1
|
Dos Santos F, Steer PJ, Johnson MR. Maternal cytokine and cellular adhesion molecules profile in pregnant women with and without congenital heart disease. Cytokine 2025; 188:156886. [PMID: 39933224 DOI: 10.1016/j.cyto.2025.156886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 03/31/2024] [Accepted: 02/05/2025] [Indexed: 02/13/2025]
Abstract
INTRODUCTION Despite corrective surgery, patients with congenital heart disease (CHD) have residual regions of disturbed oscillatory blood flow which can induce upregulation of proinflammatory cytokines and adhesion molecules. Data on cytokine and cellular adhesion molecule (CAM) profiles in low risk pregnancy and pregnancy complicated by CHD are limited. The objective of this work was to study the profile of IL-6, IL-10, TNFα, ghrelin, GROα, and ICAM/VCAM in pregnancy in women with and without CHD and test the hypothesis that the circulating levels of these are correlated with obstetric outcomes. METHODS Prospective study of women ≥18 years carrying a singleton low risk pregnancy low-risk (LR-group) and pregnant women with CHD (CHD-group). Study visits were conducted between 10 and 14, 18-22 and 30-34 weeks' gestation with blood sampling for immune profiling using the Bio-Plex® Multiplex Immunoassay. The immune profile was investigated in both groups and correlated with obstetric outcomes. This study was approved by the Health Research Authority and the London South East Research Ethics Committee (REC reference: 17/LO/0970). RESULTS Forty-five samples in 30 participants with CHD and 45 gestational age-matched samples from 34 low-risk pregnant women were analysed. Women in the CHD-group delivered earlier (38 + 6 weeks vs 39 + 4 weeks, p = 0.005) and had smaller babies (2940 g, 30.0 centile vs 3415 g, 63.5 centile, p < 0.001). There were no significant differences in the levels of IL-6, IL-10 or TNFαin both groups across trimesters. Levels of GROα increased in both groups but less so in the CHD group. Levels of ghrelin decreased in both groups but less so in the CHD group. Levels of ICAM decreased as pregnancy progressed in the CHD group. Levels of VCAM increased in both groups but more significantly in the CHD-group (second trimester, p < 0.001; third trimester, p = 0.045). Women in the CHD-group had higher levels if IL-6 (p = 0.005) and ICAM (p < 0.001) lower levels of IL-10 in the third trimester (p = 0.018). There was a significant positive association between levels of IL-6 in the first trimester and birthweight centiles (rs = 1.00, p < 0.001) in the CHD group. CONCLUSION There was minimal fluctuation in the levels of the studies cytokines during pregnancy with exception of GROα and ghrelin, both implicated in fetal growth. Women with CHD had higher levels of proinflammatory cytokines and ICAM in the first trimester, and lower levels of IL-10 in the third trimester, suggesting a more proinflammatory state. High levels of VCAM in the CHD group could be secondary to endothelial activation.
Collapse
Affiliation(s)
- Francois Dos Santos
- Imperial College London, Chelsea and Westminster Hospital, 3rd floor, Academic O&G, 369 Fulham Road, SW10 9NH, London, United Kingdom.
| | - Philip J Steer
- Imperial College London, Chelsea and Westminster Hospital, 3rd floor, Academic O&G, 369 Fulham Road, SW10 9NH, London, United Kingdom.
| | - Mark R Johnson
- Imperial College London, Chelsea and Westminster Hospital, 3rd floor, Academic O&G, 369 Fulham Road, SW10 9NH, London, United Kingdom.
| |
Collapse
|
2
|
Xie Z, Chen Z, Chai Y, Yao W, Ma G. Unveiling the placental bacterial microbiota: implications for maternal and infant health. Front Physiol 2025; 16:1544216. [PMID: 40161970 PMCID: PMC11949977 DOI: 10.3389/fphys.2025.1544216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
The human placenta is a unique organ that forms under specific physiological conditions and plays a crucial role in nutrient and metabolite exchange between the mother and fetus. Research on the placenta is important for understanding maternal-fetal diseases. Traditionally, the placenta was considered "sterile," but advancements in detection techniques have revealed the presence of a low level of microorganisms. This discovery challenges the traditional notion that the uterine placenta is sterile. The revelation of this truth marks a significant breakthrough in medical research, prompting more researchers to focus on this vital organ, the placenta. Placental microbial communities may originate from the oral, vaginal, and intestinal microbiota of expectant mothers. These microorganisms may reach the maternal-fetal interface, collectively shaping the placental microbiota and contributing to the composition of normal placental microbial communities. Abnormal placental microbial communities may be associated with some pregnancy complications and fetal developmental issues such as preterm birth, gestational hypertension, fetal growth restriction, and gestational diabetes mellitus. Intervention strategies targeting microbial communities, which include modulation of placental microbiota composition or function, such as probiotics, may help prevent or treat complications related to abnormal placental microbiota during pregnancy.
Collapse
Affiliation(s)
- Zhuojun Xie
- General Medicine Department, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - Zhongsheng Chen
- Department of Colorectal Cancer Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Chai
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Wang Yao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Guangyu Ma
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
3
|
Yoshino O, Ono Y. The physiological role of macrophages in reproductive organs. Reprod Med Biol 2025; 24:e12637. [PMID: 39959577 PMCID: PMC11827100 DOI: 10.1002/rmb2.12637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 02/04/2025] [Indexed: 02/18/2025] Open
Abstract
Background Macrophages are essential immune cells critical to reproductive physiology. They regulate key processes such as follicular development, ovulation, and luteinization in the ovaries. Macrophages are also involved in endometrial remodeling, immune tolerance, and placentation in the uterus. Methods This review examined the biological characteristics of macrophages and their role in ovarian, uterine, and fallopian tube physiology. It focused on findings from both animal and human studies to provide a comprehensive understanding of macrophage functions. Main Findings In the ovaries, M1 macrophages play a role in folliculogenesis and ovulation through the inflammatory and angiogenic pathways. Macrophages also maintain the corpus luteum and vascular integrity. In the uterus, macrophages regulate tissue repair and remodeling during the menstrual cycle and play a critical role in implantation by maintaining immune tolerance and supporting decidualization. Dysregulation of the M1/M2 balance can cause implantation failure. In the fallopian tubes, macrophages mediate tissue repair and immune responses. Macrophage polarization dynamically adapts to physiological and pathological conditions in all reproductive organs highlighting the functional plasticity of these cells. Conclusion Macrophage polarization and functions are pivotal in maintaining reproductive health. Hence, understanding the role of macrophages in various reproductive organs provides a foundation for developing new therapies.
Collapse
Affiliation(s)
- Osamu Yoshino
- Department of Obstetrics and GynecologyUniversity of YamanashiYamanashiJapan
| | - Yosuke Ono
- Department of Obstetrics and GynecologyUniversity of YamanashiYamanashiJapan
| |
Collapse
|
4
|
Einenkel R, Ehrhardt J, Zygmunt M, Muzzio DO. Less is more! Low amount of Fusobacterium nucleatum supports macrophage-mediated trophoblast functions in vitro. Front Immunol 2024; 15:1447190. [PMID: 39176096 PMCID: PMC11338817 DOI: 10.3389/fimmu.2024.1447190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024] Open
Abstract
F. nucleatum, involved in carcinogenesis of colon carcinomas, has been described as part of the commensal flora of the female upper reproductive tract. Although its contribution to destructive inflammatory processes is well described, its role as commensal uterine bacteria has not been thoroughly investigated. Since carcinogenesis shares similar mechanisms with early pregnancy development (including proliferation, invasion, blood supply and the induction of tolerance), these mechanisms induced by F. nucleatum could play a role in early pregnancy. Additionally, implantation and placentation require a well-balanced immune activation, which might be suitably managed by the presence of a limited amount of bacteria or bacterial residues. We assessed the effect of inactivated F. nucleatum on macrophage-trophoblast interactions. Monocytic cells (THP-1) were polarized into M1, M2a or M2c macrophages by IFN-γ, IL-4 or TGF-β, respectively, and subsequently treated with inactivated fusobacteria (bacteria:macrophage ratio of 0.1 and 1). Direct effects on macrophages were assessed by viability assay, flow cytometry (antigen presentation molecules and cytokines), qPCR (cytokine expression), in-cell Western (HIF and P-NF-κB) and ELISA (VEGF secretion). The function of first trimester extravillous trophoblast cells (HTR-8/SVneo) in response to macrophage-conditioned medium was microscopically assessed by migration (scratch assay), invasion (sprouting assay) and tube formation. Underlying molecular changes were investigated by ELISA (VEGF secretion) and qPCR (matrix-degrading factors and regulators). Inflammation-primed macrophages (M1) as well as high bacterial amounts increased pro-inflammatory NF-κB expression and inflammatory responses. Subsequently, trophoblast functions were impaired. In contrast, low bacterial stimulation caused an increased HIF activation and subsequent VEGF-A secretion in M2c macrophages. Accordingly, there was an increase of trophoblast tube formation. Our results suggest that a low-mass endometrial/decidual microbiome can be tolerated and while it supports implantation and further pregnancy processes.
Collapse
Affiliation(s)
| | | | | | - Damián Oscar Muzzio
- Department of Gynecology and Obstetrics, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
5
|
Bezemer RE, Faas MM, van Goor H, Gordijn SJ, Prins JR. Decidual macrophages and Hofbauer cells in fetal growth restriction. Front Immunol 2024; 15:1379537. [PMID: 39007150 PMCID: PMC11239338 DOI: 10.3389/fimmu.2024.1379537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/14/2024] [Indexed: 07/16/2024] Open
Abstract
Placental macrophages, which include maternal decidual macrophages and fetal Hofbauer cells, display a high degree of phenotypical and functional plasticity. This provides these macrophages with a key role in immunologically driven events in pregnancy like host defense, establishing and maintaining maternal-fetal tolerance. Moreover, placental macrophages have an important role in placental development, including implantation of the conceptus and remodeling of the intrauterine vasculature. To facilitate these processes, it is crucial that placental macrophages adapt accordingly to the needs of each phase of pregnancy. Dysregulated functionalities of placental macrophages are related to placental malfunctioning and have been associated with several adverse pregnancy outcomes. Although fetal growth restriction is specifically associated with placental insufficiency, knowledge on the role of macrophages in fetal growth restriction remains limited. This review provides an overview of the distinct functionalities of decidual macrophages and Hofbauer cells in each trimester of a healthy pregnancy and aims to elucidate the mechanisms by which placental macrophages could be involved in the pathogenesis of fetal growth restriction. Additionally, potential immune targeted therapies for fetal growth restriction are discussed.
Collapse
Affiliation(s)
- Romy Elisa Bezemer
- Department of Obstetrics and Gynecology, University Medical Center Groningen, Groningen, Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, Netherlands
| | - Marijke M Faas
- Department of Obstetrics and Gynecology, University Medical Center Groningen, Groningen, Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, Netherlands
| | - Sanne Jehanne Gordijn
- Department of Obstetrics and Gynecology, University Medical Center Groningen, Groningen, Netherlands
| | - Jelmer R Prins
- Department of Obstetrics and Gynecology, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
6
|
Yang L, Xu HR, Zhang X, Shi Y, Shi JX, Chen QQ, Shen XR, He YP, Tang JN, Gu WW, Wang J. Increased miR-3074-5p expression promotes M1 polarization and pyroptosis of macrophages via ERα/NLRP3 pathway and induces adverse pregnancy outcomes in mice. Cell Death Discov 2024; 10:171. [PMID: 38600077 PMCID: PMC11006911 DOI: 10.1038/s41420-024-01941-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/30/2024] [Accepted: 04/03/2024] [Indexed: 04/12/2024] Open
Abstract
Decidual macrophages (dMϕs) play critical roles in regulation of immune-microhomeostasis at maternal-fetal interface during pregnancy, but the underlying molecular mechanisms are still unclear. In this study, it was found that litter size and fetal weight were significantly reduced, whereas the rate of embryo resorption was increased in miR-3074-5p knock-in (3074-KI) pregnant mice, compared to that of wild-type (WT) pregnant mice. Plasma levels of pro-inflammatory cytokines in 3074-KI pregnant mice were also significantly elevated compared to WT pregnant mice at GD7.5. The quantity of M1-Mϕs in uterine tissues of 3074-KI pregnant mice was significantly increased compared to WT pregnant mice at GD13.5. Estrogen receptor-α (ERα) was validated to be a target of miR-3074-5p. Either miR-3074-5p overexpression or ERα knockdown promoted transcriptional activity of NF-κB/p65, induced M1-polarization and pyroptosis of THP1-derived Mϕs, accompanied with increased intracellular levels of cleaved Caspase-1, cleaved IL-1β, NLRP3, cleaved GSDMD and ASC aggregation. Furthermore, ERα could not only bind to NLRP3 or ASC directly, but also inhibit the interaction between NLRP3 and ASC. The endometrial miR-3074-5p expression level at the middle secretory stage of repeated implantation failure (RIF) patients was significantly decreased compared to that of control fertile women. These data indicated that miR-3074-5p could promote M1 polarization and pyroptosis of Mϕs via activation of NLRP3 inflammasome by targeting ERα, and the dysregulation of miR-3074-5p expression in dMϕs might damage the embryo implantation and placentation by interfering with inflammatory microenvironment at the maternal-fetal interface during early pregnancy.
Collapse
Affiliation(s)
- Long Yang
- NHC Key Laboratory of Reproduction Regulation, Shanghai Key Lab of Health and Diease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai, 200237, China
| | - Hao-Ran Xu
- NHC Key Laboratory of Reproduction Regulation, Shanghai Key Lab of Health and Diease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai, 200237, China
| | - Xuan Zhang
- NHC Key Laboratory of Reproduction Regulation, Shanghai Key Lab of Health and Diease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai, 200237, China
| | - Yan Shi
- NHC Key Laboratory of Reproduction Regulation, Shanghai Key Lab of Health and Diease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai, 200237, China
| | - Jia-Xin Shi
- NHC Key Laboratory of Reproduction Regulation, Shanghai Key Lab of Health and Diease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai, 200237, China
| | - Qian-Qian Chen
- Reproductive Medicine Center, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xiao-Rong Shen
- NHC Key Laboratory of Reproduction Regulation, Shanghai Key Lab of Health and Diease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai, 200237, China
| | - Ya-Ping He
- NHC Key Laboratory of Reproduction Regulation, Shanghai Key Lab of Health and Diease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai, 200237, China
| | - Jia-Nan Tang
- NHC Key Laboratory of Reproduction Regulation, Shanghai Key Lab of Health and Diease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai, 200237, China
| | - Wen-Wen Gu
- NHC Key Laboratory of Reproduction Regulation, Shanghai Key Lab of Health and Diease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai, 200237, China.
| | - Jian Wang
- NHC Key Laboratory of Reproduction Regulation, Shanghai Key Lab of Health and Diease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai, 200237, China.
| |
Collapse
|
7
|
Strbo N, Rodriguez S, Padula L, Fisher E, Lyons A, Rodriguez C, Rivas K, Ibrahim M, Paidas M, Attia G. Assessment of immune cells in the uterine fluid at the time of the embryo transfer. Am J Reprod Immunol 2024; 91:e13842. [PMID: 38650366 DOI: 10.1111/aji.13842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/22/2024] [Accepted: 04/28/2024] [Indexed: 04/25/2024] Open
Abstract
PROBLEM Although endometrial receptivity is a key factor in influencing implantation in both naturally conceived and assisted reproductive technology (ART) cycles, very little is known about the endometrium milieu around the time of implantation. Previous studies have demonstrated the presence of several cytokines in the endometrium that affect implantation. However, there is lacking data about the presence of immune cell subtypes within the endometrium and in the uterine cavity at the time of implantation. METHOD OF STUDY This study was approved by the Institutional Review Board (# 225589). The study was designed as a prospective observational cohort study between May 2021 and December 2022 at a single academic-based fertility center. All patients underwent at least one In Vitro Fertilization (IVF) cycle and have frozen embryos. Twenty-four participants were recruited for this study which was conducted during the frozen embryo transfer (FET) cycle regardless of the outcome of previous cycles. Two samples were acquired from each subject, denoted as lower and upper. A trial transfer catheter was introduced under ultrasound guidance into the lower uterine segment. Upon removal, the tip was rinsed in IMDM medium containing 10% FBS (lower uterus). A transfer catheter was then loaded with the embryo that was placed in the upper uterus under ultrasound guidance. The tip of the transfer catheter was rinsed in separate aliquot of the above media (upper uterus). After centrifugation, pelleted cells were stained for the following surface markers: CD45, CD3, CD19, CD4, CD8, gamma delta TCR, CD25, CD127, CD66b, CD14, CD16, CD56 and acquired on Sony SP6800 Spectral Analyzer. RESULTS Upon staining the pelleted cells, we were able to identify viable leukocytes from samples obtained from both, upper and lower uterus (0.125 × 106 cells ± SD 0.32), (0.123 × 106 cells ± SD 0.12), respectively. Among total viable cells, there was no significant difference in both percent and number of CD45+ cells between the upper and lower uterus (9.88% ± 6.98 SD, 13.67% ± 9.79 SD, p = .198) respectively. However, there was significantly higher expression of CD3+ (p = .006), CD19+ (p = .032) and CD14+ (p = .019) cells in samples collected from upper compared to lower uterus. Within all CD3+ cells, we found that gamma delta T cells (GDT) were the major population of T cells in both upper and lower uterus. In contrast, CD8+ T cells were significantly higher in the lower uterus when compared to the upper uterus (p = .009). There was no statistically significant difference in the expression of CD4+ T cells, T regulatory cells (CD4+CD25+CD127-), NK cells (CD56+), neutrophils (CD66b+) and FcγRIII+ cells (CD16+) between upper and lower uterus. CONCLUSIONS We believe the immune milieu at the time of embryo transfer will affect implantation. Understanding the composition of immune cells will guide further research in identifying optimal immune milieus that favor implantation. Comprehensive analysis of endometrium is expected to lead to new diagnostic and therapeutic approaches to improve IVF outcomes.
Collapse
Affiliation(s)
- Natasa Strbo
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Suset Rodriguez
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology, and Infertility, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Laura Padula
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Eva Fisher
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Annabel Lyons
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Carolina Rodriguez
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Katelyn Rivas
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Mohammed Ibrahim
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology, and Infertility, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Michael Paidas
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology, and Infertility, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - George Attia
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Endocrinology, and Infertility, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
8
|
Merech F, Gori S, Calo G, Hauk V, Paparini D, Rios D, Lara B, Doga L, D'Eramo L, Squassi A, Ramhorst R, Argüello RJ, Pérez Leirós C, Vota D. Monocyte immunometabolic reprogramming in human pregnancy: contribution of trophoblast cells. Am J Physiol Endocrinol Metab 2024; 326:E215-E225. [PMID: 38117266 DOI: 10.1152/ajpendo.00357.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/04/2023] [Accepted: 12/18/2023] [Indexed: 12/21/2023]
Abstract
Immunometabolism research is uncovering the relationship between metabolic features and immune cell functions in physiological and pathological conditions. Normal pregnancy entails a fine immune and metabolic regulation of the maternal-fetal interaction to assist the energetic demands of the fetus with immune homeostasis maintenance. Here, we determined the immunometabolic status of monocytes of pregnant women compared with nonpregnant controls and its impact on monocyte anti-inflammatory functions such as efferocytosis. Monocytes from pregnant women (16-20 wk) and nonpregnant age-matched controls were studied. Single cell-based metabolic assays using freshly isolated monocytes from both groups were carried out in parallel with functional assays ex vivo to evaluate monocyte efferocytic capacity. On the other hand, various in vitro metabolic assays with human monocytes or monocyte-derived macrophages were designed to explore the effect of trophoblast cells in the profiles observed. We found that pregnancy alters monocyte metabolism and function. An increased glucose dependency and enhanced efferocytosis were detected in monocytes from pregnant women at resting states, compared with nonpregnant controls. Furthermore, monocytes display a reduced glycolytic response when stimulated with lipopolysaccharide (LPS). The metabolic profiling of monocytes at this stage of pregnancy was comparable with the immunometabolic phenotypes of human monocytes treated in vitro with human first trimester trophoblast cell conditioned media. These findings suggest that immunometabolic mechanisms are involved in the functional shaping of monocytes during pregnancy with a contribution of trophoblast cells. Results provide new clues for future hypotheses regarding pregnancies complicated by metabolic disorders.NEW & NOTEWORTHY Immunometabolism stands as a novel perspective to understand the complex regulation of the immune response and to provide small molecule-based therapies. By applying this approach to study monocytes during pregnancy, we found that these cells have a unique activation pattern. They rely more on glycolysis and show increased efferocytosis/IL-10 production, but they do not have the typical proinflammatory responses. We also present evidence that trophoblast cells can shape monocytes into this distinct immunometabolic profile.
Collapse
Affiliation(s)
- Fátima Merech
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Soledad Gori
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Guillermina Calo
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Vanesa Hauk
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Daniel Paparini
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Daiana Rios
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Brenda Lara
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Luciana Doga
- Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Luciana D'Eramo
- Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Aldo Squassi
- Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Rosanna Ramhorst
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Rafael J Argüello
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille University, CNRS, INSERM, Marseille, France
| | - Claudia Pérez Leirós
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Daiana Vota
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires (UBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
9
|
Wu Y, Teh YC, Chong SZ. Going Full TeRM: The Seminal Role of Tissue-Resident Macrophages in Organ Remodeling during Pregnancy and Lactation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:513-521. [PMID: 38315948 DOI: 10.4049/jimmunol.2300560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/23/2023] [Indexed: 02/07/2024]
Abstract
During pregnancy and lactation, the uterus and mammary glands undergo remarkable structural changes to perform their critical reproductive functions before reverting to their original dormant state upon childbirth and weaning, respectively. Underlying this incredible plasticity are complex remodeling processes that rely on coordinated decisions at both the cellular and tissue-subunit levels. With their exceptional versatility, tissue-resident macrophages play a variety of supporting roles in these organs during each stage of development, ranging from maintaining immune homeostasis to facilitating tissue remodeling, although much remains to be discovered about the identity and regulation of individual macrophage subsets. In this study, we review the increasingly appreciated contributions of these immune cells to the reproductive process and speculate on future lines of inquiry. Deepening our understanding of their interactions with the parenchymal or stromal populations in their respective niches may reveal new strategies to ameliorate complications in pregnancy and breastfeeding, thereby improving maternal health and well-being.
Collapse
Affiliation(s)
- Yixuan Wu
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Ye Chean Teh
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Shu Zhen Chong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| |
Collapse
|
10
|
Paul M, Ain R. Evaluation of Molecular Interactions and Cellular Dynamics at the Maternal-Fetal Interface During Placental Morphogenesis. Methods Mol Biol 2024; 2728:45-76. [PMID: 38019391 DOI: 10.1007/978-1-0716-3495-0_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Molecular events at the maternal-fetal interface establish successful pregnancies. Identifying and characterizing the heterogeneous cell population and their cross-talk at the cellular and molecular levels are essential to expand our knowledge on the progression and maintenance of pregnancy. In this chapter, we briefly discuss the organization of maternal-fetal interface in mice/rats and humans. We illustrate methods for studying the cell composition using flow cytometry, immunocytochemical and biochemical studies, intercellular interaction using co-culture system and spheroid assay, and function of trophoblast cells using ELISA, RNA sequencing, mass spectrometry (MS) to analyze the proteome, invasion assay, and scratch wound assay.
Collapse
Affiliation(s)
- Madhurima Paul
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Rupasri Ain
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India.
| |
Collapse
|
11
|
Kouthouridis S, Sotra A, Khan Z, Alvarado J, Raha S, Zhang B. Modeling the Progression of Placental Transport from Early- to Late-Stage Pregnancy by Tuning Trophoblast Differentiation and Vascularization. Adv Healthc Mater 2023; 12:e2301428. [PMID: 37830445 PMCID: PMC11468690 DOI: 10.1002/adhm.202301428] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/18/2023] [Indexed: 10/14/2023]
Abstract
The early-stage placental barrier is characterized by a lack of fetal circulation and by a thick trophoblastic barrier, whereas the later-stage placenta consists of vascularized chorionic villi encased in a thin, differentiated trophoblast layer, ideal for nutrient transport. In this work, predictive models of early- and late-stage placental transport are created using blastocyst-derived placental stem cells (PSCs) by modulating PSC differentiation and model vascularization. PSC differentiation results in a thinner, fused trophoblast layer, as well as an increase in human chorionic gonadotropin secretion, barrier permeability, and secretion of certain inflammatory cytokines, which are consistent with in vivo findings. Further, gene expression confirms this shift toward a differentiated trophoblast subtype. Vascularization results in a molecule type- and size-dependent change in dextran and insulin permeability. These results demonstrate that trophoblast differentiation and vascularization have critical effects on placental barrier permeability and that this model can be used as a predictive measure to assess fetal toxicity of xenobiotic substances at different stages of pregnancy.
Collapse
Affiliation(s)
- Sonya Kouthouridis
- Department of Chemical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Alexander Sotra
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Zaim Khan
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Justin Alvarado
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Sandeep Raha
- Department of Pediatrics and the Graduate Programme in Medical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Boyang Zhang
- Department of Chemical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| |
Collapse
|
12
|
Merrill AK, Sobolewski M, Susiarjo M. Exposure to endocrine disrupting chemicals impacts immunological and metabolic status of women during pregnancy. Mol Cell Endocrinol 2023; 577:112031. [PMID: 37506868 PMCID: PMC10592265 DOI: 10.1016/j.mce.2023.112031] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/12/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023]
Affiliation(s)
- Alyssa K Merrill
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, NY, USA
| | - Marissa Sobolewski
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, NY, USA
| | - Martha Susiarjo
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, NY, USA.
| |
Collapse
|
13
|
A Comparative Review of Pregnancy and Cancer and Their Association with Endoplasmic Reticulum Aminopeptidase 1 and 2. Int J Mol Sci 2023; 24:ijms24043454. [PMID: 36834865 PMCID: PMC9965492 DOI: 10.3390/ijms24043454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
The fundamental basis of pregnancy and cancer is to determine the fate of the survival or the death of humanity. However, the development of fetuses and tumors share many similarities and differences, making them two sides of the same coin. This review presents an overview of the similarities and differences between pregnancy and cancer. In addition, we will also discuss the critical roles that Endoplasmic Reticulum Aminopeptidase (ERAP) 1 and 2 may play in the immune system, cell migration, and angiogenesis, all of which are essential for fetal and tumor development. Even though the comprehensive understanding of ERAP2 lags that of ERAP1 due to the lack of an animal model, recent studies have shown that both enzymes are associated with an increased risk of several diseases, including pregnancy disorder pre-eclampsia (PE), recurrent miscarriages, and cancer. The exact mechanisms in both pregnancy and cancer need to be elucidated. Therefore, a deeper understanding of ERAP's role in diseases can make it a potential therapeutic target for pregnancy complications and cancer and offer greater insight into its impact on the immune system.
Collapse
|
14
|
Ku CW, Ong LS, Goh JP, Allen J, Low LW, Zhou J, Tan TC, Lee YH. Defects in protective cytokine profiles in spontaneous miscarriage in the first trimester. F&S SCIENCE 2023; 4:36-46. [PMID: 36096448 DOI: 10.1016/j.xfss.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 09/04/2022] [Accepted: 09/06/2022] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To study differences in cytokine expression profiles between women with ongoing pregnancy and those experiencing spontaneous miscarriage, among women who presented with threatened miscarriage before week 16 of gestation. DESIGN Prospective cohort study. SETTING Academic hospital. PATIENT(S) In this prospective cohort study, 155 pregnant women, comprising normal pregnant women recruited from antenatal clinics (n = 97) and women with threatened miscarriage recruited from an emergency walk-in clinic (n = 58). INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Sixty-five serum cytokines quantified using multiplex immunoassay correlated with miscarriage outcomes. RESULT(S) Among women presenting with threatened miscarriage, those who eventually miscarried had significantly lower levels of interleukin (IL)-2, IL-12p70, IL-17A, B-cell-activating factor, B lymphocyte chemoattractant, basic nerve growth factor, interferon-γ, tumor necrosis factor-related apoptosis-inducing ligand, thymic stromal lymphopoietin, and tumor necrosis factor-α and higher levels of vascular endothelial growth factor A, IL-21, and stromal cell-derived factor 1α than those with ongoing pregnancy. Comparisons between normal pregnancies and women with threatened miscarriage who eventually miscarried revealed significant differences across 7 cytokines: B-cell-activating factor; B lymphocyte chemoattractant; basic nerve growth factor; IL-17A; fractalkine/CX3CL1; vascular endothelial growth factor A; and CCL22. Vascular endothelial growth factor A exhibited a negative correlation with the progesterone level (r = -0.270). The cluster of significant cytokines alludes to T cell proliferation, B-cell proliferation, natural killer cell-mediated cytotoxicity, and apoptosis as important pathways that determine pregnancy outcomes. Bioinformatic analysis further revealed alteration of the suppressor of cytokine signaling proteins family of Janus kinase-signal transducer and activator of transcription signaling axis by cytokines as a plausible key molecular mechanism in spontaneous miscarriage. CONCLUSION(S) This study demonstrates that the regulated balance between the proinflammatory and anti-inflammatory pathways is crucial to maintaining pregnancy. A better understanding of the cytokines associated with immunomodulatory effects may lead to novel targets for the prediction and treatment of spontaneous miscarriage.
Collapse
Affiliation(s)
- Chee Wai Ku
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore; Duke-NUS Medical School, Singapore
| | | | - Jody Paige Goh
- Division of Obstetrics and Gynecology, KK Women's and Children's Hospital, Singapore
| | | | - Louise Wenyi Low
- Division of Obstetrics and Gynecology, KK Women's and Children's Hospital, Singapore; Obstetrics and Gynecology-Academic Clinical Program, Duke-NUS Medical School, Singapore
| | - Jieliang Zhou
- Division of Obstetrics and Gynecology, KK Women's and Children's Hospital, Singapore; Obstetrics and Gynecology-Academic Clinical Program, Duke-NUS Medical School, Singapore
| | - Thiam Chye Tan
- Division of Obstetrics and Gynecology, KK Women's and Children's Hospital, Singapore; Obstetrics and Gynecology-Academic Clinical Program, Duke-NUS Medical School, Singapore
| | - Yie Hou Lee
- Obstetrics and Gynecology-Academic Clinical Program, Duke-NUS Medical School, Singapore; Translational 'Omics and Biomarkers Group, KK Research Centre, KK Women's and Children's Hospital, Singapore.
| |
Collapse
|
15
|
Something Smells Fishy: How Lipid Mediators Impact the Maternal-Fetal Interface and Neonatal Development. Biomedicines 2023; 11:biomedicines11010171. [PMID: 36672679 PMCID: PMC9855822 DOI: 10.3390/biomedicines11010171] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
Normal pregnancy relies on inflammation for implantation, placentation, and parturition, but uncontrolled inflammation can lead to poor maternal and infant outcomes. Maternal diet is one modifiable factor that can impact inflammation. Omega-3 and -6 fatty acids obtained through the diet are metabolized into bioactive compounds that effect inflammation. Recent evidence has shown that the downstream products of omega-3 and -6 fatty acids may influence physiology during pregnancy. In this review, the current knowledge relating to omega-3 and omega-6 metabolites during pregnancy will be summarized.
Collapse
|
16
|
Comins-Boo A, Valdeolivas L, Pérez-Pla F, Cristóbal I, Subhi-Issa N, Domínguez-Soto Á, Pilar-Suárez L, Gasca-Escorial P, Calvo-Urrutia M, Fernández-Arquero M, Herráiz MÁ, Corbí Á, Sánchez-Ramón S. Immunophenotyping of peripheral blood monocytes could help identify a baseline pro-inflammatory profile in women with recurrent reproductive failure. J Reprod Immunol 2022; 154:103735. [PMID: 36063657 DOI: 10.1016/j.jri.2022.103735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 07/23/2022] [Accepted: 08/19/2022] [Indexed: 12/14/2022]
Abstract
Recurrent pregnancy loss (RPL) and recurrent implantation failure (RIF) are two well-defined clinical entities, but the role of the monocytes in their pathophysiology needs to be clarified. This study aimed to evaluate the role of the three monocyte subsets (classical, intermediate, and non-classical) and relevant cytokines/chemokines in a cohort of RPL and RIF women to better characterize a baseline proinflammatory profile that could define inflammatory pathophysiology in these two different conditions. We evaluated 108 non-pregnant women: 53 RPL, 24 RIF, and 31 fertile healthy controls (HC). Multiparametric flow cytometry was used to quantify the frequency of surface chemokine receptors (CCR2, CCR5, and CX3CR1) on the monocyte subsets. Cytokines were assessed in plasma samples using a multiplex assay. The CX3CR1+ and CCR5+ intermediate monocytes were significantly higher in RPL and RIF compared to HC. A significant positive correlation was observed between CX3CR1+ intermediate monocytes and IL-17A (P = .03, r = 0.43). The Boruta algorithm followed by a multivariate logistic regression model was used to select the most relevant variables that could help define RPL and RIF: in RPL were CX3CR1 non-classical monocytes, TGF-β1, and CCR5 intermediate monocytes; in RIF: CCR5 intermediate monocytes and TGF-β3. The combination of these variables could predict RPL and RIF with 90 % and 82 %, respectively. Our study suggests that a combination of specific blood monocyte subsets and cytokines could aid in identifying RPL and RIF women with a pro-inflammatory profile. These findings could provide a more integrated understanding of these pathologies. Further investigation and validation in independent cohorts are warranted.
Collapse
Affiliation(s)
- Alejandra Comins-Boo
- Department of Immunology, IML, and IdSSC, Hospital Clínico San Carlos, Madrid, Spain; Department of Immunology, Ophthalmology, and ENT, School of Medicine, Complutense University School of Medicine, Madrid, Spain
| | - Lorena Valdeolivas
- Department of Immunology, IML, and IdSSC, Hospital Clínico San Carlos, Madrid, Spain; Department of Immunology, Ophthalmology, and ENT, School of Medicine, Complutense University School of Medicine, Madrid, Spain
| | - Fernando Pérez-Pla
- Department of Applied Mathematics and Computational Science, University of Cantabria, Spain
| | - Ignacio Cristóbal
- Department of Obstetrics and Gynecology, Hospital Clínico San Carlos, Madrid, Spain
| | - Nabil Subhi-Issa
- Department of Immunology, IML, and IdSSC, Hospital Clínico San Carlos, Madrid, Spain; Department of Immunology, Ophthalmology, and ENT, School of Medicine, Complutense University School of Medicine, Madrid, Spain
| | - Ángeles Domínguez-Soto
- Molecular Microbiology and Infection Biology Department, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Lydia Pilar-Suárez
- Department of Obstetrics and Gynecology, Hospital Clínico San Carlos, Madrid, Spain
| | - Pilar Gasca-Escorial
- Department of Obstetrics and Gynecology, Hospital Clínico San Carlos, Madrid, Spain
| | - Marta Calvo-Urrutia
- Department of Obstetrics and Gynecology, Hospital Clínico San Carlos, Madrid, Spain
| | - Miguel Fernández-Arquero
- Department of Immunology, IML, and IdSSC, Hospital Clínico San Carlos, Madrid, Spain; Department of Immunology, Ophthalmology, and ENT, School of Medicine, Complutense University School of Medicine, Madrid, Spain
| | - Miguel Ángel Herráiz
- Department of Obstetrics and Gynecology, Hospital Clínico San Carlos, Madrid, Spain
| | - Ángel Corbí
- Molecular Microbiology and Infection Biology Department, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Silvia Sánchez-Ramón
- Department of Immunology, IML, and IdSSC, Hospital Clínico San Carlos, Madrid, Spain; Department of Immunology, Ophthalmology, and ENT, School of Medicine, Complutense University School of Medicine, Madrid, Spain.
| |
Collapse
|
17
|
Crump LS, Kines KT, Richer JK, Lyons TR. Breast cancers co-opt normal mechanisms of tolerance to promote immune evasion and metastasis. Am J Physiol Cell Physiol 2022; 323:C1475-C1495. [PMID: 36189970 PMCID: PMC9662806 DOI: 10.1152/ajpcell.00189.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 11/22/2022]
Abstract
Normal developmental processes, such as those seen during embryonic development and postpartum mammary gland involution, can be reactivated by cancer cells to promote immune suppression, tumor growth, and metastatic spread. In mammalian embryos, paternal-derived antigens are at risk of being recognized as foreign by the maternal immune system. Suppression of the maternal immune response toward the fetus, which is mediated in part by the trophoblast, is critical to ensure embryonic survival and development. The postpartum mammary microenvironment also exhibits immunosuppressive mechanisms accompanying the massive cell death and tissue remodeling that occurs during mammary gland involution. These normal immunosuppressive mechanisms are paralleled during malignant transformation, where tumors can develop neoantigens that may be recognized as foreign by the immune system. To circumvent this, tumors can dedifferentiate and co-opt immune-suppressive mechanisms normally utilized during fetal tolerance and postpartum mammary involution. In this review, we discuss those similarities and how they can inform our understanding of cancer progression and metastasis.
Collapse
Affiliation(s)
- Lyndsey S Crump
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Kelsey T Kines
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center, Aurora, Colorado
| | - Traci R Lyons
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center, Aurora, Colorado
| |
Collapse
|
18
|
Denizli M, Capitano ML, Kua KL. Maternal obesity and the impact of associated early-life inflammation on long-term health of offspring. Front Cell Infect Microbiol 2022; 12:940937. [PMID: 36189369 PMCID: PMC9523142 DOI: 10.3389/fcimb.2022.940937] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/23/2022] [Indexed: 12/02/2022] Open
Abstract
The prevalence of obesity is increasingly common in the United States, with ~25% of women of reproductive age being overweight or obese. Metaflammation, a chronic low grade inflammatory state caused by altered metabolism, is often present in pregnancies complicated by obesity. As a result, the fetuses of mothers who are obese are exposed to an in-utero environment that has altered nutrients and cytokines. Notably, both human and preclinical studies have shown that children born to mothers with obesity have higher risks of developing chronic illnesses affecting various organ systems. In this review, the authors sought to present the role of cytokines and inflammation during healthy pregnancy and determine how maternal obesity changes the inflammatory landscape of the mother, leading to fetal reprogramming. Next, the negative long-term impact on offspring’s health in numerous disease contexts, including offspring’s risk of developing neuropsychiatric disorders (autism, attention deficit and hyperactive disorder), metabolic diseases (obesity, type 2 diabetes), atopy, and malignancies will be discussed along with the potential of altered immune/inflammatory status in offspring as a contributor of these diseases. Finally, the authors will list critical knowledge gaps in the field of developmental programming of health and diseases in the context of offspring of mothers with obesity, particularly the understudied role of hematopoietic stem and progenitor cells.
Collapse
Affiliation(s)
- Merve Denizli
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Indiana University School of Medicine, Indianapolis IN, United States
| | - Maegan L. Capitano
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis IN, United States
| | - Kok Lim Kua
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Indiana University School of Medicine, Indianapolis IN, United States
- *Correspondence: Kok Lim Kua,
| |
Collapse
|
19
|
Sieg W, Kiewisz J, Podolak A, Jakiel G, Woclawek-Potocka I, Lukaszuk J, Lukaszuk K. Inflammation-Related Molecules at the Maternal-Fetal Interface during Pregnancy and in Pathologically Altered Endometrium. Curr Issues Mol Biol 2022; 44:3792-3808. [PMID: 36135172 PMCID: PMC9497515 DOI: 10.3390/cimb44090260] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/04/2022] [Accepted: 08/15/2022] [Indexed: 11/23/2022] Open
Abstract
The blastocyst expresses paternally derived alloantigens and induces inflammation during implantation. However, it is necessary for the onset of pregnancy. An abnormal response might result in a pathological course of pregnancy or pregnancy failure. On the other hand, a state of maternal immune tolerance is necessary to ensure the normal development of pregnancy by suppressing inflammatory processes. This article discusses recognized mechanisms and the significance of inflammatory processes for embryo implantation and pregnancy establishment. We would also like to present disorders involving excessive inflammatory response and their influence on events occurring during embryo implantation. The chain of correlation between the processes responsible for embryo implantation and the subsequent physiological course of pregnancy is complicated. Many of those interrelationships are still yet to be discovered. Undoubtedly, their recognition will give hope to infertile couples for the emergence of new treatments that will increase the chance of giving birth to a healthy child.
Collapse
Affiliation(s)
| | - Jolanta Kiewisz
- Department of Human Histology and Embryology, Medical Faculty, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-561 Olsztyn, Poland
| | - Amira Podolak
- Department of Obstetrics and Gynecology Nursing, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Grzegorz Jakiel
- Invicta Research and Development Center, 81-740 Sopot, Poland
- The Center of Postgraduate Medical Education, 1st Department of Obstetrics and Gynecology, University of Gdansk, 01-004 Warsaw, Poland
| | - Izabela Woclawek-Potocka
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland
| | - Jakub Lukaszuk
- Invicta Research and Development Center, 81-740 Sopot, Poland
| | - Krzysztof Lukaszuk
- Invicta Research and Development Center, 81-740 Sopot, Poland
- Department of Obstetrics and Gynecology Nursing, Medical University of Gdansk, 80-210 Gdansk, Poland
| |
Collapse
|
20
|
Krstic J, Deutsch A, Fuchs J, Gauster M, Gorsek Sparovec T, Hiden U, Krappinger JC, Moser G, Pansy K, Szmyra M, Gold D, Feichtinger J, Huppertz B. (Dis)similarities between the Decidual and Tumor Microenvironment. Biomedicines 2022; 10:1065. [PMID: 35625802 PMCID: PMC9138511 DOI: 10.3390/biomedicines10051065] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/21/2022] [Accepted: 04/24/2022] [Indexed: 02/05/2023] Open
Abstract
Placenta-specific trophoblast and tumor cells exhibit many common characteristics. Trophoblast cells invade maternal tissues while being tolerated by the maternal immune system. Similarly, tumor cells can invade surrounding tissues and escape the immune system. Importantly, both trophoblast and tumor cells are supported by an abetting microenvironment, which influences invasion, angiogenesis, and immune tolerance/evasion, among others. However, in contrast to tumor cells, the metabolic, proliferative, migrative, and invasive states of trophoblast cells are under tight regulatory control. In this review, we provide an overview of similarities and dissimilarities in regulatory processes that drive trophoblast and tumor cell fate, particularly focusing on the role of the abetting microenvironments.
Collapse
Affiliation(s)
- Jelena Krstic
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| | - Alexander Deutsch
- Division of Hematology, Medical University of Graz, Stiftingtalstrasse 24, 8010 Graz, Austria; (A.D.); (K.P.); (M.S.)
| | - Julia Fuchs
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
- Division of Biophysics, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria
| | - Martin Gauster
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| | - Tina Gorsek Sparovec
- Department of Obstetrics and Gynecology, Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria; (T.G.S.); (U.H.); (D.G.)
| | - Ursula Hiden
- Department of Obstetrics and Gynecology, Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria; (T.G.S.); (U.H.); (D.G.)
| | - Julian Christopher Krappinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| | - Gerit Moser
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| | - Katrin Pansy
- Division of Hematology, Medical University of Graz, Stiftingtalstrasse 24, 8010 Graz, Austria; (A.D.); (K.P.); (M.S.)
| | - Marta Szmyra
- Division of Hematology, Medical University of Graz, Stiftingtalstrasse 24, 8010 Graz, Austria; (A.D.); (K.P.); (M.S.)
| | - Daniela Gold
- Department of Obstetrics and Gynecology, Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria; (T.G.S.); (U.H.); (D.G.)
| | - Julia Feichtinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| | - Berthold Huppertz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria; (J.K.); (J.F.); (M.G.); (J.C.K.); (G.M.); (B.H.)
| |
Collapse
|
21
|
Jiang Y, Huang F, Chai X, Yuan W, Ding H, Wu X. The role of IP-10 and its receptor CXCR3 in early pregnancy. Mol Immunol 2021; 140:59-69. [PMID: 34655920 DOI: 10.1016/j.molimm.2021.09.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 09/03/2021] [Accepted: 09/27/2021] [Indexed: 01/01/2023]
Abstract
The local immune microenvironment of the uterus plays an important role in a successful pregnancy. IP-10 (CXCL10) has been extensively studied in many immune-related diseases. However, the immune role of IP-10 in early pregnancy has not been fully recognized. This study mainly investigated the role of pro-inflammatory chemokine IP-10 in pregnancy. The levels of IP-10 and its receptor chemokine receptor 3 (CXCR3) were lower in the decidual tissues of an abortion-prone mice than in normal pregnant mice. Meantime, the expression of IP-10 and CXCR3 was higher in the decidual tissues of early pregnant women than in the endometrial tissues of non-pregnant women. IP-10 promoted the production of interleukin 17 (IL-17) and interferon gamma (IFN-γ), and also promoted the migration and differentiation of uterine decidual T cells to type 1 T helper (Th1) cells and Th17 cells. The abortion rate of early pregnant mice increased but the number of CD49b+, CD11b+, and CD3ε+ cells in the decidual tissues decreased upon treatment with anti-IP-10 antibody. Moreover, anti IP-10 antibody decreased the expression of RANTES but increased the expression of anti-inflammatory cytokines IL-6 and IL-10. A successful pregnancy requires the participation of IP-10. IP-10 participates in formation of the pro-inflammatory immune microenvironment during early pregnancy by regulating the distribution of immune cells and promoting the production of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Ying Jiang
- Department of Obstetrics & Gynecology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 430011, China
| | - Fengying Huang
- Department of Obstetrics & Gynecology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 430011, China
| | - Xiaoshan Chai
- Department of Obstetrics & Gynecology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 430011, China
| | - Wen Yuan
- Department of Obstetrics & Gynecology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 430011, China
| | - Hui Ding
- Department of Obstetrics & Gynecology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 430011, China
| | - Xianqing Wu
- Department of Obstetrics & Gynecology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 430011, China.
| |
Collapse
|
22
|
You Y, Stelzl P, Joseph DN, Aldo PB, Maxwell AJ, Dekel N, Liao A, Whirledge S, Mor G. TNF-α Regulated Endometrial Stroma Secretome Promotes Trophoblast Invasion. Front Immunol 2021; 12:737401. [PMID: 34790194 PMCID: PMC8591203 DOI: 10.3389/fimmu.2021.737401] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/14/2021] [Indexed: 01/01/2023] Open
Abstract
Successful implantation requires the coordinated migration and invasion of trophoblast cells from out of the blastocyst and into the endometrium. This process relies on signals produced by cells in the maternal endometrium. However, the relative contribution of stroma cells remains unclear. The study of human implantation has major technical limitations, therefore the need of in vitro models to elucidate the molecular mechanisms. Using a recently described 3D in vitro models we evaluated the interaction between trophoblasts and human endometrial stroma cells (hESC), we assessed the process of trophoblast migration and invasion in the presence of stroma derived factors. We demonstrate that hESC promotes trophoblast invasion through the generation of an inflammatory environment modulated by TNF-α. We also show the role of stromal derived IL-17 as a promoter of trophoblast migration through the induction of essential genes that confer invasive capacity to cells of the trophectoderm. In conclusion, we describe the characterization of a cellular inflammatory network that may be important for blastocyst implantation. Our findings provide a new insight into the complexity of the implantation process and reveal the importance of inflammation for embryo implantation.
Collapse
Affiliation(s)
- Yuan You
- C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, United States
| | - Patrick Stelzl
- Department for Gynecology, Obstetrics and Gynecological Endocrinology, Kepler University Hospital Linz, Johannes Kepler University Linz, Linz, Austria
| | - Dana N Joseph
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, United States
| | - Paulomi B Aldo
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, United States
| | - Anthony J Maxwell
- C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, United States
| | - Nava Dekel
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
| | - Aihua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shannon Whirledge
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, United States
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
23
|
da Silva Castro A, Angeloni MB, de Freitas Barbosa B, de Miranda RL, Teixeira SC, Guirelli PM, de Oliveira FC, José da Silva R, Franco PS, Ribeiro M, Milian ICB, de Oliveira Gomes A, Ietta F, Júnior SF, Mineo TWP, Mineo JR, de Oliveira Simões Alves CM, Ferro EAV. BEWO trophoblast cells and Toxoplasma gondii infection modulate cell death mechanisms in THP-1 monocyte cells by interference in the expression of death receptor and intracellular proteins. Tissue Cell 2021; 73:101658. [PMID: 34597888 DOI: 10.1016/j.tice.2021.101658] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 02/07/2023]
Abstract
Crosstalk between trophoblast and monocytes is essential for gestational success, and it can be compromised in congenital toxoplasmosis. Cell death is one of the mechanisms involved in the maintenance of pregnancy, and this study aimed to evaluate the role of trophoblast in the modulation of monocyte cell death in the presence or absence of Toxoplasma gondii infection. THP-1 cells were stimulated with supernatants of BeWo cells and then infected or not with T. gondii. The supernatants were collected and analyzed for the secretion of human Fas ligand, and cells were used to determine cell death and apoptosis, cell death receptor, and intracellular proteins expression. Cell death and apoptosis index were higher in uninfected THP-1 cells stimulated with supernatants of BeWo cells; however, apoptosis index was reduced by T. gondii infection. Macrophage migration inhibitory factor (MIF) and transforming growth factor (TGF)-β1, secreted by BeWo cells, altered the cell death and apoptosis rates in THP-1 cells. In infected THP-1 cells, the expression of Fas/CD95 and secretion of FasL was significantly higher; however, caspase 3 and phosphorylated extracellular-signal-regulated kinase (ERK1/2) were downregulated. Results suggest that soluble factors secreted by BeWo cells induce cell death and apoptosis in THP-1 cells, and Fas/CD95 can be involved in this process. On the other hand, T. gondii interferes in the mechanism of cell death and inhibits THP-1 cell apoptosis, which can be associated with active caspase 3 and phosphorylated ERK1/2. In conclusion, our results showed that human BeWo trophoblast cells and T. gondii infection modulate cell death in human THP-1 monocyte cells.
Collapse
Affiliation(s)
- Andressa da Silva Castro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, MG, Brazil
| | - Mariana Bodini Angeloni
- School of Medicine, Healthy Sciences Special Academic Unit, University of Goiás-Jataí, Jataí, GO, Brazil
| | - Bellisa de Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, MG, Brazil
| | - Renata Lima de Miranda
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, MG, Brazil
| | - Samuel Cota Teixeira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, MG, Brazil
| | - Pâmela Mendonça Guirelli
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, MG, Brazil
| | - Fernanda Chaves de Oliveira
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, MG, Brazil
| | - Rafaela José da Silva
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, MG, Brazil
| | - Priscila Silva Franco
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, MG, Brazil
| | - Mayara Ribeiro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, MG, Brazil
| | - Iliana Claudia Balga Milian
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, MG, Brazil
| | - Angélica de Oliveira Gomes
- Laboratory of Cell Biology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Francesca Ietta
- Department of Life Science, University of Siena, Siena, Italy
| | | | - Tiago Wilson Patriarca Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Federal University of Uberlandia, Uberlândia, MG, Brazil
| | - José Roberto Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Federal University of Uberlandia, Uberlândia, MG, Brazil
| | | | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, MG, Brazil.
| |
Collapse
|
24
|
Heusler M, Einenkel R, Ehrhardt J, Muzzio DO, Zygmunt M. Low Abundance Fusobacterium Nucleatum Supports Early Pregnancy Development - An In Vitro Study. Front Immunol 2021; 12:698045. [PMID: 34531854 PMCID: PMC8438310 DOI: 10.3389/fimmu.2021.698045] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/13/2021] [Indexed: 01/12/2023] Open
Abstract
Pregnancy success depends greatly on a balanced immune homeostasis. The detection of bacterial components in the upper reproductive tract in non-pregnant and pregnant women raised questions on its possible beneficial role in reproductive health. The local conditions that allow the presence of bacteria to harmonize with the establishment of pregnancy are still unknown. Among the described bacterial species in endometrial and placental samples, Fusobacterium nucleatum was found. It has been observed that F. nucleatum can induce tumorigenesis in colon carcinoma, a process that shares several features with embryo implantation. We propose that low concentrations of F. nucleatum may improve trophoblast function without exerting destructive responses. Inactivated F. nucleatum and E. coli were incubated with the trophoblastic cell lines HTR8/SVneo, BeWo, and JEG-3. Viability, proliferation, migratory capacity, invasiveness and the secretion of chemokines, other cytokines and matrix metalloproteinases were assessed. The presence of F. nucleatum significantly induced HTR8/SVneo invasion, accompanied by the secretion of soluble mediators (CXCL1, IL-6 and IL-8) and metalloproteinases (MMP-2 and MMP-9). However, as concentrations of F. nucleatum increased, these did not improve invasiveness, hindered migration, reduced cell viability and induced alterations in the cell cycle. Part of the F. nucleatum effects on cytokine release were reverted with the addition of a TLR4 blocking antibody. Other effects correlated with the level of expression of E-cadherin on the different cell lines tested. Low amounts of F. nucleatum promote invasion of HTR8/SVneo cells and induce the secretion of important mediators for pregnancy establishment. Some effects were independent of LPS and correlated with the expression of E-cadherin on trophoblasts.
Collapse
Affiliation(s)
- Martha Heusler
- Department of Obstetrics and Gynecology, University of Greifswald, Greifswald, Germany
| | - Rebekka Einenkel
- Department of Obstetrics and Gynecology, University of Greifswald, Greifswald, Germany
| | - Jens Ehrhardt
- Department of Obstetrics and Gynecology, University of Greifswald, Greifswald, Germany
| | - Damián Oscar Muzzio
- Department of Obstetrics and Gynecology, University of Greifswald, Greifswald, Germany
| | - Marek Zygmunt
- Department of Obstetrics and Gynecology, University of Greifswald, Greifswald, Germany
| |
Collapse
|
25
|
Peralta MB, Cainelli S, Stassi AF, Angeli E, Renna MS, Signorini ML, Gareis NC, Durante L, Rey F, Ortega HH, Salvetti NR, Velázquez MML. Association between phagocytic activity of monocytes and days to conception after parturition in dairy cows when considering the hormonal and metabolic milieu. Anim Reprod Sci 2021; 232:106818. [PMID: 34343817 DOI: 10.1016/j.anireprosci.2021.106818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 07/22/2021] [Accepted: 07/24/2021] [Indexed: 02/04/2023]
Abstract
The nutritional conditions and immune status of dairy cows affect reproductive performance. This study was conducted with the aim to analyze the phagocytic activity (PA) and phagocytic capacity (PC) of circulating monocytes after the period of transition from pregnancy to lactation, to evaluate possible associations with duration of time period to conception following parturition. Results indicated PA was not associated with duration of time period to conception following parturition. In contrast, cows with a lesser PC conceived earlier (98 ± 9 days in milk, DIM) than those with a greater PC (168 ± 15 DIM). Based on these results, to analyze the association of the hormonal and metabolic milieu with the PA and PC, the animals were grouped considering the days to conception following parturition. In the group with the greater number of days to conception (>168 DIM), the PA was associated with concentrations of progesterone and beta-hydroxybutyrate (BHB) at 90 DIM and glucose at 120 DIM, whereas PC was associated with the concentrations of progesterone, cortisol and glucose at 90 DIM, non-esterified fatty acids (NEFA) at 120 DIM, 17β-estradiol at 150 DIM, and 17β-estradiol and BHB at 180 DIM. Overall, these results represent a new perspective related to the reproductive performance of dairy cows. The modifications of cellular functions may be useful for predicting the onset of health complications in dairy cows and to manage cows in ways that result in an enhanced fertility during the subsequent lactational period.
Collapse
Affiliation(s)
- M B Peralta
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL)/Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Esperanza, Santa Fe, Argentina
| | - S Cainelli
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL)/Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Esperanza, Santa Fe, Argentina
| | - A F Stassi
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL)/Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Esperanza, Santa Fe, Argentina; Facultad de Ciencias Veterinarias del Litoral, Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
| | - E Angeli
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL)/Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Esperanza, Santa Fe, Argentina; Facultad de Ciencias Veterinarias del Litoral, Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
| | - M S Renna
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL)/Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Esperanza, Santa Fe, Argentina; Facultad de Ciencias Veterinarias del Litoral, Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
| | - M L Signorini
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Instituto Nacional de Tecnología Agropecuaria EEA Rafaela, Ruta 34 Km 227, Rafaela, Santa Fe, Argentina
| | - N C Gareis
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL)/Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Esperanza, Santa Fe, Argentina; Facultad de Ciencias Veterinarias del Litoral, Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
| | - L Durante
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL)/Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Esperanza, Santa Fe, Argentina; Facultad de Ciencias Veterinarias del Litoral, Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
| | - F Rey
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL)/Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Esperanza, Santa Fe, Argentina; Facultad de Ciencias Veterinarias del Litoral, Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
| | - H H Ortega
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL)/Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Esperanza, Santa Fe, Argentina; Facultad de Ciencias Veterinarias del Litoral, Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
| | - N R Salvetti
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL)/Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Esperanza, Santa Fe, Argentina; Facultad de Ciencias Veterinarias del Litoral, Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
| | - M M L Velázquez
- Laboratorio de Biología Celular y Molecular Aplicada, Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), Universidad Nacional del Litoral (UNL)/Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Esperanza, Santa Fe, Argentina; Facultad de Ciencias Veterinarias del Litoral, Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina.
| |
Collapse
|
26
|
Cui L, Jin X, Xu F, Wang S, Liu L, Li X, Lin H, Du M. Circadian rhythm-associated Rev-erbα modulates polarization of decidual macrophage via the PI3K/Akt signaling pathway. Am J Reprod Immunol 2021; 86:e13436. [PMID: 33934423 DOI: 10.1111/aji.13436] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/07/2021] [Accepted: 04/28/2021] [Indexed: 12/14/2022] Open
Abstract
PROBLEM Circadian rhythms are involved not only in the repair and regeneration of the immune system, but may also be associated with regulation of inflammation and immune responses. Rev-erbα could constitute a link between immunity and circadian rhythms since it is a transcription factor that regulates circadian rhythms and has functions in multiple physiological and pathological processes. Decidual macrophages (dMφs) play crucial roles in immune balance at the maternal-fetal interface, and abnormal macrophage polarization is related to adverse pregnancy outcomes, such as infertility, recurrent spontaneous abortion, and preterm labor. However, whether Rev-erbα could modulate the polarization of macrophages is unknown. METHODS OF STUDY In this study, we analyzed the phenotype of dMφs and the expression of Rev-erbα in dMφs from normal pregnancies and miscarriages. The effect of Rev-erbα on macrophage polarization was evaluated by its knockdown or pharmacological activation. The mechanism by which the Rev-erbα agonist SR9009 regulates macrophage polarization was also estimated. RESULTS A type-1 macrophage (M1)-like dominance was observed in dMφs from human miscarriages, with a decreased expression of Rev-erbα compared to that from normal pregnancies. Rev-erbα knockdown promoted M1 polarization in macrophages differentiated from the THP1 cell line, whereas pharmacological activation of Rev-erbα by SR9009 induced type-2 macrophage (M2)-like polarization in dMφs. Furthermore, we found that SR9009 induced M2 polarization in macrophages differentiated from the U937 cell line via the PI3K/Akt signaling pathway. CONCLUSION Rev-erbα may play an essential role in macrophage polarization. These findings might help elucidate the role of Rev-erbα in regulating the differentiation and functions of macrophages and suggest a therapeutic target for pregnancy loss and pregnancy complications.
Collapse
Affiliation(s)
- Liyuan Cui
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Xueling Jin
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Feng Xu
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Songcun Wang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Lu Liu
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Xinyi Li
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Haiyan Lin
- State Key Laboratory of Stem Cell and Reproductive Biology & Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Meirong Du
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| |
Collapse
|
27
|
Paparini DE, Grasso E, Fernandez LDC, Merech F, Weingrill‐Barbano R, Correa‐Silva S, Izbizky G, Abasolo JI, Hauk V, Ramhorst R, Bevilaqcua E, Pérez Leirós C. Decidual factors and vasoactive intestinal peptide guide monocytes to higher migration, efferocytosis and wound healing in term human pregnancy. Acta Physiol (Oxf) 2021; 232:e13579. [PMID: 33210807 DOI: 10.1111/apha.13579] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/03/2020] [Accepted: 11/16/2020] [Indexed: 12/14/2022]
Abstract
AIM To explore the functional profile of circulating monocytes and decidual macrophages at term human pregnancy and their contribution to tissue repair upon stimulation ex vivo with decidual factors and the vasoactive intestinal peptide (VIP). METHODS Peripheral blood monocytes were isolated from pregnant and non-pregnant volunteers and tested in vitro with decidual explants from term placenta and VIP. The effect of VIP on decidual explants and the effect of its conditioned media on monocytes or decidual macrophages isolated by magnetic beads was carried out by RT-qPCR and ELISA for cytokines expression and release. Migration assays were performed in transwell systems. Efferocytosis was assessed in monocytes or decidual macrophages with CFSE-labelled autologous apoptotic neutrophils and quantified by flow cytometry. Monocyte and decidual macrophages wound healing capacity was evaluated using human endometrial stromal cell monolayers. Immunohistochemistry was performed in serial tissue sections of different placentas. RESULTS VIP is expressed in the villi as well as in trophoblast giant cells distributed within the decidua of term placenta. VIP induced the expression of antiinflmammatory markers and monocyte chemoattractant CCL2 and CCL3 in decidual tissues. Monocytes presented higher migration towards decidual explants than CD4 and CD8 cells. VIP-conditioned monocytes displayed an enhanced efferocytosis and wound healing capacity comparable to that of decidual macrophages. Moreover limited efferocytosis of pregnant women monocytes was restored by VIP-induced decidual factors. CONCLUSION Results show the conditioning of monocytes by decidual factors and VIP to sustain processes required for tissue repair and homeostasis maintenance in term placenta.
Collapse
Affiliation(s)
- Daniel Esteban Paparini
- Universidad de Buenos Aires Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Instituto de Química Biológica de la Factultad de Ciencias Exactas y Naturales (IQUIBICEN) Buenos Aires Argentina
| | - Esteban Grasso
- Universidad de Buenos Aires Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Instituto de Química Biológica de la Factultad de Ciencias Exactas y Naturales (IQUIBICEN) Buenos Aires Argentina
- Institute of Biomedical Sciences Department of Cell and Developmental Biology University of São Paulo São Paulo Brazil
| | - Laura del Carmen Fernandez
- Universidad de Buenos Aires Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Instituto de Química Biológica de la Factultad de Ciencias Exactas y Naturales (IQUIBICEN) Buenos Aires Argentina
| | - Fátima Merech
- Universidad de Buenos Aires Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Instituto de Química Biológica de la Factultad de Ciencias Exactas y Naturales (IQUIBICEN) Buenos Aires Argentina
| | - Rodrigo Weingrill‐Barbano
- Institute of Biomedical Sciences Department of Cell and Developmental Biology University of São Paulo São Paulo Brazil
| | - Simone Correa‐Silva
- Institute of Biomedical Sciences Department of Cell and Developmental Biology University of São Paulo São Paulo Brazil
| | - Gustavo Izbizky
- Obstetric Service Hospital Italiano de Buenos Aires Buenos Aires Argentina
| | | | - Vanesa Hauk
- Universidad de Buenos Aires Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Instituto de Química Biológica de la Factultad de Ciencias Exactas y Naturales (IQUIBICEN) Buenos Aires Argentina
| | - Rosanna Ramhorst
- Universidad de Buenos Aires Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Instituto de Química Biológica de la Factultad de Ciencias Exactas y Naturales (IQUIBICEN) Buenos Aires Argentina
| | - Estela Bevilaqcua
- Institute of Biomedical Sciences Department of Cell and Developmental Biology University of São Paulo São Paulo Brazil
| | - Claudia Pérez Leirós
- Universidad de Buenos Aires Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Instituto de Química Biológica de la Factultad de Ciencias Exactas y Naturales (IQUIBICEN) Buenos Aires Argentina
| |
Collapse
|
28
|
Hurtado-Díaz M, Riojas-Rodríguez H, Rothenberg SJ, Schnaas-Arrieta L, Kloog I, Just A, Hernández-Bonilla D, Wright RO, Téllez-Rojo MM. Prenatal PM 2.5 exposure and neurodevelopment at 2 years of age in a birth cohort from Mexico city. Int J Hyg Environ Health 2021; 233:113695. [PMID: 33582606 DOI: 10.1016/j.ijheh.2021.113695] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND Recent studies have reported that air pollution exposure may have neurotoxic properties. OBJECTIVE To examine longitudinal associations between prenatal particles less than 2.5 μm in diameter (PM2.5) exposure and neurodevelopment during the first two years of children's life. METHODS Analysis was conducted in PROGRESS, a longitudinal birth cohort between 2007 and 2013 in Mexico City. We used satellite data to predict daily PM2.5 concentrations at high spatial resolution. Multivariate mixed-effect regression models were adjusted to examine cognitive, language and motor scores in children up to 24 months of age (n = 740) and each trimester-specific and whole pregnancy exposure to PM2.5. RESULTS Models adjusted by child sex, gestational age, birth weight, smoking and mother's IQ, showed that each increase of 1 μg/m3 of PM2.5 was associated with a decreased language function of -0.38 points (95% CI: -0.77, -0.01). PM2.5 exposure at third trimester of pregnancy contributed most to the observed association. CONCLUSION Our findings suggest that language development up to 24 months of age may be particularly sensitive to PM2.5 exposure during pregnancy.
Collapse
Affiliation(s)
- Magali Hurtado-Díaz
- Center for Population Health Research, National Institute of Public Health, Av. Universidad No. 655, Col. Sta. Ma. Ahuacatitlán, 62100, Cuernavaca, Morelos, Mexico.
| | - Horacio Riojas-Rodríguez
- Center for Population Health Research, National Institute of Public Health, Av. Universidad No. 655, Col. Sta. Ma. Ahuacatitlán, 62100, Cuernavaca, Morelos, Mexico.
| | - Stephen J Rothenberg
- Center for Population Health Research, National Institute of Public Health, Av. Universidad No. 655, Col. Sta. Ma. Ahuacatitlán, 62100, Cuernavaca, Morelos, Mexico.
| | - Lourdes Schnaas-Arrieta
- Department of Developmental Neurobiology, National Institute of Perinatology, Montes Urales 800 Col. Virreyes Deleg, Miguel Hidalgo D.F, C.P. 11000, USA.
| | - Itai Kloog
- Department of Geography and Environmental Development, Ben-Gurion University of the Negev (BGU), Beer Sheva, Israel.
| | - Allan Just
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, 17 East 102 Street, Floor 3, Room 131, New York, NY, 10029, USA.
| | - David Hernández-Bonilla
- Center for Population Health Research, National Institute of Public Health, Av. Universidad No. 655, Col. Sta. Ma. Ahuacatitlán, 62100, Cuernavaca, Morelos, Mexico.
| | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, 17 East 102 Street Floor 3 West Room D3-110, New York, 10029, NY, USA.
| | - Martha Ma Téllez-Rojo
- Center for Nutrition and Health Research, National Institute of Public Health, Av. Universidad No. 655, Col. Sta. Ma. Ahuacatitlán, 61200, Cuernavaca, Morelos, Mexico.
| |
Collapse
|
29
|
Wang R, Huang X, Ma C, Zhang H. Toxicological Effects of BPDE on Dysfunctions of Female Trophoblast Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1300:151-160. [PMID: 33523433 DOI: 10.1007/978-981-33-4187-6_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are widely spread persistent environmental toxicants. Its typical representative benzo[a]pyrene (BaP) is a human carcinogen. BaP can pass through the placental barrier and is finally metabolized into benzo[a]pyren-7, 8-dihydrodiol-9, 10-epoxide (BPDE). BPDE can form DNA adducts, which directly affect the female reproductive health. Based on the special physiological functions of trophoblast cells and its important effect on normal pregnancy, this chapter describes the toxicity and molecular mechanism of BPDE-induced dysfunctions of trophoblast cells. By affecting the invasion, migration, apoptosis, proliferation, inflammation, and hormone secretion of trophoblast cells, BPDE causes diseases such as choriocarcinoma, intrauterine growth restriction, eclampsia, and abortion. In the end, it is expected to provide a scientific basis and prevention approach for women's reproductive health and decision-making basis for the formulation of environmental health standards.
Collapse
Affiliation(s)
- Rong Wang
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health, Sichuan University, Chengdu, China
| | - Xinying Huang
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health, Sichuan University, Chengdu, China
| | - Chenglong Ma
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health, Sichuan University, Chengdu, China
| | - Huidong Zhang
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health, Sichuan University, Chengdu, China.
| |
Collapse
|
30
|
Dunk CE, van Dijk M, Choudhury R, Wright TJ, Cox B, Leavey K, Harris LK, Jones RL, Lye SJ. Functional Evaluation of STOX1 (STORKHEAD-BOX PROTEIN 1) in Placentation, Preeclampsia, and Preterm Birth. Hypertension 2020; 77:475-490. [PMID: 33356399 DOI: 10.1161/hypertensionaha.120.15619] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Revaluation of the association of the STOX1 (STORKHEAD_BOX1 PROTEIN 1) transcription factor mutation (Y153H, C allele) with the early utero-vascular origins of placental pathology is warranted. To investigate if placental STOX1 Y153H genotype affects utero-vascular remodeling-compromised in both preterm birth and preeclampsia-we utilized extravillous trophoblast (EVT) explant and placental decidual coculture models, transfection of STOX1 wild-type and mutant plasmids into EVT-like trophoblast cell lines, and a cohort of 75 placentas from obstetric pathologies. Primary EVT and HTR8/SVneo cells carrying STOX1 Y153H secreted lower levels of IL (interleukin) 6, and IL-8, and higher CXCL16 (chemokine [C-X-C motif] ligand 16) and TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) than wild-type EVT and Swan71 cells. Media from wild-type EVT or Swan71 cells transfected with wild-type STOX1 stimulated: endothelial chemokine expression, angiogenesis, and decidual natural killer cell and monocyte migration. In contrast, Y153H EVT conditioned medium, Swan71 transfected with the Y153H plasmid, or HTR8/SVneo media had no effect. Genotyping of placental decidual cocultures demonstrated association of the placental STOX1 CC allele with failed vascular remodeling. Decidual GG NODAL R165H increased in failed cocultures carrying the placental CC alleles of STOX1. Multivariate analysis of the placental cohort showed that the STOX1 C allele correlated with premature birth, with or without severe early-onset preeclampsia, and small for gestational age babies. In conclusion, placental STOX1 Y153H is a precipitating factor in preterm birth and placental preeclampsia due to defects in early utero-placental development.
Collapse
Affiliation(s)
- Caroline E Dunk
- From the Research Centre for Women's and Infants' Health, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Canada (C.E.D., S.J.L.)
| | - Marie van Dijk
- Reproductive Biology Laboratory, Amsterdam University Medical Centers, the Netherlands (M.V.D.)
| | - Ruhul Choudhury
- Maternal and Fetal Health Research Centre, Faculty of Biology Medicine and Health (R.C., L.K.H., R.L.J.), University of Manchester, United Kingdom.,Academic Health Science Centre, St Mary's Hospital, Manchester, United Kingdom (R.C., L.K.H., R.L.J.)
| | - Thomas J Wright
- Department of Ophthalmology, Kensington Eye Institute (T.J.W.), University of Toronto, Canada
| | - Brian Cox
- Department of Physiology, Faculty of Medicine (B.C., K.L., S.J.L.), University of Toronto, Canada
| | - Katherine Leavey
- Department of Physiology, Faculty of Medicine (B.C., K.L., S.J.L.), University of Toronto, Canada
| | - Lynda K Harris
- Maternal and Fetal Health Research Centre, Faculty of Biology Medicine and Health (R.C., L.K.H., R.L.J.), University of Manchester, United Kingdom.,Division of Pharmacy and Optometry (L.K.H.), University of Manchester, United Kingdom.,Academic Health Science Centre, St Mary's Hospital, Manchester, United Kingdom (R.C., L.K.H., R.L.J.)
| | - Rebecca L Jones
- Maternal and Fetal Health Research Centre, Faculty of Biology Medicine and Health (R.C., L.K.H., R.L.J.), University of Manchester, United Kingdom.,Academic Health Science Centre, St Mary's Hospital, Manchester, United Kingdom (R.C., L.K.H., R.L.J.)
| | - Stephen J Lye
- From the Research Centre for Women's and Infants' Health, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Canada (C.E.D., S.J.L.).,Department of Physiology, Faculty of Medicine (B.C., K.L., S.J.L.), University of Toronto, Canada.,Department of Obstetrics and Gynaecology, Faculty of Medicine (S.J.L.), University of Toronto, Canada
| |
Collapse
|
31
|
Favaro RR, Murrieta-Coxca JM, Gutiérrez-Samudio RN, Morales-Prieto DM, Markert UR. Immunomodulatory properties of extracellular vesicles in the dialogue between placental and immune cells. Am J Reprod Immunol 2020; 85:e13383. [PMID: 33251688 DOI: 10.1111/aji.13383] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/18/2020] [Indexed: 12/16/2022] Open
Abstract
Extracellular vesicle (EV)-mediated communication has been implicated in the cooperative alliance between trophoblast and immune cells toward maternal tolerance and placentation. Syncytiotrophoblast cells secrete EVs directly into the maternal circulation, which are taken up by immune cells, endothelial cells, and other cell types. Initial evidence also shows that EVs produced by immune cells are, in turn, incorporated by trophoblast cells and modulate placental responses. Non-coding RNAs (ncRNAs), proteins, and lipid mediators transported in EVs are able to influence proliferation, differentiation, cytokine production, and immunological responses of recipient cells. The molecular alphabet and cellular targets involved in this dialogue are being revealed. Nevertheless, several questions regarding the whole content, surface markers, and biological functions of EVs still remain to be investigated in both physiological and pathological conditions. Analysis of circulating EVs in maternal blood has the potential to serve as a minimally invasive approach to monitoring placental functions and immunological features of pregnancy, aiding in the diagnostics of complications. This review addresses the immunomodulatory properties of EVs and their tasks in the communication between placental and immune cells.
Collapse
Affiliation(s)
- Rodolfo R Favaro
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| | - Jose Martín Murrieta-Coxca
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany.,RNA Bioinformatics, High Throughput Analysis, Faculty of Mathematics and Computer Science, Friedrich-Schiller-University Jena, Jena, Germany
| | | | | | - Udo R Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| |
Collapse
|
32
|
Xie J, Zhu H, Chang HM, Klausen C, Dong M, Leung PCK. GDF8 Promotes the Cell Invasiveness in Human Trophoblasts by Upregulating the Expression of Follistatin-Like 3 Through the ALK5-SMAD2/3 Signaling Pathway. Front Cell Dev Biol 2020; 8:573781. [PMID: 33195207 PMCID: PMC7655915 DOI: 10.3389/fcell.2020.573781] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/28/2020] [Indexed: 11/30/2022] Open
Abstract
Growth differentiation factor 8 (GDF8) and its antagonist follistatin-like 3 (FSTL3) are expressed in the placenta during early pregnancy. These two factors may have a role to play in the regulation of normal placentation. However, whether GDF8 can regulate the expression of FSTL3 in human trophoblasts remains to be elucidated. In this study, we aimed to investigate the effects of GDF8 on the expression of FSTL3 and the underlying molecular mechanisms using human trophoblasts as a study model. Our results showed that GDF8 significantly upregulates the expression and production of FSTL3, which further promotes cell invasiveness in immortalized extravillous cytotrophoblast cells and primary extravillous cytotrophoblast cells obtained from human first-trimester placentae. Additionally, using an siRNA-mediated knockdown approach, we found that this regulatory effect is most likely mediated by the ALK5-Sma- and Mad-related protein (SMAD)2/3-induced signaling pathway. These findings deepen our understanding of the functional roles of GDF8 and FSTL3 in the regulation of cell invasiveness of trophoblasts.
Collapse
Affiliation(s)
- Jiamin Xie
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Hua Zhu
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Christian Klausen
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Minyue Dong
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Women's Reproductive Health of Zhejiang Province, Hangzhou, China.,Key Laboratory of Reproductive Genetics, Ministry of Education, Hangzhou, China
| | - Peter C K Leung
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
33
|
Arefieva A, Nikolaeva M, Stepanova E, Krechetova L, Golubeva E, Tetruashvili N, Sukhikh G. Association of CD200 expression in paternal lymphocytes with female Th1/Th2 balance and pregnancy establishment at immunotherapy of recurrent spontaneous abortion. Am J Reprod Immunol 2020; 85:e13355. [PMID: 33015886 DOI: 10.1111/aji.13355] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 09/11/2020] [Accepted: 09/28/2020] [Indexed: 12/20/2022] Open
Abstract
PROBLEM We hypothesized that expression of transmembrane glycoprotein CD200 on paternal lymphocytes used for pre-gestational lymphocyte immunotherapy (LIT) of recurrent spontaneous abortion (RSA) can suppress the pro-inflammatory Th1-type immunity required for successful implantation. To reveal the association between CD200 expression, female immune background after LIT, and pregnancy establishment, we have performed this work. METHOD OF STUDY Pre-gestational alloimmunizations were given to 37 women using paternal peripheral blood leukocytes, combined with additional alloimmunizations in case of pregnancy. Lymphocyte phenotypes were determined by flow cytometry. Cytokines produced by mitogen-stimulated female peripheral blood cells were estimated by FlowCytomix™ technology. RESULTS We have shown that 78.4% (29/37) of women became pregnant within 12 menstrual cycles after pre-gestational LIT. Pregnancy establishment depends on the intensity of CD200 expression, which is significantly higher on the CD200+ lymphocytes administered to women who later did not achieve pregnancy (P < .05). The expression of CD200 negatively correlates with the ratios of Th1/Th2 cytokines produced by female peripheral blood cells (P < .05) and positively correlates with the frequency of female circulating regulatory T cells after LIT (P < .05). The ROC analysis showed that the intensity of CD200 expression and the Th1/Th2 ratios are the significant predictors of pregnancy establishment after pre-gestational LIT (P < .05 and P < .01, respectively). CONCLUSION Elevated CD200 expression on allogeneic lymphocytes most likely suppresses the pro-inflammatory Th1-type immunity needed for successful embryo implantation. Therefore, a personalized approach of LIT should be applied to avoid negative effects of such immunomodulation on pregnancy establishment.
Collapse
Affiliation(s)
- Alla Arefieva
- Laboratory of Clinical Immunology, National Medical Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Marina Nikolaeva
- Laboratory of Clinical Immunology, National Medical Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Elena Stepanova
- Laboratory of Clinical Immunology, National Medical Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Lubov Krechetova
- Laboratory of Clinical Immunology, National Medical Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Elena Golubeva
- Laboratory of Clinical Immunology, National Medical Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Nana Tetruashvili
- Department of Pregnancy Loss Prevention and Therapy, National Medical Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Gennady Sukhikh
- Laboratory of Clinical Immunology, National Medical Research Center for Obstetrics, Gynecology and Perinatology of Ministry of Healthcare of Russian Federation, Moscow, Russia.,First Moscow State Medical University named after I.M. Sechenov, Moscow, Russia
| |
Collapse
|
34
|
Cornish EF, Filipovic I, Åsenius F, Williams DJ, McDonnell T. Innate Immune Responses to Acute Viral Infection During Pregnancy. Front Immunol 2020; 11:572567. [PMID: 33101294 PMCID: PMC7556209 DOI: 10.3389/fimmu.2020.572567] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Abstract
Immunological adaptations in pregnancy allow maternal tolerance of the semi-allogeneic fetus but also increase maternal susceptibility to infection. At implantation, the endometrial stroma, glands, arteries and immune cells undergo anatomical and functional transformation to create the decidua, the specialized secretory endometrium of pregnancy. The maternal decidua and the invading fetal trophoblast constitute a dynamic junction that facilitates a complex immunological dialogue between the two. The decidual and peripheral immune systems together assume a pivotal role in regulating the critical balance between tolerance and defense against infection. Throughout pregnancy, this equilibrium is repeatedly subjected to microbial challenge. Acute viral infection in pregnancy is associated with a wide spectrum of adverse consequences for both mother and fetus. Vertical transmission from mother to fetus can cause developmental anomalies, growth restriction, preterm birth and stillbirth, while the mother is predisposed to heightened morbidity and maternal death. A rapid, effective response to invasive pathogens is therefore essential in order to avoid overwhelming maternal infection and consequent fetal compromise. This sentinel response is mediated by the innate immune system: a heritable, highly evolutionarily conserved system comprising physical barriers, antimicrobial peptides (AMP) and a variety of immune cells—principally neutrophils, macrophages, dendritic cells, and natural killer cells—which express pattern-receptors that detect invariant molecular signatures unique to pathogenic micro-organisms. Recognition of these signatures during acute infection triggers signaling cascades that enhance antimicrobial properties such as phagocytosis, secretion of pro-inflammatory cytokines and activation of the complement system. As well as coordinating the initial immune response, macrophages and dendritic cells present microbial antigens to lymphocytes, initiating and influencing the development of specific, long-lasting adaptive immunity. Despite extensive progress in unraveling the immunological adaptations of pregnancy, pregnant women remain particularly susceptible to certain acute viral infections and continue to experience mortality rates equivalent to those observed in pandemics several decades ago. Here, we focus specifically on the pregnancy-induced vulnerabilities in innate immunity that contribute to the disproportionately high maternal mortality observed in the following acute viral infections: Lassa fever, Ebola virus disease (EVD), dengue fever, hepatitis E, influenza, and novel coronavirus infections.
Collapse
Affiliation(s)
- Emily F Cornish
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, United Kingdom
| | - Iva Filipovic
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Fredrika Åsenius
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, United Kingdom
| | - David J Williams
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, United Kingdom
| | - Thomas McDonnell
- Department of Biochemical Engineering, University College London, London, United Kingdom
| |
Collapse
|
35
|
Gordon SM, Nishiguchi MA, Chase JM, Mani S, Mainigi MA, Behrens EM. IFNs Drive Development of Novel IL-15-Responsive Macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:1113-1124. [PMID: 32690654 PMCID: PMC7415599 DOI: 10.4049/jimmunol.2000184] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/08/2020] [Indexed: 11/19/2022]
Abstract
Disruption in homeostasis of IL-15 is linked to poor maternal and fetal outcomes during pregnancy. The only cells described to respond to IL-15 at the early maternal-fetal interface have been NK cells. We now show a novel population of macrophages, evident in several organs but enriched in the uterus of mice and humans, expressing the β-chain of the IL-15R complex (CD122) and responding to IL-15. CD122+ macrophages (CD122+Macs) are morphologic, phenotypic, and transcriptomic macrophages that can derive from bone marrow monocytes. CD122+Macs develop in the uterus and placenta with kinetics that mirror IFN activity at the maternal-fetal interface. M-CSF permits macrophages to express CD122, and IFNs are sufficient to drive expression of CD122 on macrophages. Neither type I nor type II IFNs are required to generate CD122+Macs, however. In response to IL-15, CD122+Macs activate the ERK signaling cascade and enhance production of proinflammatory cytokines after stimulation with the TLR9 agonist CpG. Finally, we provide evidence of human cells that phenocopy murine CD122+Macs in secretory phase endometrium during the implantation window and in first-trimester uterine decidua. Our data support a model wherein IFNs local to the maternal-fetal interface direct novel IL-15-responsive macrophages with the potential to mediate IL-15 signals critical for optimal outcomes of pregnancy.
Collapse
Affiliation(s)
- Scott M Gordon
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA 19104
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Mailyn A Nishiguchi
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Julie M Chase
- Division of Rheumatology, Children's Hospital of Philadelphia, Philadelphia, PA 19104; and
| | - Sneha Mani
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA 19104
| | - Monica A Mainigi
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, PA 19104
| | - Edward M Behrens
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104;
- Division of Rheumatology, Children's Hospital of Philadelphia, Philadelphia, PA 19104; and
| |
Collapse
|
36
|
Zhang YH, Aldo P, You Y, Ding J, Kaislasuo J, Petersen JF, Lokkegaard E, Peng G, Paidas MJ, Simpson S, Pal L, Guller S, Liu H, Liao AH, Mor G. Trophoblast-secreted soluble-PD-L1 modulates macrophage polarization and function. J Leukoc Biol 2020; 108:983-998. [PMID: 32386458 DOI: 10.1002/jlb.1a0420-012rr] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/22/2022] Open
Abstract
Decidual macrophages are in close contact with trophoblast cells during placenta development, and an appropriate crosstalk between these cellular compartments is crucial for the establishment and maintenance of a healthy pregnancy. During different phases of gestation, macrophages undergo dynamic changes to adjust to the different stages of fetal development. Trophoblast-secreted factors are considered the main modulators responsible for macrophage differentiation and function. However, the phenotype of these macrophages induced by trophoblast-secreted factors and the factors responsible for their polarization has not been elucidated. In this study, we characterized the phenotype and function of human trophoblast-induced macrophages. Using in vitro models, we found that human trophoblast-educated macrophages were CD14+ CD206+ CD86- and presented an unusual transcriptional profile in response to TLR4/LPS activation characterized by the expression of type I IFN-β expression. IFN-β further enhances the constitutive production of soluble programmed cell death ligand 1 (PD-L1) from trophoblast cells. PD-1 blockage inhibited trophoblast-induced macrophage differentiation. Soluble PD-L1 (sPD-L1) was detected in the blood of pregnant women and increased throughout the gestation. Collectively, our data suggest the existence of a regulatory circuit at the maternal fetal interface wherein IFN-β promotes sPD-L1 expression/secretion by trophoblast cells, which can then initiate a PD-L1/PD-1-mediated macrophage polarization toward an M2 phenotype, consequently decreasing inflammation. Macrophages then maintain the expression of sPD-L1 by the trophoblasts through IFN-β production induced through TLR4 ligation.
Collapse
Affiliation(s)
- Yong-Hong Zhang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA.,Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Paulomi Aldo
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | - Yuan You
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Jiahui Ding
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Janina Kaislasuo
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Obstetrics and Gynecology, University of Helsinki and the Helsinki University Hospital, Helsinki, Finland
| | - Jesper F Petersen
- Department of Obstetrics and Gynecology, North Zealand Hospital, Hilleroed, Denmark
| | - Ellen Lokkegaard
- Department of Obstetrics and Gynecology, North Zealand Hospital, Hilleroed, Denmark
| | - Gang Peng
- Department of Biostatistics, School of Public Health, Yale University, New Haven, Connecticut, USA
| | - Michael J Paidas
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Samantha Simpson
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | - Lubna Pal
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | - Seth Guller
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | - Hong Liu
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Ai Hua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Gil Mor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA.,C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
37
|
Cappelletti M, Presicce P, Kallapur SG. Immunobiology of Acute Chorioamnionitis. Front Immunol 2020; 11:649. [PMID: 32373122 PMCID: PMC7177011 DOI: 10.3389/fimmu.2020.00649] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 03/23/2020] [Indexed: 12/19/2022] Open
Abstract
Acute chorioamnionitis is characterized by neutrophilic infiltration and inflammation at the maternal fetal interface. It is a relatively common complication of pregnancy and can have devastating consequences including preterm labor, maternal infections, fetal infection/inflammation, fetal lung, brain, and gastrointestinal tract injury. In this review, we will discuss current understanding of the pathogenesis, immunobiology, and mechanisms of this condition. Most commonly, acute chorioamnionitis is a result of ascending infection with relatively low-virulence organisms such as the Ureaplasma species. Furthermore, recent vaginal microbiome studies suggest that there is a link between vaginal dysbiosis, vaginal inflammation, and ascending infection. Although less common, microorganisms invading the maternal-fetal interface via hematogenous route (e.g., Zika virus, Cytomegalovirus, and Listeria) can cause placental villitis and severe fetal inflammation and injury. We will provide an overview of the knowledge gleaned from different animal models of acute chorioamnionitis and the role of different immune cells in different maternal-fetal compartments. Lastly, we will discuss how infectious agents can break the maternal tolerance of fetal allograft during pregnancy and highlight the novel future therapeutic approaches.
Collapse
Affiliation(s)
- Monica Cappelletti
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | - Pietro Presicce
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | - Suhas G Kallapur
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
38
|
Tagoma A, Haller‐Kikkatalo K, Roos K, Oras A, Kirss A, Ilonen J, Uibo R. Interleukin‐7, T helper 1, and regulatory T‐cell activity‐related cytokines are increased during the second trimester of healthy pregnancy compared to non‐pregnant women. Am J Reprod Immunol 2019; 82:e13188. [DOI: 10.1111/aji.13188] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 08/02/2019] [Accepted: 08/28/2019] [Indexed: 12/17/2022] Open
Affiliation(s)
- Aili Tagoma
- Department of Immunology Faculty of Medicine Institute of Biomedicine and Translational Medicine University of Tartu Tartu Estonia
| | - Kadri Haller‐Kikkatalo
- Department of Immunology Faculty of Medicine Institute of Biomedicine and Translational Medicine University of Tartu Tartu Estonia
| | - Kristine Roos
- Department of Immunology Faculty of Medicine Institute of Biomedicine and Translational Medicine University of Tartu Tartu Estonia
| | - Astrid Oras
- Department of Immunology Faculty of Medicine Institute of Biomedicine and Translational Medicine University of Tartu Tartu Estonia
| | - Anne Kirss
- Women's Clinic Tartu University Hospital Tartu Estonia
| | - Jorma Ilonen
- Immunogenetics Laboratory Institute of Biomedicine University of Turku Turku Finland
- Clinical Microbiology Turku University Hospital Turku Finland
| | - Raivo Uibo
- Department of Immunology Faculty of Medicine Institute of Biomedicine and Translational Medicine University of Tartu Tartu Estonia
| |
Collapse
|
39
|
Fan M, Li X, Gao X, Dong L, Xin G, Chen L, Qiu J, Xu Y. LPS Induces Preeclampsia-Like Phenotype in Rats and HTR8/SVneo Cells Dysfunction Through TLR4/p38 MAPK Pathway. Front Physiol 2019; 10:1030. [PMID: 31507429 PMCID: PMC6718930 DOI: 10.3389/fphys.2019.01030] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/25/2019] [Indexed: 12/20/2022] Open
Abstract
Accumulating evidence has shown that preeclampsia (PE) was associated with an aberrant maternal-fetal inflammatory response. In the present study, we first found that in human PE placentas levels of toll-like receptor 4 (TLR4), phosphorylated p38 mitogen-activated protein kinase (p-p38 MAPK) and inflammatory cytokines IL-6 and MCP-1 were significantly upregulated. Next, we demonstrated a notable increase in systolic blood pressure (SBP) and proteinuria in lipopolysaccharide (LPS)-treated pregnant rats and concomitant high levels of TLR4 and p-p38 in these PE-like rat placentas, which led to aberrant overexpression of both IL-6 and MCP-1, as well as deficient trophoblast invasion and spiral artery (SA) remodeling, and these abnormalities were ameliorated by SB203580, a reported inhibitor of p38. In vitro we further confirmed that LPS triggered the activation of TLR4/p38 signaling pathway, which promoted trophoblast apoptosis and damaged trophoblastic invasion via downstream effectors IL-6 and MCP-1; these mutations were rectified by silencing this signaling pathway. These findings elaborated potential mechanisms that aberrant TLR4/p38 signaling might contribute to PE and LPS-induced PE-like symptom by damaging trophoblast invasion and SA remodeling via activating inflammatory cytokines including IL-6 and MCP-1.
Collapse
Affiliation(s)
- Minghua Fan
- Department of Obstetrics and Gynecology, The Second Hospital of Shandong University, Jinan, China
| | - Xiaobing Li
- Department of Laboratory Medicine, Institute of Basic Medicine, Shandong First Medical University and Academy of Medical Sciences, Jinan, China
| | - Xiaolin Gao
- Department of Obstetrics and Gynecology, The Second Hospital of Shandong University, Jinan, China
| | - Lihua Dong
- Department of Obstetrics and Gynecology, The Second Hospital of Shandong University, Jinan, China
| | - Gang Xin
- Department of Obstetrics and Gynecology, The Second Hospital of Shandong University, Jinan, China
| | - Liqun Chen
- Department of Nephrology, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianqing Qiu
- Department of Obstetrics and Gynecology, The Second Hospital of Shandong University, Jinan, China
| | - Yongping Xu
- Department of Obstetrics and Gynecology, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
40
|
Kaminski VDL, Ellwanger JH, Chies JAB. Extracellular vesicles in host-pathogen interactions and immune regulation - exosomes as emerging actors in the immunological theater of pregnancy. Heliyon 2019; 5:e02355. [PMID: 31592031 PMCID: PMC6771614 DOI: 10.1016/j.heliyon.2019.e02355] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 06/30/2019] [Accepted: 08/19/2019] [Indexed: 02/06/2023] Open
Abstract
This review correlates and summarizes the role of the maternal-fetal interface in the immune tolerance of the fetus and the processes that lead to infection avoidance, emphasizing the participation of exosomes and other extracellular vesicles in both situations. Exosomes are released into the extracellular medium by several cell types and are excellent carriers of biomolecules. Host-derived exosomes and the transport of pathogen-derived molecules by exosomes impact infections in different ways. The interactions of exosomes with the maternal immune system are pivotal to a favorable gestational outcome. In this review, we highlight the potential role of exosomes in the establishment of an adequate milieu that enables embryo implantation and discuss the participation of exosomes released at the maternal-fetal interface during the establishment of an immune-privileged compartment for fetal development. The placenta is a component where important strategies are used to minimize the risk of infection. To present a contrast, we also discuss possible mechanisms used by pathogens to cross the maternal-fetal interface. We review the processes, mechanisms, and potential consequences of dysregulation in all of the abovementioned phenomena. Basic information about exosomes and their roles in viral immune evasion is also presented. The interactions between extracellular vesicles and bacteria, fungi, parasites and proteinaceous infectious agents are addressed. The discovery of the placental microbiota and the implications of this new microbiota are also discussed, and current proposals that explain fetal/placental colonization by both pathogenic and commensal microbes are addressed. The comprehension of such interactions will help us to understand the immune dynamics of human pregnancy and the mechanisms of immune evasion used by different pathogens.
Collapse
Affiliation(s)
| | | | - José Artur Bogo Chies
- Laboratório de Imunobiologia e Imunogenética, Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, RS, Brazil
| |
Collapse
|
41
|
CXCL9/CXCL10 angiostasis CXC-chemokines in parallel with the CXCL12 as an angiogenesis CXC-chemokine are variously expressed in pre-eclamptic-women and their neonates. Pregnancy Hypertens 2019; 17:36-42. [DOI: 10.1016/j.preghy.2019.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 04/14/2019] [Accepted: 05/02/2019] [Indexed: 12/31/2022]
|
42
|
Schumacher A, Sharkey DJ, Robertson SA, Zenclussen AC. Immune Cells at the Fetomaternal Interface: How the Microenvironment Modulates Immune Cells To Foster Fetal Development. THE JOURNAL OF IMMUNOLOGY 2019; 201:325-334. [PMID: 29987001 DOI: 10.4049/jimmunol.1800058] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/06/2018] [Indexed: 12/23/2022]
Abstract
Immune cells adapt their phenotypic and functional characteristics in response to the tissue microenvironment within which they traffic and reside. The fetomaternal interface, consisting of placental trophoblasts and the maternal decidua, is a highly specialized tissue with a unique and time-limited function: to nourish and support development of the semiallogeneic fetus and protect it from inflammatory or immune-mediated injury. It is therefore important to understand how immune cells within these tissues are educated and adapt to fulfill their biological functions. This review article focuses on the local regulatory mechanisms ensuring that both innate and adaptive immune cells appropriately support the early events of implantation and placental development through direct involvement in promoting immune tolerance of fetal alloantigens, suppressing inflammation, and remodeling of maternal uterine vessels to facilitate optimal placental function and fetal growth.
Collapse
Affiliation(s)
- Anne Schumacher
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg 39108, Germany; and
| | - David J Sharkey
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, South Australia 5005, Australia
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, South Australia 5005, Australia
| | - Ana C Zenclussen
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg 39108, Germany; and
| |
Collapse
|
43
|
Azizi R, Soltani-Zangbar MS, sheikhansari G, Pourmoghadam Z, Mehdizadeh A, Mahdipour M, Sandoghchian S, Danaii S, Koushaein L, Samadi Kafil H, Yousefi M. Metabolic syndrome mediates inflammatory and oxidative stress responses in patients with recurrent pregnancy loss. J Reprod Immunol 2019; 133:18-26. [DOI: 10.1016/j.jri.2019.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/09/2019] [Accepted: 05/07/2019] [Indexed: 12/11/2022]
|
44
|
Piechowski J. Plausibility of trophoblastic-like regulation of cancer tissue. Cancer Manag Res 2019; 11:5033-5046. [PMID: 31213916 PMCID: PMC6549421 DOI: 10.2147/cmar.s190932] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 04/30/2019] [Indexed: 01/15/2023] Open
Abstract
Background: Thus far, a well-established logical pattern of malignancy does not exist. The current approach to cancer properties is primarily descriptive with usually, for each of them, extensive analyses of the underlying associated biomolecular mechanisms. However, this remains a catalog and it would be valuable to determine the organizational chart that could account for their implementation, hierarchical links and input into tumor regulation. Hypothesis: Striking phenotypic similarities exist between trophoblast (invasive and expanding early placenta) and cancer regarding cell functions, logistics of development, means of protection and capacity to hold sway over the host organism. The concept of cancer cell trophoblastic-like transdifferentiation appears to be a rational proposal in an attempt to explain this analogy and provide a consistent insight into how cancer cells are functioning. Should this concept be validated, it could pave the way to promising research and therapeutic perspectives given that the trophoblastic properties are vital for the tumor while they are permanently epigenetically turned off in normal cells. Specifically targeting expression of the trophoblastic master genes could thereby be envisaged to jeopardize the tumor and its metastases without, in principle, inducing adverse side effects in the healthy tissues. Conclusion: A wide set of functional features of cancer tissue regulation, including some apparently paradoxical facts, was reviewed. Cancer cell misuse of physiological trophoblastic functions can clearly account for them, which identifies trophoblastic-like transdifferentiation as a likely key component of malignancy and makes it a potential relevant anticancer target.
Collapse
|
45
|
Einenkel R, Zygmunt M, Muzzio DO. Microorganisms in the healthy upper reproductive tract: from denial to beneficial assignments for reproductive biology. Reprod Biol 2019; 19:113-118. [PMID: 31023521 DOI: 10.1016/j.repbio.2019.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/22/2019] [Accepted: 04/12/2019] [Indexed: 12/25/2022]
Abstract
Contrary to the traditional assumption of a sterile uterus, the number of studies characterizing microbial entities in the healthy upper reproductive tract (endometrial cavity, including follicular fluid and placenta) have been on the increase. Substantial data has been accumulated correlating microbial composition with fertility outcome. In this context, the presence of certain taxa was associated to an improved reproductive success. A summarization for the evidence of these molecular mechanisms through which bacteria may affect developmental processes during pregnancy is presented and discussed with special focus placed upon the immunological aspects.
Collapse
Affiliation(s)
- Rebekka Einenkel
- Department of Obstetrics and Gynecology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany
| | - Marek Zygmunt
- Department of Obstetrics and Gynecology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany
| | - Damián Oscar Muzzio
- Department of Obstetrics and Gynecology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475, Greifswald, Germany.
| |
Collapse
|
46
|
IL-36 Cytokines: Regulators of Inflammatory Responses and Their Emerging Role in Immunology of Reproduction. Int J Mol Sci 2019; 20:ijms20071649. [PMID: 30987081 PMCID: PMC6479377 DOI: 10.3390/ijms20071649] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 03/28/2019] [Accepted: 03/28/2019] [Indexed: 01/01/2023] Open
Abstract
The IL-36 subfamily of cytokines has been recently described as part of the IL-1 superfamily. It comprises three pro-inflammatory agonists (IL-36α, IL-36β, and IL-36γ), their receptor (IL-36R), and one antagonist (IL-36Ra). Although expressed in a variety of cells, the biological relevance of IL-36 cytokines is most evident in the communication between epithelial cells, dendritic cells, and neutrophils, which constitute the common triad responsible for the initiation, maintenance, and expansion of inflammation. The immunological role of IL-36 cytokines was initially described in studies of psoriasis, but novel evidence demonstrates their involvement in further immune and inflammatory processes in physiological and pathological situations. Preliminary studies have reported a dynamic expression of IL-36 cytokines in the female reproductive tract throughout the menstrual cycle, as well as their association with the production of immune mediators and cellular recruitment in the vaginal microenvironment contributing to host defense. In pregnancy, alteration of the placental IL-36 axis has been reported upon infection and pre-eclampsia suggesting its pivotal role in the regulation of maternal immune responses. In this review, we summarize current knowledge regarding the regulatory mechanisms and biological actions of IL-36 cytokines, their participation in different inflammatory conditions, and the emerging data on their potential role in normal and complicated pregnancies.
Collapse
|
47
|
Yockey LJ, Iwasaki A. Interferons and Proinflammatory Cytokines in Pregnancy and Fetal Development. Immunity 2018; 49:397-412. [PMID: 30231982 PMCID: PMC6152841 DOI: 10.1016/j.immuni.2018.07.017] [Citation(s) in RCA: 346] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 05/13/2018] [Accepted: 07/26/2018] [Indexed: 12/15/2022]
Abstract
Successful pregnancy requires carefully-coordinated communications between the mother and fetus. Immune cells and cytokine signaling pathways participate as mediators of these communications to promote healthy pregnancy. At the same time, certain infections or inflammatory conditions in pregnant mothers cause severe disease and have detrimental impacts on the developing fetus. In this review, we examine evidence for the role of maternal and fetal immune responses affecting pregnancy and fetal development, both under homeostasis and following infection. We discuss immune responses that are necessary to promote healthy pregnancy and those that lead to congenital disorders and pregnancy complications, with a particular emphasis on the role of interferons and cytokines. Understanding the contributions of the immune system in pregnancy and fetal development provides important insights into the pathogenesis underlying maternal and fetal diseases and sheds insights on possible targets for therapy.
Collapse
Affiliation(s)
- Laura J Yockey
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
48
|
Hauk V, Vota D, Gallino L, Calo G, Paparini D, Merech F, Ochoa F, Zotta E, Ramhorst R, Waschek J, Leirós CP. Trophoblast VIP deficiency entails immune homeostasis loss and adverse pregnancy outcome in mice. FASEB J 2018; 33:1801-1810. [PMID: 30204500 DOI: 10.1096/fj.201800592rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Immune homeostasis maintenance throughout pregnancy is critical for normal fetal development. Trophoblast cells differentiate into an invasive phenotype and contribute to the transformation of maternal arteries and the functional shaping of decidual leukocyte populations. Insufficient trophoblast invasion, inadequate vascular remodeling, and a loss of immunologic homeostasis are associated with pregnancy complications, such as preeclampsia and intrauterine growth restriction. Vasoactive intestinal peptide (VIP) is a pleiotropic neuropeptide synthetized in trophoblasts at the maternal-placental interface. It regulates the function of trophoblast cells and their interaction with decidual leukocytes. By means of a murine model of pregnancy in normal maternal background with VIP-deficient trophoblast cells, here we demonstrate that trophoblast VIP is critical for trophoblast function: VIP gene haploinsufficiency results in lower matrix metalloproteinase 9 expression, and reduced migration and invasion capacities. A reduced number of regulatory T cells at the implantation sites along with a lower expression of proangiogenic and antiinflammatory markers were also observed. Findings detected in the implantation sites at early stages were followed by an abnormal placental structure and lower fetal weight. This effect was overcome by VIP treatment of the early pregnant mice. Our results support the relevance of trophoblast-synthesized VIP as a critical factor in vivo for trophoblast-cell function and immune homeostasis maintenance in mouse pregnancy.-Hauk, V., Vota, D., Gallino, L., Calo, G., Paparini, D., Merech, F., Ochoa, F., Zotta, E., Ramhorst, R., Waschek, J., Leirós, C. P. Trophoblast VIP deficiency entails immune homeostasis loss and adverse pregnancy outcome in mice.
Collapse
Affiliation(s)
- Vanesa Hauk
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)
| | - Daiana Vota
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)
| | - Lucila Gallino
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)
| | - Guillermina Calo
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)
| | - Daniel Paparini
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)
| | - Fátima Merech
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)
| | - Federico Ochoa
- Departamento de Ciencias Fisiológicas, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO-Houssay), Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires, Argentina
| | - Elsa Zotta
- Departamento de Ciencias Fisiológicas, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO-Houssay), Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires, Argentina.,Catedra de Fisiopatología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rosanna Ramhorst
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)
| | - James Waschek
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, The David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Claudia Pérez Leirós
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN)
| |
Collapse
|
49
|
Small for gestational age: Case definition & guidelines for data collection, analysis, and presentation of maternal immunisation safety data. Vaccine 2018; 35:6518-6528. [PMID: 29150057 PMCID: PMC5710996 DOI: 10.1016/j.vaccine.2017.01.040] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 01/13/2017] [Indexed: 12/13/2022]
|
50
|
Pavlov O, Selutin A, Pavlova O, Selkov S. Macrophages are a source of IL-17 in the human placenta. Am J Reprod Immunol 2018; 80:e13016. [DOI: 10.1111/aji.13016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/12/2018] [Indexed: 12/18/2022] Open
Affiliation(s)
- Oleg Pavlov
- Department of Immunology and Cell Interaction; D.O. Ott Research Institute for Obstetrics, Gynecology and Reproduction; St. Petersburg Russia
| | - Aleksandr Selutin
- Department of Immunology and Cell Interaction; D.O. Ott Research Institute for Obstetrics, Gynecology and Reproduction; St. Petersburg Russia
| | - Oksana Pavlova
- Department of Histology, Cytology and Embryology; Pavlov First Saint Petersburg State Medical University; St. Petersburg Russia
| | - Sergei Selkov
- Department of Immunology and Cell Interaction; D.O. Ott Research Institute for Obstetrics, Gynecology and Reproduction; St. Petersburg Russia
| |
Collapse
|