1
|
Aktemur G, Çakır BT, Karabay G, Filiz AA, Seyhanlı Z, Sucu ST, Tonyalı NV, Çelen Ş. Second-Trimester Inflammatory Markers in Predicting Fetal Growth Restriction: A Retrospective Analysis. Am J Reprod Immunol 2025; 93:e70047. [PMID: 39821967 DOI: 10.1111/aji.70047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/11/2024] [Accepted: 01/06/2025] [Indexed: 01/19/2025] Open
Abstract
PROBLEM Fetal growth restriction (FGR) is a critical pregnancy complication linked to increased perinatal morbidity and mortality. Inflammation plays a key role in FGR's pathophysiology, and systemic inflammation markers may serve as predictors. This study evaluates the role of various inflammation indices; systemic immune-inflammation index (SII), systemic inflammatory response index (SIRI), pan-immune-inflammation value (PIV), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), monocyte-to-lymphocyte ratio (MLR), lymphocyte-to-platelet ratio (LMR), monocyte-to-platelet ratio (MPR), aggregate systemic inflammation index (AISI), systemic coagulation inflammation index (SCII), and immature granulocyte percentage (IG%) in predicting FGR. METHODS OF STUDY This retrospective study included 403 pregnant women treated at Ankara Etlik City Hospital between August 2022 and May 2024. The study population comprised 202 women with uncomplicated pregnancies (control group) and 201 women diagnosed with FGR per the Delphi Consensus Criteria. Second-trimester blood samples were used to calculate the inflammatory indices. RESULTS SII and NLR levels were significantly higher in the FGR group compared to controls (p = 0.020, p = 0.028, respectively). However, no significant associations were found between these indices and adverse neonatal outcomes. Cut-off values for SII and NLR were 896 and 3.91, respectively, with moderate sensitivity and specificity. CONCLUSIONS SII and NLR, measured in the second trimester, may be useful in predicting FGR. Although these indices did not correlate with adverse neonatal outcomes, further prospective studies with larger populations are needed to validate their clinical utility.
Collapse
Affiliation(s)
- Gizem Aktemur
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Betül Tokgöz Çakır
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Gülşan Karabay
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Ahmet Arif Filiz
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Zeynep Seyhanlı
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Serap Topkara Sucu
- Department of Obstetrics and Gynecology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Nazan Vanlı Tonyalı
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| | - Şevki Çelen
- Department of Obstetrics and Gynecology, Division of Perinatology, Ankara Etlik City Hospital, Ankara, Turkey
| |
Collapse
|
2
|
Suaidi NA, Alshawsh MA, Hoe SZ, Mokhtar MH, Md Zin SR. Impact of xylene exposure during organogenesis on foeto-placental efficiency and foetal viability: Exploring its association with oxidative stress-induced inflammation and apoptosis in utero. Toxicol Ind Health 2024; 40:692-710. [PMID: 39308155 DOI: 10.1177/07482337241286569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
The potential maternal and foetal toxicity resulting from exposure to xylene at or below the allowable limit of 100 ppm during gestation is not thoroughly studied. The aim of this study was to investigate maternal and foetal outcomes following prenatal exposure to xylene during organogenesis. Pregnant Sprague Dawley (SD) rats were administered intraperitoneal (IP) corn oil (vehicle), 100, 500, and 1000 parts per million (ppm) of xylene from gestational day (GD) 6 until GD17. Clinical signs, maternal weight gain, and food consumption were recorded daily. A caesarean hysterectomy was performed on GD21 to assess the reproductive and foetal outcomes. Exposure to 1000 ppm of xylene caused a significant decrease in the maternal body weight and food consumption, and an increase in intrauterine foetal deaths. Foetal assessment revealed a significant decrease in foetal weight in both male and female foetuses of female rats treated with 500 and 1000 ppm. Male placental weight was significantly decreased in all xylene-treated groups, while 1000 ppm xylene significantly decreased female placental weight. Histologically, marked uterine inflammatory lesions, fibrosis of the liver and renal tissues, as well as increased placental glycogen content were observed. Immunohistochemistry revealed a significant increase in lipid peroxidation and apoptotic markers. Thus, the foeto-maternal toxicities of xylene have been shown to be mediated by a systemic inflammatory response that exacerbates intrauterine oxidative stress and impairs foeto-placental transfer, leading to an increase in foetal mortality.
Collapse
Affiliation(s)
- Noor Asyikin Suaidi
- Department of Anatomy, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Mohammed Abdullah Alshawsh
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- School of Clinical Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - See-Ziau Hoe
- Department of Physiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Mohd Helmy Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Siti Rosmani Md Zin
- Department of Anatomy, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
3
|
Yount CS, Scheible K, Thurston SW, Qiu X, Ge Y, Hopke PK, Lin Y, Miller RK, Murphy SK, Brunner J, Barrett E, O'Connor TG, Zhang J, Rich DQ. Short term air pollution exposure during pregnancy and associations with maternal immune markers. ENVIRONMENTAL RESEARCH 2024; 260:119639. [PMID: 39034020 PMCID: PMC11421383 DOI: 10.1016/j.envres.2024.119639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Air pollution exposure during pregnancy has been associated with numerous adverse pregnancy, birth, and child health outcomes. One proposed mechanism underlying these associations is maternal immune activation and dysregulation. We examined associations between PM2.5 and NO2 exposure during pregnancy and immune markers within immune function groups (TH1, TH2, TH17, Innate/Early Activation, Regulatory, Homeostatic, and Proinflammatory), and examined whether those associations changed across pregnancy. METHODS In a pregnancy cohort study (n = 290) in Rochester, New York, we measured immune markers (using Luminex) in maternal plasma up to 3 times during pregnancy. We estimated ambient PM2.5 and NO2 concentrations at participants' home addresses using a spatial-temporal model. Using mixed effects models, we estimated changes in immune marker concentrations associated with interquartile range increases in PM2.5 (2.88 μg/m3) and NO2 (7.82 ppb) 0-6 days before blood collection, and assessed whether associations were different in early, mid, and late pregnancy. RESULTS Increased NO2 concentrations were associated with higher maternal immune markers, with associations observed across TH1, TH2, TH17, Regulatory, and Homeostatic groups of immune markers. Furthermore, the largest increases in immune markers associated with each 7.82 ppb increase in NO2 concentration were in late pregnancy (e.g., IL-23 = 0.26 pg/ml, 95% CI = 0.07, 0.46) compared to early pregnancy (e.g., IL-23 = 0.08 pg/ml, 95% CI = -0.11, 0.26). CONCLUSIONS Results were suggestive of NO2-related immune activation. Increases in effect sizes from early to mid to late pregnancy may be due to changes in immune function over the course of pregnancy. These findings provide a basis for immune activation as a mechanism for previously observed associations between air pollution exposure during pregnancy and reduced birthweight, fetal growth restriction, and pregnancy complications.
Collapse
Affiliation(s)
- C S Yount
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, USA
| | - K Scheible
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - S W Thurston
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA
| | - X Qiu
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA
| | - Y Ge
- Nicholas School of the Environment & Duke Global Health Institute, Duke University, Durham, NC, USA
| | - P K Hopke
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, USA; Center for Air and Aquatic Resources Engineering and Sciences, Clarkson University, Potsdam, NY, USA
| | - Y Lin
- Nicholas School of the Environment & Duke Global Health Institute, Duke University, Durham, NC, USA
| | - R K Miller
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA; Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - S K Murphy
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - J Brunner
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA
| | - E Barrett
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, USA; Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA; Department of Biostatistics and Epidemiology, Rutgers University School of Public Health, Piscataway, NJ, USA
| | - T G O'Connor
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA; Department of Psychology, University of Rochester, Rochester, NY, USA; Department of Psychiatry, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA; Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - J Zhang
- Nicholas School of the Environment & Duke Global Health Institute, Duke University, Durham, NC, USA
| | - D Q Rich
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, USA; Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA; Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
4
|
Ma Y, Deng X, Shen R, Zhang H, Qian Y. Unveiling immune tolerance pathways in preeclampsia placenta: implications for molecular targets and discovery of potential biomarkers. Front Endocrinol (Lausanne) 2024; 15:1385154. [PMID: 38894741 PMCID: PMC11182985 DOI: 10.3389/fendo.2024.1385154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/13/2024] [Indexed: 06/21/2024] Open
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.
Collapse
Affiliation(s)
- Yantuanjin Ma
- Institute of Biomedical Engineering, Kunming Medical University, Kunming, China
- Kunming Medical University, Kunming, China
| | - Xingli Deng
- Kunming Medical University, Kunming, China
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ru Shen
- Kunming Medical University, Kunming, China
| | - Hongqing Zhang
- Kunming Medical University, Kunming, China
- Clinical Medical Research Center for Obstetrics and Gynecology (Yunnan Joint Key Laboratory), Kunming City of Maternal and Child Health Hospital, Kunming City of Women and Children, Kunming, China
| | - Yuan Qian
- Kunming Medical University, Kunming, China
- Clinical Medical Research Center for Obstetrics and Gynecology (Yunnan Joint Key Laboratory), Kunming City of Maternal and Child Health Hospital, Kunming City of Women and Children, Kunming, China
| |
Collapse
|
5
|
Jia B, Tang L, Liu H, Chen W, Chen Q, Zhong M, Yin A. Potential roles of the interactions between gut microbiota and metabolites in LPS-induced intrauterine inflammation (IUI) and associated preterm birth (PTB). J Transl Med 2024; 22:7. [PMID: 38167140 PMCID: PMC10762855 DOI: 10.1186/s12967-023-04603-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/06/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Prenatal exposure to intrauterine inflammation (IUI) is a crucial event in preterm birth (PTB) pathophysiology, increasing the incidence of neurodevelopmental disorders. Gut microbiota and metabolite profile alterations have been reported to be involved in PTB pathophysiology. METHOD AND RESULTS In this study, IUI-exposed PTB mouse model was established and verified by PTB rate and other perinatal adverse reactions; LPS-indued IUI significantly increased the rates of PTB, apoptosis and inflammation in placenta tissue samples. LPS-induced IUI caused no significant differences in species richness and evenness but significantly altered the species abundance distribution. Non-targeted metabolomics analysis indicated that the metabolite profile of the preterm mice was altered, and differential metabolites were associated with signaling pathways including pyruvate metabolism. Furthermore, a significant positive correlation between Parasutterella excrementihominis and S4572761 (Nb-p-coumaroyltryptamine) and Mreference-1264 (pyruvic acid), respectively, was observed. Lastly, pyruvic acid treatment partially improved LPS-induced IUI phenotypes and decreased PTB rates and decreased the apoptosis and inflammation in placenta tissue samples. CONCLUSION This study revealed an association among gut microbiota dysbiosis, metabolite profile alterations, and LPS-induced IUI and PTB in mice models. Our investigation revealed the possible involvement of gut microbiota in the pathophysiology of LPS-induced IUI and PTB, which might be mediated by metabolites such as pyruvic acid. Future studies should be conducted to verify the findings through larger sample-sized animal studies and clinical investigations.
Collapse
Affiliation(s)
- Bei Jia
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Avenue, Guangzhou, 510515, People's Republic of China
| | - Lijun Tang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Avenue, Guangzhou, 510515, People's Republic of China
| | - Huibing Liu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Avenue, Guangzhou, 510515, People's Republic of China
| | - Wenqian Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Avenue, Guangzhou, 510515, People's Republic of China
| | - Qian Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Avenue, Guangzhou, 510515, People's Republic of China
| | - Mei Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Avenue, Guangzhou, 510515, People's Republic of China
| | - Ailan Yin
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Northern Avenue, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
6
|
Shah NM, Charani E, Ming D, Cheah FC, Johnson MR. Antimicrobial stewardship and targeted therapies in the changing landscape of maternal sepsis. JOURNAL OF INTENSIVE MEDICINE 2024; 4:46-61. [PMID: 38263965 PMCID: PMC10800776 DOI: 10.1016/j.jointm.2023.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/04/2023] [Accepted: 07/30/2023] [Indexed: 01/25/2024]
Abstract
Pregnant and postnatal women are a high-risk population particularly prone to rapid progression to sepsis with significant morbidity and mortality worldwide. Moreover, severe maternal infections can have a serious detrimental impact on neonates with almost 1 million neonatal deaths annually attributed to maternal infection or sepsis. In this review we discuss the susceptibility of pregnant women and their specific physiological and immunological adaptations that contribute to their vulnerability to sepsis, the implications for the neonate, as well as the issues with antimicrobial stewardship and the challenges this poses when attempting to reach a balance between clinical care and urgent treatment. Finally, we review advancements in the development of pregnancy-specific diagnostic and therapeutic approaches and how these can be used to optimize the care of pregnant women and neonates.
Collapse
Affiliation(s)
- Nishel M Shah
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Esmita Charani
- Health Protection Research Unit in Healthcare Associated Infection and Antimicrobial Resistance, Imperial College London, London, UK
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Damien Ming
- Department of Infectious Diseases, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Fook-Choe Cheah
- Department of Paediatrics, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Mark R Johnson
- Department of Metabolism, Digestion and Reproduction, Imperial College London, Chelsea and Westminster Hospital, London, UK
| |
Collapse
|
7
|
Magawa S, Nii M, Enomoto N, Tamaishi Y, Takakura S, Maki S, Ishida M, Osato K, Kondo E, Sakuma H, Ikeda T. COVID-19 during pregnancy could potentially affect placental function. J Matern Fetal Neonatal Med 2023; 36:2265021. [PMID: 37806776 DOI: 10.1080/14767058.2023.2265021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 09/25/2023] [Indexed: 10/10/2023]
Abstract
OBJECTIVE COVID-19 is an ongoing pandemic and has been extensively studied. However, the effects of COVID-19 during pregnancy, particularly on placental function, have not been verified. In this study, we used blood oxygen level-dependent magnetic resonance imaging (BOLD-MRI) to evaluate whether COVID-19 incidence during pregnancy has any lasting effects with respect to placental oxygenation. METHODS This is a case-control study, in which eight cases of singleton pregnancies before 30 weeks gestation with COVID-19 mothers were included. Placental oxygenation was evaluated using BOLD-MRI after 32 weeks of gestation. BOLD-MRI was consecutively performed under normoxia (21% O2), hyperoxia (100% O2), and normoxia for 4 min each. Individual placental time-activity curves were evaluated to calculate the peak score (peakΔR2*) and the time from the start of maternal oxygen administration to the time of peakΔR2* (time to peakΔR2*). Eighteen COVID-19-free normal pregnancies from a previous study were used as the control group. RESULTS No significant differences were found between the two groups regarding maternal background, number of days of delivery, birth weight, and placental weight. The parameter peakΔR2* was significantly decreased in the COVID-19 group (8 ± 3 vs. 5 ± 1, p < .001); however, there was no significant difference in time to peakΔR2* (458 ± 74 s vs. 471 ± 33 s, p = .644). CONCLUSIONS In this study, BOLD-MRI was used to evaluate placental oxygenation during pregnancy in COVID-19-affected patients. COVID-19 during pregnancy decreased placental oxygenation even post-illness, but had no effect on fetal growth; further investigation of the possible effects of COVID-19 on the fetus and mother is warranted.
Collapse
Affiliation(s)
- Shoichi Magawa
- Department of Obstetrics and Gynecology, Mie University Faculty of Medicine, Tsu, Japan
| | - Masafumi Nii
- Department of Obstetrics and Gynecology, Mie University Faculty of Medicine, Tsu, Japan
| | - Naosuke Enomoto
- Department of Obstetrics and Gynecology, Mie University Faculty of Medicine, Tsu, Japan
| | - Yuya Tamaishi
- Department of Obstetrics and Gynecology, Mie University Faculty of Medicine, Tsu, Japan
| | - Sho Takakura
- Department of Obstetrics and Gynecology, Mie University Faculty of Medicine, Tsu, Japan
| | - Shintaro Maki
- Department of Obstetrics and Gynecology, Mie University Faculty of Medicine, Tsu, Japan
| | - Masaki Ishida
- Department of Radiology, Mie University Faculty of Medicine, Tsu, Japan
| | - Kazuhiro Osato
- Department of Obstetrics and Gynecology, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Eiji Kondo
- Department of Obstetrics and Gynecology, Mie University Faculty of Medicine, Tsu, Japan
| | - Hajime Sakuma
- Department of Radiology, Mie University Faculty of Medicine, Tsu, Japan
| | - Tomoaki Ikeda
- Department of Obstetrics and Gynecology, Mie University Faculty of Medicine, Tsu, Japan
| |
Collapse
|
8
|
El-Beltagy AEFBM, Bakr SM, Mekhaimer SSG, Ghanem NF, Attaallah A. Zinc-nanoparticles alleviate the ovarian damage induced by bacterial lipopolysaccharide (LPS) in pregnant rats and their fetuses. Histochem Cell Biol 2023; 160:453-475. [PMID: 37495867 PMCID: PMC10624724 DOI: 10.1007/s00418-023-02222-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/03/2023] [Indexed: 07/28/2023]
Abstract
Lipopolysaccharide (LPS) is an endotoxin derived from the cell wall of Gram-negative bacteria. LPS exposure during early gestation is associated with adverse effects on the placenta as well as on developmental outcomes, including embryonic resorption, fetal death, congenital teratogenesis, and fetal growth retardation. This work aimed to explore the adverse effects of LPS injected at an early stage of gestation on the gonads of pregnant rats and the ovaries of their pups and the role of zinc nanoparticles (Zn-NPs) against these adverse effects. Twenty-four pregnant rats were used in this study. They were divided at gestation day 4 into four groups (n = 6): control, Zn-NPs (20 mg/kg orally from gestation day E14 till the end of weaning), LPS (50 µg/kg at gestation days E7 and E9), and LPS + Zn-NPs group. The body weight and placenta weight were recorded at gestational day 16. At postnatal day 21 (weaning), the mothers rats and their offspring were sacrificed and immediately dissected to remove the ovaries and uteri from the mothers and the ovaries from their offspring for subsequent biochemical, histological, and immunohistochemical investigations. The obtained results revealed that LPS exposure during early gestation caused severe histopathological alterations in the placenta, uterus, and ovaries of mothers, as well as in the ovaries of their pups. Also, the uterine and ovarian sections displayed a positive reaction for caspase-3 antibody and a negative reaction for Bcl-2 antibody, which reflects the apoptotic effect of LPS. Additionally, remarkable reductions in the levels of antioxidants (superoxide dismutase and catalase) and significant increases in malondialdehyde (MDA) levels were recorded in the serum of LPS-treated mothers and in the ovarian tissues of their offspring. Further biochemical analysis of the ovarian tissues from LPS-maternally treated offspring showed a significant increase in the levels of caspase-3, TNF-α, and TGF-β1, but a significant decrease in the level of IGF-1. On the other hand, treatment of mothers with Zn-NPs from day 14 of gestation until the weaning day (21st day postnatal) successfully ameliorated most of the deleterious histopathological, immunohistochemical, and biochemical changes induced by LPS.
Collapse
Affiliation(s)
| | - Samaa M Bakr
- Zoology Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Samah S G Mekhaimer
- Zoology Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Noura F Ghanem
- Zoology Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Amany Attaallah
- Zoology Department, Faculty of Science, Damanhour University, Damanhour, Egypt
| |
Collapse
|
9
|
Ahmed M, Casanova NG, Zaghloul N, Gupta A, Rodriguez M, Robbins IR, Kempf CL, Sun X, Song JH, Hernon VR, Sammani S, Camp SM, Moreira A, Hsu CD, Garcia JGN. The eNAMPT/TLR4 inflammatory cascade drives the severity of intra-amniotic inflammation in pregnancy and predicts infant outcomes. Front Physiol 2023; 14:1129413. [PMID: 37415908 PMCID: PMC10319582 DOI: 10.3389/fphys.2023.1129413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 06/01/2023] [Indexed: 07/08/2023] Open
Abstract
Introduction: Intra-amniotic inflammation (IAI) or chorioamnionitis is a common complication of pregnancy producing significant maternal morbidity/mortality, premature birth and neonatal risk of chronic lung diseases such as bronchopulmonary dysplasia (BPD). We examined eNAMPT (extracellular nicotinamide phosphoribosyltransferase), a critical inflammatory DAMP and TLR4 ligand, as a potential therapeutic target to reduce IAI severity and improve adverse fetal/neonatal outcomes. Methods: Blood/tissue samples were examined in: 1) women with histologically-proven chorioamnionitis, 2) very low birth weight (VLBW) neonates, and 3) a preclinical murine pregnancy model of IAI. Groups of pregnant IAI-exposed mice and pups were treated with an eNAMPT-neutralizing mAb. Results: Human placentas from women with histologically-proven chorioamnionitis exhibited dramatic NAMPT expression compared to placentas without chorioamnionitis. Increased NAMPT expression in whole blood from VLBW neonates (day 5) significantly predicted BPD development. Compared to untreated LPS-challenged murine dams (gestational day 15), pups born to eNAMPT mAb-treated dams (gestational days 15/16) exhibited a > 3-fold improved survival, reduced neonate lung eNAMPT/cytokine levels, and reduced development and severity of BPD and pulmonary hypertension (PH) following postnatal exposure to 100% hyperoxia days 1-14. Genome-wide gene expression studies of maternal uterine and neonatal cardiac tissues corroborated eNAMPT mAb-induced reductions in inflammatory pathway genes. Discussion: The eNAMPT/TLR4 inflammatory pathway is a highly druggable contributor to IAI pathobiology during pregnancy with the eNAMPT-neutralizing mAb a novel therapeutic strategy to decrease premature delivery and improve short- and long-term neonatal outcomes. eNAMPT blood expression is a potential biomarker for early prediction of chronic lung disease among premature neonates.
Collapse
Affiliation(s)
- Mohamed Ahmed
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Nancy G. Casanova
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Nahla Zaghloul
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Akash Gupta
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Marisela Rodriguez
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Ian R. Robbins
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Carrie L. Kempf
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Xiaoguang Sun
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Jin H. Song
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Vivian Reyes Hernon
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Saad Sammani
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Sara M. Camp
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Alvaro Moreira
- Department of Pediatrics, UT Health San Antonio, Long School of Medicine, San Antonio, TX, United States
| | - Chaur-Dong Hsu
- Department of Obstetrics and Gynecology, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Joe G. N. Garcia
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| |
Collapse
|
10
|
Hu M, Zhang Y, Lu Y, Han J, Guo T, Cui P, Brännström M, Shao LR, Billig H. Regulatory mechanisms of HMGB1 and its receptors in polycystic ovary syndrome-driven gravid uterine inflammation. FEBS J 2023; 290:1874-1906. [PMID: 36380688 PMCID: PMC10952262 DOI: 10.1111/febs.16678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/23/2022] [Accepted: 11/15/2022] [Indexed: 11/17/2022]
Abstract
High-mobility group box 1 (HMGB1) is critical for inflammatory homeostasis and successful pregnancy, and there is a strong association among elevated levels of HMGB1, polycystic ovary syndrome (PCOS), chronic inflammation and pregnancy loss. However, the mechanisms responsible for PCOS-driven regulation of uterine HMGB1 and its candidate receptors [toll-like receptor (TLR) 2 and 4] and inflammatory responses during pregnancy remain unclear. In this study, we found a gestational stage-dependent decrease in uterine HMGB1 and TLR4 protein abundance in rats during normal pregnancy. We demonstrated that increased expression of HMGB1, TLR2 and TLR4 proteins was associated with activation of inflammation-related signalling pathways in the gravid uterus exposed to 5α-dihydrotestosterone and insulin, mimicking the clinical features (hyperandrogenism and insulin resistance) of PCOS and this elevation was completely inhibited by treatment with the androgen receptor (AR) antagonist flutamide. Interestingly, acute exposure to lipopolysaccharide suppressed HMGB1, TLR4 and inflammation-related protein abundance but did not affect androgen levels or AR expression in the gravid uterus with viable fetuses. Furthermore, immunohistochemical analysis revealed that, in addition to being localized predominately in the nuclear compartment, HMGB1 immunoreactivity was also detected in the cytoplasm in the PCOS-like rat uterus, PCOS endometrium and pregnant rat uterus with haemorrhagic and resorbed fetuses, possibly via activation of nuclear factor κB signalling. These results suggest that both AR-dependent and AR-independent mechanisms contribute to the modulation of HMGB1/TLR2/TLR4-mediated uterine inflammation. We propose that the elevation of HMGB1 and its receptors and disruption of the pro-/anti-inflammatory balance in the gravid uterus may participate in the pathophysiology of PCOS-associated pregnancy loss.
Collapse
Affiliation(s)
- Min Hu
- Department of Traditional Chinese MedicineThe First Affiliated Hospital of Guangzhou Medical UniversityChina
- Institute of Integrated Traditional Chinese Medicine and Western MedicineGuangzhou Medical UniversityChina
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgSweden
| | - Yuehui Zhang
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgSweden
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated HospitalHeilongjiang University of Chinese MedicineHarbinChina
| | - Yaxing Lu
- Department of Traditional Chinese MedicineThe First Affiliated Hospital of Guangzhou Medical UniversityChina
- Institute of Integrated Traditional Chinese Medicine and Western MedicineGuangzhou Medical UniversityChina
| | - Jing Han
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated HospitalHeilongjiang University of Chinese MedicineHarbinChina
| | - Tingting Guo
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated HospitalHeilongjiang University of Chinese MedicineHarbinChina
| | - Peng Cui
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgSweden
- Department of Obstetrics and GynecologyShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineChina
| | - Mats Brännström
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Sahlgrenska AcademyUniversity of GothenburgSweden
| | - Linus R. Shao
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgSweden
| | - Håkan Billig
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska AcademyUniversity of GothenburgSweden
| |
Collapse
|
11
|
Lodge-Tulloch NA, Toews AJ, Atallah A, Cotechini T, Girard S, Graham CH. Cross-Generational Impact of Innate Immune Memory Following Pregnancy Complications. Cells 2022; 11:3935. [PMID: 36497193 PMCID: PMC9741472 DOI: 10.3390/cells11233935] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Pregnancy complications can have long-term negative effects on the health of the affected mothers and their children. In this review, we highlight the underlying inflammatory etiologies of common pregnancy complications and discuss how aberrant inflammation may lead to the acquisition of innate immune memory. The latter can be described as a functional epigenetic reprogramming of innate immune cells following an initial exposure to an inflammatory stimulus, ultimately resulting in an altered response following re-exposure to a similar inflammatory stimulus. We propose that aberrant maternal inflammation associated with complications of pregnancy increases the cross-generational risk of developing noncommunicable diseases (i.e., pregnancy complications, cardiovascular disease, and metabolic disease) through a process mediated by innate immune memory. Elucidating a role for innate immune memory in the cross-generational health consequences of pregnancy complications may lead to the development of novel strategies aimed at reducing the long-term risk of disease.
Collapse
Affiliation(s)
| | - Alexa J. Toews
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Aline Atallah
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Tiziana Cotechini
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Charles H. Graham
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada
| |
Collapse
|
12
|
Ushida T, Cotechini T, Protopapas N, Atallah A, Collyer C, Toews AJ, Macdonald-Goodfellow SK, Tse MY, Winn LM, Pang SC, Adams MA, Othman M, Kotani T, Kajiyama H, Graham CH. Aberrant inflammation in rat pregnancy leads to cardiometabolic alterations in the offspring and intrauterine growth restriction in the F2 generation. J Dev Orig Health Dis 2022; 13:706-718. [PMID: 35593438 DOI: 10.1017/s2040174422000265] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Children of women with pre-eclampsia have increased risk of cardiovascular (CV) and metabolic disease in adult life. Furthermore, the risk of pregnancy complications is higher in daughters born to women affected by pre-eclampsia than in daughters born after uncomplicated pregnancies. While aberrant inflammation contributes to the pathophysiology of pregnancy complications, including pre-eclampsia, the contribution of maternal inflammation to subsequent risk of CV and metabolic disease as well as pregnancy complications in the offspring remains unclear. Here, we demonstrate that 24-week-old female rats (F1) born to dams (F0) exposed to lipopolysaccharide (LPS) during pregnancy (to induce inflammation) exhibited mild systolic dysfunction, increased cardiac growth-related gene expression, altered glucose tolerance, and coagulopathy; whereas male F1 offspring exhibited altered glucose tolerance and increased visceral fat accumulation compared with F1 sex-matched offspring born to saline-treated dams. Both male and female F1 offspring born to LPS-treated dams had evidence of anemia. Fetuses (F2) from F1 females born to LPS-treated dams were growth restricted, and this reduction in fetal growth was associated with increased CD68 positivity (indicative of macrophage presence) and decreased expression of glucose transporter-1 in their utero-placental units. These results indicate that abnormal maternal inflammation can contribute to increased risk of CV and metabolic disease in the offspring, and that the effects of inflammation may cross generations. Our findings provide evidence in support of early screening for CV and metabolic disease, as well as pregnancy complications in offspring affected by pre-eclampsia or other pregnancy complications associated with aberrant inflammation.
Collapse
Affiliation(s)
- Takafumi Ushida
- Department of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tiziana Cotechini
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Nicole Protopapas
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Aline Atallah
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Charlotte Collyer
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Alexa J Toews
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | | - M Yat Tse
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Louise M Winn
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Stephen C Pang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Michael A Adams
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Maha Othman
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
- School of Baccalaureate Nursing, St. Lawrence College, Kingston, Ontario, Canada
| | - Tomomi Kotani
- Department of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Charles H Graham
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
13
|
Maternal Underweight and Obesity Are Associated with Placental Pathologies in Human Pregnancy. Reprod Sci 2022; 29:3425-3448. [PMID: 35739350 DOI: 10.1007/s43032-022-00983-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/19/2022] [Indexed: 12/14/2022]
Abstract
Maternal underweight and obesity are prevalent conditions, associated with chronic, low-grade inflammation, poor fetal development, and long-term adverse outcomes for the child. The placenta senses and adapts to the pregnancy environment in an effort to support optimal fetal development. However, the mechanisms driving these adaptations, and the resulting placental phenotypes, are poorly understood. We hypothesised that maternal underweight and obesity would be associated with increased prevalence of placental pathologies in term and preterm pregnancies. Data from 12,154 pregnancies were obtained from the Collaborative Perinatal Project, a prospective cohort study conducted from 1959 to 1974. Macro- and microscopic placental pathologies were analysed across maternal prepregnancy body mass index (BMI) to assess differences in the presence of pathologies among underweight, overweight, and obese BMI groups compared to normal weight reference BMI at term and preterm. Placental pathologies were also assessed across fetal sex. Pregnancies complicated by maternal obesity had placentae with increased fetal inflammation at preterm, and increased inflammation of maternal gestational tissues at term. In term pregnancies, increasing maternal BMI associated with increased maternal vascular malperfusion (MVM), odds of an appropriately mature placenta for gestational age, and placental weight, and decreased placental efficiency. Male placentae, independent of maternal BMI, had increased inflammation, MVM, and placental efficiency than female placentae, particularly at term. Maternal underweight and obesity are not inert conditions for the placenta, and the histomorphological changes driven by suboptimal maternal BMI may serve as indicators of adversities experienced in utero and potential predictors of future health trajectories.
Collapse
|
14
|
Avci S, Kuscu N, Durkut B, Kilinc L, Ustunel I, Celik-Ozenci C. Altered expression of Notch signaling, Tlr receptors, and surfactant protein expression after prostaglandin inhibition may be associated with the delayed labor in LPS-induced mice. J Assist Reprod Genet 2022; 39:1531-1544. [PMID: 35538257 DOI: 10.1007/s10815-022-02515-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 05/02/2022] [Indexed: 11/28/2022] Open
Abstract
PURPOSE This study aims to investigate whether indomethacin (IND) delays preterm birth by regulating the Notch pathway, Tlr receptors, and Sp-A in the placenta in lipopolysaccharide (LPS)-induced preterm labor (PTL) model. METHODS CD-1 mice were distributed to the pregnant control (PC), Sham, PBS, IND (2 mg/kg; i.p.), LPS (25 μg/100 μl; intrauterine), and LPS + IND groups. The injections were performed on day 14.5 of pregnancy. Placentae were collected on day 15.5 of pregnancy, and immunohistochemical analyzes were performed. Differences in staining intensities between the Cox-1, Notch-1 (N1), Dll-1, Jagged-2 (Jag-2), Tlr-2, and Tlr-4 proteins were compared. RESULTS Preterm labor rates were 100% and 66% (preterm delivery delayed 5 h) in the LPS and LPS + IND groups, respectively. In LPS-treated mice, a general morphological deterioration was observed in the placenta. Total placental mid-sagittal measurement was significantly reduced in the LPS-treated group, while it was similar to the PC group in the LPS + IND group. Cox-1 expression in the LZ increased, and Sp-A expression decreased after LPS injection, and IND administration diminished this increase. N1 expression increased in the labyrinth zone (LZ) and the junctional zone (JZ). Dll-1 and Jag-2 expression increased in the JZ after LPS injection (p < 0.0001). IND administration diminished Tlr-2 expression in the LZ and Tlr-4 expression in the JZ after LPS injection. CONCLUSION In conclusion, PG (prostaglandin) inhibition may alter Notch signaling, Tlr, and Sp-A protein expression and may be associated with delayed labor in LPS-induced mice.
Collapse
Affiliation(s)
- Sema Avci
- Department of Histology and Embryology, School of Medicine, Alanya Alaaddin Keykubat University, Alanya, Turkey
| | - Nilay Kuscu
- Department of Histology and Embryology, School of Medicine, Akdeniz University, Antalya, Turkey
| | - Begum Durkut
- Department of Histology and Embryology, School of Medicine, Akdeniz University, Antalya, Turkey
| | - Leyla Kilinc
- Department of Histology and Embryology, School of Medicine, Akdeniz University, Antalya, Turkey
| | - Ismail Ustunel
- Department of Histology and Embryology, School of Medicine, Akdeniz University, Antalya, Turkey
| | - Ciler Celik-Ozenci
- Department of Histology and Embryology, School of Medicine, Koc University, Istanbul, Turkey. .,Koç University Research Center for Translational Medicine (KUTTAM), Koc University, Istanbul, Turkey.
| |
Collapse
|
15
|
Ikumi NM, Matjila M. Preterm Birth in Women With HIV: The Role of the Placenta. Front Glob Womens Health 2022; 3:820759. [PMID: 35392117 PMCID: PMC8982913 DOI: 10.3389/fgwh.2022.820759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/08/2022] [Indexed: 01/12/2023] Open
Abstract
Maternal HIV infection is associated with an increased risk of preterm birth (PTB). However, the mechanisms underlying this increased risk in women with HIV remain poorly understood. In this regard, it is well-established that labor is an inflammatory process and premature activation of the pro-inflammatory signals (associated with labor) can result in preterm labor which can subsequently lead to PTB. HIV infection is known to cause severe immune dysregulation within its host characterized by altered immune profiles, chronic inflammation and eventually, the progressive failure of the immune system. The human placenta comprises different immune cell subsets, some of which play an important role during pregnancy including participating in the inflammatory processes that accompany labor. It is therefore plausible that HIV/antiretroviral therapy (ART)-associated immune dysregulation within the placental microenvironment may underlie the increased risk of PTB reported in women with HIV. Here, we review evidence from studies that point toward the placental origin of spontaneous PTB and discuss possible ways maternal HIV infection and/or ART could increase this risk. We focus on key cellular players in the maternal decidua including natural killer cells, CD4+ T cells including CD4+ regulatory T cells, CD8+ T cells as well as macrophages.
Collapse
|
16
|
Shukla V, Soares MJ. Modeling Trophoblast Cell-Guided Uterine Spiral Artery Transformation in the Rat. Int J Mol Sci 2022; 23:ijms23062947. [PMID: 35328368 PMCID: PMC8950824 DOI: 10.3390/ijms23062947] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 12/20/2022] Open
Abstract
The rat possesses hemochorial placentation with deep intrauterine trophoblast cell invasion and trophoblast-guided uterine spiral artery remodeling, which resembles human placentation. Uterine spiral arteries are extensively remodeled to deliver sufficient supply of maternal blood and nutrients to the developing fetus. Inadequacies in these key processes negatively impact fetal growth and development. Recent innovations in genome editing combined with effective phenotyping strategies have provided new insights into placental development. Application of these research approaches has highlighted both conserved and species-specific features of hemochorial placentation. The review provides foundational information on rat hemochorial placental development and function during physiological and pathological states, especially as related to the invasive trophoblast cell-guided transformation of uterine spiral arteries. Our goal is to showcase the utility of the rat as a model for in vivo mechanistic investigations targeting regulatory events within the uterine-placental interface.
Collapse
Affiliation(s)
- Vinay Shukla
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Correspondence: (V.S.); (M.J.S.)
| | - Michael J. Soares
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy, Kansas City, MO 64108, USA
- Correspondence: (V.S.); (M.J.S.)
| |
Collapse
|
17
|
Salazar MD, Wang WJ, Skariah A, He Q, Field K, Nixon M, Reed R, Dambaeva S, Beaman K, Gilman-Sachs A, Kwak-Kim J. Post-hoc evaluation of peripheral blood natural killer cell cytotoxicity in predicting the risk of recurrent pregnancy losses and repeated implantation failures. J Reprod Immunol 2022; 150:103487. [DOI: 10.1016/j.jri.2022.103487] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 12/04/2021] [Accepted: 01/16/2022] [Indexed: 12/24/2022]
|
18
|
Zhou Q, Xiong Y, Qu B, Bao A, Zhang Y. DNA Methylation and Recurrent Pregnancy Loss: A Mysterious Compass? Front Immunol 2021; 12:738962. [PMID: 34745108 PMCID: PMC8566749 DOI: 10.3389/fimmu.2021.738962] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/04/2021] [Indexed: 12/24/2022] Open
Abstract
Recurrent pregnancy loss (RPL) is a common and severe pathological pregnancy, whose pathogenesis is not fully understood. With the development of epigenetics, the study of DNA methylation, provides a new perspective on the pathogenesis and therapy of RPL. The abnormal DNA methylation of imprinted genes, placenta-specific genes, immune-related genes and sperm DNA may, directly or indirectly, affect embryo implantation, growth and development, leading to the occurrence of RPL. In addition, the unique immune tolerogenic microenvironment formed at the maternal-fetal interface has an irreplaceable effect on the maintenance of pregnancy. In view of these, changes in the cellular components of the maternal-fetal immune microenvironment and the regulation of DNA methylation have attracted a lot of research interest. This review summarizes the research progress of DNA methylation involved in the occurrence of RPL and the regulation of the maternal-fetal immune microenvironment. The review provides insights into the personalized diagnosis and treatment of RPL.
Collapse
Affiliation(s)
- Qi Zhou
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yunhe Xiong
- Urology Department, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bing Qu
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Anyu Bao
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan Zhang
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
19
|
Wu H, You Q, Jiang Y, Mu F. Tumor necrosis factor inhibitors as therapeutic agents for recurrent spontaneous abortion (Review). Mol Med Rep 2021; 24:847. [PMID: 34643255 DOI: 10.3892/mmr.2021.12487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 08/31/2021] [Indexed: 11/05/2022] Open
Abstract
Recurrent spontaneous abortion (RSA) is a troublesome pregnancy disorder that manifests as sequential early pregnancy losses; its causes are diverse and complex. Among the known possible causes of RSA, the development of an immune disorder in response to the embryo appears to be the most pronounced. The imbalance between immune rejection and immune tolerance contributes to pregnancy loss in females with RSA, wherein the abnormal ratio of T helper (Th)1 cell‑related cytokines [predominantly tumor necrosis factor (TNF)‑α] and Th2 cell‑related cytokines is a strong risk factor for RSA. TNF‑α is a pro‑inflammatory cytokine and TNF inhibitors have been effective in the treatment of various autoimmune diseases, such as ankylosing spondylitis, and inflammatory diseases, such as ulcerative colitis. Based on their immunomodulatory properties, TNF inhibitors have been used in the treatment of RSA to reduce the immune rejection rate and improvement in pregnancy outcomes has been observed in females suffering from RSA who were treated with TNF inhibitors. The aim of the present review was to interpret the involvement of TNF‑α in the immunological disorder underlying RSA and summarize the clinical outcomes of TNF inhibitor treatment in patients with RSA.
Collapse
Affiliation(s)
- Hong Wu
- Department of Integrated TCM and Western Medicine, Southwest Hospital, Army Medical University, Chongqing 400038, P.R. China
| | - Qingxia You
- Department of Integrated TCM and Western Medicine, Southwest Hospital, Army Medical University, Chongqing 400038, P.R. China
| | - Yi Jiang
- Department of Integrated TCM and Western Medicine, Southwest Hospital, Army Medical University, Chongqing 400038, P.R. China
| | - Fangxiang Mu
- Department of Reproductive Medicine, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| |
Collapse
|
20
|
Qi L, Bi S, Fu J, Fu Y, Li L, Zhao L. Efficacy of Intrauterine Perfusion of Cyclosporin A for Intractable Recurrent Spontaneous Abortion Patients With Endometrial Alloimmune Disorders: A Randomized Controlled Trial. Front Physiol 2021; 12:737878. [PMID: 34552510 PMCID: PMC8450415 DOI: 10.3389/fphys.2021.737878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/13/2021] [Indexed: 11/13/2022] Open
Abstract
Objective To explore the therapeutic efficacy of intrauterine perfusion of cyclosporin A (CsA) in intractable recurrent spontaneous abortion (RSA) patients with endometrial alloimmune dysfunction. Methods This is a randomized controlled trial (RCT) of patients with intractable RSA with endometrial alloimmune disorders. A total of 201 women were enrolled, all of whom had at least 3 serial abortions and endometrial alloimmune dysfunction. Participants were randomly assigned to two groups. The CsA group (n = 101) received intrauterine infusion of 250 mg CsA on the 3rd and 7th days after menstruation for 2 menstrual cycles, while the placebo group (n = 100) received placebo. The birth of healthy, deformity-free babies was the main study outcome. Results In total, 75 (74.26%) women in the CsA group and 59 (59.00%) women in the placebo group gave birth to healthy babies [P = 0.01, OR = 2.01; 95% CI (1.10∼3.65)]. Compared to the placebo group, the CsA group had dramatically lower endometrial CD56+ cell and CD57+ cell concentrations at the luteal phase of the second menstrual cycle (P < 0.05). Conclusion Intrauterine perfusion of CsA was confirmed to be a promising approach for the treatment of intractable alloimmune RSA.
Collapse
Affiliation(s)
- Lijuan Qi
- Department of Obstetrics, Qingdao Jinhua Hospital, Qingdao, China
| | - Shuqin Bi
- Department of Obstetrics, Qingdao Jinhua Hospital, Qingdao, China
| | - Jinhua Fu
- Department of Obstetrics, Qingdao Jinhua Hospital, Qingdao, China
| | - Yinghui Fu
- Department of Obstetrics, Qingdao Jinhua Hospital, Qingdao, China
| | - Lin Li
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Long Zhao
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
21
|
Dang Y, Souchet C, Moresi F, Jeljeli M, Raquillet B, Nicco C, Chouzenoux S, Lagoutte I, Marcellin L, Batteux F, Doridot L. BCG-trained innate immunity leads to fetal growth restriction by altering immune cell profile in the mouse developing placenta. J Leukoc Biol 2021; 111:1009-1020. [PMID: 34533228 DOI: 10.1002/jlb.4a0720-458rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Trained immunity is a new concept illustrating that innate immune cells are able to undergo a long-term metabolic and epigenetic reprogramming after infection or vaccination, thus displaying either a pro- or an anti-inflammatory phenotype during a sequential unrelated challenge. Innate immune cells such as natural killer (NK) cells and macrophages constitute a large part of the decidual leukocyte population at the maternal-fetal interface, playing an important role in placental development and as such in fetal growth and development. In this study, we hypothesized that training the innate immune cells before pregnancy could have an impact on pregnancy. To test this hypothesis, we used CBA/J x DBA/2 mouse model to investigate pregnancy outcomes and leukocyte population at the maternal-fetal interface. Although we were not able to show a beneficial effect of LPS-tolerogenic training on fetal resorption, Bacillus Calmette-Guérin (BCG) training, known to prime innate immune cells to be proinflammatory, led to fetal growth restriction, without aggravating the fetal resorption rate. We also found that BCG training led to less NK cells and macrophages at the maternal-fetal interface at the early stage of placentation (E9.5), associated with a down-regulation of Ccr3 and Lif mRNA expression. This induced altered leucocyte population profile can be an explanation for the subsequent fetal growth restriction. These data suggest that preconceptional infections-induced trained immunity could influence pregnancy outcomes.
Collapse
Affiliation(s)
- Yipu Dang
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France
| | - Camille Souchet
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France
| | - Fabiana Moresi
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France
| | - Mohamed Jeljeli
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France.,Service d'immunologie Biologique, AP-HP, Hôpital Universitaire Paris Centre, F-75014 Paris, France, Paris, France
| | - Bruno Raquillet
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France
| | - Carole Nicco
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France
| | - Sandrine Chouzenoux
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France
| | - Isabelle Lagoutte
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France
| | - Louis Marcellin
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France.,Département de Gynécologie Obstétrique II et Médecine de la Reproduction, AP-HP, Hôpital Universitaire Paris Centre, F-75014 Paris, France, Paris, France
| | - Frederic Batteux
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France.,Service d'immunologie Biologique, AP-HP, Hôpital Universitaire Paris Centre, F-75014 Paris, France, Paris, France
| | - Ludivine Doridot
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France
| |
Collapse
|
22
|
Li M, Sun F, Qian J, Chen L, Li D, Wang S, Du M. Tim-3/CTLA-4 pathways regulate decidual immune cells-extravillous trophoblasts interaction by IL-4 and IL-10. FASEB J 2021; 35:e21754. [PMID: 34191338 DOI: 10.1096/fj.202100142r] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/21/2021] [Accepted: 06/09/2021] [Indexed: 11/11/2022]
Abstract
To obtain a successful pregnancy, the establishment of maternal-fetal tolerance and successful placentation are required to be established. Disruption of this immune balance and/or inadequate placental perfusion is believed to be associated with a lot of pregnancy-related complications, such as recurrent spontaneous abortion, pre-eclampsia, and fetal intrauterine growth restriction. Extravillous trophoblasts (EVTs) have the unique ability to instruct decidual immune cells (DICs) to develop a regulatory phenotype for fetal tolerance. Utilizing immortalized human first trimester extravillous trophoblast cells and primary EVTs, we found that DICs promote EVT function and placental development. We have previously shown that checkpoints T-cell immunoglobulin mucin-3 (Tim-3) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) are important for DIC function. In the present study, we showed that blockade of Tim-3 and CTLA-4 pathways leaded to the abnormal DICs-EVTs interaction, poor placental development, and increased fetal loss. Treatment with IL-4 and IL-10 could rescue the adverse effects of targeting Tim-3 and CTLA-4 on the pregnancy outcome. Hence, the reproductive safety must be a criterion considered in the assessment of immuno-therapeutic agents. In addition, IL-4 and IL-10 may represent novel therapeutic strategies to prevent pregnancy loss induced by checkpoint inhibition.
Collapse
Affiliation(s)
- Mengdie Li
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, P.R. China
| | - Fengrun Sun
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, P.R. China
| | - Jinfeng Qian
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, P.R. China
| | - Lanting Chen
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, P.R. China
| | - Dajin Li
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, P.R. China
| | - Songcun Wang
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, P.R. China
| | - Meirong Du
- Laboratory for Reproductive Immunology, Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, P.R. China
| |
Collapse
|
23
|
Wedn AM, El-Bassossy HM, Eid AH, El-Mas MM. Modulation of preeclampsia by the cholinergic anti-inflammatory pathway: Therapeutic perspectives. Biochem Pharmacol 2021; 192:114703. [PMID: 34324867 DOI: 10.1016/j.bcp.2021.114703] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/12/2022]
Abstract
The cholinergic anti-inflammatory pathway (CAP) is vital for the orchestration of the immune and inflammatory responses under normal and challenged conditions. Over the past two decades, peripheral and central circuits of CAP have been shown to be critically involved in dampening the inflammatory reaction in a wide array of inflammatory disorders. Additionally, emerging evidence supports a key role for CAP in the regulation of the female reproductive system during gestation as well as in the advent of serious pregnancy-related inflammatory insults such as preeclampsia (PE). Within this framework, the modulatory action of CAP encompasses the perinatal maternal and fetal adverse consequences that surface due to antenatal PE programming. Albeit, a considerable gap still exists in our knowledge of the precise cellular and molecular underpinnings of PE/CAP interaction, which hampered global efforts in safeguarding effective preventive or therapeutic measures against PE complications. Here, we summarize reports in the literature regarding the roles of peripheral and reflex cholinergic neuroinflammatory pathways of nicotinic acetylcholine receptors (nAChRs) in reprogramming PE complications in mothers and their progenies. The possible contributions of α7-nAChRs, cholinesterases, immune cells, adhesion molecules, angiogenesis, and endothelial dysfunction to the interaction have also been reviewed.
Collapse
Affiliation(s)
- Abdalla M Wedn
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Hany M El-Bassossy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar; Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Mahmoud M El-Mas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Kuwait.
| |
Collapse
|
24
|
Akerele OA, Manning SJ, Dixon SE, Lacey AE, Cheema SK. Maternal omega-3 fatty acids maintained positive maternal lipids and cytokines profile, and improved pregnancy outcomes of C57BL/6 mice. J Nutr Biochem 2021; 98:108813. [PMID: 34242722 DOI: 10.1016/j.jnutbio.2021.108813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 10/20/2022]
Abstract
Omega (n)-3 polyunsaturated fatty acids (PUFA) are known to regulate lipid metabolism and inflammation; however, the regulation of maternal lipid metabolism and cytokines profile by n-3 PUFA during different gestation stages, and its impact on fetal sustainability is not known. We investigated the effects of maternal diet varying in n-3 PUFA prior to, and during gestation, on maternal metabolic profile, placental inflammatory cytokines, and fetal outcomes. Female C57BL/6 mice were fed either a high, low or very low (9, 3 or 1% w/w n-3 PUFA) diet, containing n-6:n-3 PUFA of 5:1, 20:1 and 40:1, respectively for two weeks before mating, and throughout pregnancy. Animals were sacrificed prior to mating (NP), and during pregnancy at gestation days 6.5, 12.5 and 18.5. Maternal metabolic profile, placental cytokines and fetal outcomes were determined. Our results show for the first time that a maternal diet high in n-3 PUFA prevented dyslipidemia in NP mice, and maintained the expected lipid profile during pregnancy. However, females fed the very low n-3 PUFA diet became hyperlipidemic prior to pregnancy, and carried this profile into pregnancy. Maternal diet high in n-3 PUFA maintained maternal plasma progesterone and placental pro-inflammatory cytokines profile, and sustained fetal numbers throughout pregnancy, while females fed the low and very-low n-3 PUFA diet had fewer fetuses. Our findings demonstrate the importance of maternal diet before, and during pregnancy, to maintain maternal metabolic profile and fetus sustainability. These findings are important when designing dietary strategies to optimize maternal metabolism during pregnancy for successful pregnancy outcome.
Collapse
Affiliation(s)
- Olatunji Anthony Akerele
- Department of Biochemistry, Memorial University of Newfoundland, Newfoundland and Labrador, Canada
| | - Sarah Jane Manning
- Department of Biochemistry, Memorial University of Newfoundland, Newfoundland and Labrador, Canada
| | - Sarah Emily Dixon
- Department of Biochemistry, Memorial University of Newfoundland, Newfoundland and Labrador, Canada
| | - Amelia Estelle Lacey
- Department of Biochemistry, Memorial University of Newfoundland, Newfoundland and Labrador, Canada
| | - Sukhinder Kaur Cheema
- Department of Biochemistry, Memorial University of Newfoundland, Newfoundland and Labrador, Canada.
| |
Collapse
|
25
|
Kozai K, Iqbal K, Moreno-Irusta A, Scott RL, Simon ME, Dhakal P, Fields PE, Soares MJ. Protective role of IL33 signaling in negative pregnancy outcomes associated with lipopolysaccharide exposure. FASEB J 2021; 35:e21272. [PMID: 33423320 DOI: 10.1096/fj.202001782rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/15/2020] [Accepted: 11/30/2020] [Indexed: 01/21/2023]
Abstract
Interleukin 33 (IL33) signaling has been implicated in the establishment and maintenance of pregnancy and in pregnancy disorders. The goal of this project was to evaluate the role of IL33 signaling in rat pregnancy. The rat possesses hemochorial placentation with deep intrauterine trophoblast invasion; features also characteristic of human placentation. We generated and characterized a germline mutant rat model for IL33 using CRISPR/Cas9 genome editing. IL33 deficient rats exhibited deficits in lung responses to an inflammatory stimulus (Sephadex G-200) and to estrogen-induced uterine eosinophilia. Female rats deficient in IL33 were fertile and exhibited pregnancy outcomes (gestation length and litter size) similar to wild-type rats. Placental weight was adversely affected by the disruption of IL33 signaling. A difference in pregnancy-dependent adaptations to lipopolysaccharide (LPS) exposure was observed between wild-type and IL33 deficient pregnancies. Pregnancy in wild-type rats treated with LPS did not differ significantly from pregnancy in vehicle-treated wild-type rats. In contrast, LPS treatment decreased fetal survival rate, fetal and placental weights, and increased fetal growth restriction in IL33 deficient rats. In summary, a new rat model for investigating IL33 signaling has been established. IL33 signaling participates in the regulation of placental development and protection against LPS-induced fetal and placental growth restriction.
Collapse
Affiliation(s)
- Keisuke Kozai
- Institute for Reproduction and Perinatal Research, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas, KS, USA
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas, KS, USA
| | - Ayelen Moreno-Irusta
- Institute for Reproduction and Perinatal Research, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas, KS, USA
| | - Regan L Scott
- Institute for Reproduction and Perinatal Research, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas, KS, USA
| | - Mikaela E Simon
- Institute for Reproduction and Perinatal Research, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas, KS, USA
| | - Pramod Dhakal
- Institute for Reproduction and Perinatal Research, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas, KS, USA
| | - Patrick E Fields
- Institute for Reproduction and Perinatal Research, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas, KS, USA
| | - Michael J Soares
- Institute for Reproduction and Perinatal Research, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas, KS, USA.,Department of Pediatrics, University of Kansas Medical Center, Kansas, KS, USA.,Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas, KS, USA.,Center for Perinatal Research, Children's Mercy Research Institute, Children's Mercy, Kansas, MO, USA
| |
Collapse
|
26
|
Wang H, Li F, Miao M, Yu Y, Ji H, Liu H, Huang R, Obel C, Zhang J, Li J. Maternal spontaneous abortion and the risk of attention-deficit/hyperactivity disorder in offspring: a population-based cohort study. Hum Reprod 2021; 35:1211-1221. [PMID: 32340041 DOI: 10.1093/humrep/deaa035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 02/03/2020] [Accepted: 02/11/2020] [Indexed: 12/19/2022] Open
Abstract
STUDY QUESTION Is a maternal history of spontaneous abortion (SA) associated with an increased risk of attention-deficit/hyperactivity disorder (ADHD) in offspring? SUMMARY ANSWER Our results suggest an association between maternal history of SA and ADHD in offspring, with the risk increasing with the number of maternal SA and highest in the firstborn children whose mothers had had recurrent SAs after adjusting for a number of potential confounders. WHAT IS KNOWN ALREADY A history of SA has been associated with more complications in next pregnancies and adverse childbirth outcomes, which are risk factors for ADHD in the offspring. However, no previous study has investigated whether maternal SA increases risk of ADHD in the offspring. STUDY DESIGN, SIZE, DURATION This population-based study included all live-born children in Denmark from 1 January 1995 to 31 December 2012 (n = 1 062 667). All children were followed from 3 years of age until the day of ADHD diagnosis, death, emigration or 31 December 2016, whichever came first. PARTICIPANTS/MATERIALS, SETTING, METHODS There were 130 206 (12.2%) children born to mothers who had at least one SA. Cox proportional hazards regression models were used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs). MAIN RESULTS AND THE ROLE OF CHANCE During a median follow-up of 9.4 years (interquartile range, 5.4-14.3), 25 747 children were diagnosed with ADHD. Overall, children of mothers with a history of SA had an increased rate of ADHD (HR, 1.11; 95% CI, 1.07 to 1.15). The HRs increased with the number of maternal SA, 1.09 (95% CI, 1.05 to 1.13) for one SA and 1.22 (95% CI, 1.12 to 1.33) for at least two SAs, respectively. These findings were consistent when we took into consideration a number of factors, such as maternal socioeconomic status, type of SA, birth order, parental history of psychiatric disorders, pregnancy characteristics and adverse birth outcomes. LIMITATIONS, REASONS FOR CAUTION Misclassification of SA was possible as we used population-based register data to capture maternal history of SA. However, any misclassification of maternal history of SA would be non-differential with regard to the diagnosis of ADHD in offspring, which generally leads to underestimation of the associations. Furthermore, probabilistic sensitivity analysis suggested that only 1% of change in the estimate may have been due to misclassification of SA. WIDER IMPLICATIONS OF THE FINDINGS SA is quite frequent (varying from 15 to 20%), and a small increase of neurodevelopmental problems in offspring could have major public health implications. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by grants from the National Natural Science Foundation of China (No. 81703237, No. 81530086 and No. 81761128035), National Key Research and Development Program (2018YFC1002801, 2016YFC1000505), Shanghai Municipal Commission of Health and Family Planning (No. 2017ZZ02026, No. 2017EKHWYX-02), the Novo Nordisk Foundation (NNF18OC0052029), the Danish Council for Independent Research (DFF-6110-00019), the Nordic Cancer Union (176673, 186200 and R217-A13234-18-S65), Karen Elise Jensens Fond (2016) and Xinhua Hospital of Shanghai Jiao Tong University School of Medicine (2018YJRC03). All authors report no conflict of interest. TRIAL REGISTRATION NUMBER NA.
Collapse
Affiliation(s)
- Hui Wang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Li
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Maohua Miao
- NHC Key Lab. of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, China
| | - Yongfu Yu
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Honglei Ji
- NHC Key Lab. of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai, China
| | - Hui Liu
- School of Public Health/Medical Informatics Center, Peking University, Beijing, China
| | - Rong Huang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Carsten Obel
- Unit of Mental Public Health, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Jun Zhang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiong Li
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
27
|
A pilot study on investigating the role of Salvia miltiorrhiza in fetal growth restriction. Biosci Rep 2021; 40:224149. [PMID: 32406912 PMCID: PMC7269916 DOI: 10.1042/bsr20201222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/06/2020] [Accepted: 05/14/2020] [Indexed: 11/17/2022] Open
Abstract
To date there is no effective treatment for pregnancies complicated by fetal growth restriction (FGR). Salvia miltiorrhiza, a traditional Chinese herb has been shown to promote blood flow and improve microcirculatory disturbance. In this pilot study, we evaluated whether S. miltiorrhiza can potentially become a possible therapy for FGR. Nineteen pregnant women with FGR were treated with S. miltiorrhiza and ATP supplementation for an average of 7 days, and 17 cases received ATP supplementation as controls. The estimated fetal weights (EFWs) were measured by ultrasound after treatment, and the birthweights were recorded after birth. After treatment with S. miltiorrhiza, 7 (37%) FGR cases showed an increase in EFW to above the 10th percentile, compared with 4 (23%) FGR cases in controls (odds ratio: 1.896, 95% confidence limits (CLs): 0.44-8.144). At delivery, 10 (53%) FGR cases in the treatment group delivered babies with a birthweight above the 10th percentile, compared with 6 (35%) FGR cases in the control group (odds ratio: 2.037, 95% CL: 0.532-7.793); 80 or 64% FGR cases in the treatment group showed an increase in fetal abdominal circumference (AC) or biparietal diameter (BPD) above the 10th percentile before delivery. While 44 or 30% FGR cases in the control group showed an increase in AC or BPD. No improvement of head circumference (HC) or femur length (FL) was seen. These pilot data suggest the need for multicenter randomized clinical trials on the potential of S. miltiorrhiza to improve perinatal outcome in pregnant women complicated by FGR.
Collapse
|
28
|
Wei Y, Zhang C, Fan G, Meng L. Organoids as Novel Models for Embryo Implantation Study. Reprod Sci 2021; 28:1637-1643. [PMID: 33650092 DOI: 10.1007/s43032-021-00501-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 02/14/2021] [Indexed: 10/22/2022]
Abstract
In the last decade, organoids have become emerging novel models for biomedical research. Organoids are small, self-organized three-dimensional (3D) tissue cultures derived from stem cells that mimic certain tissues or organs. In reproductive medicine, researchers have generated numerous organoids including blastoid (blastocyst organoid), endometrial organoid, and trophoblast organoid. These organdies provide useful models for studying the embryo implantation mechanism through observation of cell differentiation, gene expression, and epigenetic profiles at the implantation stage. As in vitro tissue models, organoids could be coupled with many other frontier technologies such as gene editing and genomic sequencing. However, the main drawback of organoids is that they do not fully mimic their counterparts in vivo tissues. Furthermore, there is a consensus of research ethics on organoids that may limit the types of studies that scientists perform with. Nevertheless, all discoveries and efforts surrounding organoids still greatly benefit therapy development for reproductive clinics.
Collapse
Affiliation(s)
- Yubao Wei
- Institute of Reproductive Medicine, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, 450003, China.
| | - Cuilian Zhang
- Institute of Reproductive Medicine, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, 450003, China.
| | - Guoping Fan
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Li Meng
- Incinta Fertility Center, Los Angeles, CA, 90503, USA
| |
Collapse
|
29
|
Rampersaud AM, Dunk CE, Lye SJ, Renaud SJ. Palmitic acid induces inflammation in placental trophoblasts and impairs their migration toward smooth muscle cells through plasminogen activator inhibitor-1. Mol Hum Reprod 2020; 26:850-865. [PMID: 32898274 DOI: 10.1093/molehr/gaaa061] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 08/21/2020] [Indexed: 12/12/2022] Open
Abstract
A critical component of early human placental development includes migration of extravillous trophoblasts (EVTs) into the decidua. EVTs migrate toward and displace vascular smooth muscle cells (SMCs) surrounding several uterine structures, including spiral arteries. Shallow trophoblast invasion features in several pregnancy complications including preeclampsia. Maternal obesity is a risk factor for placental dysfunction, suggesting that factors within an obese environment may impair early placental development. Herein, we tested the hypothesis that palmitic acid, a saturated fatty acid circulating at high levels in obese women, induces an inflammatory response in EVTs that hinders their capacity to migrate toward SMCs. We found that SMCs and SMC-conditioned media stimulated migration and invasion of an EVT-like cell line, HTR8/SVneo. Palmitic acid impaired EVT migration and invasion toward SMCs, and induced expression of several vasoactive and inflammatory mediators in EVTs, including endothelin, interleukin (IL)-6, IL-8 and PAI1. PAI1 was increased in plasma of women with early-onset preeclampsia, and PAI1-deficient EVTs were protected from the anti-migratory effects of palmitic acid. Using first trimester placental explants, palmitic acid exposure decreased EVT invasion through Matrigel. Our findings reveal that palmitic acid induces an inflammatory response in EVTs and attenuates their migration through a mechanism involving PAI1. High levels of palmitic acid in pathophysiological situations like obesity may impair early placental development and predispose to placental dysfunction.
Collapse
Affiliation(s)
- Amanda M Rampersaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Caroline E Dunk
- Research Centre for Women's and Infants' Health, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Stephen J Lye
- Research Centre for Women's and Infants' Health, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Stephen J Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Children's Health Research Institute, Lawson Health Research Institute, London, ON, Canada
| |
Collapse
|
30
|
Wang G, Huang Y, Hu T, Zhang B, Tang Z, Yao R, Huang Y, Fan X, Ni X. Contribution of placental 11β-HSD2 to the pathogenesis of preeclampsia. FASEB J 2020; 34:15379-15399. [PMID: 32978833 DOI: 10.1096/fj.202001003rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Preeclampsia, a major human pregnancy-specific disorder, leads to maternal and fetal morbidity and mortality. Here we reported that 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2), an enzyme that degrades active glucocorticoids, is one of the key factors that contributes to preeclampsia development. In the pregnant rat model, we firstly confirmed that administration of 11β-HSD2 inhibitor carbenoxolone (CBX) subcutaneously or by placenta-targeted delivery system could lead to a decrease in placental 11β-HSD2 expression and activity and an increase in corticosterone level in placenta and maternal circulation. Then, we showed that subcutaneous administration and placenta-targeted delivery of CBX resulted in the hallmark of preeclampsia-like features including hypertension, proteinuria, renal damages as well as elevated circulatory soluble fms-like tyrosine kinase 1 (sFlt1) and increased sFlt1/placental growth factor (PlGF) ratio in pregnant rats. These animals displayed decreased trophoblast invasion in uterus, impaired spiral artery remodeling, and reduced placental blood flow. Preeclampsia-like features could also be induced by administration of dexamethasone in pregnant rats. In the cultured human trophoblast models, we found that cortisol only inhibited migration and invasion of the extravillous trophoblasts with 11β-HSD2 knockdown, and promoted sFlt1 release in the cultured syncytiotrophoblasts with 11β-HSD2 knockdown. Furthermore, we elucidated that cortisol stimulated a disintegrin and metalloprotease (ADAM)17 expression in placentas, thereby promoting sFlt1 release in placenta. Collectively, our study provided the evidence that placental 11β-HSD2 dysfunction plays a key role in the development of preeclampsia and immediately identified innovative target to counteract preeclampsia.
Collapse
Affiliation(s)
- Gang Wang
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, China.,Department of Physiology, Second Military Medical University, Shanghai, China
| | - Yan Huang
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, China.,Department of Physiology, Second Military Medical University, Shanghai, China
| | - Tianxiao Hu
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Baozhen Zhang
- Center for Reproduction and Health Development, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhengshan Tang
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, China
| | - Ruojing Yao
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, China
| | - Ying Huang
- Maternity and Child Health Hospital of Pudong New District, Shanghai, China
| | - Xiujun Fan
- Center for Reproduction and Health Development, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xin Ni
- Department of Gynecology and Obstetrics and Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, China.,Department of Physiology, Second Military Medical University, Shanghai, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, China
| |
Collapse
|
31
|
Evans MB, Nobles CJ, Kim K, Hill MJ, DeCherney AH, Silver RM, Mumford SL, Sjaarda LA, Perkins NJ, Schisterman EF. Low-dose aspirin in reproductive health: effects on menstrual cycle characteristics. Fertil Steril 2020; 114:1263-1270. [PMID: 32896390 DOI: 10.1016/j.fertnstert.2020.06.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/01/2020] [Accepted: 06/11/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVE To estimate the effect of daily 81 mg low-dose aspirin (LDA) on menstrual cycle length and hormone profiles. DESIGN Secondary analysis of a trial evaluating the effect of daily LDA or placebo on live birth among women with one or two previous pregnancy losses. SETTING University medical centers. PATIENT(S) A total of 915 regularly menstruating women who had at least one menstrual cycle (3,190 total cycles) in which pregnancy did not occur. INTERVENTION(S) Randomized allocation to LDA versus placebo. MAIN OUTCOME MEASURE(S) Menstrual cycle length and follicular and luteal phases were measured. Urinary pregnanediol glucuronide, follicle-stimulating hormone, luteinizing hormone, and estrone-3-glucuronide were assessed up to six times during the first two cycles. Generalized estimating equations estimated relative risk of short (<25th percentile: <27 days) and long (>75th percentile: ≥32 days) versus normal cycle length. Linear mixed models estimated mean hormone level differences with weights used to account for multiple cycles contributed per participant. RESULT(S) There were no significant differences in total menstrual cycle, follicular phase, or luteal phase length between LDA and placebo groups. LDA posed no greater risk of having a short versus normal-length or long versus normal-length follicular phase, or having a short versus normal-length or long versus normal-length luteal phase. There were no significant differences in hormone levels across the menstrual cycle between the LDA and placebo groups. CONCLUSION(S) Daily LDA use did not result in any changes to menstrual cycle, follicular phase, or luteal phase length or hormone levels across the menstrual cycle compared with placebo. CLINICAL TRIAL REGISTRATION NUMBER NCT00467363.
Collapse
Affiliation(s)
- M Blake Evans
- Program in Reproductive Endocrinology and Gynecology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Carrie J Nobles
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Keewan Kim
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Micah J Hill
- Program in Reproductive Endocrinology and Gynecology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Alan H DeCherney
- Program in Reproductive Endocrinology and Gynecology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Robert M Silver
- Department of Obstetrics and Gynecology, University of Utah School of Medicine, Salt Lake City, Utah
| | - Sunni L Mumford
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Lindsey A Sjaarda
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Neil J Perkins
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Enrique F Schisterman
- Program in Reproductive Endocrinology and Gynecology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
32
|
Wang W, Sung N, Gilman-Sachs A, Kwak-Kim J. T Helper (Th) Cell Profiles in Pregnancy and Recurrent Pregnancy Losses: Th1/Th2/Th9/Th17/Th22/Tfh Cells. Front Immunol 2020; 11:2025. [PMID: 32973809 PMCID: PMC7461801 DOI: 10.3389/fimmu.2020.02025] [Citation(s) in RCA: 249] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/27/2020] [Indexed: 12/15/2022] Open
Abstract
During pregnancy, various immune effectors and molecules participating in the immune-microenvironment establish specific maternal tolerance toward the semi-allogeneic fetus. Activated maternal immune effectors by the trophoblast antigens, such as T helper (Th), T cytotoxic (Tc), T regulatory (Treg), and B cells, are involved in the regulation of adaptive immunity. Recognition of active signal through the T cell receptors stimulate the differentiation of naive CD3+CD4+ T cells into specific T cell subsets, such as Th1, Th2, Th9, Th17, Th22, and follicular Th cells (Tfh). Each of these subsets has a significant and distinct role in human pregnancy. Th1 immunity, characterized by immune-inflammatory responses, becomes dominant during the peri-implantation period, and the “controlled” Th1 immunity benefits the invading trophoblasts rather than harm. Quickly after the placental implantation, the early inflammatory Th1 immunity is shifted to the Th2 anti-inflammatory immune responses. The predominant Th2 immunity, which overrules the Th1 immunity at the placental implantation site, protects a fetus by balancing Th1 immunity and accommodate fetal and placental development. Moreover, Treg and Th9 cells regulate local inflammatory immune responses, potentially detrimental to the fetus. Th17 cells induce protective immunity against extracellular microbes during pregnancy. However, excessive Th17 immunity may induce uncontrolled neutrophil infiltration at the maternal-fetal interface. Other Th cell subsets such as Tfh cells, also contribute to pregnancy by setting up favorable humoral immunity during pregnancy. However, dysregulation of Th cell immunity during pregnancy may result in obstetrical complications, such as recurrent pregnancy losses (RPL) and preeclampsia (PE). With this review, we intend to deliver a comprehensive overview of CD4+ Th cell subsets, including Th1, Th2, Th9, Th17, Th22, and Tfh cells, in human pregnancy by reviewing their roles in normal and pathological pregnancies.
Collapse
Affiliation(s)
- Wenjuan Wang
- Reproductive Medicine and Immunology, Obstetrics and Gynecology, Department of Clinical Sciences, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States.,Reproductive Medicine Center, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Nayoung Sung
- Reproductive Medicine and Immunology, Obstetrics and Gynecology, Department of Clinical Sciences, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Alice Gilman-Sachs
- Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States.,Clinical Immunology Laboratory, Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Joanne Kwak-Kim
- Reproductive Medicine and Immunology, Obstetrics and Gynecology, Department of Clinical Sciences, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States.,Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|
33
|
Abstract
Pregnancy is a natural process that poses an immunological challenge because non-self fetus must be accepted. During the pregnancy period, the fetus as 'allograft' inherits maternal and also paternal antigens. For successful and term pregnancy, the fetus is tolerated and nurtured enjoying immune privileges that minimize the risk of being rejected by maternal immune system. Multiple mechanisms contribute to tolerate the semi-allogeneic fetus. Here, we summarize the recent progresses on how the maternal immune system actively collaborates to maintain the immune balance and maternal-fetal tolerance.
Collapse
Affiliation(s)
- Xiaopeng Li
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jiayi Zhou
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Min Fang
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,International College, University of the Chinese Academy of Sciences, Beijing, China
| | - Bolan Yu
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
34
|
Cappelletti M, Presicce P, Kallapur SG. Immunobiology of Acute Chorioamnionitis. Front Immunol 2020; 11:649. [PMID: 32373122 PMCID: PMC7177011 DOI: 10.3389/fimmu.2020.00649] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 03/23/2020] [Indexed: 12/19/2022] Open
Abstract
Acute chorioamnionitis is characterized by neutrophilic infiltration and inflammation at the maternal fetal interface. It is a relatively common complication of pregnancy and can have devastating consequences including preterm labor, maternal infections, fetal infection/inflammation, fetal lung, brain, and gastrointestinal tract injury. In this review, we will discuss current understanding of the pathogenesis, immunobiology, and mechanisms of this condition. Most commonly, acute chorioamnionitis is a result of ascending infection with relatively low-virulence organisms such as the Ureaplasma species. Furthermore, recent vaginal microbiome studies suggest that there is a link between vaginal dysbiosis, vaginal inflammation, and ascending infection. Although less common, microorganisms invading the maternal-fetal interface via hematogenous route (e.g., Zika virus, Cytomegalovirus, and Listeria) can cause placental villitis and severe fetal inflammation and injury. We will provide an overview of the knowledge gleaned from different animal models of acute chorioamnionitis and the role of different immune cells in different maternal-fetal compartments. Lastly, we will discuss how infectious agents can break the maternal tolerance of fetal allograft during pregnancy and highlight the novel future therapeutic approaches.
Collapse
Affiliation(s)
- Monica Cappelletti
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | - Pietro Presicce
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | - Suhas G Kallapur
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
35
|
Presicce P, Cappelletti M, Senthamaraikannan P, Ma F, Morselli M, Jackson CM, Mukherjee S, Miller LA, Pellegrini M, Jobe AH, Chougnet CA, Kallapur SG. TNF-Signaling Modulates Neutrophil-Mediated Immunity at the Feto-Maternal Interface During LPS-Induced Intrauterine Inflammation. Front Immunol 2020; 11:558. [PMID: 32308656 PMCID: PMC7145904 DOI: 10.3389/fimmu.2020.00558] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/11/2020] [Indexed: 01/10/2023] Open
Abstract
Accumulation of activated neutrophils at the feto-maternal interface is a defining hallmark of intrauterine inflammation (IUI) that might trigger an excessive immune response during pregnancy. Mechanisms responsible of this massive neutrophil recruitment are poorly investigated. We have previously showed that intraamniotic injection of LPS in rhesus macaques induced a neutrophil predominant inflammatory response similar to that seen in human IUI. Here, we demonstrate that anti-TNF antibody (Adalimumab) inhibited ~80% of genes induced by LPS involved in inflammatory signaling and innate immunity in chorio-decidua neutrophils. Consistent with the gene expression data, TNF-blockade decreased LPS-induced neutrophil accumulation and activation at the feto-maternal interface. We also observed a reduction in IL-6 and other pro-inflammatory cytokines but not prostaglandins concentrations in the amniotic fluid. Moreover, TNF-blockade decreased mRNA expression of inflammatory cytokines in the chorio-decidua but not in the uterus, suggesting that inhibition of TNF-signaling decreased the inflammation in a tissue-specific manner within the uterine compartment. Taken together, our results demonstrate a predominant role for TNF-signaling in modulating the neutrophilic infiltration at the feto-maternal interface during IUI and suggest that blockade of TNF-signaling could be considered as a therapeutic approach for IUI, the major leading cause of preterm birth.
Collapse
Affiliation(s)
- Pietro Presicce
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Monica Cappelletti
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Paranthaman Senthamaraikannan
- Division of Neonatology/Pulmonary Biology, Cincinnati Children's Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Feiyang Ma
- Department of Molecular, Cell and Developmental Biology Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Marco Morselli
- Department of Molecular, Cell and Developmental Biology Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Institute for Quantitative and Computational Biosciences-Collaboratory, University of California, Los Angeles, Los Angeles, CA, United States
| | - Courtney M Jackson
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Shibabrata Mukherjee
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Lisa A Miller
- California National Primate Research Center, University of California, Davis, Davis, CA, United States.,Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Institute for Quantitative and Computational Biosciences-Collaboratory, University of California, Los Angeles, Los Angeles, CA, United States
| | - Alan H Jobe
- Division of Neonatology/Pulmonary Biology, Cincinnati Children's Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Claire A Chougnet
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Suhas G Kallapur
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
36
|
Fu J, Li L, Qi L, Zhao L. A randomized controlled trial of etanercept in the treatment of refractory recurrent spontaneous abortion with innate immune disorders. Taiwan J Obstet Gynecol 2020; 58:621-625. [PMID: 31542082 DOI: 10.1016/j.tjog.2019.07.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2019] [Indexed: 10/26/2022] Open
Abstract
OBJECTIVE To evaluate the effectiveness of etanercept in the treatment of refractory recurrent spontaneous abortion with innate immune disorders. MATERIALS AND METHODS A randomized controlled trial in patients with refractory innate immune RSA was conducted in our hospital. 188 patients were selected, all with at least 4 consecutive miscarriages and caused by innate immunity disorders. Patients were randomly allocated into 2 groups. One group (n = 95) used etanercept 25 mg per week starting from the first day after menstruation, while the other (n = 93) with placebo. Delivery of a healthy baby without malformations was regarded as the primary outcome. RESULTS In etanercept group, 85 (89.47%) patients delivered a healthy baby, while in placebo group, this number was only 67 (72.04%) [P = 0.01, OR = 3.30; 95% CI(1.49~7.32)]. Significantly lower levels of TNF-α and NK cell activity were observed in gestation weeks 4-10 in etanercept group versus placebo group (P < 0.05). CONCLUSION The results provide a proof of principle that etanercept can be an attractive therapeutic strategy for refractory innate immune RSA.
Collapse
Affiliation(s)
- Jinhua Fu
- Department of Obstetrics, Qingdao Jinhua Hospital, Qingdao, 266200, China; Department of Obstetrics, Weifang People's Hospital, Weifang, 261041, China
| | - Lin Li
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Lijuan Qi
- Department of Obstetrics, Qingdao Jinhua Hospital, Qingdao, 266200, China
| | - Long Zhao
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
37
|
Thromboelastography testing in mice following blood collection from facial vein and cardiac puncture. Blood Coagul Fibrinolysis 2020; 30:366-369. [PMID: 31318718 DOI: 10.1097/mbc.0000000000000836] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
: Blood collection is critical for mouse research studies particularly in hemostatic testing. Cardiac puncture; a standard effective method requires anesthesia and is a terminal procedure while facial vein technique allows multiple collections. Thromboelastography (TEG) is a global hemostasis test, provides a dynamic real-time picture of coagulation. However, TEG experiments in mice require large number of animals and may not allow pre/postinterventions assessment. In this study, we aimed to investigate the feasibility of facial vein sampling for TEG analysis as an alternative to cardiac puncture and examined the impact on coagulation results. Blood samples were obtained from a total of 10 C57BL/6 and CD-1 mice via cardiac puncture and a total of another eight mice of similar strains via facial vein sampling. We compared TEG parameters in both methods using descriptive statistics and the Student t test. Results show no significant difference in any of the TEG parameters between cardiac and facial vein blood indicating the two methods are comparable. Facial vein sampling provides a less costly alternative to cardiac puncture. It is a suitable blood collection method for pre/postinterventions or follow-up studies and it better addresses reduction and refinement goals in mouse studies. A larger study to evaluate the sex or strain and genetic background differences will be valuable.
Collapse
|
38
|
Baines KJ, Rampersaud AM, Hillier DM, Jeyarajah MJ, Grafham GK, Eastabrook G, Lacefield JC, Renaud SJ. Antiviral Inflammation during Early Pregnancy Reduces Placental and Fetal Growth Trajectories. THE JOURNAL OF IMMUNOLOGY 2019; 204:694-706. [PMID: 31882516 DOI: 10.4049/jimmunol.1900888] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/28/2019] [Indexed: 12/22/2022]
Abstract
Many viruses are detrimental to pregnancy and negatively affect fetal growth and development. What is not well understood is how virus-induced inflammation impacts fetal-placental growth and developmental trajectories, particularly when inflammation occurs in early pregnancy during nascent placental and embryo development. To address this issue, we simulated a systemic virus exposure in early pregnant rats (gestational day 8.5) by administering the viral dsRNA mimic polyinosinic:polycytidylic acid (PolyI:C). Maternal exposure to PolyI:C induced a potent antiviral response and hypoxia in the early pregnant uterus, containing the primordial placenta and embryo. Maternal PolyI:C exposure was associated with decreased expression of the maternally imprinted genes Mest, Sfrp2, and Dlk1, which encode proteins critical for placental growth. Exposure of pregnant dams to PolyI:C during early pregnancy reduced fetal growth trajectories throughout gestation, concomitant with smaller placentas, and altered placental structure at midgestation. No detectable changes in placental hemodynamics were observed, as determined by ultrasound biomicroscopy. An antiviral response was not evident in rat trophoblast stem (TS) cells following exposure to PolyI:C, or to certain PolyI:C-induced cytokines including IL-6. However, TS cells expressed high levels of type I IFNR subunits (Ifnar1 and Ifnar2) and responded to IFN-⍺ by increasing expression of IFN-stimulated genes and decreasing expression of genes associated with the TS stem state, including Mest IFN-⍺ also impaired the differentiation capacity of TS cells. These results suggest that an antiviral inflammatory response in the conceptus during early pregnancy impacts TS cell developmental potential and causes latent placental development and reduced fetal growth.
Collapse
Affiliation(s)
- Kelly J Baines
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
| | - Amanda M Rampersaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
| | - Dendra M Hillier
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
| | - Mariyan J Jeyarajah
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
| | - Grace K Grafham
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1
| | - Genevieve Eastabrook
- Department of Obstetrics and Gynaecology, University of Western Ontario, London, Ontario, Canada N6H 5W9.,Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada N6C 2V5
| | - James C Lacefield
- Department of Electrical and Computer Engineering, School of Biomedical Engineering, University of Western Ontario, London, Ontario, Canada N6A 3K7.,Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada N6A 3K7; and.,Robarts Research Institute, University of Western Ontario, London, Ontario, Canada N6A 5B7
| | - Stephen J Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada N6A 5C1; .,Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada N6C 2V5
| |
Collapse
|
39
|
Tok A, Ozer A, Kanat-Pektas M, Aral M, Sakalli H, Aydogdu S, Yutan-Kaya E, Sager H. The role of omentin in early pregnancy losses. J OBSTET GYNAECOL 2019; 40:107-110. [PMID: 31495295 DOI: 10.1080/01443615.2019.1606179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
This prospective case-control study aimed to investigate the role of omentin, an anti-inflammatory adipokine in early pregnancy losses. The study comprised 47 women with spontaneous miscarriage at a gestational age of 8-12 weeks and 36 healthy pregnant women, matched for age, body mass index and gestational age, gravdity and parity. A significant negative correlation was determined between plasma omentin concentrations and body weight (r= -0.242, p = .027) and gestational age (r= -0.249, p = .023). Although not statistically, the women with spontaneous miscarriage had higher plasma concentrations of omentin compared to those with healthy pregnancies (7.798 ± 3.453 ng/ml vs. 7.200 ± 3.442 ng/ml, p = .435). This finding might support the hypothesis that increased inflammation plays a role in the etiopathogenesis of early pregnancy losses. These results revealed the potential use of omentin to predict unhealthy pregnancies.Impact statementWhat is already known on the subject of the paper? The exact mechanism of early pregnancy loss with euploid foetal karyotype has not been elucidated yet. An alteration in the physiological inflammatory response of pregnancy might be one of the mechanisms responsible for miscarriage.What does this study add? To the best of our knowledge, this is the first study to investigate the role of omentin in early pregnancy loss. The results obtained from this current study could be used to clarify the relationship between inflammatory processes and miscarriage.What are the implications for clinical practice and/or further research? Identification of the role of omentin in the process of early pregnancy losses would be helpful in order to design further studies to determine the feasibility of using omentin as a serum marker to predict the risk of miscarriage in early pregnancies. Additionally, understanding of the etiopathogenesis of early pregnancy losses with euploid karyotype will give a lead to further researches which could focus on exploring new interventions to detect and treat altered inflammation in early pregnancies.
Collapse
Affiliation(s)
- Abdullah Tok
- Department of Obstetrics and Gynecology, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| | - Alev Ozer
- Department of Obstetrics and Gynecology, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| | - Mine Kanat-Pektas
- Department of Obstetrics and Gynecology, Afyon Kocatepe University, Afyonkarahisar, Turkey
| | - Murat Aral
- Department of Microbiology, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| | - Hilal Sakalli
- Department of Obstetrics and Gynecology, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| | - Sakir Aydogdu
- Department of Obstetrics and Gynecology, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| | - Esra Yutan-Kaya
- Department of Microbiology, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| | - Hatice Sager
- Department of Biochemistry, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| |
Collapse
|
40
|
Hu J, Zhang J, Zhu B. Protective effect of metformin on a rat model of lipopolysaccharide-induced preeclampsia. Fundam Clin Pharmacol 2019; 33:649-658. [PMID: 31334867 DOI: 10.1111/fcp.12501] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 07/05/2019] [Accepted: 07/18/2019] [Indexed: 12/23/2022]
Abstract
Recent in vitro and clinical studies have found that metformin (MET) may play a preventive or therapeutic role in preeclampsia (PE) and may be a candidate drug for the prevention and/or treatment of PE. In this study, we used lipopolysaccharide (LPS) to induce a PE-like rat model and investigated the intervention effect of MET from the perspectives of clinical manifestations, placental morphology, serum marker for placental injury, systemic inflammatory response and oxidative/nitrative stress, and placental nuclear factor-κB (NF-κB) signaling. The results showed that MET improved LPS-induced hypertension, proteinuria, fetal growth restriction (FGR) and stillbirth, alleviated placental injury and decreased maternal serum marker alpha-fetoprotein (MS-AFP) level; MET suppressed LPS-induced TNF-α and IL-6 productions, reduced oxidative/nitrative stress as evidenced by increased superoxide dismutase (SOD) activity, decreased inducible nitric oxide synthase (iNOS) activity, and decreased levels of malondialdehyde (MDA) and nitric oxide (NO); MET inhibited LPS-induced NF-κB activation in placentas. Based on these findings, it can be concluded that MET is beneficial to the PE-like rat model by protecting placentas from injury, suppressing systemic inflammatory response and oxidative/nitrative stress, and inhibiting placental NF-κB signaling pathway. MET is a promising drug for prevention and/or treatment of PE.
Collapse
Affiliation(s)
- Jilin Hu
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China.,Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Jinman Zhang
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China.,National Health Commission's Key Laboratory for Healthy Births in Western China, Department of Obstetrics and Gynecology, First People's Hospital of Yunnan Province, Kunming, 650032, China
| | - Baosheng Zhu
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China.,National Health Commission's Key Laboratory for Healthy Births in Western China, Department of Obstetrics and Gynecology, First People's Hospital of Yunnan Province, Kunming, 650032, China
| |
Collapse
|
41
|
Yu L, Wang T, Que R, Yang J, Wang Z, Jiang X, Wang L. The potentially protective role of ATP-binding cassette transporters in preeclampsia via Nrf2. Pregnancy Hypertens 2019; 18:21-28. [PMID: 31445436 DOI: 10.1016/j.preghy.2019.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 07/25/2019] [Accepted: 08/09/2019] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Preeclampsia (PE) is a severe placental syndrome that likely results from placental oxidative stress and inflammation, and can lead to maternal hypertension and premature delivery. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) activates several genes involved in antioxidant defense in the placentae, along with the ATP-binding cassette (ABC) transporters which regulate substrate flow between maternal and fetal circulation. Although several ABC transporters are down-regulated in PE, their exact mechanistic role is poorly understood. METHODS& RESULTS: In this study, we compared the levels of major ABC transporters and NRF2 in placentae of healthy full-term pregnant women and those with early and term onset PE. We found a significant decrease in the levels of Nrf2 and several ABC transporters in the placentae of early onset compared to term onset PE. In addition, women with term onset PE showed improved post-partum parameters (lower blood pressure, and greater placental and neonatal weights) compared to those with early onset PE. Mechanistically, Nrf2 knockdown/knockout downregulated the genes for ABC transporters and antioxidant enzymes, and upregulated pro-inflammatory factors, whereas Nrf2 upregulation had the opposite effects. CONCLUSIONS Nrf2 protects the placenta against PE by activating the ABC transporter-mediated efflux, indicating a novel target in PE therapy.
Collapse
Affiliation(s)
- Lu Yu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Ting Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China; Department of Pharmacy, Sichuan Cancer Hospital & Institution, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Rongcheng Que
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jillian Yang
- Department of Pharmaceutical Sciences, College of Pharmacy, Marshall B. Ketchum University, Fullerton, CA 92831, USA
| | - Zhijun Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, Marshall B. Ketchum University, Fullerton, CA 92831, USA
| | - Xuehua Jiang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Ling Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
42
|
Sharami SH, Gholipour M, Milani F, Kazemnejad E, Heirati SFD, Ranjbar ZA. The Association between Dyslipidemia and Preterm Birth: A Prospective Cohort Study in The North of Iran. Endocr Metab Immune Disord Drug Targets 2019; 20:227-233. [PMID: 31142254 DOI: 10.2174/1871530319666190529090517] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 04/25/2019] [Accepted: 04/30/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Dyslipidemia is recognized as an important factor in the incidence of Preterm Birth (PTB). The early diagnosis of factors affecting PTB is important in the reduction of maternalneonatal complications; therefore, we aimed to evaluate the association between dyslipidemia and PTB in women from the Guilan province of Iran. METHODS The current investigation was a prospective cohort study on 378 pregnant women in Rasht city (Guilan province), during 2018-2019. Samples were randomly selected among pregnant women who referred to Al-Zahra hospital. Association of the lipid profiles: Total Cholesterol (TC), High- Density Lipoprotein (HDLC), Low-Density Lipoprotein (LDLC), and Triglycerides (TG) with PTB was assessed using the Chi-square, Fisher tests and logistic regression analysis. RESULTS Our findings showed that of the evaluated lipid profiles, TG and TC had the highest predictive power with AUC =0.833 (95٪, CI: 0.736-0.930) and 0.772 (95%, CI: 0.676-0.867), respectively; also, their sensitivity and specificity were 83.3%, 70.2% and 83.3%, 66.1%, respectively. Moreover, abnormal LDL concentrations increase the risk of PTB by two folds (P < 0.05). CONCLUSION It seems that by controlling the lipid profiles of pregnant women, the risk of PTB could be reduced.
Collapse
Affiliation(s)
- Seyedeh H Sharami
- Department of Obstetrics & Gynecology, Reproductive Health Research Center, Azzhra Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mahboobeh Gholipour
- Department of Cardiology, Cardiovascular Diseases Research Center, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Forozan Milani
- Department of Obstetrics & Gynecology, Reproductive Health Research Center, Azzhra Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ehsan Kazemnejad
- Department of Biostatictis, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Seyedeh F D Heirati
- Department of Obstetrics & Gynecology, Reproductive Health Research Center, Azzhra Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Zahra A Ranjbar
- Department of Endocrinology and Metabolism, Reproductive Health Research center, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
43
|
Jørgensen N, Persson G, Hviid TVF. The Tolerogenic Function of Regulatory T Cells in Pregnancy and Cancer. Front Immunol 2019; 10:911. [PMID: 31134056 PMCID: PMC6517506 DOI: 10.3389/fimmu.2019.00911] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 04/09/2019] [Indexed: 12/12/2022] Open
Abstract
Regulatory T cells, a subpopulation of suppressive T cells, are potent mediators of self-tolerance and essential for the suppression of triggered immune responses. The immune modulating capacity of these cells play a major role in both transplantation, autoimmune disease, allergy, cancer and pregnancy. During pregnancy, low numbers of regulatory T cells are associated with pregnancy failure and pregnancy complications such as pre-eclampsia. On the other hand, in cancer, low numbers of immunosuppressive T cells are correlated with better prognosis. Hence, maternal immune tolerance toward the fetus during pregnancy and the escape from host immunosurveillance by cancer seem to be based on similar immunological mechanisms being highly dependent on the balance between immune activation and suppression. As regulatory T cells hold a crucial role in several biological processes, they may also be promising subjects for therapeutic use. Especially in the field of cancer, cell therapy and checkpoint inhibitors have demonstrated that immune-based therapies have a very promising potential in treatment of human malignancies. However, these therapies are often accompanied by adverse autoimmune side effects. Therefore, expanding the knowledge to recognize the complexities of immune regulation pathways shared across different immunological scenarios is extremely important in order to improve and develop new strategies for immune-based therapy. The intent of this review is to highlight the functional characteristics of regulatory T cells in the context of mechanisms of immune regulation in pregnancy and cancer, and how manipulation of these mechanisms potentially may improve therapeutic options.
Collapse
Affiliation(s)
| | | | - Thomas Vauvert F. Hviid
- Department of Clinical Biochemistry, Centre for Immune Regulation and Reproductive Immunology (CIRRI), The ReproHealth Consortium ZUH, Zealand University Hospital, and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
44
|
Oral cholecalciferol supplementation alleviates lipopolysaccharide-induced preterm delivery partially through regulating placental steroid hormones and prostaglandins in mice. Int Immunopharmacol 2019; 69:235-244. [DOI: 10.1016/j.intimp.2019.01.052] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 01/24/2019] [Accepted: 01/30/2019] [Indexed: 12/19/2022]
|
45
|
Akerele OA, Cheema SK. A diet enriched in longer chain omega-3 fatty acids reduced placental inflammatory cytokines and improved fetal sustainability of C57BL/6 mice. Prostaglandins Leukot Essent Fatty Acids 2018; 137:43-51. [PMID: 30293596 DOI: 10.1016/j.plefa.2018.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/28/2018] [Accepted: 08/30/2018] [Indexed: 10/28/2022]
Abstract
Omega (n)-3 polyunsaturated fatty acids (PUFA) are important regulators of inflammatory response that may impact pregnancy outcome. The effects of breeding chow diets containing n-3 PUFA from either fish oil (FO) or soybean oil (SO) were investigated on tissue fatty acid composition, inflammatory cytokines and pregnancy outcome. Female C57BL/6 mice (7 weeks old) were fed FO or SO diets for 2 weeks before mating and throughout pregnancy. Animals were sacrificed before and during pregnancy at day 6.5, 12.5 and 18.5. The FO diet increased the incorporation of n-3 PUFA in placenta, with a concomitant decrease in the concentration of pro-inflammatory cytokines. The FO diet increased the mRNA expression of placental specific PUFA transporter, which coincided with accretion of n-3 PUFA in fetal brain. Sites of fetal resorption were noticeable in the SO group but not in the FO group. N-3 PUFA may improve fetal sustainability via altering cytokine levels.
Collapse
Affiliation(s)
- O A Akerele
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL A1B 3X9, Canada
| | - S K Cheema
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, NL A1B 3X9, Canada.
| |
Collapse
|
46
|
Wu Y, Qi F, Song D, He Z, Zuo Z, Yang Y, Liu Q, Hu S, Wang X, Zheng X, Yang J, Yuan Q, Zou J, Guo K, Yao Z. Prenatal influenza vaccination rescues impairments of social behavior and lamination in a mouse model of autism. J Neuroinflammation 2018; 15:228. [PMID: 30103815 PMCID: PMC6090662 DOI: 10.1186/s12974-018-1252-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/11/2018] [Indexed: 12/14/2022] Open
Abstract
Background Prenatal infection is a substantial risk factor for neurodevelopmental disorders such as autism in offspring. We have previously reported that influenza vaccination (VAC) during early pregnancy contributes to neurogenesis and behavioral function in offspring. Results Here, we probe the efficacy of VAC pretreatment on autism-like behaviors in a lipopolysaccharide (LPS)-induced maternal immune activation (MIA) mouse model. We show that VAC improves abnormal fetal brain cytoarchitecture and lamination, an effect associated with promotion of intermediate progenitor cell differentiation in MIA fetal brain. These beneficial effects are sufficient to prevent social deficits in adult MIA offspring. Furthermore, whole-genome analysis suggests a strong interaction between Ikzf1 (IKAROS family zinc-finger 1) and neuronal differentiation. Intriguingly, VAC rescues excessive microglial Ikzf1 expression and attenuates microglial inflammatory responses in the MIA fetal brain. Conclusions Our study implies that a preprocessed influenza vaccination prevents maternal bacterial infection from causing neocortical lamination impairments and autism-related behaviors in offspring. Electronic supplementary material The online version of this article (10.1186/s12974-018-1252-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yingying Wu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Fangfang Qi
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Dan Song
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Zitian He
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Zejie Zuo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Yunjie Yang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Qiongliang Liu
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Saisai Hu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Xiao Wang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Xiaona Zheng
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Junhua Yang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Qunfang Yuan
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Juntao Zou
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Kaihua Guo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China
| | - Zhibin Yao
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China. .,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, Guangzhou, 510080, China.
| |
Collapse
|
47
|
Soares MJ, Iqbal K, Kozai K. Hypoxia and Placental Development. Birth Defects Res 2018; 109:1309-1329. [PMID: 29105383 DOI: 10.1002/bdr2.1135] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/04/2017] [Indexed: 12/17/2022]
Abstract
Hemochorial placentation is orchestrated through highly regulated temporal and spatial decisions governing the fate of trophoblast stem/progenitor cells. Trophoblast cell acquisition of specializations facilitating invasion and uterine spiral artery remodeling is a labile process, sensitive to the environment, and represents a process that is vulnerable to dysmorphogenesis in pathologic states. Hypoxia is a signal guiding placental development, and molecular mechanisms directing cellular adaptations to low oxygen tension are integral to trophoblast cell differentiation and placentation. Hypoxia can also be used as an experimental tool to investigate regulatory processes controlling hemochorial placentation. These developmental processes are conserved in mouse, rat, and human placentation. Consequently, elements of these developmental events can be modeled and hypotheses tested in trophoblast stem cells and in genetically manipulated rodents. Hypoxia is also a consequence of a failed placenta, yielding pathologies that can adversely affect maternal adjustments to pregnancy, fetal health, and susceptibility to adult disease. The capacity of the placenta for adaptation to environmental challenges highlights the importance of its plasticity in safeguarding a healthy pregnancy. Birth Defects Research 109:1309-1329, 2017.© 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas.,Fetal Health Research, Children's Research Institute, Children's Mercy, Kansas City, Missouri
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Keisuke Kozai
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
48
|
RNA binding protein, tristetraprolin in a murine model of recurrent pregnancy loss. Oncotarget 2018; 7:72486-72502. [PMID: 27732963 PMCID: PMC5341924 DOI: 10.18632/oncotarget.12539] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 09/26/2016] [Indexed: 01/23/2023] Open
Abstract
Recurrent pregnancy loss is a major reproductive pathology affecting 1-5% of pregnant women worldwide. A distinct feature of this reproductive pathology is involvement of key inflammatory cytokines and transcription factors such as tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6) and nuclear factor kappa beta (NF-κB). Special classes of RNA-binding proteins regulate the transcripts of many of these important cytokines and regulatory factors via binding to the 3' untranslated regions (UTRs) and/or poly(A) tail and destabilizing/stabilizing the transcript. The tristetraprolin (TTP/ZFP36) family have been found to be potent destabilizers of the aforementioned inflammatory and cellular response cytokines. The aim of this study was to evaluate whether tristetraprolin is expressed in the placenta and involved in modulating inflammation in mouse model of lipopolysaccharide (LPS)-induced fetal loss. In this study, Swiss-albino mice were injected with LPS at gestational day 15.5 and placental tissues were harvested 6 hours post-LPS injection. Histopathology and immunohistochemistry analyses clearly revealed cellular stress and death in LPS treated placentas compared to controls. TTP protein was downregulated, while targets TNF-α and IL-6 were upregulated in LPS group compared to controls. We observed increased TTP nuclear immunolocalization corresponding with higher NF-κB nuclear localization in trophoblasts from LPS treated placentas. Our results suggest that RNA-binding proteins such as TTP are expressed and perhaps involved in the modulation of inflammation-induced pregnancy pathologies.
Collapse
|
49
|
He B, Yang X, Li Y, Huang D, Xu X, Yang W, Dai Y, Zhang H, Chen Z, Cheng W. TLR9 (Toll-Like Receptor 9) Agonist Suppresses Angiogenesis by Differentially Regulating VEGFA (Vascular Endothelial Growth Factor A) and sFLT1 (Soluble Vascular Endothelial Growth Factor Receptor 1) in Preeclampsia. Hypertension 2018; 71:671-680. [PMID: 29437897 DOI: 10.1161/hypertensionaha.117.10510] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 11/09/2017] [Accepted: 01/11/2018] [Indexed: 01/19/2023]
Abstract
Preeclampsia is a common pregnancy-specific disorder characterized by elevated blood pressure and proteinuria. Activation of the maternal immune system and impaired placental angiogenesis are thought to contribute to the pathogenesis of preeclampsia. TLR9 (Toll-like receptor 9) plays a role in innate immunity, defending the organism against infection. The purpose of this study was to determine whether TLR9 inhibits angiogenesis at the fetomaternal interface under conditions of preeclampsia. We confirmed the downregulation of VEGFA (vascular endothelial growth factor A) and upregulation of TLR9 and sFLT1 (soluble vascular endothelial growth factor receptor 1) in placentas from preeclamptic women. Then, we established a mouse model with preeclampsia-like symptoms using the synthetic TLR9 agonist CpG (cytidine-phosphate-guanosine)-ODN (oligodeoxynucleotide; ODN1826). We observed the downregulation of VEGFA and the upregulation of sFLT1 in placentas from the preeclampsia-like animal model and in trophoblasts treated with CpG-ODN (ODN2006). In addition, silencing TLR9 promoted the migration and invasion of HTR8/SVneo cells. In conclusion, TLR9 is capable of robustly suppressing angiogenesis by differentially regulating the expression of VEGFA and sFLT1 at the fetomaternal interface, potentially contributing to the development of preeclampsia.
Collapse
Affiliation(s)
- Biwei He
- From the International Peace Maternity and Child Health Hospital (B.H., X.Y., Y.L., D.H., W.Y., H.Z., W.C.), Institute of Embryo-Fetal Original Adult Disease (X.Y.), and Department of Obstetrics and Gynecology, Xinhua Hospital (X.X.), School of Medicine, Shanghai Jiao Tong University, China; State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science (Y.D., Z.C.); and School of Life Science and Technology, Shanghai Tech University, China (Z.C.)
| | - Xingyu Yang
- From the International Peace Maternity and Child Health Hospital (B.H., X.Y., Y.L., D.H., W.Y., H.Z., W.C.), Institute of Embryo-Fetal Original Adult Disease (X.Y.), and Department of Obstetrics and Gynecology, Xinhua Hospital (X.X.), School of Medicine, Shanghai Jiao Tong University, China; State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science (Y.D., Z.C.); and School of Life Science and Technology, Shanghai Tech University, China (Z.C.)
| | - Yamei Li
- From the International Peace Maternity and Child Health Hospital (B.H., X.Y., Y.L., D.H., W.Y., H.Z., W.C.), Institute of Embryo-Fetal Original Adult Disease (X.Y.), and Department of Obstetrics and Gynecology, Xinhua Hospital (X.X.), School of Medicine, Shanghai Jiao Tong University, China; State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science (Y.D., Z.C.); and School of Life Science and Technology, Shanghai Tech University, China (Z.C.)
| | - Ding Huang
- From the International Peace Maternity and Child Health Hospital (B.H., X.Y., Y.L., D.H., W.Y., H.Z., W.C.), Institute of Embryo-Fetal Original Adult Disease (X.Y.), and Department of Obstetrics and Gynecology, Xinhua Hospital (X.X.), School of Medicine, Shanghai Jiao Tong University, China; State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science (Y.D., Z.C.); and School of Life Science and Technology, Shanghai Tech University, China (Z.C.)
| | - Xin Xu
- From the International Peace Maternity and Child Health Hospital (B.H., X.Y., Y.L., D.H., W.Y., H.Z., W.C.), Institute of Embryo-Fetal Original Adult Disease (X.Y.), and Department of Obstetrics and Gynecology, Xinhua Hospital (X.X.), School of Medicine, Shanghai Jiao Tong University, China; State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science (Y.D., Z.C.); and School of Life Science and Technology, Shanghai Tech University, China (Z.C.)
| | - Wenjun Yang
- From the International Peace Maternity and Child Health Hospital (B.H., X.Y., Y.L., D.H., W.Y., H.Z., W.C.), Institute of Embryo-Fetal Original Adult Disease (X.Y.), and Department of Obstetrics and Gynecology, Xinhua Hospital (X.X.), School of Medicine, Shanghai Jiao Tong University, China; State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science (Y.D., Z.C.); and School of Life Science and Technology, Shanghai Tech University, China (Z.C.)
| | - Yan Dai
- From the International Peace Maternity and Child Health Hospital (B.H., X.Y., Y.L., D.H., W.Y., H.Z., W.C.), Institute of Embryo-Fetal Original Adult Disease (X.Y.), and Department of Obstetrics and Gynecology, Xinhua Hospital (X.X.), School of Medicine, Shanghai Jiao Tong University, China; State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science (Y.D., Z.C.); and School of Life Science and Technology, Shanghai Tech University, China (Z.C.)
| | - Huijuan Zhang
- From the International Peace Maternity and Child Health Hospital (B.H., X.Y., Y.L., D.H., W.Y., H.Z., W.C.), Institute of Embryo-Fetal Original Adult Disease (X.Y.), and Department of Obstetrics and Gynecology, Xinhua Hospital (X.X.), School of Medicine, Shanghai Jiao Tong University, China; State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science (Y.D., Z.C.); and School of Life Science and Technology, Shanghai Tech University, China (Z.C.)
| | - Zhengjun Chen
- From the International Peace Maternity and Child Health Hospital (B.H., X.Y., Y.L., D.H., W.Y., H.Z., W.C.), Institute of Embryo-Fetal Original Adult Disease (X.Y.), and Department of Obstetrics and Gynecology, Xinhua Hospital (X.X.), School of Medicine, Shanghai Jiao Tong University, China; State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science (Y.D., Z.C.); and School of Life Science and Technology, Shanghai Tech University, China (Z.C.)
| | - Weiwei Cheng
- From the International Peace Maternity and Child Health Hospital (B.H., X.Y., Y.L., D.H., W.Y., H.Z., W.C.), Institute of Embryo-Fetal Original Adult Disease (X.Y.), and Department of Obstetrics and Gynecology, Xinhua Hospital (X.X.), School of Medicine, Shanghai Jiao Tong University, China; State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Science (Y.D., Z.C.); and School of Life Science and Technology, Shanghai Tech University, China (Z.C.).
| |
Collapse
|
50
|
Li M, Gao Y, Yong L, Huang D, Shen J, Liu M, Ren C, Hou X. Molecular signature and functional analysis of uterine ILCs in mouse pregnancy. J Reprod Immunol 2017; 123:48-57. [PMID: 28915450 DOI: 10.1016/j.jri.2017.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 09/01/2017] [Accepted: 09/08/2017] [Indexed: 02/07/2023]
Abstract
In addition to natural killer cells, other innate lymphoid cells have recently been identified in the mouse and human uterus, but their roles in successful pregnancy remain poorly defined. In this study, we examined the dynamic changes of uterine innate lymphoid cells throughout pregnancy in mice. We found that the total number of uterine innate lymphoid cells markedly increased at early-gestation. Among the three groups of uterine innate lymphoid cells, the number of the group 2 uterine innate lymphoid cells increased the most during pregnancy. We also determined that the depletion of uterine innate lymphoid cells in Rag1-/- mice resulted in impaired uterine spiral artery remodeling. These results suggest that uterine innate lymphoid cells may play an important role in mouse reproduction.
Collapse
Affiliation(s)
- Minmin Li
- Department of Microbiology and Parasitology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Yimin Gao
- Department of Microbiology and Parasitology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Liang Yong
- Department of Microbiology and Parasitology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Dake Huang
- Department of Microbiology and Parasitology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Jijia Shen
- Department of Microbiology and Parasitology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China; Anhui Provincial Laboratory of Microbiology and Parasitology, Hefei, Anhui, China
| | - Miao Liu
- Department of Microbiology and Parasitology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China; Anhui Provincial Laboratory of Microbiology and Parasitology, Hefei, Anhui, China
| | - Cuiping Ren
- Department of Microbiology and Parasitology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China; Anhui Provincial Laboratory of Microbiology and Parasitology, Hefei, Anhui, China
| | - Xin Hou
- Department of Microbiology and Parasitology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China; Anhui Provincial Laboratory of Microbiology and Parasitology, Hefei, Anhui, China.
| |
Collapse
|