2,916 results on '"Placenta immunology"'
Search Results
2. Interferon regulatory factor 1 mediated inhibition of Treg cell differentiation induces maternal-fetal immune imbalance in preeclampsia.
- Author
-
Ma Y, Ye S, Liu Y, Zhao X, Wang Y, and Wang Y
- Subjects
- Humans, Female, Pregnancy, Adult, Forkhead Transcription Factors metabolism, Forkhead Transcription Factors genetics, Placenta immunology, Placenta metabolism, Th17 Cells immunology, Immune Tolerance, Cells, Cultured, Pre-Eclampsia immunology, Interferon Regulatory Factor-1 metabolism, Interferon Regulatory Factor-1 genetics, T-Lymphocytes, Regulatory immunology, Cell Differentiation, Trophoblasts immunology, Trophoblasts metabolism
- Abstract
The establishment and maintenance of a successful pregnancy rely heavily on maternal-fetal immune tolerance. Inflammatory and immune mechanisms during pregnancy bear a resemblance to those observed in tumor progression, with Treg cells exhibiting similar immunoregulatory functions in both contexts. Interferon regulatory factor 1 (IRF1) is implicated in modulating the immune milieu within tumors and influencing regulatory T (Treg) cell differentiation. However, the precise association between IRF1 and the onset of preeclampsia (PE) remains unclear. In our investigation, we identified trophoblasts as a significant source of IRF1 expression at the maternal-fetal interface through immunofluorescence analysis. Moreover, heightened levels of IRF1 expression were detected in both placental tissues and peripheral blood samples obtained from PE patients, concomitant with an imbalance in the Th17/Treg ratio. In the peripheral circulation, a notable inverse correlation was observed between IRF1 mRNA levels and Foxp3 mRNA, a transcription factor specific to Treg cells. IRF1 mRNA expression showed a positive association with systolic blood pressure and a negative association with serum albumin levels. Furthermore, co-culturing naïve T cells with supernatants from HTR-8/SV neo cells overexpressing IRF1 resulted in diminished differentiation of T cells into Treg cells. In summary, our study indicates elevated IRF1 expression in the peripheral blood and trophoblast cells of PE patients. Elevated IRF1 in trophoblast cells hinders the differentiation of maternal Treg cells, disrupting maternal-fetal immune tolerance and contributing to PE pathogenesis. Additionally, IRF1 expression correlates with disease severity, suggesting its potential as a novel sensitive target in PE., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. Regulators of placental antibody transfer through a modeling lens.
- Author
-
Wessel RE and Dolatshahi S
- Subjects
- Humans, Pregnancy, Female, Immunoglobulin G immunology, Infant, Newborn, Animals, Vaccines immunology, Models, Immunological, Vaccination, Placenta immunology, Placenta metabolism, Maternal-Fetal Exchange immunology, Immunity, Maternally-Acquired
- Abstract
Infants are vulnerable to infections owing to a limited ability to mount a humoral immune response and their tolerogenic immune phenotype, which has impeded the success of newborn vaccination. Transplacental transfer of IgG from mother to fetus provides crucial protection in the first weeks of life, and maternal immunization has recently been implemented as a public health strategy to protect newborns against serious infections. Despite their early success, current maternal vaccines do not provide comparable protection across pregnancies with varying gestational lengths and placental and maternal immune features, and they do not account for the dynamic interplay between the maternal immune response and placental transfer. Moreover, progress toward the rational design of maternal vaccines has been hindered by inadequacies of existing experimental models and safety challenges of investigating longitudinal dynamics of IgG transfer in pregnant humans. Alternatively, in silico mechanistic models are a logical framework to disentangle the processes regulating placental antibody transfer. This Review synthesizes current literature through a mechanistic modeling lens to identify placental and maternal regulators of antibody transfer, their clinical covariates, and knowledge gaps to guide future research. We also describe opportunities to use integrated modeling and experimental approaches toward the rational design of vaccines against existing and emerging neonatal pathogen threats., (© 2024. Springer Nature America, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
4. The rebalancing of the immune system at the maternal-fetal interface ameliorates autism-like behavior in adult offspring.
- Author
-
Shen C, Zhu X, Chang H, Li C, Hou M, Chen L, Lu Chen, Zhou Z, Ji M, and Xu Z
- Subjects
- Animals, Female, Pregnancy, Mice, Macrophages immunology, Macrophages metabolism, Poly I-C pharmacology, Mice, Inbred C57BL, Behavior, Animal, Prenatal Exposure Delayed Effects immunology, Autism Spectrum Disorder immunology, Immune System, Male, Maternal-Fetal Exchange immunology, T-Lymphocytes, Regulatory immunology, Autistic Disorder immunology, Placenta immunology, Placenta metabolism
- Abstract
Maternal immune activation (MIA) is critical for imparting neuropathology and altered behaviors in offspring; however, maternal-fetal immune cell populations have not been thoroughly investigated in MIA-induced autism spectrum disorders (ASDs). Here, we report the single-cell transcriptional landscape of placental cells in both PBS- and poly(I:C)-induced MIA dams. We observed a decrease in regulatory T (Treg) cells but an increase in the M1 macrophage population at the maternal-fetal interface in MIA dams. Based on the Treg-targeting approach, we investigate an immunoregulatory protein, the helminth-derived heat shock protein 90α (Sjp90α), that induces maternal Treg cells and subsequently rescues the autism-like behaviors in adult offspring. Furthermore, in vivo depletion of maternal macrophages attenuates placental inflammatory reaction and reverses behavioral abnormalities in adult offspring. Notably, Sjp90α induces CD4
+ T cell differentiation via scavenger receptor A (SR-A) on the macrophage in vitro. Our findings suggest a maternal Treg-targeted approach to alleviate MIA-induced autism-like behavior in adult offspring., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
5. Gestational diabetes exacerbates intrauterine microbial exposure induced intestinal microbiota change in offspring contributing to increased immune response.
- Author
-
Liu J, Chen Y, Laurent I, Yang P, Xiao X, and Li X
- Subjects
- Pregnancy, Animals, Female, Mice, Humans, Fetal Blood, Prenatal Exposure Delayed Effects immunology, Prenatal Exposure Delayed Effects microbiology, Lipopolysaccharides, Escherichia coli, RNA, Ribosomal, 16S, Lactobacillus, Male, Mice, Inbred C57BL, Animals, Newborn, Diabetes, Gestational microbiology, Diabetes, Gestational immunology, Gastrointestinal Microbiome, Placenta immunology, Placenta microbiology
- Abstract
Background: maternal health during pregnancy can affect the intestinal microbial community of offspring, but currently the impact of intrauterine environmental changes resulting from gestational diabetes mellitus (GDM) on the microbiota of offspring as well as its interaction with the immune system remains unclear., Aims: to explore the impact of intrauterine microbial exposure during pregnancy of gestational diabetes mellitus on the development of neonate's intestinal microbiota and activation of immune responses., Methods: Levels of lipopolysaccharides in cord blood from GDM and expression of microbial recognition-related proteins in the placenta were measured. To evaluate embryonic intestinal colonization, pregnant mice with GDM were administered with labeled Escherichia coli or Lactobacillus. The intestinal colonization of pups was analyzed through 16S rRNA gene sequencing and labeled microbial culture. Additionally, memory T lymphocyte and dendritic cell co-culture experiments were conducted to elucidate the immune memory of intestinal microbes during the embryonic stages., Result: Gestational diabetes mellitus led to elevated umbilical cord blood LPS level and increased GFP labeled Escherichia coli in the offspring's intestine after gestational microbial exposure. The mouse model of GDM exhibited increased immune markers including TLR4, TLR5, IL-22 and IL-23 in the placenta and a recall response from memory T cells in offspring's intestines, with similar observations found in human experiments. Furthermore, reduced intestinal microbiome diversity and an increased ratio of Firmicutes/Bacteroidetes was found in GDM progeny, with the stability of bacterial colonization been interfered., Conclusions: Our investigation has revealed a noteworthy correlation between gestational diabetes and intrauterine microbial exposure, as well as alterations in the neonatal microbiota and activation of immune responses. These findings highlight the gestational diabetes's role on offspring's gut microbiota and immune system interactions with early-life pathogen exposure., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. Scoping Review of Microbiota Dysbiosis and Risk of Preeclampsia.
- Author
-
Jordan MM, Amabebe E, Khanipov K, and Taylor BD
- Subjects
- Humans, Pregnancy, Female, Microbiota, Gastrointestinal Microbiome, Mouth microbiology, Pre-Eclampsia microbiology, Pre-Eclampsia immunology, Dysbiosis, Placenta microbiology, Placenta immunology, Vagina microbiology
- Abstract
Limited studies have investigated the role of the microbiota in hypertensive disorders of pregnancy (HDP), particularly preeclampsia, which often results in preterm birth. We evaluated 23 studies that explored the relationship between gut, vaginal, oral, or placental microbiotas and HDP. Scopus, ProQuest Health Research Premium Collection, ProQuest Nursing & Allied Health Database, EBSCO, and Ovid were searched for relevant literature. Majority (18) of studies focused on the gut microbiota, and far fewer examined the oral cavity (3), vagina (3), and placenta (1). One study examined the gut, oral, and vaginal microbiotas. The consensus highlights a potential role for microbiota dysbiosis in preeclampsia and HDP. Especially in the third trimester, preeclampsia is associated with gut dysbiosis-deficient in beneficial species of Akkermansia, Bifidobacterium, and Coprococcus but enriched with pathogenic Campylobacterota and Candidatus Saccharibacteria, with low community α-diversity. Similarly, the preeclamptic vaginal and oral microbiotas are enriched with bacterial vaginosis and periodontal disease-associated species, respectively. The trend is also observed in the placenta, which is colonized by gastrointestinal, respiratory tract, and periodontitis-related pathogens. Consequently, a chronic proinflammatory state that adversely impacts placentation is implicated. These observations however require more mechanistic studies to establish the timing of the preceding immune dysfunction and any causality., (© 2024 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
7. On placental and lactational transfer of IgG-based therapeutic proteins - Current understanding and knowledge gaps from a clinical pharmacology perspective.
- Author
-
Guinn D, Kratz K, Baisden K, Ridge S, McClymont S, Fletcher EP, Johnson T, and Wang YM
- Subjects
- Humans, Female, Pregnancy, Milk, Human immunology, Milk, Human chemistry, Milk, Human metabolism, Pharmacology, Clinical methods, Infant, Newborn, Infant, Immunoglobulin G immunology, Lactation, Placenta metabolism, Placenta immunology, Maternal-Fetal Exchange immunology
- Abstract
Maternal medication use may expose the developing fetus through placental transfer or the infant through lactational transfer. Because pregnant and lactating individuals have been historically excluded from early drug development trials, there is often limited to no human data available to inform pharmacokinetics (PK) and safety in these populations at the time of drug approval. We describe the known mechanisms of placental or lactational transfer of IgG-based therapeutic proteins and use clinical examples to highlight the potential for fetal or infant exposure during pregnancy and lactation. Placental transfer of IgG-based therapeutic proteins may result in systemic exposure to the developing fetus. A lactational transfer may be associated with local gastrointestinal (GI) exposure in the infant and may also result in systemic exposure, although data are very limited as proteins have shown instability in the GI tract. Understanding of PK and pharmacodynamic (PD) effects of IgG-based therapeutic proteins in infants exposed in utero as well as the potential exposure through human milk and its clinical implications is critical for developing treatment strategies for pregnant or lactating individuals. We share the current knowledge gaps and considerations for future evaluations to inform PK, PD, and the safety of IgG-based therapeutic proteins for safe use during pregnancy and lactation. With the increasing use of IgG-based therapeutic proteins in treating chronic diseases during pregnancy and lactation, there is a need to improve the quantity and quality of data to inform the safe use in pregnant and lactating individuals., (© 2024 The Author(s). Clinical and Translational Science published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.)
- Published
- 2024
- Full Text
- View/download PDF
8. Hypertension and Cognitive Dysfunction in a Pregnant Rat Model of PE; a Role for CD4+ T Cells.
- Author
-
Deer E, Herrock O, Simmons K, Campbell N, Amaral L, Zheng B, Morris R, Wallace K, Cleveland EH, Belk S, Dodd C, and LaMarca B
- Subjects
- Animals, Female, Pregnancy, Rats, Humans, Hypertension immunology, Adult, Placenta immunology, CD4-Positive T-Lymphocytes immunology, Pre-Eclampsia immunology, Cognitive Dysfunction immunology, COVID-19 immunology, COVID-19 complications, Disease Models, Animal, Rats, Nude, SARS-CoV-2 immunology
- Abstract
Objective: Preeclampsia (PE) is associated with hypertension (HTN) during pregnancy and activated CD4+ T cells, inflammatory cytokines, and autoantibodies to the angiotensin II type I receptor (AT1-AA). Having had COVID-19 (CV) during pregnancy is associated with an increased incidence of a PE-like phenotype. Both PE and CV have long-lasting neurological implications and studies show that nonpregnant COVID patients produce AT1-AA. We have shown that CD4+ T cells from PE women cause a PE phenotype in nude athymic rats. In this study, we sought to examine the role of CD4+ T cells from PE with a CV History (Hx) to contribute to a PE phenotype and to determine the importance of CD4+ T cells in cognitive dysfunction during pregnancy., Methods: At delivery, blood and placentas were collected, and one million placental CD4+ T cells from each PE and each normotensive patient, with (NT) or without (NP) a CV (Hx) during pregnancy, were isolated, purified, and injected i.p. into a gestational day (GD) 12 pregnant nude athymic rat (one patient/rat). At GD19, blood pressure (MAP) and circulating factors were assessed in recipient rats. Cognitive function and memory were assessed using Novel Object Recognition and Barnes Maze tests, respectively. Placental ACE-2 activity and AT1-AA were measured from COVID Hx patients. A one- or two-way ANOVA with Bonferroni's multiple comparisons test was used for statistical analysis., Results: Blood pressure was increased in patients with PE, with or without COVID, compared to NT patients. There were no significant changes in placental ACE activity in patients with COVID Hx with or without PE. AT1-AA was elevated in PE patients and in both PE and NT COVID Hx compared to control NP. In pregnant recipient rats, MAP increased in CV Hx PE (113 ± 2, n = 8) compared to CV Hx NT (101 ± 5, n = 6). PE and PE CV Hx CD4+ T Cell recipient rats exhibited impaired memory and cognitive dysfunction (p < 0.05), compared to control groups. Recipient rats of PE CV Hx CD4+ T cells had elevated AT1-AA compared to NT CV Hx recipients. Both COVID Hx groups and recipients of PE CD4+ T cells had elevated TNF alpha compared to NP., Conclusion: Our findings indicate that pregnant patients with a Hx of COVID during pregnancy produce AT1-AA, with or without PE. Recipients of CD4+ T cells from PE with or without a CV Hx during pregnancy cause HTN and elevated AT1-AA. TNF-α is elevated in PE and in CV Hx NT and PE recipients. Interestingly, recipients of T cells from PE patients with or without a Hx of CV had worse cognitive function during pregnancy, compared to recipient rats of NP CD4+ T cells. These data demonstrate the importance of CD4+ T cells in HTN and impaired neurological function during PE in the presence or absence of a prior COVID-19 infection during pregnancy., (© 2024 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
9. Quantitative Modeling to Characterize Maternal Inflammatory Response of Histologic Chorioamnionitis in Placental Membranes.
- Author
-
Chou T, Senkow KJ, Nguyen MB, Patel PV, Sandepudi K, Cooper LA, and Goldstein JA
- Subjects
- Female, Humans, Pregnancy, Neutrophils immunology, Neural Networks, Computer, Adult, Machine Learning, Chorioamnionitis immunology, Chorioamnionitis pathology, Placenta immunology, Placenta pathology, Inflammation immunology, Inflammation pathology
- Abstract
Problem: The placental membranes are a key barrier to fetal and uterine infection. Inflammation of the membranes, diagnosed as maternal inflammatory response (MIR) or alternatively as acute chorioamnionitis, is associated with adverse maternal-fetal outcomes. MIR is staged 1-3, with higher stages indicating more hazardous inflammation. However, the diagnosis relies upon subjective evaluation and has not been deeply characterized. The goal of this work is to develop a cell classifier for eight placental membrane cells and quantitatively characterize MIR1-2., Method of Study: Hematoxylin and eosin (H&E)-stained placental membrane slides were digitized. A convolutional neural network was trained on a dataset of hand-annotated and machine learning-identified cells. Overall cell class-level metrics were calculated. The model was applied to 20 control, 20 MIR1, and 23 MIR2 placental membrane cases. MIR cell composition and neutrophil distribution were assessed via density and Ripley's cross K-function. Clinical data were compared to neutrophil density and distribution., Results: The classification model achieved a test-set accuracy of 0.845, with high precision and recall for amniocytes, decidual cells, endothelial cells, and trophoblasts. Using this model to classify 53 073 cells from healthy and MIR1-2 placental membranes, we found that (1) MIR1-2 have higher neutrophil density and fewer decidual cells and trophoblasts, (2) Neutrophils colocalize heavily around decidual cells in healthy placental membranes and around trophoblasts in MIR1, (3) Neutrophil density impacts distribution in MIR, and (4) Neutrophil metrics correlate with features of clinical chorioamnionitis., Conclusions: This paper introduces cell classification into the placental membranes and quantifies cell composition and neutrophil spatial distributions in MIR., (© 2024 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
10. SARS-CoV-2 infection vs. vaccination during pregnancy: the placenta leads the way.
- Author
-
Pérez-Latorre L and Ramilo O
- Subjects
- Humans, Pregnancy, Female, Immunity, Maternally-Acquired, Antibodies, Viral immunology, Immunization, Passive methods, COVID-19 prevention & control, COVID-19 immunology, SARS-CoV-2 immunology, Pregnancy Complications, Infectious prevention & control, Pregnancy Complications, Infectious immunology, COVID-19 Vaccines immunology, COVID-19 Vaccines administration & dosage, Placenta virology, Placenta immunology, Vaccination methods
- Abstract
Purpose of Review: To understand the characteristics and determinants of transplacental antibody transfer against SARS-CoV-2 and to compare the differences between SARS-CoV-2 infection and vaccination., Recent Findings: The need for information during the COVID-19 pandemic and the exclusion of pregnant women from randomized clinical trials have led to a vast amount of clinical data primarily based on observational studies with diverse design and sample analyses that yield variable results. This review aims to critically and comprehensively integrate the relevant knowledge related to transplacental transfer of antibodies against SARS-CoV-2, emphasizing the differences between infection and vaccination., Summary: Passive immunization is key to conferring protection to the infant during their first months of life. Understanding the mechanisms of transplacental antibody transfer during SARS-CoV-2 infection and vaccination, and their associated protection will allow optimizing the implementation of well tolerated and effective preventive strategies for both pregnant women and infants., (Copyright © 2024 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
11. Integrated transcriptomic analysis and machine learning for characterizing diagnostic biomarkers and immune cell infiltration in fetal growth restriction.
- Author
-
Wei X, Liu Z, Cai L, Shi D, Sun Q, Zhang L, Zhou F, and Sun L
- Subjects
- Humans, Female, Pregnancy, Placenta metabolism, Placenta immunology, Placenta pathology, Nomograms, Adult, Fetal Growth Retardation genetics, Fetal Growth Retardation diagnosis, Fetal Growth Retardation immunology, Machine Learning, Gene Expression Profiling, Biomarkers, Transcriptome, Triggering Receptor Expressed on Myeloid Cells-1 genetics
- Abstract
Background: Fetal growth restriction (FGR) occurs in 10% of pregnancies worldwide. Placenta dysfunction, as one of the most common causes of FGR, is associated with various poor perinatal outcomes. The main objectives of this study were to screen potential diagnostic biomarkers for FGR and to evaluate the function of immune cell infiltration in the process of FGR., Methods: Firstly, differential expression genes (DEGs) were identified in two Gene Expression Omnibus (GEO) datasets, and gene set enrichment analysis was performed. Diagnosis-related key genes were identified by using three machine learning algorithms (least absolute shrinkage and selection operator, random forest, and support vector machine model), and the nomogram was then developed. The receiver operating characteristic curve, calibration curve, and decision curve analysis curve were used to verify the validity of the diagnostic model. Using cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT), the characteristics of immune cell infiltration in placental tissue of FGR were evaluated and the candidate key immune cells of FGR were screened. In addition, this study also validated the diagnostic efficacy of TREM1 in the real world and explored associations between TREM1 and various clinical features., Results: By overlapping the genes selected by three machine learning algorithms, four key genes were identified from 290 DEGs, and the diagnostic model based on the key genes showed good predictive performance (AUC = 0.971). The analysis of immune cell infiltration indicated that a variety of immune cells may be involved in the development of FGR, and nine candidate key immune cells of FGR were screened. Results from real-world data further validated TREM1 as an effective diagnostic biomarker (AUC = 0.894) and TREM1 expression was associated with increased uterine artery PI (UtA-PI) (p-value = 0.029)., Conclusion: Four candidate hub genes (SCD, SPINK1, TREM1, and HIST1H2BB) were identified, and the nomogram was constructed for FGR diagnosis. TREM1 was not only associated with a variety of key immune cells but also correlated with increased UtA-PI. The results of this study could provide some new clues for future research on the prediction and treatment of FGR., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Wei, Liu, Cai, Shi, Sun, Zhang, Zhou and Sun.)
- Published
- 2024
- Full Text
- View/download PDF
12. The role of aryl hydrocarbon receptors in nutrient metabolism and immune regulation at the maternal-fetal interface.
- Author
-
Li Y, Yu X, Shi J, Zhao J, and Li L
- Subjects
- Pregnancy, Female, Humans, Animals, Nutrients metabolism, Receptors, Aryl Hydrocarbon metabolism, Maternal-Fetal Exchange immunology, Placenta metabolism, Placenta immunology
- Abstract
The maternal-fetal interface is composed of the placenta, which is affiliated with the fetus, and the maternal decidua. During pregnancy, the placenta is mainly responsible for nutrient transport and immune tolerance maintenance, which plays a key role in fetal growth and development and pregnancy maintenance. The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that exists in various cell types at the maternal-fetal interface and is involved in multiple cellular processes. Recent studies have highlighted the role of AhR in regulating various physiological processes, including glucose and lipid metabolism, as well as tryptophan metabolism and immune responses, within non-pregnant tissues. This review shifts focus towards understanding how AhR modulation impacts metabolism and immune regulation at the maternal-fetal interface. This may implicate the development of pregnancy-related complications and the potential target of the AhR pathway for therapeutic strategies against poor pregnancy outcomes., Competing Interests: Declaration of competing interest We have no conflicts of interest to disclose., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. Fetal MAVS and type I IFN signaling pathways control ZIKV infection in the placenta and maternal decidua.
- Author
-
Alippe Y, Wang L, Coskun R, Muraro SP, Zhao FR, Elam-Noll M, White JM, Vota DM, Hauk VC, Gordon JI, Handley SA, and Diamond MS
- Subjects
- Female, Animals, Pregnancy, Mice, Mice, Inbred C57BL, Mice, Knockout, Immunity, Innate, Pregnancy Complications, Infectious immunology, Pregnancy Complications, Infectious virology, Disease Models, Animal, Interferon Type I metabolism, Interferon Type I immunology, Signal Transduction immunology, Adaptor Proteins, Signal Transducing metabolism, Adaptor Proteins, Signal Transducing genetics, Placenta immunology, Placenta virology, Placenta metabolism, Zika Virus Infection immunology, Zika Virus Infection virology, Zika Virus immunology, Zika Virus physiology, Decidua immunology, Decidua virology, Decidua metabolism, Fetus immunology, Fetus virology, Trophoblasts immunology, Trophoblasts virology, Trophoblasts metabolism, Receptor, Interferon alpha-beta genetics, Receptor, Interferon alpha-beta metabolism
- Abstract
The contribution of placental immune responses to congenital Zika virus (ZIKV) syndrome remains poorly understood. Here, we leveraged a mouse model of ZIKV infection to identify mechanisms of innate immune restriction exclusively in the fetal compartment of the placenta. ZIKV principally infected mononuclear trophoblasts in the junctional zone, which was limited by mitochondrial antiviral-signaling protein (MAVS) and type I interferon (IFN) signaling mechanisms. Single nuclear RNA sequencing revealed MAVS-dependent expression of IFN-stimulated genes (ISGs) in spongiotrophoblasts but not in other placental cells that use alternate pathways to induce ISGs. ZIKV infection of Ifnar1-/- or Mavs-/- placentas was associated with greater infection of the adjacent immunocompetent decidua, and heterozygous Mavs+/- or Ifnar1+/- dams carrying immunodeficient fetuses sustained greater maternal viremia and tissue infection than dams carrying wild-type fetuses. Thus, MAVS-IFN signaling in the fetus restricts ZIKV infection in junctional zone trophoblasts, which modulates dissemination and outcome for both the fetus and the pregnant mother., (© 2024 Alippe et al.)
- Published
- 2024
- Full Text
- View/download PDF
14. Additional breastfeeding data essential to better understand kinetics of A (H1N1) pdm09 strain-specific antibodies induced by placental transfer and natural infection in children from birth to 3 years of age.
- Author
-
Li Y
- Subjects
- Humans, Female, Infant, Pregnancy, Infant, Newborn, Child, Preschool, Placenta metabolism, Placenta immunology, Breast Feeding, Influenza, Human immunology, Influenza A Virus, H1N1 Subtype immunology, Antibodies, Viral blood, Antibodies, Viral immunology
- Abstract
Competing Interests: I declare no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
15. IGF2BP3 participates in the pathogenesis of recurrent spontaneous abortion by regulating ferroptosis.
- Author
-
Dai F, Zhang Y, Deng Z, Zhang J, Wang R, Chen J, Yang D, Mao S, Liu H, Cheng Y, and Hu M
- Subjects
- Humans, Female, Pregnancy, Animals, Mice, Cell Movement genetics, Cell Line, Placenta metabolism, Placenta pathology, Placenta immunology, Adult, Apoptosis immunology, Ferroptosis immunology, Abortion, Habitual metabolism, Abortion, Habitual pathology, Abortion, Habitual immunology, Trophoblasts metabolism, Trophoblasts pathology, RNA-Binding Proteins metabolism, RNA-Binding Proteins genetics
- Abstract
The aberrant invasive capability of trophoblast cells is widely acknowledged as a primary mechanism underlying RSA. Recently, IGF2BP3 has been implicated in various cancers due to its influence on cellular invasion and migration. However, whether IGF2BP3 involve in the occurrence of RSA and the specific functions it assumes in the development of RSA remain elusive. In our study, we firstly collected villous tissues from RSA and those with normal pregnancies individuals to performed Protein sequencing and then detected the expression of IGF2BP3 through Western blot, qRT-PCR and immunohistochemistry. Secondly, we analyzed the single-cell data (GSE214607) to assess the expression of IGF2BP3 in invasive EVT trophoblasts. Thirdly, we utilized lentivirus technology to establish HTR-8/SVneo cell lines with stable IGF2BP3 knockdown and RNA-seq analysis was employed to investigate the GO functional pathway enrichment of IGF2BP3. Meanwhile, the effect of IGF2BP3 knockdown on trophoblast cells apoptosis, migration, and ferroptosis was evaluated through functional experiments. Additionally, LPS-induced abortion animal model was constructed to evaluate IGF2BP3 expression in placental tissues. A significant downregulation of IGF2BP3 was observed in the villous tissues of RSA patient, a finding corroborated by subsequent single cell sequencing results. Furthermore, it suggested that IGF2BP3 may be involved in the migration and apoptotic processes of trophoblast cells. Mechanistic research indicated that IGF2BP3 knockdown could compromise GPX4 mRNA stability, leading to the promotion of ferroptosis. Finally, our investigation observed the down-regulation of IGF2BP3 expression in placental villous tissues of an LPS-induced abortion animal model. Our findings revealed that IGF2BP3 was downregulated in the villous tissues of RSA patients. Mechanically, down-regulation of IGF2BP3 may induce RSA by promoting GPX4-mediated ferroptosis and inhibiting trophoblast invasion and migration. Our study may provide new targets and research directions for the pathogenesis of RSA., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
16. Shoutai Wan treatment upregulates the expression of TNFAIP3 and improves T cell immune tolerance at maternal-fetal interface.
- Author
-
Du L, Pan D, Huang H, Liu Q, Yang Y, and Jiang G
- Subjects
- Animals, Female, Pregnancy, Mice, Male, Disease Models, Animal, Signal Transduction drug effects, Signal Transduction immunology, Placenta immunology, Placenta drug effects, Placenta metabolism, Up-Regulation drug effects, Humans, Maternal-Fetal Exchange immunology, Tumor Necrosis Factor alpha-Induced Protein 3 metabolism, Tumor Necrosis Factor alpha-Induced Protein 3 genetics, Immune Tolerance drug effects, Mice, Inbred DBA, Mice, Inbred CBA, Drugs, Chinese Herbal pharmacology, NF-kappa B metabolism
- Abstract
Shoutai Wan (STW) is a traditional Chinese medicine formula used to treat various conditions. The objective of this study was to evaluate the impact of STW on the abortion rate in the URSA mouse model and elucidate its underlying molecular mechanisms. Female CBA/J mice were mated with male DBA/2 mice to establish the URSA model. Network pharmacological analysis was employed to investigate the potential molecular mechanisms of STW. Hematoxylin-eosin staining, immunofluorescence, and ELISA were performed to examine placental microenvironmental changes, protein expression related to TNFAIP3 and the NF-κB signaling pathway. Treatment with STW reduced the abortion rate in URSA model mice and improved trophoblast development. TNFAIP3 was identified as a potential target of STW for treating URSA, as STW enhanced TNFAIP3 protein expression while decreasing IL-6 and TNF-α secretion in the placenta. Moreover, STW upregulated TNFAIP3 protein expression and Foxp3 mRNA levels, increased the production of anti-inflammatory cytokines such as IL-10 and TGF-β1, and decreased p-NF-κB expression in CD4+ cells at the placenta. The findings of this study indicate that STW treatment reduces the abortion rate in the URSA mouse model. These effects are likely mediated by increased TNFAIP3 expression and decreased NF-κB signaling pathway activity at the maternal-fetal interface. These molecular changes may contribute to the regulation of T cell immunity and immune tolerance during pregnancy., Competing Interests: Declaration of Competing Interest The authors declare that they have no competing interests., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
17. The influence of the oral microbiota in full-term pregnant women on immune regulation during pregnancy.
- Author
-
Yang P, Ma G, Lu T, Zhou J, Fan H, Zhang X, Fan S, and Xiao X
- Subjects
- Humans, Female, Pregnancy, Adult, Mouth microbiology, Mouth immunology, Interleukin-5 immunology, Interleukin-5 metabolism, Microbiota immunology, Placenta immunology, Placenta microbiology
- Abstract
Background: This study aims to conduct a preliminary exploration of the correlation between the oral microbiota of full-term pregnant women and both local placental immunity and the systemic immune system of the mother., Methods: A total of 26 pregnant women participated in this study, with samples collected from oral swabs, placental tissue, and peripheral venous blood. High-throughput sequencing was used to examine the oral microbial community. Flow cytometry was employed to assess immune cells in placental tissue and peripheral venous blood. ELISA and Luminex liquid bead chip technology were utilized to detect cytokines in both placental tissue and peripheral venous blood., Results: In placental tissue, The oral microbial community is primarily negatively correlated with placental CD3+CD4+CD8+T cells and positively correlated with placental IL-5. In the peripheral blood, The oral microbial community is primarily positively correlated with maternal systemic immune parameters, including CD3+CD4+ T cells and the CD4+/CD8+ ratio, as well as positively correlated with peripheral IL-18., Conclusions: The oral microbiota of full-term pregnant women participates in the regulatory function of the maternal immune system. Meanwhile, the oral microbial community may also be an important factor mediating local immune regulation in the placenta., Competing Interests: Declaration of Competing Interest The authors declare that there is no conflict of interest regarding the publication of this paper., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
18. Antiviral and Immunomodulatory Effects of Interferon Lambda at the Maternal-Fetal Interface.
- Author
-
Dedloff MR and Lazear HM
- Subjects
- Humans, Pregnancy, Female, Animals, Pregnancy Complications, Infectious immunology, Pregnancy Complications, Infectious virology, Interferon Lambda, Maternal-Fetal Exchange immunology, Decidua immunology, Virus Diseases immunology, Virus Diseases virology, Endometrium immunology, Endometrium virology, Antiviral Agents, Interleukins immunology, Interleukins metabolism, Interferons immunology, Placenta immunology, Placenta virology
- Abstract
Interferon lambda (IFN-λ, type III IFN, IL-28/29) is a family of antiviral cytokines that are especially important at barrier sites, including the maternal-fetal interface. Recent discoveries have identified important roles for IFN-λ during pregnancy, particularly in the context of congenital infections. Here, we provide a comprehensive review of the activity of IFN-λ at the maternal-fetal interface, highlighting cell types that produce and respond to IFN-λ in the placenta, decidua, and endometrium. Further, we discuss the role of IFN-λ during infections with congenital pathogens including Zika virus, human cytomegalovirus, rubella virus, and Listeria monocytogenes . We discuss advances in experimental models that can be used to fill important knowledge gaps about IFN-λ-mediated immunity.
- Published
- 2024
- Full Text
- View/download PDF
19. Progestogen-driven B7-H4 contributes to onco-fetal immune tolerance.
- Author
-
Yu J, Yan Y, Li S, Xu Y, Parolia A, Rizvi S, Wang W, Zhai Y, Xiao R, Li X, Liao P, Zhou J, Okla K, Lin H, Lin X, Grove S, Wei S, Vatan L, Hu J, Szumilo J, Kotarski J, Freeman ZT, Skala S, Wicha M, Cho KR, Chinnaiyan AM, Schon S, Wen F, Kryczek I, Wang S, Chen L, and Zou W
- Subjects
- Animals, Female, Humans, Mice, Pregnancy, Breast Neoplasms immunology, Breast Neoplasms genetics, Breast Neoplasms metabolism, Receptors, Progesterone metabolism, Transcription Factors metabolism, Cell Line, Tumor, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Mice, Inbred C57BL, Placenta metabolism, Placenta immunology, V-Set Domain-Containing T-Cell Activation Inhibitor 1 metabolism, Immune Tolerance, Progestins pharmacology, Progestins metabolism, Progesterone metabolism
- Abstract
Immune tolerance mechanisms are shared in cancer and pregnancy. Through cross-analyzing single-cell RNA-sequencing data from multiple human cancer types and the maternal-fetal interface, we found B7-H4 (VTCN1) is an onco-fetal immune tolerance checkpoint. We showed that genetic deficiency of B7-H4 resulted in immune activation and fetal resorption in allogeneic pregnancy models. Analogously, B7-H4 contributed to MPA/DMBA-induced breast cancer progression, accompanied by CD8
+ T cell exhaustion. Female hormone screening revealed that progesterone stimulated B7-H4 expression in placental and breast cancer cells. Mechanistically, progesterone receptor (PR) bound to a newly identified -58 kb enhancer, thereby mediating B7-H4 transcription via the PR-P300-BRD4 axis. PR antagonist or BRD4 degrader potentiated immunotherapy in a murine B7-H4+ breast cancer model. Thus, our work unravels a mechanistic and biological connection of a female sex hormone (progesterone) to onco-fetal immune tolerance via B7-H4 and suggests that the PR-P300-BRD4 axis is targetable for treating B7-H4+ cancer., Competing Interests: Declaration of interests W.Z. has served as a scientific advisor or consultant for Cstone, NextCure, and Hanchorbio. S. Wang is a co-founder and paid consultant of Ascentage Pharma Group International and owns stock in Ascentage. L.C. has been a scientific founder, consultant, and/or board observer for NextCure, Normunity, Tayu, Zai Lab, Tpioneer, Vcanbio, OncoC4, and GenomiCare and has sponsored research funds from NextCure, Normunity, and DynamiCure. This research is conducted independently and has not received resources from and is unrelated to the scientific and commercial pursuits of these industrial entities, including NextCure., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
20. In Situ Analyses of Placental Inflammatory Response to SARS-CoV-2 Infection in Cases of Mother-Fetus Vertical Transmission.
- Author
-
Morotti D, Tabano S, Gaudioso G, Radaelli T, Croci GA, Bianchi N, Ghirardi G, Gianatti A, Patanè L, Poletti de Chaurand V, Schwartz DA, Hagazi MAAA, and Grizzi F
- Subjects
- Adult, Female, Humans, Infant, Newborn, Pregnancy, Cytokines metabolism, Cytokines genetics, Gene Expression Profiling, Inflammation genetics, Inflammation immunology, Inflammation virology, Transcriptome, COVID-19 immunology, COVID-19 transmission, COVID-19 virology, Infectious Disease Transmission, Vertical, Placenta immunology, Placenta metabolism, Placenta virology, Pregnancy Complications, Infectious immunology, Pregnancy Complications, Infectious virology, SARS-CoV-2 immunology
- Abstract
It has been shown that vertical transmission of the SARS-CoV-2 strain is relatively rare, and there is still limited information on the specific impact of maternal SARS-CoV-2 infection on vertical transmission. The current study focuses on a transcriptomics analysis aimed at examining differences in gene expression between placentas from mother-newborn pairs affected by COVID-19 and those from unaffected controls. Additionally, it investigates the in situ expression of molecules involved in placental inflammation. The Papa Giovanni XXIII Hospital in Bergamo, Italy, has recorded three instances of intrauterine transmission of SARS-CoV-2. The first two cases occurred early in the pandemic and involved pregnant women in their third trimester who were diagnosed with SARS-CoV-2. The third case involved an asymptomatic woman in her second trimester with a twin pregnancy, who unfortunately delivered two stillborn fetuses due to the premature rupture of membranes. Transcriptomic analysis revealed significant differences in gene expression between the placentae of COVID-19-affected mother/newborn pairs and two matched controls. The infected and control placentae were matched for gestational age. According to the Benjamani-Hochberg method, 305 genes met the criterion of an adjusted p -value of less than 0.05, and 219 genes met the criterion of less than 0.01. Up-regulated genes involved in cell signaling (e.g., CCL20, C3, MARCO) and immune response (e.g., LILRA3, CXCL10, CD48, CD86, IL1RN, IL-18R1) suggest their potential role in the inflammatory response to SARS-CoV-2. RNAscope
® technology, coupled with image analysis, was utilized to quantify the surface area covered by SARS-CoV-2, ACE2, IL-1β, IL-6, IL-8, IL-10, and TNF-α on both the maternal and fetal sides of the placenta. A non-statistically significant gradient for SARS-CoV-2 was observed, with a higher surface coverage on the fetal side (2.42 ± 3.71%) compared to the maternal side (0.74 ± 1.19%) of the placenta. Although not statistically significant, the surface area covered by ACE2 mRNA was higher on the maternal side (0.02 ± 0.04%) compared to the fetal side (0.01 ± 0.01%) of the placenta. IL-6 and IL-8 were more prevalent on the fetal side (0.03 ± 0.04% and 0.06 ± 0.08%, respectively) compared to the maternal side (0.02 ± 0.01% and 0.02 ± 0.02%, respectively). The mean surface areas of IL-1β and IL-10 were found to be equal on both the fetal (0.04 ± 0.04% and 0.01 ± 0.01%, respectively) and maternal sides of the placenta (0.04 ± 0.05% and 0.01 ± 0.01%, respectively). The mean surface area of TNF-α was found to be equal on both the fetal and maternal sides of the placenta (0.02 ± 0.02% and 0.02 ± 0.02%, respectively). On the maternal side, ACE-2 and all examined interleukins, but not TNF-α, exhibited an inverse mRNA amount compared to SARS-CoV-2. On the fetal side, ACE-2, IL-6 and IL-8 were inversely correlated with SARS-CoV-2 (r = -0.3, r = -0.1 and r = -0.4, respectively), while IL-1β and IL-10 showed positive correlations (r = 0.9, p = 0.005 and r = 0.5, respectively). TNF-α exhibited a positive correlation with SARS-CoV-2 on both maternal (r = 0.4) and fetal sides (r = 0.9) of the placenta. Further research is needed to evaluate the correlation between cell signaling and immune response genes in the placenta and the vertical transmission of SARS-CoV-2. Nonetheless, the current study extends our comprehension of the molecular and immunological factors involved in SARS-CoV-2 placental infection underlying maternal-fetal transmission.- Published
- 2024
- Full Text
- View/download PDF
21. Virtual crossmatching reveals upregulation of placental HLA-Class II in chronic histiocytic intervillositis.
- Author
-
Brady CA, Ford LB, Moss C, Zou Z, Crocker IP, and Heazell AEP
- Subjects
- Humans, Female, Pregnancy, Adult, Placenta pathology, Placenta metabolism, Placenta immunology, Up-Regulation, Placenta Diseases pathology, Placenta Diseases immunology, Placenta Diseases metabolism, Chorionic Villi metabolism, Chorionic Villi pathology, Chorionic Villi immunology, Trophoblasts metabolism, Trophoblasts pathology, Trophoblasts immunology, Chronic Disease, Histocompatibility Antigens Class II metabolism
- Abstract
Chronic histiocytic intervillositis (CHI) is a recurrent placental lesion where maternal macrophages infiltrate the intervillous space. Its cause is unknown, though due to similarities to rejected allografts one hypothesis is that CHI represents maternal-fetal rejection. Here, virtual crossmatching was applied to healthy pregnancies and those with a history of CHI. Anti-HLA antibodies, measured by Luminex, were present in slightly more controls than CHI (8/17 (47.1%) vs 5/14 (35.7%)), but there was no significant difference in levels of sensitisation or fetal specific antibodies. Quantification of immunohistochemical staining for HLA-Class II was increased in syncytiotrophoblast of placentas with CHI (Grade 0.44 [IQR 0.1-0.7]) compared to healthy controls (0.06 [IQR 0-0.2]) and subsequent pregnancies (0.13 [IQR 0-0.3]) (P = 0.0004). HLA-Class II expression was positively related both to the severity of CHI (r = 0.67) and C4d deposition (r = 0.48). There was no difference in overall C4d and HLA-Class I immunostaining. Though increased anti-HLA antibodies were not evident in CHI, increased expression of HLA-Class II at the maternal-fetal interface suggests that they may be relevant in its pathogenesis. Further investigation of antibodies immediately after diagnosis is warranted in a larger cohort of CHI cases to better understand the role of HLA in its pathophysiology., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
22. Placental Pathologic Features and Perinatal Outcomes in Pregnant Woman With Autoimmune Connective Tissue Disease.
- Author
-
Zheng A, Zheng Y, Li D, Li X, Tong X, and Wang F
- Subjects
- Humans, Female, Pregnancy, Adult, Antiphospholipid Syndrome pathology, Antiphospholipid Syndrome immunology, Infant, Newborn, Connective Tissue Diseases pathology, Connective Tissue Diseases immunology, Premature Birth, Undifferentiated Connective Tissue Diseases immunology, Undifferentiated Connective Tissue Diseases pathology, Cesarean Section, Placenta pathology, Placenta immunology, Pregnancy Outcome, Pregnancy Complications immunology, Lupus Erythematosus, Systemic pathology
- Abstract
Introduction: We aimed to investigate the association between perinatal outcomes and placental pathological features in pregnant women with ACTD, including systemic lupus erythematosus (SLE), antiphospholipid antibody syndrome (APS), and undifferentiated connective tissue disease (UCTD)., Materials and Methods: Placental tissue from SLE (n = 44), APS (n = 45), and UCTD (n = 45) were included, and contemporaneous deliveries of placenta were served as a control group (n = 46) between September 2015 and March 2021. The placental histopathology was evaluated using the Manual of Human Placental Pathology and classified according to the Amsterdam consensus framework., Results: SLE pregnant women have a higher rate of cesarean section (61.40%), premature birth (24.56%), and SGA (26.32%) when compared to control group (p = 0.008, p = 0.005, and p = 0.000, respectively). The rate of vascular malperfusion, inflammatory-immune lesions, and other placental lesions in the SLE group was 47.73%, 56.82%, and 63.64%, which were higher than the control group (p = 0.000, p = 0.000, and p = 0.006, respectively). In the meantime, the incidence of inflammatory-immune lesions in the APS group (42.22%, p = 0.004) and vascular malperfusion in the UCTD group (37.78%, p = 0.007) were increased when compared to the control group., Conclusions: SLE appeared to confer increased risk for a wide range of adverse perinatal outcomes. We determined elevated placental histopathology risk for most women with ACTD, including vascular maldevelopment, vascular malperfusion, and inflammatory-immune lesions., (© 2024 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
23. Unraveling the role of sulfiredoxin-1 in early-onset preeclampsia: A key player in trophoblast ferroptosis.
- Author
-
Wei J, Qiu D, Yang X, Wang J, Shi M, Sun L, Lu X, Wang C, Liu H, and Li R
- Subjects
- Female, Pregnancy, Humans, Animals, Mice, Placenta metabolism, Placenta pathology, Placenta immunology, Adult, Disease Models, Animal, Pre-Eclampsia immunology, Pre-Eclampsia pathology, Pre-Eclampsia metabolism, Ferroptosis immunology, Trophoblasts metabolism, Trophoblasts pathology, Oxidoreductases Acting on Sulfur Group Donors metabolism
- Abstract
Preeclampsia (PE) significantly contributes to obstetric complications and maternal mortality, yet its pathogenesis and mechanisms are not well understood. Sulfiredoxin-1 (SRXN1) is known for its antioxidant activity and its role in defending against oxidative stress; it is also linked to various cancers. However, the role of SRXN1 in PE remains unclear. Our study found a significant decrease in SRXN1 levels in the serum and placental tissues of patients with early-onset preeclampsia (EOPE). Similarly, a PE-like mouse model showed reduced SRXN1 expression. Our in vitro experiments showed that reducing SRXN1 impaired trophoblast viability, decreased invasion and migration, and led to cell death, primarily through ferroptosis. These results are consistent with analyses of placental tissues from EOPE patients. In summary, lower SRXN1 levels during pregnancy contribute to trophoblast ferroptosis, potentially affecting the development and progression of EOPE., Competing Interests: Declaration of Competing Interest All authors reviewed and critically edited the manuscript.The authors declare no competing interests., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
24. Altered placental macrophage numbers and subsets in pregnancies complicated with intrahepatic cholestasis of pregnancy (ICP) compared to healthy pregnancies.
- Author
-
Brenøe JE, van Hoorn EGM, Beck L, Bulthuis M, Bezemer RE, Gordijn SJ, Schoots MH, and Prins JR
- Subjects
- Humans, Female, Pregnancy, Adult, Case-Control Studies, Ursodeoxycholic Acid therapeutic use, Cholestasis, Intrahepatic pathology, Cholestasis, Intrahepatic metabolism, Cholestasis, Intrahepatic blood, Cholestasis, Intrahepatic immunology, Pregnancy Complications pathology, Pregnancy Complications immunology, Placenta pathology, Placenta metabolism, Placenta immunology, Macrophages immunology, Macrophages pathology, Macrophages metabolism
- Abstract
Introduction: Intrahepatic cholestasis of pregnancy (ICP) can result in adverse outcomes for both mother and fetus. Inflammatory (M1 subset) or anti-inflammatory (M2 subset) macrophage polarisation is associated with various complications of pregnancy. However, the influence of ICP on macrophage numbers and polarisation remains unknown. This study analyses macrophage density and distribution in placentas of patients with ICP compared to controls. Clinical parameters were correlated to macrophage distribution and ursodeoxycholic acid use (UDCA)., Methods: This study included routinely collected placental tissue samples of 42 women diagnosed with ICP and of 50 control pregnancies. Immunohistochemical staining was performed on placental tissue using CD68 antibody as a pan-macrophage marker, CD206 antibody as an M2 and HLA-DR antibody as an M1 macrophage marker. Macrophage density (cells/mm
2 ) and distribution (CD206+ /CD68+ or CD206+ /CD68+ HLA-DR+ ) in both decidua (maternal tissue) and villous parenchyma (fetal tissue) were compared between groups. Macrophage density and distribution were correlated to clinical parameters for ICP patients., Results: The density of CD68+ macrophages differed significantly between groups in villous parenchyma. In both decidua and villous parenchyma, CD206+ /CD68+ ratio was significantly lower in ICP patients compared to controls (p = 0.003 and p=<0.001, respectively). No difference was found based on UDCA use or in CD68+ HLA-DR+ cell density. Significant correlations were found between macrophage density and peak serum bile acids and liver enzymes., Discussion: In ICP patients, an immune shift was observed in both decidual and villous tissue, indicated by a lower CD206+ /CD68+ ratio. ICP seems to affect placental tissue, however more research is required to understand its consequences., Competing Interests: Declaration of competing interest None., (Copyright © 2024 The Authors. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
25. Effects of Maternal SARS-CoV-2 Infection During Pregnancy on Fetal Development.
- Author
-
Li J, Yao J, and Yang Z
- Subjects
- Humans, Pregnancy, Female, Premature Birth immunology, Placenta virology, Placenta immunology, Pregnancy Outcome, COVID-19 immunology, COVID-19 transmission, Pregnancy Complications, Infectious immunology, Pregnancy Complications, Infectious virology, SARS-CoV-2 immunology, Fetal Development immunology, Infectious Disease Transmission, Vertical
- Abstract
Background: The severe acute respiratory syndrome coronavirus (SARS-CoV-2) outbreak in 2019 has necessitated investigating its potential adverse effects on pregnancy outcomes and fetal development., Objective: This study aimed to review the evidence on the impact of SARS-CoV-2 infection during pregnancy on fetal outcomes., Method of Study: Literatures since the outbreak of COVID-19 from PubMed and Web of Science were summarized in this narrative review, to show the effects of maternal SARS-CoV-2 infection during pregnancy on fetal development., Results: SARS-CoV-2 infection during pregnancy can be transmitted vertically through the placenta, both in utero and perinatally, affecting the maternal-fetal immune interface and placental function. Viral infections during pregnancy have been linked to central nervous system development impairments and disorders such as autism. Changes in the structure and function of the respiratory, immune, and visceral systems have also been reported. SARS-CoV-2 infection during pregnancy has been linked with increased risks of stillbirth and preterm birth. However, the mechanisms involved remain unclear and may include cytokine storms, macrophage mediation, genetic mutations, methylation, and other epigenetic changes. Exploring the protective effects of antiviral treatment and other interventions in animal and clinical studies may help improve outcomes., Conclusion: SARS-CoV-2 infection during pregnancy activates the maternal-fetal immune interface through vertical transmission, and has short- and long-term effects on fetal development, including the central nervous system. Future long-term studies may help provide evidence that can inform interventions to reduce the risk of adverse outcomes., (© 2024 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
26. Exosomes-mediated transmission of standard bovine viral diarrhea strain OregonC24Va in bovine trophoblast cells.
- Author
-
Liang Y, Liu B, Xiao L, Ren S, Sheng X, Qi X, Zhang Z, Yuan N, Guo K, and Wang X
- Subjects
- Animals, Cattle, Female, Pregnancy, Placenta virology, Placenta immunology, Cells, Cultured, Virus Replication, Trophoblasts virology, Trophoblasts immunology, Exosomes metabolism, Exosomes virology, Bovine Virus Diarrhea-Mucosal Disease transmission, Bovine Virus Diarrhea-Mucosal Disease virology, Bovine Virus Diarrhea-Mucosal Disease immunology, Diarrhea Viruses, Bovine Viral physiology, Diarrhea Viruses, Bovine Viral immunology, Autophagy
- Abstract
Bovine viral diarrhoea virus (BVDV) can infect cows on days 30-110 of gestation and crossing the placental barrier, resulting in persistently infected (PI) and causing significant economic losses to dairy farming. Bovine placental trophoblast cells (BTCs) are the major cells in the early chorionic tissue of the placenta and play important roles in placental resistance to viral transmission. In this study, we have confirmed that BTCs is among a groups of cell types those could be infected by BVDV in vivo, and BVDV infection stimulates the autophagic responses in BTCs and promotes the release of exosomes. Meanwhile, the exosomes derived from BTCs can be used by BVDV to spread between placental trophoblast cells, and this mode of transmission cannot be blocked by antibodies against the BVDV E2 protein, whereas the replication and spread of BVDV in BTCs can be blocked by inhibiting autophagy and exosomogenesis. Our study provides a theoretical and practical basis for scientific prediction and intervention of reproductive disorders caused by BVDV infection in cows of different gestation periods from a novel perspective., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
27. Immunological mechanisms in preeclampsia: A narrative review.
- Author
-
Javandoust Gharehbagh F, Soltani-Zangbar MS, and Yousefzadeh Y
- Subjects
- Humans, Pregnancy, Female, Immune Tolerance, Cytokines metabolism, Cytokines immunology, Animals, Inflammation immunology, Pre-Eclampsia immunology, Placenta immunology
- Abstract
Maternal immunologic mechanisms for tolerance are essential for a successful pregnancy because they prevent maladaptive immune responses to the placenta and semi-allogeneic fetus and promote fetal growth. Preeclampsia is a major global cause of fetal mortality and morbidity. It is characterized by new-onset hypertension and proteinuria that occurs at twenty weeks of pregnancy or later. Preeclampsia is defined by a rise in cytokines that are pro-inflammatory and antiangiogenic components in the fetoplacental unit and the vascular endothelium of pregnant women, as well as an excessive and increasing stimulation of the immune system. Crucially, inflammation can result in low birth weight and inadequate placental perfusion in neonates. Preeclampsia, which is ultimately connected to inflammatory responses, can be impacted by several immunological mechanisms. Our goal in this work was to compile the most recent research on the pathoimmunology of preeclampsia, including studies on angiogenic variables and, in particular, immunological components., Competing Interests: Declaration of Competing Interest The authors declare no conflict of interest., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
28. The protective effects of Ferrostatin-1 against inflammation-induced preterm birth and fetal brain injury.
- Author
-
Chen C, Zhu S, Fu T, Chen Y, and Chen D
- Subjects
- Animals, Female, Pregnancy, Mice, Humans, Inflammation immunology, Inflammation drug therapy, Disease Models, Animal, Mice, Inbred C57BL, Premature Birth immunology, Premature Birth prevention & control, Cyclohexylamines pharmacology, Cyclohexylamines administration & dosage, Placenta pathology, Placenta immunology, Placenta drug effects, Brain Injuries prevention & control, Brain Injuries immunology, Brain Injuries etiology, Brain Injuries drug therapy, Brain Injuries pathology, Phenylenediamines pharmacology, Phenylenediamines administration & dosage, Lipopolysaccharides immunology, Ferroptosis drug effects
- Abstract
Introduction: Recent studies have suggested the involvement of ferroptosis in preterm birth. Despite compelling evidence, the underlying mechanism remains unknown. This investigation aimed to determine the therapeutic effects of Ferrostatin-1 (Fer-1), an inhibitor of ferroptosis, in preterm birth and fetal brain injury., Methods: Human placenta samples and clinical data of participants were collected to ascertain whether placental ferroptosis was associated with preterm birth. Lipopolysaccharide (LPS)-induced preterm birth mouse model was used to examine the protective effects of Fer-1 on preterm birth. Fetal brain tissues and offspring mice at 5 and 8 weeks were studied to determine the effects of Fer-1 on the cognitive function of offspring., Results: We examined the mechanism of spontaneous preterm birth and discovered that placental ferroptosis was associated with preterm birth. Fer-1 inhibited preterm birth by ameliorating placental ferroptosis and maternal inflammation, thus improving LPS-induced intrauterine inflammation to maintain pregnancy. Antenatal administration of Fer-1 prevented LPS-induced fetal brain damage in the acute phase and improved long-term neurodevelopmental impairments by improving placental neuroendocrine signaling and maintaining placental function., Conclusion: Fer-1 inhibited preterm birth and fetal brain injury by inhibiting maternal inflammation and improving placental function. Our findings provide a novel therapeutic strategy for preterm birth., Competing Interests: Declaration of Competing Interest We wish to confirm that there are no known conflicts of interest associated with this publication and there has been no significant financial support for this work that could have influenced its outcome., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
29. From Womb to World: Exploring the Immunological Connections between Mother and Child.
- Author
-
Cherayil BJ and Jain N
- Subjects
- Humans, Pregnancy, Female, Infant, Newborn, Placenta immunology, Immune System immunology, Pregnancy Complications, Infectious immunology, Maternal-Fetal Exchange immunology, Mothers, Child, Fetus immunology, Prenatal Exposure Delayed Effects immunology, Vaccination
- Abstract
Mother and child are immunologically interconnected by mechanisms that we are only beginning to understand. During pregnancy, multiple molecular and cellular factors of maternal origin are transferred across the placenta and influence the development and function of the fetal and newborn immune system. Altered maternal immune states arising from pregnancy-associated infections or immunizations have the potential to program offspring immune function in ways that may have long-term health consequences. In this study, we review current literature on the impact of prenatal infection and vaccination on the developing immune system, highlight knowledge gaps, and look to the horizon to envision maternal interventions that could benefit both the mother and her child., (Copyright © 2024 The Authors.)
- Published
- 2024
- Full Text
- View/download PDF
30. The expression of TIM-3 and Gal-9 on macrophages and Hofbauer cells in the placenta of preeclampsia patients.
- Author
-
Mittelberger J, Seefried M, Löb S, Kuhn C, Franitza M, Garrido F, Ditsch N, Jeschke U, and Dannecker C
- Subjects
- Humans, Pregnancy, Female, Adult, Antigens, CD metabolism, Antigens, Differentiation, Myelomonocytic metabolism, Receptors, Cell Surface metabolism, Chorionic Villi immunology, Chorionic Villi metabolism, Chorionic Villi pathology, Male, Hepatitis A Virus Cellular Receptor 2 metabolism, Pre-Eclampsia immunology, Pre-Eclampsia metabolism, Pre-Eclampsia pathology, Macrophages immunology, Macrophages metabolism, Galectins metabolism, Placenta immunology, Placenta metabolism, Placenta pathology
- Abstract
Preeclampsia is a disorder of pregnancy characterized by endothelial dysfunction, abnormal placentation, systemic inflammation, and altered immune reaction. The aim of this study was to investigate the immune checkpoint molecules TIM-3 and Gal-9 on macrophages and Hofbauer cells (HBC) in the placenta of preeclampsia patients. Immunohistochemistry and Immunofluorescence was used to characterize the expression of the macrophage markers CD68 and CD163, CK7 and the proteins TIM-3 and Gal-9 in the placentas of preeclampsia patients comparing it to the placentas of healthy pregnancies. Double immunofluorescence staining (TIM-3 with CD3/CD19/CD56) was used to analyze the TIM-3 expression on other immune cells (T cells, B cells, NK cells) within the chorionic villi. The expression of TIM-3 on decidual macrophages did not significantly differ between the preeclamptic and the control group (p = 0.487). When looking at the different offspring we saw an upregulation of TIM-3 expression on decidual macrophages in preeclamptic placentas with female offspring (p = 0.049). On Hofbauer cells within the chorionic villi, the TIM-3 expression was significantly downregulated in preeclamptic cases without a sex-specific difference (p < 0.001). Looking at the protein Gal-9 the expression was proven to be downregulated both, on decidual macrophages (p = 0.003) and on Hofbauer cells (p = 0.002) within preeclamptic placentas compared to healthy controls. This was only significant in male offspring (p < 0.001 and p = 0.013) but not in female offspring (p = 0.360 and p = 0.068). While TIM-3 expression within the extravillious trophoblast and the syncytiotrophoblast was significantly downregulated (p < 0.001 and p = 0.012) in preeclampsia, the expression of Gal-9 was upregulated in (p < 0.001 and p < 0.001) compared to healthy controls. The local variations of the immune checkpoint molecules TIM-3 and Gal-9 in the placenta may contribute to the inflammation observed in preeclamptic patients. It could therefore contribute to the pathogenesis and be an important target in the treatment of preeclampsia., Competing Interests: Declaration of Competing Interest N.D. reports funding from MSD, Novartis, Pfizer, Roche, AstraZeneca, TEVA, Mentor, and MCI Healthcare. U.J. received travel money from pfm. C.D. is funded by Roche, AstraZeneca, TEVA, Mentor, and MCI Healthcare. All other authors declare no conflict of interest., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
31. IL-33 Signaling Inhibition Leads to a Preeclampsia-Like Phenotype in Pregnant Rats.
- Author
-
Wang X, Shields CA, Thompson D, McKay J, Wilson R, Robbins MK, Glenn H, Fontenot M, Williams JM, and Cornelius DC
- Subjects
- Female, Pregnancy, Animals, Rats, Rats, Sprague-Dawley, Th17 Cells immunology, Disease Models, Animal, T-Lymphocytes, Regulatory immunology, Humans, Oxidative Stress, Placenta immunology, Placenta metabolism, Blood Pressure, Interleukin-1 Receptor-Like 1 Protein metabolism, Interleukin-33 metabolism, Pre-Eclampsia immunology, Signal Transduction
- Abstract
Problem: Preeclampsia (PE) is a hypertensive pregnancy disorder that is a leading cause of maternal and fetal morbidity and mortality characterized by maternal vascular dysfunction, oxidative stress, chronic immune activation, and excessive inflammation. No cure exists beyond delivery of the fetal-placental unit and the mechanisms driving pathophysiology are not fully understood. However, aberrant immune responses have been extensively characterized in clinical studies and shown to mediate PE pathophysiology in animal studies. One pathway that may mediate aberrant immune responses in PE is deficiencies in the IL-33 signaling pathway. In this study, we aim to investigate the impact of IL-33 signaling inhibition on cNK, T
H 17, and TReg populations, vascular function, and maternal blood pressure during pregnancy., Method of Study: In this study, IL-33 signaling was inhibited using two different methods: intraperitoneal administration of recombinant ST2 (which acts as a decoy receptor for IL-33) and administration of a specific IL-33 neutralizing antibody. Maternal blood pressure, uterine artery resistance index, renal and placental oxidative stress, cNK, TH 17, and TReg populations, various cytokines, and pre-proendothelin-1 levels were measured., Results: IL-33 signaling inhibition increased maternal blood pressure, uterine artery resistance, placental and renal oxidative stress. IL-33 signaling inhibition also increased placental cNK and TH 17 and renal TH 17 cells while decreasing placental TReg populations. IL-33 neutralization increased circulating cNK and TH 17s and decreased circulating TReg s in addition to increasing pre-proendothelin-1 levels., Conclusions: Data presented in this study demonstrate a role for IL-33 signaling in controlling vascular function and maternal blood pressure during pregnancy possibly by mediating innate and adaptive immune inflammatory responses, identifying the IL-33 signaling pathway as a potential therapeutic target for managing preeclampsia., (© 2024 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)- Published
- 2024
- Full Text
- View/download PDF
32. Activated and Naïve Allogenic Human Placental Mesenchymal Stromal Cells Exert an Immunomodulatory Effect on Hidradenitis Suppurativa Patient Peripheral Blood Mononuclear Cells.
- Author
-
Jariene V, Valiukevicius P, Maciulaitis J, Kuzaityte U, Insodaite R, Ciapiene I, Maciulaitis R, and Valiukeviciene S
- Subjects
- Humans, Female, Pregnancy, Adult, Immunomodulation, Cytokines metabolism, Cells, Cultured, Coculture Techniques, Cell Proliferation drug effects, Leukocytes, Mononuclear metabolism, Leukocytes, Mononuclear immunology, Mesenchymal Stem Cells immunology, Mesenchymal Stem Cells metabolism, Placenta immunology, Placenta cytology, Placenta metabolism
- Abstract
This pilot study aimed to evaluate the immunomodulatory effect of placental mesenchymal stem/stromal cells (MSCs) on peripheral blood mononuclear cells (PBMCs) from patients with hidradenitis suppurativa (HS). Blood samples were collected from 3 healthy and 3 patients with HS. Isolated PBMCs were stained with carboxyfluorescein succinimidyl ester (CFSE) and stimulated with phorbol 12-myristate 13-acetate (PMA)/Ionomycin solution. The PBMCs of patients with HS were co-cultured with naïve MSCs (n-MSCs), activated with tumor necrosis factor (TNF)-α (10 ng/mL) and interferon (IFN)-γ (10 ng/mL) MSCs (a-MSCs), or adalimumab (30 μg/mL). The division index (proliferation inhibition) of PBMCs was analyzed by flow cytometry using the Proliferation Modeling tool after 5 days of coculture. The relative inflammatory gene expression dynamics and cytokine secretion were quantified in triplicate using real-time polymerase chain reaction (PCR) and Luminex assays. PBMCs from the HS control group showed statistically significant increases in interleukin (IL)-6 and IFN-γ cytokine concentrations and IL-17A gene expression when compared with healthy subjects. Statistically significant reduction of the division index was found in the a-MSCs group ( P = 0.04). Also, the Luminex assay revealed significantly reduced proinflammatory cytokine concentrations of IL-9 ( P = 0.022) and IL-17A ( P = 0.022) in the a-MSCs group with the same trend of numerical lowering in n-MSCs group when compared to HS control. The results of real-time PCR revealed a numerical increase in the expression of the IL-1β, IL-36α , and TNF-α genes in both the a-MSCs and n-MSCs groups compared with the HS control. In conclusion, our findings suggest that MSCs can effectively curb PBMCs proliferation and suppress the production of inflammatory cytokines. Moreover, the preactivation of MSCs with IFN-γ and TNF-α before use can enhance their therapeutic effectiveness. Nevertheless, a larger sample size is imperative to validate these results.
- Published
- 2024
- Full Text
- View/download PDF
33. Protective role of macrophages from maternal-fetal interface in unvaccinated coronavirus disease 2019 pregnant women.
- Author
-
Gay L, Madariaga Zarza S, Abou Atmeh P, Rouvière MS, Andrieu J, Richaud M, Boumaza A, Miquel L, Diallo AB, Bechah Y, Otmani Idrissi M, La Scola B, Olive D, Resseguier N, Bretelle F, Mezouar S, and Mege JL
- Subjects
- Humans, Female, Pregnancy, Adult, Antigens, CD immunology, Antigens, Differentiation, Myelomonocytic, Receptors, Cell Surface immunology, Receptors, Cell Surface metabolism, Virus Internalization, COVID-19 immunology, COVID-19 virology, Placenta immunology, Placenta virology, Macrophages immunology, Macrophages virology, Pregnancy Complications, Infectious virology, Pregnancy Complications, Infectious immunology, SARS-CoV-2 immunology
- Abstract
Pregnant women represent a high-risk population for Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) infection. The presence of SARS-CoV-2 has been reported in placenta from infected pregnant women, but whether the virus influences placenta immune response remains unclear. We investigated the properties of maternal-fetal interface macrophages (MFMs) in a cohort of unvaccinated women who contracted coronavirus disease 2019 (COVID-19) during their pregnancy. We reported an infiltration of CD163
+ macrophages in placenta from COVID-19 women 19 whereas lymphoid compartment was not affected. Isolated MFMs exhibited nonpolarized activated signature (NOS2, IDO1, IFNG, TNF, TGFB) mainly in women infected during the second trimester of pregnancy. COVID-19 during pregnancy primed MFM to produce type I and III interferon response to SARS-CoV-2 (Wuhan and δ strains), that were unable to elicit this in MFMs from healthy pregnant women. COVID-19 also primed SARS-CoV-2 internalization by MFM in an angiotensin-converting enzyme 2-dependent manner. Activation and recall responses of MFMs were influenced by fetal sex. Collectively, these findings support a role for MFMs in the local immune response to SARS-CoV-2 infection, provide a basis for protective placental immunity in COVID-19, and highlight the interest of vaccination in pregnant women., (© 2024 The Author(s). Journal of Medical Virology published by Wiley Periodicals LLC.)- Published
- 2024
- Full Text
- View/download PDF
34. Decidual macrophages and Hofbauer cells in fetal growth restriction.
- Author
-
Bezemer RE, Faas MM, van Goor H, Gordijn SJ, and Prins JR
- Subjects
- Pregnancy, Humans, Female, Animals, Fetal Growth Retardation immunology, Macrophages immunology, Decidua immunology, Placenta immunology
- 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., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Bezemer, Faas, van Goor, Gordijn and Prins.)
- Published
- 2024
- Full Text
- View/download PDF
35. Gravidity influences distinct transcriptional profiles of maternal and fetal placental macrophages at term.
- Author
-
Ozarslan N, Robinson JF, Buarpung S, Kim MY, Ansbro MR, Akram J, Montoya DJ, Kamya MR, Kakuru A, Dorsey G, Rosenthal PJ, Cheng G, Feeney ME, Fisher SJ, and Gaw SL
- Subjects
- Female, Pregnancy, Humans, Gene Expression Profiling, Fetus immunology, Adult, Monocytes immunology, Monocytes metabolism, Macrophages immunology, Macrophages metabolism, Placenta immunology, Placenta metabolism, Transcriptome
- Abstract
Introduction: Maternal intervillous monocytes (MIMs) and fetal Hofbauer cells (HBCs) are myeloid-derived immune cells at the maternal-fetal interface. Maternal reproductive history is associated with differential risk of pregnancy complications. The molecular phenotypes and roles of these distinct monocyte/macrophage populations and the influence of gravidity on these phenotypes has not been systematically investigated., Methods: Here, we used RNA sequencing to study the transcriptional profiles of MIMs and HBCs in normal term pregnancies., Results: Our analyses revealed distinct transcriptomes of MIMs and HBCs. Genes involved in differentiation and cell organization pathways were more highly expressed in MIMs vs. HBCs. In contrast, HBCs had higher expression of genes involved in inflammatory responses and cell surface receptor signaling. Maternal gravidity influenced monocyte programming, as expression of pro-inflammatory molecules was significantly higher in MIMs from multigravidae compared to primigravidae. In HBCs, multigravidae displayed enrichment of gene pathways involved in cell-cell signaling and differentiation., Discussion: Our results demonstrated that MIMs and HBCs have highly divergent transcriptional signatures, reflecting their distinct origins, locations, functions, and roles in inflammatory responses. Furthermore, maternal gravidity influences the gene signatures of MIMs and HBCs, potentially modulating the interplay between tolerance and trained immunity. The phenomenon of reproductive immune memory may play a novel role in the differential susceptibility of primigravidae to pregnancy complications., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Ozarslan, Robinson, Buarpung, Kim, Ansbro, Akram, Montoya, Kamya, Kakuru, Dorsey, Rosenthal, Cheng, Feeney, Fisher and Gaw.)
- Published
- 2024
- Full Text
- View/download PDF
36. Emerging role of C5aR2: novel insights into the regulation of uterine immune cells during pregnancy.
- Author
-
Froehlich F, Landerholm K, Neeb J, Meß AK, Seiler DL, Tilburgs T, and Karsten CM
- Subjects
- Female, Animals, Pregnancy, Mice, Humans, Placenta immunology, Placenta metabolism, Complement C5a immunology, Complement C5a metabolism, Mice, Inbred C57BL, Interferon-gamma metabolism, Interferon-gamma immunology, Receptor, Anaphylatoxin C5a genetics, Receptor, Anaphylatoxin C5a metabolism, Uterus immunology, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, Mice, Knockout
- Abstract
Pregnancy is a fascinating immunological phenomenon because it allows allogeneic fetal and placental tissues to survive inside the mother. As a component of innate immunity with high inflammatory potential, the complement system must be tightly regulated during pregnancy. Dysregulation of the complement system plays a role in pregnancy complications including pre-eclampsia and intrauterine growth restriction. Complement components are also used as biomarkers for pregnancy complications. However, the mechanisms of detrimental role of complement in pregnancy is poorly understood. C5a is the most potent anaphylatoxin and generates multiple immune reactions via two transmembrane receptors, C5aR1 and C5aR2. C5aR1 is pro-inflammatory, but the role of C5aR2 remains largely elusive. Interestingly, murine NK cells have been shown to express C5aR2 without the usual co-expression of C5aR1. Furthermore, C5aR2 appears to regulate IFN-γ production by NK cells in vitro . As IFN-γ produced by uterine NK cells is one of the major factors for the successful development of a vital pregnancy, we investigated the role anaphylatoxin C5a and its receptors in the establishment of pregnancy and the regulation of uterine NK cells by examinations of murine C 5ar2
-/- pregnancies and human placental samples. C 5ar2-/- mice have significantly reduced numbers of implantation sites and a maternal C5aR2 deficiency results in increased IL-12, IL-18 and IFN-γ mRNA expression as well as reduced uNK cell infiltration at the maternal-fetal interface. Human decidual leukocytes have similar C5a receptor expression patterns showing clinical relevance. In conclusion, this study identifies C5aR2 as a key contributor to dNK infiltration and pregnancy success., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Froehlich, Landerholm, Neeb, Meß, Seiler, Tilburgs and Karsten.)- Published
- 2024
- Full Text
- View/download PDF
37. Endocytosis at the maternal-fetal interface: balancing nutrient transport and pathogen defense.
- Author
-
Fan M, Wu H, Sferruzzi-Perri AN, Wang YL, and Shao X
- Subjects
- Humans, Pregnancy, Female, Animals, Biological Transport, Nutrients metabolism, Immune Tolerance, Placenta immunology, Placenta metabolism, Endocytosis immunology, Maternal-Fetal Exchange immunology
- Abstract
Endocytosis represents a category of regulated active transport mechanisms. These encompass clathrin-dependent and -independent mechanisms, as well as fluid phase micropinocytosis and macropinocytosis, each demonstrating varying degrees of specificity and capacity. Collectively, these mechanisms facilitate the internalization of cargo into cellular vesicles. Pregnancy is one such physiological state during which endocytosis may play critical roles. A successful pregnancy necessitates ongoing communication between maternal and fetal cells at the maternal-fetal interface to ensure immunologic tolerance for the semi-allogenic fetus whilst providing adequate protection against infection from pathogens, such as viruses and bacteria. It also requires transport of nutrients across the maternal-fetal interface, but restriction of potentially harmful chemicals and drugs to allow fetal development. In this context, trogocytosis, a specific form of endocytosis, plays a crucial role in immunological tolerance and infection prevention. Endocytosis is also thought to play a significant role in nutrient and toxin handling at the maternal-fetal interface, though its mechanisms remain less understood. A comprehensive understanding of endocytosis and its mechanisms not only enhances our knowledge of maternal-fetal interactions but is also essential for identifying the pathogenesis of pregnancy pathologies and providing new avenues for therapeutic intervention., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Fan, Wu, Sferruzzi-Perri, Wang and Shao.)
- Published
- 2024
- Full Text
- View/download PDF
38. Pathogenesis of viral infections during pregnancy.
- Author
-
Creisher PS and Klein SL
- Subjects
- Pregnancy, Female, Humans, Animals, Placenta virology, Placenta immunology, Infectious Disease Transmission, Vertical, Disease Models, Animal, Pregnancy Complications, Infectious immunology, Pregnancy Complications, Infectious virology, Virus Diseases immunology, Virus Diseases transmission
- Abstract
SUMMARYViral infections during pregnancy are associated with significant adverse perinatal and fetal outcomes. Pregnancy is a unique immunologic and physiologic state, which can influence control of virus replication, severity of disease, and vertical transmission. The placenta is the organ of the maternal-fetal interface and provides defense against microbial infection while supporting the semi-allogeneic fetus via tolerogenic immune responses. Some viruses, such as cytomegalovirus, Zika virus, and rubella virus, can breach these defenses, directly infecting the fetus and having long-lasting consequences. Even without direct placental infection, other viruses, including respiratory viruses like influenza viruses and severe acute respiratory syndrome coronavirus 2, still cause placental damage and inflammation. Concentrations of progesterone and estrogens rise during pregnancy and contribute to immunological adaptations, placentation, and placental development and play a pivotal role in creating a tolerogenic environment at the maternal-fetal interface. Animal models, including mice, nonhuman primates, rabbits, and guinea pigs, are instrumental for mechanistic insights into the pathogenesis of viral infections during pregnancy and identification of targetable treatments to improve health outcomes of pregnant individuals and offspring., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
39. IL-1 and TNF mediates IL-6 signaling at the maternal-fetal interface during intrauterine inflammation.
- Author
-
Presicce P, Roland C, Senthamaraikannan P, Cappelletti M, Hammons M, Miller LA, Jobe AH, Chougnet CA, DeFranco E, and Kallapur SG
- Subjects
- Female, Pregnancy, Humans, Animals, Chorioamnionitis immunology, Chorioamnionitis metabolism, Chorioamnionitis veterinary, Lipopolysaccharides immunology, Interleukin-1 metabolism, Adult, Obstetric Labor, Premature immunology, Obstetric Labor, Premature metabolism, Inflammation immunology, Inflammation metabolism, Interleukin 1 Receptor Antagonist Protein metabolism, Interleukin 1 Receptor Antagonist Protein pharmacology, Placenta metabolism, Placenta immunology, Interleukin-6 metabolism, Signal Transduction, Macaca mulatta, Tumor Necrosis Factor-alpha metabolism
- Abstract
Introduction: IL6 signaling plays an important role in triggering labor and IL6 is an established biomarker of intrauterine infection/inflammation (IUI) driven preterm labor (PTL). The biology of IL6 during IUI at the maternal-fetal interface was investigated in samples from human subjects and non-human primates (NHP)., Methods: Pregnant women with histologic chorioamnionitis diagnosed by placenta histology were recruited (n=28 term, n=43 for preterm pregnancies from 26-36 completed weeks of gestation). IUI was induced in Rhesus macaque by intraamniotic injection of lipopolysachharide (LPS, n=23). IL1 signaling was blocked using Anakinra (human IL-1 receptor antagonist, n=13), and Tumor necrosis factor (TNF) signaling was blocked by anti TNF-antibody (Adalimumab n=14). The blockers were given before LPS. All animals including controls (intraamniotic injection of saline n=27), were delivered 16h after LPS/saline exposure at about 80% gestation., Results: IUI induced a robust expression of IL6 mRNAs in the fetal membranes (chorion-amnion-decidua tissue) both in humans (term and preterm) and NHP. The major sources of IL6 mRNA expression were the amnion mesenchymal cells (AMC) and decidua stroma cells. Additionally, during IUI in the NHP, ADAM17 (a protease that cleaves membrane bound IL6 receptor (IL6R) to release a soluble form) and IL6R mRNA increased in the fetal membranes, and the ratio of IL6 and soluble forms of IL6R, gp130 increased in the amniotic fluid signifying upregulation of IL6 trans-signaling. Both IL1 and TNF blockade suppressed LPS-induced IL6 mRNAs in the AMC and variably decreased elements of IL6 trans-signaling., Discussion: These data suggest that IL1 and TNF blockers may be useful anti-inflammatory agents via suppression of IL6 signaling at the maternal-fetal interface., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Presicce, Roland, Senthamaraikannan, Cappelletti, Hammons, Miller, Jobe, Chougnet, DeFranco and Kallapur.)
- Published
- 2024
- Full Text
- View/download PDF
40. Unveiling immune tolerance pathways in preeclampsia placenta: implications for molecular targets and discovery of potential biomarkers.
- Author
-
Ma Y, Deng X, Shen R, Zhang H, and Qian Y
- Subjects
- Humans, Pregnancy, Female, Gene Expression Profiling, Computational Biology methods, Transcriptome, Adult, Pre-Eclampsia immunology, Pre-Eclampsia genetics, Placenta metabolism, Placenta immunology, Immune Tolerance, Biomarkers metabolism
- Abstract
During pregnancy, there is a link between disruption of maternal immune tolerance and preeclampsia, but the molecular mechanisms that regulate maternal and fetal immune tolerance remain unclear. This study employs bioinformatics to identify new markers related to placental immune tolerance and explore their potential role in predicting preeclampsia. Analyzing preeclampsia-related gene expression profiles in the Gene Expression Omnibus (GEO) dataset reveals 211 differentially expressed genes (DEGs) in the placenta, mainly influencing immune cell differentiation and response pathways. Employing weighted gene co-expression network analysis (WGCNA) and lasso regression, four potential target genes (ANKRD37, CRH, LEP, SIGLEC6) are identified for potential prediction of preeclampsia. Validation using the GSE4707 dataset confirmed the diagnostic and predictive potential of these candidate genes. RT-qPCR verified up-regulation in the placenta, while ELISA showed their correlation with immune tolerance factors associated with placental immune tolerance. As a result of this study, identifies potential biomarkers associated with placental immunity and contributes to understanding the molecular mechanism of preeclampsia., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Ma, Deng, Shen, Zhang and Qian.)
- Published
- 2024
- Full Text
- View/download PDF
41. Pulmonary maternal immune activation does not cross the placenta but leads to fetal metabolic adaptation.
- Author
-
Hansen SSK, Krautz R, Rago D, Havelund J, Stigliani A, Færgeman NJ, Prézelin A, Rivière J, Couturier-Tarrade A, Akimov V, Blagoev B, Elfving B, Neess D, Vogel U, Khodosevich K, Hougaard KS, and Sandelin A
- Subjects
- Female, Pregnancy, Animals, Docosahexaenoic Acids metabolism, Suppressor of Cytokine Signaling 3 Protein metabolism, Suppressor of Cytokine Signaling 3 Protein genetics, Mice, Inflammation immunology, Inflammation metabolism, Mice, Inbred C57BL, Adaptation, Physiological immunology, Fetal Development immunology, Maternal-Fetal Exchange immunology, Interleukin-6 metabolism, Interleukin-6 immunology, Placenta metabolism, Placenta immunology, Fetus immunology, Fetus metabolism, Lung immunology, Lung metabolism, Lipopolysaccharides, Liver metabolism, Liver immunology
- Abstract
The fetal development of organs and functions is vulnerable to perturbation by maternal inflammation which may increase susceptibility to disorders after birth. Because it is not well understood how the placenta and fetus respond to acute lung- inflammation, we characterize the response to maternal pulmonary lipopolysaccharide exposure across 24 h in maternal and fetal organs using multi-omics, imaging and integrative analyses. Unlike maternal organs, which mount strong inflammatory immune responses, the placenta upregulates immuno-modulatory genes, in particular the IL-6 signaling suppressor Socs3. Similarly, we observe no immune response in the fetal liver, which instead displays metabolic changes, including increases in lipids containing docosahexaenoic acid, crucial for fetal brain development. The maternal liver and plasma display similar metabolic alterations, potentially increasing bioavailability of docosahexaenoic acid for the mother and fetus. Thus, our integrated temporal analysis shows that systemic inflammation in the mother leads to a metabolic perturbation in the fetus., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
42. Angiogenic factors and the lectin pathway of complement in women with secondary recurrent pregnancy loss.
- Author
-
Krog MC, Flachs EM, Kolte AM, de Jager W, Meyaard L, Christiansen OB, Steffensen R, Vomstein K, Garred P, and Nielsen HS
- Subjects
- Humans, Female, Pregnancy, Adult, Vascular Endothelial Growth Factor A metabolism, Vascular Endothelial Growth Factor A blood, Angiopoietin-2 metabolism, Angiopoietin-2 immunology, Angiopoietin-2 blood, C-Reactive Protein metabolism, C-Reactive Protein analysis, Angiopoietin-1 blood, Angiopoietin-1 metabolism, Serum Amyloid P-Component metabolism, Ficolins, Cohort Studies, Placenta immunology, Placenta metabolism, Placenta pathology, Pregnancy Outcome, Angiogenesis Inducing Agents metabolism, Complement Activation immunology, Abortion, Habitual immunology, Abortion, Habitual blood, Complement Pathway, Mannose-Binding Lectin immunology, Lectins metabolism, Lectins blood, Lectins immunology, Mannose-Binding Lectin blood, Mannose-Binding Lectin metabolism, Vascular Endothelial Growth Factor Receptor-1 metabolism, Vascular Endothelial Growth Factor Receptor-1 blood
- Abstract
The poor remodeling of placental spiral arteries seen in preeclampsia is also discussed to contribute to recurrent pregnancy loss (RPL) preceded by abnormal angiogenesis and excessive complement activation. Low levels of Mannose-binding-lectin (MBL), a pattern recognition molecule (PRM) of the lectin pathway, have been found in women with RPL. We propose that pregnancy loss is connected to defective angiogenesis with reperfusion damage in the placenta and decreased levels of PRM in the lectin pathway in women with RPL. In this cohort study, we investigate the angiogenic factors and the lectin complement pathway in early pregnancy and their time-dependent relationship with pregnancy outcomes in 76 women with secondary RPL (sRPL) who have at least four prior pregnancy losses and a live birth. We evaluated levels of Angiopoietin-1 (Ang-1), Angiopoietin-2 (Ang-2), Vascular Endothelial Growth Factor (VEGF), soluble fms-like tyrosine kinase-1 (sFlt-1), and the PRMs, MBL, ficolin-1, -2, -3 and an additional soluble PRM, Pentraxin-3, during the 5th, 6th, and 7th gestational weeks. Our results showed that, compared to live births, pregnancies that ended in loss were associated with elevated VEGF levels and decreased levels of the Ang-2/Ang-1 ratio. Also, increasing levels of ficolin-2 were significantly associated with pregnancy loss, with MBL showing no association. Our research suggests that women with sRPL may have inadequate placentation with impaired angiogenesis in pregnancies ending in a loss., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
43. Differential proteomics of circulating extracellular vesicles of placental origin isolated from women with early-onset preeclampsia reveal aberrant innate immune and hemostasis processes.
- Author
-
Rao A, Subedi R, Kundu I, Idicula-Thomas S, Shinde U, Bansal V, Balsarkar G, Mayadeo N, Das DK, Balasinor N, and Madan T
- Subjects
- Humans, Female, Pregnancy, Adult, Biomarkers metabolism, Extracellular Vesicles metabolism, Extracellular Vesicles immunology, Pre-Eclampsia immunology, Pre-Eclampsia metabolism, Immunity, Innate, Placenta metabolism, Placenta immunology, Proteomics, Hemostasis
- Abstract
Problem: Early-onset preeclampsia (EOPE) is a severe gestational hypertensive disorder with significant feto-maternal morbidity and mortality due to uteroplacental insufficiency. Circulating extracellular vesicles of placental origin (EV-P) are known to be involved in the pathophysiology of EOPE and might serve as an ideal reservoir for its specific biomarkers. Therefore, we aimed to characterize and perform comparative proteomics of circulating EV-P from healthy pregnant and EOPE women before delivery., Method of Study: The EV-P from both groups were isolated using immunoaffinity and were characterized using transmission electron microscopy, dynamic light scattering, nanoparticle tracking analysis, and immunoblotting. Following IgG albumin depletion, the pooled proteins that were isolated from EV-P of both groups were subjected to quantitative TMT proteomics., Results: Circulating term EV-P isolated from both groups revealed ∼150 nm spherical vesicles containing CD9 and CD63 along with placental PLAP and HLA-G proteins. Additionally, the concentration of EOPE-derived EV-P was significantly increased. A total of 208 proteins were identified, with 26 among them being differentially abundant in EV-P of EOPE women. This study linked the pathophysiology of EOPE to 19 known and seven novel proteins associated with innate immune responses such as complement and TLR signaling along with hemostasis and oxygen homeostasis., Conclusion: The theory suggesting circulating EVs of placental origin could mimic molecular information from the parent organ-"the placenta"-is strengthened by this study. The findings pave the way for possible discovery of novel prognostic and predictive biomarkers as well as provide insight into the mechanisms driving the pathogenesis of EOPE., (© 2024 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
44. Pyroptosis is involved in the immune microenvironment regulation of unexplained recurrent miscarriage.
- Author
-
Wang J, Nuray U, Yan H, Xu Y, Fang L, Li R, Zhou X, and Zhang H
- Subjects
- Humans, Female, Pregnancy, Gene Expression Profiling, Gene Regulatory Networks, Gene Ontology, Placenta metabolism, Placenta immunology, Transcriptome, Cellular Microenvironment genetics, Cellular Microenvironment immunology, Gene Expression Regulation, Pyroptosis genetics, Abortion, Habitual genetics, Abortion, Habitual immunology
- Abstract
Unexplained recurrent miscarriage (URM) is a common pregnancy complication with few effective therapies. Moreover, little is known regarding the role of pyroptosis in the regulation of the URM immune microenvironment. To address this issue, gene expression profiles of publicly available placental datasets GSE22490 and GSE76862 were downloaded from the Gene Expression Omnibus database. Pyroptosis-related differentially expressed genes were identified and a total of 16 differentially expressed genes associated with pyroptosis were detected, among which 1 was upregulated and 15 were downregulated. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses indicated that the functionally enriched modules and pathways of these genes are closely related to immune and inflammatory responses. Four hub genes were identified: BTK, TLR8, NLRC4, and TNFSF13B. BTK, TLR8, and TNFSF13B were highly connected with immune cells, according to the correlation analysis of four hub genes and 20 different types of immune cells (p < 0.05). The four hub genes were used as research objects to construct the interaction networks. Chorionic villus tissue was used for quantitative real-time polymerase chain reaction and western blot to confirm the expression levels of hub genes, and the results showed that the expression of the four hub genes was significantly decreased in the chorionic villus tissue in the URM group. Collectively, the present study indicates that perhaps pyroptosis is essential to the diversity and complexity of the URM immune microenvironment, and provides a theoretical basis and research ideas for subsequent target gene verification and mechanism research., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
45. The interplay between extracellular NAMPT and inflammatory cytokines in preeclampsia.
- Author
-
Nunes PR, Pereira DA, Passeti LFP, Coura LLF, Gomes KB, Sandrim VC, and Luizon MR
- Subjects
- Humans, Pregnancy, Female, Placenta immunology, Placenta metabolism, Inflammation Mediators metabolism, Inflammation Mediators blood, Inflammation immunology, Pre-Eclampsia immunology, Pre-Eclampsia blood, Nicotinamide Phosphoribosyltransferase blood, Nicotinamide Phosphoribosyltransferase metabolism, Cytokines blood, Cytokines metabolism, Biomarkers blood
- Abstract
Preeclampsia (PE) is the major cause of maternal-fetal mortality and morbidity. Its pathophysiology is not elucidated, but there is evidence for the role of visfatin/nicotinamide phosphoribosyl transferase (NAMPT), mainly due to its relation to endothelial dysfunction, a hallmark of PE. However, there is heterogeneous data regarding visfatin/NAMPT in healthy pregnancy (HP) and PE. Therefore, we performed a search on MEDLINE/PubMed using the terms "visfatin and preeclampsia" and "NAMPT and preeclampsia, and we selected 23 original articles: 12 articles reported increased levels in PE compared to HP, only four articles showed lower levels and eight articles did not find differences regarding visfatin/NAMPT in the groups studied. It is widely acknowledged that levels detected in plasma, serum, or placenta can be influenced by the size of the population and sample analyzed, as well as genetic factors. We further discussed the correlations of visfatin/NAMPT with clinical biomarkers in PE and inflammatory pathways. Considering the common inflammatory mechanisms between PE and visfatin/NAMPT, few studies have recently performed serum or plasma dosages. In conclusion, further studies are needed to highlight the potential role of visfatin/NAMPT in the pathophysiology of PE. This will provide comparative evidence to establish it as a biomarker for disease outcomes and treatment., Competing Interests: Declaration of Competing Interest The authors declare that no financial or non-financial interests are directly or indirectly related to the work submitted for peer review., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
46. Placental expression of inflammatory Galectin-12 is associated with gestational diabetes.
- Author
-
Buschmann C, Unverdorben L, Knabl J, Hutter S, Meister S, Beyer S, Burgmann M, Zati Zehni A, Schmoeckel E, Kessler M, Jeschke U, Eggersmann TK, Mahner S, Kolben T, and Ganster F
- Subjects
- Adult, Female, Humans, Male, Pregnancy, Inflammation immunology, Inflammation metabolism, Diabetes, Gestational immunology, Diabetes, Gestational metabolism, Galectins metabolism, Placenta metabolism, Placenta immunology, Placenta pathology, Trophoblasts metabolism, Trophoblasts pathology, Trophoblasts immunology
- Abstract
Objectives: Gestational diabetes mellitus (GDM) is a growing health concern. Since members of the galectin-family are identified to play a role in the pathogenesis of GDM, we determined galectin-12 as an essential protein due to its influence in lipolysis and inflammation processes. This study investigates the expression of galectin-12 in the placentas of women with GDM., Study Design: The study population includes 40 expectant women suffering from GDM and 40 healthy controls. The expression of galectin-12 in the syncytiotrophoblast (SCT) and the extra villous trophoblast (EVT) of the placenta was analyzed by immunohistological staining and double immunofluorescence. Immunoreactivity Score (IRS) was used for evaluation., Results: The results demonstrate a significant overexpression of galectin-12 in the nucleus of the SCT and the EVT of placentas with GDM compared to the healthy control group. Additionally, double immunofluorescence visualizes corresponding results with an overexpression of galectin-12 in the extra villous trophoblast of GDM placentas representing maternal cells., Conclusion: This study identifies galectin-12 to be associated with the process of gestational diabetes mellitus. These findings are in correspondence with the involvement of galectin-12 in inflammatory processes. Maternal BMI and male sex seem to be confounder for the expression of galectin-12 in the nuclear syncytiotrophoblast, but not in other parts of the investigated placental areas. Further investigations are necessary to verify the correlation between gestational diabetes mellitus and the expression of galectin-12 in the placenta and to further elucidate its distinct role., Competing Interests: Declaration of Competing Interest Thomas Kolben: holds stock of Bayer AG. Sven Mahner: Research support, advisory board, honoraria and travel expenses from AbbVie, AstraZeneca, Clovis, Eisai, GlaxoSmithKline, Medac, MSD, Novartis, Olympus, PharmaMar, Pfizer, Roche, Sensor Kinesis, Teva, Tesaro. The other authors declare no conflict of interest., (Copyright © 2024. Published by Elsevier B.V.)
- Published
- 2024
- Full Text
- View/download PDF
47. Progesterone-mediated remodeling of the maternal-fetal interface by a PGRMC1-dependent mechanism.
- Author
-
Wang F, Ferreira LMR, Mazzanti A, Yu H, Gu B, Meissner TB, Li Q, and Strominger JL
- Subjects
- Humans, Female, Pregnancy, Placenta immunology, Placenta metabolism, Signal Transduction immunology, Maternal-Fetal Exchange immunology, Embryo Implantation immunology, Receptors, Progesterone metabolism, Progesterone metabolism, Progesterone pharmacology, Membrane Proteins metabolism, Membrane Proteins genetics, Trophoblasts metabolism, Trophoblasts immunology
- Abstract
Implantation and maintenance of pregnancy involve intricate immunological processes that enable the developing fetus to coexist with the maternal immune system. Progesterone, a critical hormone during pregnancy, is known to promote immune tolerance and prevent preterm labor. However, the mechanism by which progesterone mediates these effects remains unclear. In this study, we investigated the role of the non-classical progesterone receptor membrane component 1 (PGRMC1) in progesterone signaling at the maternal-fetal interface. Using JEG3 cells, a trophoblast model cell line, we observed that progesterone stimulation increased the expression of human leukocyte antigen-C (HLA-C) and HLA-G, key molecules involved in immune tolerance. We also found that progesterone upregulated the expression of the transcription factor ELF3, which is known to regulate trophoblast-specific HLA-C expression. Interestingly, JEG3 cells lacked expression of classical progesterone receptors (PRs) but exhibited high expression of PGRMC1, a finding we confirmed in primary trophoblasts by mining sc-RNA seq data from human placenta. To investigate the role of PGRMC1 in progesterone signaling, we used CRISPR/Cas9 technology to knockout PGRMC1 in JEG3 cells. PGRMC1-deficient cells showed a diminished response to progesterone stimulation. Furthermore, we found that the progesterone antagonist RU486 inhibited ELF3 expression in a PGRMC1-dependent manner, suggesting that RU486 acts as a progesterone antagonist by competing for receptor binding. Additionally, we found that RU486 inhibited cell invasion, an important process for successful pregnancy, and this inhibitory effect was dependent on PGRMC1. Our findings highlight the crucial role of PGRMC1 in mediating the immunoregulatory effects of progesterone at the maternal-fetal interface., Competing Interests: Declaration of Competing Interest The authors report no conflicts of interest., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
48. Pre-eclampsia: Re-visiting pathophysiology, role of immune cells, biomarker identification and recent advances in its management.
- Author
-
Tamil Barathi P and Mohanapriya A
- Subjects
- Animals, Female, Humans, Pregnancy, Decidua immunology, HLA-C Antigens immunology, HLA-C Antigens genetics, HLA-C Antigens metabolism, Killer Cells, Natural immunology, Placenta immunology, Placenta pathology, Receptors, KIR immunology, Receptors, KIR metabolism, Receptors, KIR genetics, Trophoblasts immunology, Biomarkers analysis, Pre-Eclampsia immunology, Pre-Eclampsia diagnosis, Pre-Eclampsia therapy
- Abstract
Pre-eclampsia (PE) is a hypertension condition that occurs exclusively during pregnancy and has the potential to impact nearly all organ systems. It is estimated to complicate approximately 2-8% of pregnancies worldwide. PE is a prominent medical disorder that poses a significant risk to pregnant mothers and their infants. This review commences by giving the most up-to- date concepts about the pathophysiology of PE. The condition involves atypical infiltration of trophoblast cells into the spiral arteries of the decidua and myometrium, resulting in an insufficient establishment of proper blood flow between the uterus and placenta. The aberrant activation of natural killer (NK) cells in both the peripheral blood and the decidua has been identified as one of the contributing factors to the development of PE. The strong evidence for the genetic etiology of PE is provided by the association between maternal killer cell immunoglobulin-like receptor (KIR) and Human Leukocyte Antigen (HLA-C) in trophoblast cells. Recent observations provide evidence that changes in the expression of anti-angiogenic factors in the placenta are the underlying cause of the clinical symptoms associated with the condition. This review also provides a comprehensive overview of the latest advancements in understanding the underlying causes of PE. It specifically highlights the emergence of new diagnostic biomarkers and their potential implications for therapeutic interventions in managing this medical condition., Competing Interests: Declaration of Competing Interest I therefore declare that the review work "Pre-eclampsia: Re-visiting Pathophysiology, Role of Immunocells in Pre-Eclampsia, Biomarker Identification, and Recent Advances in its Management" is unique. I have acknowledged and properly cited all sources of information. This manuscript has not previously been published and is not currently being considered for publication elsewhere., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
49. Blocking OLFM4/galectin-3 axis in placental polymorphonuclear myeloid-derived suppressor cells triggers intestinal inflammation in newborns.
- Author
-
Lv S, Chen M, Li Z, Huang Z, Wan S, Kuang S, Peng L, Ye J, Yang M, Li J, and He Y
- Subjects
- Animals, Female, Pregnancy, Humans, Infant, Newborn, Mice, Mice, Knockout, Enterocolitis, Necrotizing immunology, Enterocolitis, Necrotizing metabolism, Granulocyte Colony-Stimulating Factor metabolism, Granulocyte Colony-Stimulating Factor genetics, Mice, Inbred C57BL, Male, Galectins metabolism, Galectins genetics, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Intestines immunology, Intestines pathology, Placenta immunology, Placenta metabolism, Myeloid-Derived Suppressor Cells immunology, Myeloid-Derived Suppressor Cells metabolism, Fetal Growth Retardation immunology
- Abstract
Fetal growth restriction (FGR) is a major cause of premature and low-weight births, which increases the risk of necrotizing enterocolitis (NEC); however, the association remains unclear. We report a close correlation between placental polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) and NEC. Newborns with previous FGR exhibited intestinal inflammation and more severe NEC symptoms than healthy newborns. Placental PMN-MDSCs are vital regulators of fetal development and neonatal gut inflammation. Placental single-cell transcriptomics revealed that PMN-MDSCs populations and olfactomedin-4 gene (Olfm4) expression levels were significantly increased in PMN-MDSCs in later pregnancy compared to those in early pregnancy and non-pregnant females. Female mice lacking Olfm4 in myeloid cells mated with wild-type males showed FGR during pregnancy, with a decreased placental PMN-MDSCs population and expression of growth-promoting factors (GPFs) from placental PMN-MDSCs. Galectin-3 (Gal-3) stimulated the OLFM4-mediated secretion of GPFs by placental PMN-MDSCs. Moreover, GPF regulation via OLFM4 in placental PMN-MDSCs was mediated via hypoxia inducible factor-1α (HIF-1α). Notably, the offspring of mothers lacking Olfm4 exhibited intestinal inflammation and were susceptible to NEC. Additionally, OLFM4 expression decreased in placental PMN-MDSCs from pregnancies with FGR and was negatively correlated with neonatal morbidity. These results revealed that placental PMN-MDSCs contributed to fetal development and ameliorate newborn intestinal inflammation., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
50. Immune rebalancing at the maternal-fetal interface of maternal SARS-CoV-2 infection during early pregnancy.
- Author
-
Xi C, Yan Z, Bai D, Zhang Y, Wang B, Han X, Wu L, Shi X, Hu Z, Tang M, Su Z, Liu Y, Liu B, Yin J, Wang H, Li X, Zhang Y, Gao S, and Liu W
- Subjects
- Female, Pregnancy, Humans, Immune Tolerance, Trophoblasts immunology, Trophoblasts metabolism, Trophoblasts virology, Adult, Pregnancy Trimester, First immunology, Transcriptome, COVID-19 immunology, COVID-19 virology, SARS-CoV-2 immunology, Pregnancy Complications, Infectious immunology, Pregnancy Complications, Infectious virology, Placenta immunology, Placenta virology, Placenta metabolism
- Abstract
The current coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus (SARS-CoV-2) remains a threat to pregnant women. However, the impact of early pregnancy SARS-CoV-2 infection on the maternal-fetal interface remains poorly understood. Here, we present a comprehensive analysis of single-cell transcriptomics and metabolomics in placental samples infected with SARS-CoV-2 during early pregnancy. Compared to control placentas, SARS-CoV-2 infection elicited immune responses at the maternal-fetal interface and induced metabolic alterations in amino acid and phospholipid profiles during the initial weeks post-infection. However, subsequent immune cell activation and heightened immune tolerance in trophoblast cells established a novel dynamic equilibrium that mitigated the impact on the maternal-fetal interface. Notably, the immune response and metabolic alterations at the maternal-fetal interface exhibited a gradual decline during the second trimester. Our study underscores the adaptive immune tolerance mechanisms and establishment of immunological balance during the first two trimesters following maternal SARS-CoV-2 infection., (© The Author(s) 2024. Published by Oxford University Press on behalf of Higher Education Press.)
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.