Reprod. 2019, 101, 138–147. [Google Scholar] [CrossRef] [PubMed]
  14. Ono, M.; Toyoda, N.; Kagami, K.; Hosono, T.; Matsumoto, T.; Horike, S.I.; Yamazaki, R.; Nakamura, M.; Mizumoto, Y.; Fujiwara, T.; et al. Uterine Deletion of Bmal1 Impairs Placental Vascularization and Induces Intrauterine Fetal Death in Mice. Int. J. Mol. Sci. 2022, 23, 7637. [Google Scholar] [CrossRef] [PubMed]
  15. Diallo, A.B.; Coiffard, B.; Desbriere, R.; Katsogiannou, M.; Donato, X.; Bretelle, F.; Mezouar, S.; Mege, J.L. Disruption of the Expression of the Placental Clock and Melatonin Genes in Preeclampsia. Int. J. Mol. Sci. 2023, 24, 2363. [Google Scholar] [CrossRef]
  16. Sciarra, F.; Franceschini, E.; Campolo, F.; Gianfrilli, D.; Pallotti, F.; Paoli, D.; Isidori, A.M.; Venneri, M.A. Disruption of Circadian Rhythms: A Crucial Factor in the Etiology of Infertility. Int. J. Mol. Sci. 2020, 21, 3943. [Google Scholar] [CrossRef]
  17. Arjona, A.; Sarkar, D.K. Evidence supporting a circadian control of natural killer cell function. Brain Behav. Immun. 2006, 20, 469–476. [Google Scholar] [CrossRef]
  18. Wei, X.; Yang, X. The central role of natural killer cells in preeclampsia. Front. Immunol. 2023, 14, 1009867. [Google Scholar] [CrossRef]
  19. Aneman, I.; Pienaar, D.; Suvakov, S.; Simic, T.P.; Garovic, V.D.; McClements, L. Mechanisms of Key Innate Immune Cells in Early- and Late-Onset Preeclampsia. Front. Immunol. 2020, 11, 1864. [Google Scholar] [CrossRef]
  20. Boulanger, H.; Bounan, S.; Mahdhi, A.; Drouin, D.; Ahriz-Saksi, S.; Guimiot, F.; Rouas-Freiss, N. Immunologic aspects of preeclampsia. AJOG Glob. Rep. 2024, 4, 100321. [Google Scholar] [CrossRef]
  21. Du, M.; Wang, W.; Huang, L.; Guan, X.; Lin, W.; Yao, J.; Li, L. Natural killer cells in the pathogenesis of preeclampsia: A double-edged sword. J. Matern. Fetal Neonatal. Med. 2022, 35, 1028–1035. [Google Scholar] [CrossRef] [PubMed]
  22. Yang, X.; Meng, T. Killer-cell immunoglobulin-like receptor/human leukocyte antigen-C combination and ‘great obstetrical syndromes’ (Review). Exp. Ther. Med. 2021, 22, 1178. [Google Scholar] [CrossRef] [PubMed]
  23. Blais, M.E.; Dong, T.; Rowland-Jones, S. HLA-C as a mediator of natural killer and T-cell activation: Spectator or key player? Immunology 2011, 133, 1–7. [Google Scholar] [CrossRef]
  24. Saito, S. Reconsideration of the Role of Regulatory T Cells during Pregnancy: Differential Characteristics of Regulatory T Cells between the Maternal-Fetal Interface and Peripheral Sites and between Early and Late Pregnancy. Med. Princ. Pract. 2022, 31, 403–414. [Google Scholar] [CrossRef]
  25. Ou, Q.; Power, R.; Griffin, M.D. Revisiting regulatory T cells as modulators of innate immune response and inflammatory diseases. Front. Immunol. 2023, 14, 1287465. [Google Scholar] [CrossRef]
  26. Yan, Y.N.; Zhang, J.; Yang, N.; Chen, C.; Li, W. T Cell Subsets and the Expression of Related MicroRNAs in Patients with Recurrent Early Pregnancy Loss. Mediat. Inflamm. 2023, 2023, 8215567. [Google Scholar] [CrossRef] [PubMed]
  27. Sizova, O.; John, L.S.; Ma, Q.; Molldrem, J.J. Multi-faceted role of LRP1 in the immune system. Front. Immunol. 2023, 14, 1166189. [Google Scholar] [CrossRef] [PubMed]
  28. Su, Y.; Rossi, R.; De Groot, A.S.; Scott, D.W. Regulatory T cell epitopes (Tregitopes) in IgG induce tolerance in vivo and lack immunogenicity per se. J. Leukoc. Biol. 2013, 94, 377–383. [Google Scholar] [CrossRef]
  29. Cousens, L.; Najafian, N.; Martin, W.D.; De Groot, A.S. Tregitope: Immunomodulation powerhouse. Hum. Immunol. 2014, 75, 1139–1146. [Google Scholar] [CrossRef]
  30. Konstantinou, G.N. T-Cell Epitope Prediction. Methods Mol. Biol. 2017, 1592, 211–222. [Google Scholar] [CrossRef]
  31. Ning, J.; Zhang, M.; Cui, D.; Yang, H. The pathologic changes of human placental macrophages in women with hyperglycemia in pregnancy. Placenta 2022, 130, 60–66. [Google Scholar] [CrossRef] [PubMed]
  32. Mezouar, S.; Katsogiannou, M.; Ben Amara, A.; Bretelle, F.; Mege, J.L. Placental macrophages: Origin, heterogeneity, function and role in pregnancy-associated infections. Placenta 2021, 103, 94–103. [Google Scholar] [CrossRef] [PubMed]
  33. Lasch, M.; Sudan, K.; Paul, C.; Schulz, C.; Kolben, T.; Dorp, J.V.; Eren, S.; Beyer, S.; Siniscalchi, L.; Mahner, S.; et al. Isolation of Decidual Macrophages and Hofbauer Cells from Term Placenta-Comparison of the Expression of CD163 and CD80. Int. J. Mol. Sci. 2022, 23, 6113. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The concept of the pathomechanism of pre-eclampsia (PE) based on the central role of natural killer (NK) cells. (A). NORMAL PREGNANCY. A sufficiently deep invasion of extravascular trophoblast (EVT) cells into the uterine spiral arteries determines proper implantation, creating low-resistance circulation with optimal blood flow for the progression of pregnancy growth. Bold is used intentionally for better readability and clear differentiation between parts A and B of Figure 1. (B). PRE-ECLAMPSIA. A significant immune system disorder in PE patients is a change in the number and activity of NK cells. This phenomenon applies to both decidual NK (dNK) cells and peripheral (circulating; cNK) cells. The increase (↑) in the number of NK cells may be a manifestation of a compensatory mechanism in the event of the insufficient activation of NK cells, which may be caused by a change in the proportion of killer cell immunoglobulin-like receptor (KIR) expression on NK cells relative to human leukocyte antigen c (HLA-C) in defective extravillous trophoblast (EVT) cells [23]. The change in NK cell activity may result from an imbalance between NK type-1 (NK1) and NK2 cells, as pregnant women with PE have a significantly greater NK1/NK2 cell ratio than healthy pregnant women. This leads to the dominance of NK1, which is manifested by a change in the cytokine profile toward increased cytotoxic and cytolytic activity. The presence of defective EVT cells causes abnormal placentation with insufficient vessel remodeling, preventing the creation of low-resistance circulation between the spiral uterine arteries and the intervillous placental plate vessels.
Figure 1. The concept of the pathomechanism of pre-eclampsia (PE) based on the central role of natural killer (NK) cells. (A). NORMAL PREGNANCY. A sufficiently deep invasion of extravascular trophoblast (EVT) cells into the uterine spiral arteries determines proper implantation, creating low-resistance circulation with optimal blood flow for the progression of pregnancy growth. Bold is used intentionally for better readability and clear differentiation between parts A and B of Figure 1. (B). PRE-ECLAMPSIA. A significant immune system disorder in PE patients is a change in the number and activity of NK cells. This phenomenon applies to both decidual NK (dNK) cells and peripheral (circulating; cNK) cells. The increase (↑) in the number of NK cells may be a manifestation of a compensatory mechanism in the event of the insufficient activation of NK cells, which may be caused by a change in the proportion of killer cell immunoglobulin-like receptor (KIR) expression on NK cells relative to human leukocyte antigen c (HLA-C) in defective extravillous trophoblast (EVT) cells [23]. The change in NK cell activity may result from an imbalance between NK type-1 (NK1) and NK2 cells, as pregnant women with PE have a significantly greater NK1/NK2 cell ratio than healthy pregnant women. This leads to the dominance of NK1, which is manifested by a change in the cytokine profile toward increased cytotoxic and cytolytic activity. The presence of defective EVT cells causes abnormal placentation with insufficient vessel remodeling, preventing the creation of low-resistance circulation between the spiral uterine arteries and the intervillous placental plate vessels.
Ijms 25 05132 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Szukiewicz, D. Reproductive Immunology and Pregnancy 2.0. Int. J. Mol. Sci. 2024, 25, 5132. https://doi.org/10.3390/ijms25105132

AMA Style

Szukiewicz D. Reproductive Immunology and Pregnancy 2.0. International Journal of Molecular Sciences. 2024; 25(10):5132. https://doi.org/10.3390/ijms25105132

Chicago/Turabian Style

Szukiewicz, Dariusz. 2024. "Reproductive Immunology and Pregnancy 2.0" International Journal of Molecular Sciences 25, no. 10: 5132. https://doi.org/10.3390/ijms25105132

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop