1
|
Ali Khazaei H, Farzaneh F, Sarhadi S, Dehghan Haghighi J, Forghani F, Sheikhi V, Khazaei B, Asadollahi L. Comparison of serum levels of interleukin 33 in combination with serum levels of C-reactive protein, Immunoglobulin G, Immunoglobulin A, and Immunoglobulin M in recurrent pregnancy loss: A case-control study. Int J Reprod Biomed 2024; 22:317-322. [PMID: 39035635 PMCID: PMC11255461 DOI: 10.18502/ijrm.v22i4.16392] [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: 02/16/2023] [Revised: 06/19/2023] [Accepted: 08/12/2023] [Indexed: 07/23/2024] Open
Abstract
Background One of the critical cases of recurrent pregnancy loss is immunological factors, whereas obtaining effective prevention or treatment is necessary for cognition of reasons. Objective In this study, we tried to evaluate some immunological factors related to recurrent pregnancy loss. Materials and Methods This case-control study was conducted on 66 women at the age of 18-35 yr who were referred to the Clinic of Gynecology and Obstetrics, Ali Ibn Abi Taleb hospital, Zahedan, Iran, from August-December 2019. Interleukin 33 (IL-33) serum levels were measured using enzyme-linked immunosorbent assay. Immunoglobulin G, Immunoglobulin A, Immunoglobulin M (IgM), and C-reactive protein levels were measured by serology and hematology methods. Results The mean age of total participants was 30.8 ± 3.80 yr. The mean serum IL-33 in the case group was 318.5 ± 254.1 pg/ml and was lower than the control group (354.2 ± 259.9 pg/ml), which was not statistically significant (p = 0.52). The level of C-reactive protein in the case and control was not significantly different (p = 0.27), and Immunoglobulin A and Immunoglobulin G in the case and control were also not significantly different (p = 0.46, and p = 0.16, respectively), but there were significant differences (p = 0.003) between the level of the IgM in the case and control groups. Conclusion No statistically significant difference was observed in the IL-33 serum level, for at least 4-6 months after the last abortion in the case group and the final live birth in the control group. In contrast, serum levels of IgM were statistically significant. Finally, the need for more studies is felt according to the different results of the previous studies in this field.
Collapse
Affiliation(s)
- Hossein Ali Khazaei
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Farahnaz Farzaneh
- Department of Obstetrics and Gynecology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Saeedeh Sarhadi
- Health Promotion Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Community Medicine, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Javid Dehghan Haghighi
- Department of Community Medicine, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Forough Forghani
- Department of Obstetrics and Gynecology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Vahid Sheikhi
- Department of Pediatrics, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Bahman Khazaei
- School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Lida Asadollahi
- School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
2
|
Tacrolimus improved the pregnancy outcomes of patients with refractory recurrent spontaneous abortion and immune bias disorders: a randomized controlled trial. Eur J Clin Pharmacol 2023; 79:627-634. [PMID: 36912957 DOI: 10.1007/s00228-023-03473-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023]
Abstract
OBJECTIVE To investigate the effect of tacrolimus treatment on refractory recurrent spontaneous abortion (RSA) patients with elevated serum IL-33/ST2 levels. METHODS This study was a randomized controlled trial (RCT) of refractory RSA patients with elevated peripheral blood IL-33/ST2 levels or an elevated Th1/Th2 cell ratio. A total of 149 women were enrolled, each of whom had had at least 3 serial miscarriages and was confirmed to have elevated peripheral blood IL-33/ST2 levels or an elevated Th1/Th2 cell ratio. These women were randomly divided into two groups. The tacrolimus group (n = 75) received basic therapy with the addition of tacrolimus (Prograf). Tacrolimus was administered at a dose of 0.05 ~ 0.1 mg/kg/day from the end of the menstrual period to the beginning of the next menstrual period or to the 10th week of pregnancy. In contrast, basic therapy with the addition of placebo was given to the placebo group (n = 74). The main study outcome was the delivery of healthy newborns without deformities. RESULTS A total of 60 (80.00%) patients in the tacrolimus group and 47 (63.51%) patients in the placebo group delivered healthy newborns [P = 0.03, odds ratio = 2.30; 95% confidence interval (1.10 ~ 4.81)]. The peripheral blood IL-33/ST2 levels and Th1/Th2 cell ratio of the tacrolimus group were much lower than those of the placebo group (P < 0.05). CONCLUSION We validated our previous finding that serum IL-33 and sST2 concentrations are related to RSA. Immunosuppressive treatment with tacrolimus was demonstrated to be a promising method to treat refractory RSA with immune bias disorders.
Collapse
|
3
|
Abstract
When discovered in the early 2000s, interleukin-33 (IL-33) was characterized as a potent driver of type 2 immunity and implicated in parasite clearance, as well as asthma, allergy, and lung fibrosis. Yet research in other models has since revealed that IL-33 is a highly pleiotropic molecule with diverse functions. These activities are supported by elusive release mechanisms and diverse expression of the IL-33 receptor, STimulation 2 (ST2), on both immune and stromal cells. Interestingly, IL-33 also supports type 1 immune responses during viral and tumor immunity and after allogeneic hematopoietic stem cell transplantation. Yet the IL-33-ST2 axis is also critical to the establishment of systemic homeostasis and tissue repair and regeneration. Despite these recent findings, the mechanisms by which IL-33 governs the balance between immunity and homeostasis or can support both effective repair and pathogenic fibrosis are poorly understood. As such, ongoing research is trying to understand the potential reparative and regulatory versus pro-inflammatory and pro-fibrotic roles for IL-33 in transplantation. This review provides an overview of the emerging regenerative role of IL-33 in organ homeostasis and tissue repair as it relates to transplantation immunology. It also outlines the known impacts of IL-33 in commonly transplanted solid organs and covers the envisioned roles for IL-33 in ischemia-reperfusion injury, rejection, and tolerance. Finally, we give a comprehensive summary of its effects on different cell populations involved in these processes, including ST2 + regulatory T cells, innate lymphoid cell type 2, as well as significant myeloid cell populations.
Collapse
|
4
|
Maternal IL-33 critically regulates tissue remodeling and type 2 immune responses in the uterus during early pregnancy in mice. Proc Natl Acad Sci U S A 2022; 119:e2123267119. [PMID: 35994660 PMCID: PMC9436313 DOI: 10.1073/pnas.2123267119] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The pregnant uterus is an immunologically rich organ, with dynamic changes in the inflammatory milieu and immune cell function underlying key stages of pregnancy. Recent studies have implicated dysregulated expression of the interleukin-1 (IL-1) family cytokine, IL-33, and its receptor, ST2, in poor pregnancy outcomes in women, including recurrent pregnancy loss, preeclampsia, and preterm labor. How IL-33 supports pregnancy progression in vivo is not well understood. Here, we demonstrate that maternal IL-33 signaling critically regulates uterine tissue remodeling and immune cell function during early pregnancy in mice. IL-33-deficient dams exhibit defects in implantation chamber formation and decidualization, and abnormal vascular remodeling during early pregnancy. These defects coincide with delays in early embryogenesis, increased resorptions, and impaired fetal and placental growth by late pregnancy. At a cellular level, myometrial fibroblasts, and decidual endothelial and stromal cells, are the main IL-33+ cell types in the uterus during decidualization and early placentation, whereas ST2 is expressed by uterine immune populations associated with type 2 immune responses, including ILC2s, Tregs, CD4+ T cells, M2- and cDC2-like myeloid cells, and mast cells. Early pregnancy defects in IL-33-deficient dams are associated with impaired type 2 cytokine responses by uterine lymphocytes and fewer Arginase-1+ macrophages in the uterine microenvironment. Collectively, our data highlight a regulatory network, involving crosstalk between IL-33-producing nonimmune cells and ST2+ immune cells at the maternal-fetal interface, that critically supports pregnancy progression in mice. This work has the potential to advance our understanding of how IL-33 signaling may support optimal pregnancy outcomes in women.
Collapse
|
5
|
IL-33–ILC2 axis in the female reproductive tract. Trends Mol Med 2022; 28:569-582. [DOI: 10.1016/j.molmed.2022.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 02/06/2023]
|
6
|
Abstract
Our understanding of the functions of the IL-1 superfamily cytokine and damage-associated molecular pattern IL-33 continues to evolve with our understanding of homeostasis and immunity. The early findings that IL-33 is a potent driver of type 2 immune responses promoting parasite expulsion, but also inflammatory diseases like allergy and asthma, have been further supported. Yet, as the importance of a type 2 response in tissue repair and homeostasis has emerged, so has the fundamental importance of IL-33 to these processes. In this review, we outline an evolving understanding of IL-33 immunobiology, paying particular attention to how IL-33 directs a network of ST2+ regulatory T cells, reparative and regulatory macrophages, and type 2 innate lymphoid cells that are fundamental to tissue development, homeostasis, and repair. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Gaelen K. Dwyer
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Louise M. D'Cruz
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hēth R. Turnquist
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
7
|
Abstract
Chorioamnionitis or intrauterine inflammation is a frequent cause of preterm birth. Chorioamnionitis can affect almost every organ of the developing fetus. Multiple microbes have been implicated to cause chorioamnionitis, but "sterile" inflammation appears to be more common. Eradication of microorganisms has not been shown to prevent the morbidity and mortality associated with chorioamnionitis as inflammatory mediators account for continued fetal and maternal injury. Mounting evidence now supports the concept that the ensuing neonatal immune dysfunction reflects the effects of inflammation on immune programming during critical developmental windows, leading to chronic inflammatory disorders as well as vulnerability to infection after birth. A better understanding of microbiome alterations and inflammatory dysregulation may help develop better treatment strategies for infants born to mothers with chorioamnionitis.
Collapse
|
8
|
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: 8] [Impact Index Per Article: 2.0] [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
|
9
|
Gomez-Lopez N, Romero R, Garcia-Flores V, Leng Y, Miller D, Hassan SS, Hsu CD, Panaitescu B. Inhibition of the NLRP3 inflammasome can prevent sterile intra-amniotic inflammation, preterm labor/birth, and adverse neonatal outcomes†. Biol Reprod 2020; 100:1306-1318. [PMID: 30596885 DOI: 10.1093/biolre/ioy264] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/10/2018] [Accepted: 12/26/2018] [Indexed: 01/20/2023] Open
Abstract
Sterile intra-amniotic inflammation is commonly observed in patients with spontaneous preterm labor, a syndrome that commonly precedes preterm birth, the leading cause of perinatal morbidity and mortality worldwide. However, the mechanisms leading to sterile intra-amniotic inflammation are poorly understood and no treatment exists for this clinical condition. Herein, we investigated whether the alarmin S100B could induce sterile intra-amniotic inflammation by activating the NLRP3 inflammasome, and whether the inhibition of this pathway could prevent preterm labor/birth and adverse neonatal outcomes. We found that the ultrasound-guided intra-amniotic administration of S100B induced a 50% rate of preterm labor/birth and a high rate of neonatal mortality (59.7%) without altering the fetal and placental weights. Using a multiplex cytokine array and immunoblotting, we reported that S100B caused a proinflammatory response in the amniotic cavity and induced the activation of the NLRP3 inflammasome in the fetal membranes, indicated by the upregulation of the NLRP3 protein and increased release of active caspase-1 and mature IL-1β. Inhibition of the NLRP3 inflammasome via the specific inhibitor MCC950 prevented preterm labor/birth by 35.7% and reduced neonatal mortality by 26.7%. Yet, inhibition of the NLRP3 inflammasome at term did not drastically obstruct the physiological process of parturition. In conclusion, the data presented herein indicate that the alarmin S100B can induce sterile intra-amniotic inflammation, preterm labor/birth, and adverse neonatal outcomes by activating the NLRP3 inflammasome, which can be prevented by inhibiting such a pathway. These findings provide evidence that sterile intra-amniotic inflammation could be treated by targeting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Immunology, Microbiology and Biochemistry, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA.,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yaozhu Leng
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Derek Miller
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Sonia S Hassan
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Chaur-Dong Hsu
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Bogdan Panaitescu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
10
|
Lebold KM, Jacoby DB, Drake MG. Inflammatory mechanisms linking maternal and childhood asthma. J Leukoc Biol 2020; 108:113-121. [PMID: 32040236 DOI: 10.1002/jlb.3mr1219-338r] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/17/2019] [Accepted: 12/18/2019] [Indexed: 12/26/2022] Open
Abstract
Asthma is a chronic inflammatory airway disease characterized by airway hyperresponsiveness, inflammation, and remodeling. Asthma often develops during childhood and causes lifelong decrements in lung function and quality of life. Risk factors for childhood asthma are numerous and include genetic, epigenetic, developmental, and environmental factors. Uncontrolled maternal asthma during pregnancy exposes the developing fetus to inflammatory insults, which further increase the risk of childhood asthma independent of genetic predisposition. This review focuses on the role of maternal asthma in the development of asthma in offspring. We will present maternal asthma as a targetable and modifiable risk factor for childhood asthma and discuss the mechanisms by which maternal inflammation increases childhood asthma risk. Topics include how exposure to maternal asthma in utero shapes structural lung development with a special emphasis on airway nerves, how maternal type-2 cytokines such as IL-5 activate the fetal immune system, and how changes in lung and immune cell development inform responses to aero-allergens later in life. Finally, we highlight emerging evidence that maternal asthma establishes a unique "asthma signature" in the airways of children, leading to novel mechanisms of airway hyperreactivity and inflammatory cell responses.
Collapse
Affiliation(s)
- Katie M Lebold
- Division of Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - David B Jacoby
- Division of Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Matthew G Drake
- Division of Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
11
|
Ozler S, Oztas E, Guler BG, Caglar AT. Increased levels of serum IL-33 is associated with adverse maternal outcomes in placenta previa accreta. J Matern Fetal Neonatal Med 2019; 34:3192-3199. [PMID: 31608786 DOI: 10.1080/14767058.2019.1679766] [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: 10/25/2022]
Abstract
PURPOSE IL-33 is associated with invasion, proliferation, and metastasis of various cancers. The trophoblastic cells of placenta previa accreta (PPA) invade into the myometrium in a similar way to the invasion of cancers. We studied the role of IL-33 in PPA and also aimed to investigate its relation with adverse maternal outcome in this placental disorder. METHODS A total of 87 pregnant patients were enrolled in this prospective case-control study [27 with PPA, 30 with placenta previa totalis (PPT; nonadherent placenta previa), and 30 controls]. IL-33 and IL-6 levels were studied in maternal serum at late preterm gestation weeks. Multiple logistic regression analyses analyzed the risk factors which are associated with PPA and adverse maternal outcomes. Adjusted odds ratios and 95% confidence intervals were also calculated. Enzyme-linked immunosorbent assay (ELISA) method was used to determine maternal serum IL-33 and IL-6 levels. RESULTS Serum IL-33 levels were significantly higher in PPA patients when compared with both nonadherent PPT and the control groups (p = .011, p = .010). Serum IL-6 and neutrophil/lymphocyte ratio levels were significantly higher than the control group's (p = .045, p = .028). IL-33 levels and history of previous cesarean section were found to be significantly associated with PPA (OR: 1.039, 95% CI: 1.004-1.075; p = .030 and OR: 0.067, 95% CI: 0.014-0.309, p = .001, respectively). Serum IL-33 levels were positively correlated with previous cesarean section history in PPA. Increased maternal serum IL-33 levels were found to be independently associated with a cesarean hysterectomy and massive transfusion in PPA patients (OR: 1.098, 95% CI: 0.998-1.207; p = .049 and OR: 1.162 95% CI: 1.010-1.337; p = .036). CONCLUSION Increased levels of maternal serum IL-33 and history of previous cesarean section were found to be significantly associated with PPA, and also increased maternal serum IL-33 levels were related to cesarean hysterectomy and massive blood transfusion in PPA. We suggest that IL-33 may have a role in abnormal placental invasion.
Collapse
Affiliation(s)
- Sibel Ozler
- Department of Perinatology, Selcuk University Medical School, Konya, Turkey
| | - Efser Oztas
- Department of Perinatology, Eskisehir City Hospital, Eskisehir, Turkey
| | | | - Ali Turhan Caglar
- Department of Pathology, University of Health Sciences Ankara City Hospital, Ankara, Turkey
| |
Collapse
|
12
|
Richardson L, Dixon CL, Aguilera-Aguirre L, Menon R. Oxidative stress-induced TGF-beta/TAB1-mediated p38MAPK activation in human amnion epithelial cells. Biol Reprod 2018; 99:1100-1112. [PMID: 29893818 PMCID: PMC7190655 DOI: 10.1093/biolre/ioy135] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/04/2018] [Accepted: 06/07/2018] [Indexed: 02/07/2023] Open
Abstract
Term and preterm parturition are associated with oxidative stress (OS)-induced p38 mitogen-activated protein kinase (p38MAPK)-mediated fetal tissue (amniochorion) senescence. p38MAPK activation is a complex cell- and stimulant-dependent process. Two independent pathways of OS-induced p38MAPK activation were investigated in amnion epithelial cells (AECs) in response to cigarette smoke extract (CSE: a validated OS inducer in fetal cells): (1) the OS-mediated oxidation of apoptosis signal-regulating kinase (ASK)-1 bound Thioredoxin (Trx[SH]2) dissociates this complex, creating free and activated ASK1-signalosome and (2) transforming growth factor-mediated activation of (TGF)-beta-activated kinase (TAK)1 and TGF-beta-activated kinase 1-binding protein (TAB)1. AECs isolated from normal term, not-in-labor fetal membranes increased p38MAPK in response to CSE and downregulated it in response to antioxidant N-acetylcysteine. In AECs, both Trx and ASK1 were localized; however, they remained dissociated and not complexed, regardless of conditions. Silencing either ASK1 or its downstream effectors (MKK3/6) did not affect OS-induced p38MAPK activation. Conversely, OS increased TGF-beta's release from AECs and increased phosphorylation of both p38MAPK and TAB1. Silencing of TAB1, but not TAK1, prevented p38MAPK activation, which is indicative of TAB1-mediated autophosphorylation of p38MAPK, an activation mechanism seldom seen. OS-induced p38MAPK activation in AECs is ASK1-Trx signalosome-independent and is mediated by the TGF-beta pathway. This knowledge will help to design strategies to reduce p38MAPK activation-associated pregnancy risks.
Collapse
Affiliation(s)
- Lauren Richardson
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine and Perinatal Research, The University of Texas Medical Branch, Galveston, Texas, USA
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Tx, 77550
| | - Christopher Luke Dixon
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine and Perinatal Research, The University of Texas Medical Branch, Galveston, Texas, USA
| | - Leopoldo Aguilera-Aguirre
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine and Perinatal Research, The University of Texas Medical Branch, Galveston, Texas, USA
| | - Ramkumar Menon
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine and Perinatal Research, The University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
13
|
The association of IL-33 and Foxp3 gene polymorphisms with recurrent pregnancy loss in Egyptian women. Cytokine 2018; 108:115-119. [DOI: 10.1016/j.cyto.2018.03.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/11/2017] [Accepted: 03/19/2018] [Indexed: 11/23/2022]
|
14
|
Bartemes K, Chen CC, Iijima K, Drake L, Kita H. IL-33-Responsive Group 2 Innate Lymphoid Cells Are Regulated by Female Sex Hormones in the Uterus. THE JOURNAL OF IMMUNOLOGY 2017; 200:229-236. [PMID: 29133293 DOI: 10.4049/jimmunol.1602085] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 10/14/2017] [Indexed: 12/13/2022]
Abstract
Group 2 innate lymphoid cells (ILC2s) reside in multiple organs in the body, where they play roles in immunity, tissue homeostasis, and metabolic regulation. However, little is known about the regulatory mechanisms of ILC2s in different organs. Here, we identified ILC2s in the mouse uterus and found that they express cell surface molecules, including the IL-33 receptor, ST2, that are roughly comparable to those expressed by lung ILC2s. Both in vivo and in vitro treatment with IL-33 induced type 2 cytokine production in uterine ILC2s, suggesting that they respond to IL-33 in a manner similar to ILC2s in other organs. Importantly, uterine ILC2s were nearly absent in ovariectomized mice and were increased in wild-type mice by estrogen administration, whereas lung ILC2s were unaffected by both ovariectomy and estrogen administration. Likewise, a marked reduction in uterine ILC2s was observed in mice deficient in estrogen receptor α or estrogen receptor β. Furthermore, uterine ILC2s highly expressed estrogen receptor α, and in vitro culture of isolated uterine ILC2s with 17β-estradiol modified expression of a number of genes. Finally, an increased prevalence in neonatal mortality was observed in litters from dams lacking the IL-33 receptor, ST2. Taken together, our findings indicate that unlike lung IL2Cs, uterine ILC2s are regulated by female sex hormones, which may specialize them for specific physiological functions.
Collapse
Affiliation(s)
- Kathleen Bartemes
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905; and.,Department of Immunology, Mayo Clinic, Rochester, MN 55905
| | - Chien-Chang Chen
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905; and.,Department of Immunology, Mayo Clinic, Rochester, MN 55905
| | - Koji Iijima
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905; and
| | - Li Drake
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905; and
| | - Hirohito Kita
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905; and .,Department of Immunology, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
15
|
Scott LM, Bryant AH, Rees A, Down B, Jones RH, Thornton CA. Production and regulation of interleukin-1 family cytokines at the materno-fetal interface. Cytokine 2017; 99:194-202. [DOI: 10.1016/j.cyto.2017.07.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 07/06/2017] [Accepted: 07/07/2017] [Indexed: 11/26/2022]
|
16
|
Romero R, Chaemsaithong P, Tarca AL, Korzeniewski SJ, Maymon E, Pacora P, Panaitescu B, Chaiyasit N, Dong Z, Erez O, Hassan SS, Chaiworapongsa T. Maternal plasma-soluble ST2 concentrations are elevated prior to the development of early and late onset preeclampsia - a longitudinal study. J Matern Fetal Neonatal Med 2017; 31:418-432. [PMID: 28114842 DOI: 10.1080/14767058.2017.1286319] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The objectives of this study were to determine (1) the longitudinal profile of plasma soluble ST2 (sST2) concentrations in patients with preeclampsia and those with uncomplicated pregnancies; (2) whether the changes in sST2 occur prior to the diagnosis of preeclampsia; and (3) the longitudinal sST2 profile of women with early or late preeclampsia. MATERIALS AND METHODS This longitudinal nested case-control study included singleton pregnancies in the following groups: (1) uncomplicated pregnancies (n = 160); and (2) those complicated by early (<34 weeks, n = 9) and late (≥34 weeks, n = 31) preeclampsia. sST2 concentrations were determined by enzyme-linked immunosorbent assays. Mixed-effects models were used for the longitudinal analysis. RESULTS (1) Plasma sST2 concentration profiles across gestation differed significantly among cases and controls (p < 0.0001); (2) women with early preeclampsia had higher mean sST2 concentrations than controls at >22 weeks of gestation; cases with late preeclampsia had higher mean concentrations at >33 weeks of gestation (both p < 0.05); and (3) these changes started approximately 6 weeks prior to clinical diagnosis. CONCLUSIONS Maternal plasma sST2 concentrations are elevated 6 weeks prior to the clinical diagnosis of preeclampsia. An increase in the maternal plasma concentration of sST2 may contribute to an exaggerated intravascular inflammatory response and/or the Th1/Th2 imbalance in some cases.
Collapse
Affiliation(s)
- Roberto Romero
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,b Department of Obstetrics and Gynecology , University of Michigan , Ann Arbor , MI , USA.,c Department of Epidemiology and Biostatistics , Michigan State University , East Lansing , MI , USA.,d Center for Molecular Medicine and Genetics , Wayne State University , Detroit , MI , USA
| | - Piya Chaemsaithong
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Adi L Tarca
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Steven J Korzeniewski
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,c Department of Epidemiology and Biostatistics , Michigan State University , East Lansing , MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Eli Maymon
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Percy Pacora
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Bogdan Panaitescu
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Noppadol Chaiyasit
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Zhong Dong
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Offer Erez
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Sonia S Hassan
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Tinnakorn Chaiworapongsa
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH/DHHS , Bethesda , MD, and Detroit, MI , USA.,e Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| |
Collapse
|
17
|
Genetic variant in IL-33 is associated with idiopathic recurrent miscarriage in Chinese Han population. Sci Rep 2016; 6:23806. [PMID: 27026387 PMCID: PMC4812300 DOI: 10.1038/srep23806] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 03/15/2016] [Indexed: 01/19/2023] Open
Abstract
Recurrent miscarriage (RM) is the occurrence of repeated pregnancies that end in miscarriage of the fetus before 20 weeks of gestation. At least 50% of the RM patients are considered idiopathic. High IL-33 levels are critical in early pregnancy and impact the outcome of subsequent pregnancies. However, the association of polymorphisms of IL-33 with idiopathic RM is still unclear. The present study was initiated to investigate whether IL-33 polymorphisms are risk factors for idiopathic RM in Chinese Han population. Study subjects comprised of 321 cases and 384 controls. Five polymorphisms (rs10435816, rs16924159, rs16924171, rs1929992, rs1332290) in IL-33 and serum IL-33 concentrations were assessed. rs16924159 variant exhibits significant association with RM in additive and recessive genetic model (additive model P = 0.015, recessive model P = 0.007). In contrast, rs10435816, rs16924171, rs1929992 and rs1332290 are not significantly associated with RM. Serum IL-33 levels are significantly lower in RM cases than in control (173.51 ± 94.12 versus. 200.97 ± 110.06 (pg/ml), P = 4.57 × 10−4). There are lower levels of serum IL-33 in rs16924159 homozygous mutant (AA) than homozygous wild-type (GG) in this study population, including cases and control groups (172.18 ± 103.01 versus. 205.82 ± 119.01 (pg/ml), P = 0.006). Reduced IL-33 levels and rs16924159 IL-33 variant may contribute to the pathogenesis of idiopathic RM in Chinese Han population.
Collapse
|
18
|
St Louis D, Romero R, Plazyo O, Arenas-Hernandez M, Panaitescu B, Xu Y, Milovic T, Xu Z, Bhatti G, Mi QS, Drewlo S, Tarca AL, Hassan SS, Gomez-Lopez N. Invariant NKT Cell Activation Induces Late Preterm Birth That Is Attenuated by Rosiglitazone. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:1044-59. [PMID: 26740111 PMCID: PMC4724534 DOI: 10.4049/jimmunol.1501962] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/24/2015] [Indexed: 12/13/2022]
Abstract
Preterm birth (PTB) is the leading cause of neonatal morbidity and mortality worldwide. Although intra-amniotic infection is a recognized cause of spontaneous preterm labor, the noninfection-related etiologies are poorly understood. In this article, we demonstrated that the expansion of activated CD1d-restricted invariant NKT (iNKT) cells in the third trimester by administration of α-galactosylceramide (α-GalCer) induced late PTB and neonatal mortality. In vivo imaging revealed that fetuses from mice that underwent α-GalCer-induced late PTB had bradycardia and died shortly after delivery. Yet, administration of α-GalCer in the second trimester did not cause pregnancy loss. Peroxisome proliferator-activated receptor (PPAR)γ activation, through rosiglitazone treatment, reduced the rate of α-GalCer-induced late PTB and improved neonatal survival. Administration of α-GalCer in the third trimester suppressed PPARγ activation, as shown by the downregulation of Fabp4 and Fatp4 in myometrial and decidual tissues, respectively; this suppression was rescued by rosiglitazone treatment. Administration of α-GalCer in the third trimester induced an increase in the activation of conventional CD4(+) T cells in myometrial tissues and the infiltration of activated macrophages, neutrophils, and mature dendritic cells to myometrial and/or decidual tissues. All of these effects were blunted after rosiglitazone treatment. Administration of α-GalCer also upregulated the expression of inflammatory genes at the maternal-fetal interface and systemically, and rosiglitazone treatment partially attenuated these responses. Finally, an increased infiltration of activated iNKT-like cells in human decidual tissues is associated with noninfection-related preterm labor/birth. Collectively, these results demonstrate that iNKT cell activation in vivo leads to late PTB by initiating innate and adaptive immune responses and suggest that the PPARγ pathway has potential as a target for prevention of this syndrome.
Collapse
Affiliation(s)
- Derek St Louis
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201; Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health/U.S. Department of Health and Human Services, Bethesda, MD 20892 and Detroit, MI 48201
| | - Roberto Romero
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health/U.S. Department of Health and Human Services, Bethesda, MD 20892 and Detroit, MI 48201; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48825; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201
| | - Olesya Plazyo
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201; Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health/U.S. Department of Health and Human Services, Bethesda, MD 20892 and Detroit, MI 48201
| | - Marcia Arenas-Hernandez
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201; Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health/U.S. Department of Health and Human Services, Bethesda, MD 20892 and Detroit, MI 48201
| | - Bogdan Panaitescu
- Department of Pediatrics, Neonatology Division, Wayne State University School of Medicine, Detroit, MI 48201
| | - Yi Xu
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health/U.S. Department of Health and Human Services, Bethesda, MD 20892 and Detroit, MI 48201
| | - Tatjana Milovic
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Zhonghui Xu
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201; Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health/U.S. Department of Health and Human Services, Bethesda, MD 20892 and Detroit, MI 48201
| | - Gaurav Bhatti
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201; Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health/U.S. Department of Health and Human Services, Bethesda, MD 20892 and Detroit, MI 48201
| | - Qing-Sheng Mi
- Immunology Program, Henry Ford Health System, Detroit, MI 48202; Department of Dermatology, Henry Ford Health System, Detroit, MI 48202; and Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Sascha Drewlo
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Adi L Tarca
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201; Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health/U.S. Department of Health and Human Services, Bethesda, MD 20892 and Detroit, MI 48201
| | - Sonia S Hassan
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201; Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health/U.S. Department of Health and Human Services, Bethesda, MD 20892 and Detroit, MI 48201
| | - Nardhy Gomez-Lopez
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201; Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health/U.S. Department of Health and Human Services, Bethesda, MD 20892 and Detroit, MI 48201; Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI 48201
| |
Collapse
|
19
|
Campbell DE, Boyle RJ, Thornton CA, Prescott SL. Mechanisms of allergic disease - environmental and genetic determinants for the development of allergy. Clin Exp Allergy 2015; 45:844-858. [DOI: 10.1111/cea.12531] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- D. E. Campbell
- Children's Hospital Westmead; Sydney NSW Australia
- Discipline of Paediatrics and Child Health; University of Sydney; Sydney NSW Australia
| | - R. J. Boyle
- Section of Paediatrics; Faculty of Medicine; Imperial College; London UK
| | - C. A. Thornton
- Institute of Life Science; College of Medicine; Swansea University; Swansea UK
| | - S. L. Prescott
- School of Paediatrics and Child Health and Telethon KIDS Institute; c/o Princess Margaret Hospital; University of Western Australia; Perth WA Australia
| |
Collapse
|
20
|
Brown MB, von Chamier M, Allam AB, Reyes L. M1/M2 macrophage polarity in normal and complicated pregnancy. Front Immunol 2014; 5:606. [PMID: 25505471 PMCID: PMC4241843 DOI: 10.3389/fimmu.2014.00606] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 11/11/2014] [Indexed: 12/14/2022] Open
Abstract
Tissue macrophages play an important role in all stages of pregnancy, including uterine stromal remodeling (decidualization) before embryo implantation, parturition, and post-partum uterine involution. The activation state and function of utero-placental macrophages are largely dependent on the local tissue microenvironment. Thus, macrophages are involved in a variety of activities such as regulation of immune cell activities, placental cell invasion, angiogenesis, and tissue remodeling. Disruption of the uterine microenvironment, particularly during the early stages of pregnancy (decidualization, implantation, and placentation) can have profound effects on macrophage activity and subsequently impact pregnancy outcome. In this review, we will provide an overview of the temporal and spatial regulation of utero-placental macrophage activation during normal pregnancy in human beings and rodents with a focus on more recent findings. We will also discuss the role of M1/M2 dysregulation within the intrauterine environment during adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Mary B Brown
- D. H. Barron Reproductive and Perinatal Biology Research Program, Department of Infectious Disease and Pathology, College of Veterinary Medicine, University of Florida , Gainesville, FL , USA
| | - Maria von Chamier
- D. H. Barron Reproductive and Perinatal Biology Research Program, Department of Infectious Disease and Pathology, College of Veterinary Medicine, University of Florida , Gainesville, FL , USA
| | - Ayman B Allam
- D. H. Barron Reproductive and Perinatal Biology Research Program, Department of Infectious Disease and Pathology, College of Veterinary Medicine, University of Florida , Gainesville, FL , USA
| | - Leticia Reyes
- D. H. Barron Reproductive and Perinatal Biology Research Program, Department of Infectious Disease and Pathology, College of Veterinary Medicine, University of Florida , Gainesville, FL , USA
| |
Collapse
|
21
|
Lappas M. Caspase-1 activation is increased with human labour in foetal membranes and myometrium and mediates infection-induced interleukin-1β secretion. Am J Reprod Immunol 2013; 71:189-201. [PMID: 24238269 DOI: 10.1111/aji.12174] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 10/11/2013] [Indexed: 12/21/2022] Open
Abstract
PROBLEM Interleukin-1β (IL-1β) is a pro-inflammatory cytokine that is involved in human parturition, especially in the context of infection-induced preterm birth. Caspase-1 is a key component of inflammasomes, which are activated upon infection to trigger the maturation of IL-1β. METHOD OF STUDY To determine the effect of human labour on caspase-1 activation in human foetal membranes and myometrium. In addition, the mechanisms by which inflammasome activation regulates IL-1β production were also be assessed. RESULTS Higher caspase-1 gene and protein expression were detected in foetal membranes myometrium obtained from term labouring women when compared with samples taken from non labouring women. Lipopolysaccharide induced the transcription and secretion of IL-1β from foetal membranes and myometrium; both events were dependent on nuclear factor kappa B (NF-κB). However, levels of extracellular IL-1β were greatly increased by subsequent treatment with the potassium-proton ionophore Adenosine triphosphate (ATP) or nigericin; an effect that was dependent on active caspase-1. Additionally, ATP induced IL-1β secretion via the purinergic P2X7 receptor, whereas the pannexin-1 channel was required for nigericin induced IL-1β secretion. CONCLUSION Taken together, these results demonstrate that caspase-1 activation is increased with human labour in foetal membranes and myometrium, and is required for infection-induced IL-1β secretion.
Collapse
Affiliation(s)
- Martha Lappas
- Department of Obstetrics and Gynaecology, University of Melbourne, Heidelberg, Vic., Australia; Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Vic., Australia
| |
Collapse
|
22
|
Stampalija T, Chaiworapongsa T, Romero R, Tarca AL, Bhatti G, Chiang PJ, Than NG, Ferrazzi E, Hassan SS, Yeo L. Soluble ST2, a modulator of the inflammatory response, in preterm and term labor. J Matern Fetal Neonatal Med 2013; 27:111-21. [PMID: 23688338 DOI: 10.3109/14767058.2013.806894] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Intra-amniotic infection/inflammation (IAI) is causally linked with spontaneous preterm labor and delivery. The ST2L receptor and its soluble form (sST2) are capable of binding to interleukin (IL)-33, a member of the IL-1 superfamily. Members of this cytokine family have been implicated in the onset of spontaneous preterm labor in the context of infection. Soluble ST2 has anti-inflammatory properties, and plasma concentrations are elevated in systemic inflammation, such as sepsis, acute pyelonephritis in pregnancy and the fetal inflammatory response syndrome. The aims of this study were to examine: (1) whether amniotic fluid concentrations of sST2 change with IAI, preterm, and term parturition; and (2) if mRNA expression of ST2 in the chorioamniotic membranes changes with acute histologic chorioamnionitis in women who deliver preterm. METHOD A cross-sectional study was conducted to determine amniotic fluid concentrations of sST2 in: (1) women with preterm labor (PTL) who delivered at term (n=49); (2) women with PTL who delivered preterm without IAI (n=21); (3) women with PTL who delivered preterm with IAI (n=31); (4) term pregnancies not in labor (n=13); and (5) term pregnancies in labor (n=43). The amniotic fluid concentration of sST2 was determined by ELISA. The mRNA expression of ST2 in the chorioamniotic membranes of women who delivered preterm with (n=24), and without acute histologic chorioamnionitis (n=19) was determined by qRT-PCR. RESULTS (1) Patients with PTL who delivered preterm with IAI had a lower median amniotic fluid concentration of sST2 compared to those with PTL who delivered preterm without IAI [median 410 ng/mL, inter-quartile range (IQR) 152-699 ng/mL versus median 825 ng/mL, IQR 493-1216 ng/mL; p=0.0003] and those with PTL who delivered at term [median 410 ng/mL, IQR 152-699 ng/mL versus median 673 ng/mL, IQR 468-1045 ng/mL; p=0.0003]; (2) no significant differences in the median amniotic fluid concentration of sST2 were observed between patients with PTL who delivered at term and those who delivered preterm without IAI (p=0.4), and between women at term in labor and those at term not in labor (p=0.9); (3) the mean mRNA expression of ST2 was 4-fold lower in women who delivered preterm with acute histologic chorioamnionitis than in those without this lesion (p=0.008). CONCLUSIONS The median sST2 amniotic fluid concentration and mRNA expression of ST2 by chorioamniotic membranes is lower in PTL associated with IAI and acute histologic chorioamnionitis than in PTL without these conditions. Changes in the median amniotic fluid sST2 concentration are not observed in preterm and term parturition without IAI. Thus, amniotic fluid sST2 in the presence of IAI behaves differently when compared to sST2 in the plasma of individuals affected by fetal inflammatory response syndrome, acute pyelonephritis in pregnancy, and adult sepsis. Decreased concentrations of sST2 in IAI are likely to promote a pro-inflammatory response, which is important for parturition in the context of infection.
Collapse
Affiliation(s)
- Tamara Stampalija
- Perinatology Research Branch, NICHD/NIH/DHHS , Bethesda, Maryland, and Detroit, Michigan , USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Southcombe JH, Lédée N, d’Hauterive SP, Turner K, Child T, Snider JV, Redman CWG, Sargent IL, Granne I. Detection of soluble ST2 in human follicular fluid and luteinized granulosa cells. PLoS One 2013; 8:e74385. [PMID: 24040238 PMCID: PMC3769262 DOI: 10.1371/journal.pone.0074385] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 07/31/2013] [Indexed: 11/28/2022] Open
Abstract
Follicular fluid (FF) contains various cytokines that are involved with folliculogenesis, some of which have been shown to be associated with oocyte quality and the implantation potential of a resulting embryo. Several IL-1 family members have previously been identified in FF. This study investigates a newly identified member of the family, IL-33, and its receptor ST2, comparing values to those of FF Granulocyte-Colony Stimulating Factor (G-CSF) – a known predictor of Assisted Reproductive Technology (ART) success. FF was collected from patients undergoing in vitro fertilisation/intra-cytoplasmic sperm injection (IVF/ICSI) at oocyte retrieval to analyse IL-33 and sST2 expression in human follicles. sST2, but not IL-33, is highly increased in the FF compared to plasma levels (up to 7.9-fold), with higher levels in larger follicles (p<0.05). Furthermore, we identify that human luteinised granulosa cells are one possible source of the FF sST2, as these cells express and secrete sST2 when cultured ex vivo. FF associated with oocytes which when fertilised develop into good quality embryos have higher sST2 levels than those which are graded average (p<0.01). These embryos were transferred to the patient and levels of FF sST2 compared between successful and unsuccessful ICSI cycles. However unlike G-CSF, sST2 levels cannot be used to predict cycle outcome.
Collapse
Affiliation(s)
- Jennifer H. Southcombe
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, United Kingdom
- * E-mail:
| | | | - Sophie Perrier d’Hauterive
- Medicine of Experimental Reproduction and Centre de Procréation Médicalement Assistée, University of Liège, Liège, Belgium
| | - Karen Turner
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, United Kingdom
- Oxford Fertility Unit, University of Oxford, Oxford, United Kingdom
| | - Tim Child
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, United Kingdom
- Oxford Fertility Unit, University of Oxford, Oxford, United Kingdom
| | - James V. Snider
- Critical Diagnostics, San Diego, California, United States of America
| | | | - Ian L. Sargent
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, United Kingdom
| | - Ingrid Granne
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, United Kingdom
- Oxford Fertility Unit, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
24
|
Fock V, Mairhofer M, Otti GR, Hiden U, Spittler A, Zeisler H, Fiala C, Knöfler M, Pollheimer J. Macrophage-derived IL-33 is a critical factor for placental growth. THE JOURNAL OF IMMUNOLOGY 2013; 191:3734-43. [PMID: 23997215 DOI: 10.4049/jimmunol.1300490] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-33, the most recently discovered member of the IL-1 superfamily and ligand for the transmembrane form of ST2 (ST2L), has been linked to several human pathologies including rheumatoid arthritis, asthma, and cardiovascular disease. Deregulated levels of soluble ST2, the natural IL-33 inhibitor, have been reported in sera of preeclamptic patients. However, the role of IL-33 during healthy pregnancy remains elusive. In the current study, IL-33 was detected in the culture supernatants of human placental and decidual macrophages, identifying them as a major source of secreted IL-33 in the uteroplacental unit. Because flow cytometry and immunofluorescence stainings revealed membranous ST2L expression on specific trophoblast populations, we hypothesized that IL-33 stimulates trophoblasts in a paracrine manner. Indeed, BrdU incorporation assays revealed that recombinant human IL-33 significantly increased proliferation of primary trophoblasts as well as of villous cytotrophoblasts and cell column trophoblasts in placental explant cultures. These effects were fully abolished upon addition of soluble ST2. Interestingly, Western blot and immunofluorescence analyses demonstrated that IL-33 activates AKT and ERK1/2 in primary trophoblasts and placental explants. Inhibitors against PI3K (LY294002) and MEK1/2 (UO126) efficiently blocked IL-33-induced proliferation in all model systems used. In summary, with IL-33, we define for the first time, to our knowledge, a macrophage-derived regulator of placental growth during early pregnancy.
Collapse
Affiliation(s)
- Valerie Fock
- Reproductive Biology Unit, Department of Obstetrics and Fetal-Maternal Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Stampalija T, Romero R, Korzeniewski SJ, Chaemsaithong P, Miranda J, Yeo L, Dong Z, Hassan SS, Chaiworapongsa T. Soluble ST2 in the fetal inflammatory response syndrome: in vivo evidence of activation of the anti-inflammatory limb of the immune response. J Matern Fetal Neonatal Med 2013; 26:1384-93. [PMID: 23488731 DOI: 10.3109/14767058.2013.784258] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Inflammation is a mechanism of host response to infection, which can be harmful when inappropriately modulated. Soluble ST2 (sST2) is a decoy receptor of interleukin (IL)-33, and this complex modulates the balance in the Th1/Th2 immune response. Moreover, sST2 inhibits the production of pro-inflammatory cytokines in cooperation with an anti-inflammatory cytokine, IL-10. The objectives of this study were to: (1) determine whether umbilical cord plasma sST2 concentration differs between preterm neonates with and without funisitis and between those with and without the fetal inflammatory response syndrome (FIRS); and (2) evaluate the relationship between sST2 and IL-10 among neonates with funisitis and/or FIRS. METHODS Umbilical cord plasma was collected from neonates delivered prematurely due to preterm labor or preterm prelabor rupture of membranes with (n = 36), and without funisitis (n = 30). FIRS (umbilical cord IL-6 concentration ≥ 17.5 pg/mL) was identified in 29 neonates. Plasma sST2 and IL-10 concentrations were determined by enzyme linked immune sorbent assay. RESULTS The median umbilical cord plasma sST2 concentration was 6.7-fold higher in neonates with FIRS than in those without FIRS (median 44.6 ng/mL, interquartile range (IQR) 13.8-80.3 ng/mL versus median 6.7 ng/mL, IQR 5.6-20.1 ng/mL; p < 0.0001). Similarly, the median umbilical cord plasma sST2 concentration was 2.7-fold higher in neonates with funisitis than in those without funisitis (median 19.1 ng/mL; IQR 7.1-75.0 ng/mL versus median 7.2 ng/mL; IQR 5.9-23.1 ng/mL; p = 0.008). There was a strong positive correlation between sST2 and IL-10 in neonates with funisitis and/or FIRS (Spearman's Rho = 0.7, p < 0.0001). CONCLUSION FIRS and funisitis are associated with an elevation of umbilical cord plasma concentrations of soluble ST2. This protein represents an important mediator of the immune response in neonates diagnosed with FIRS by promoting an anti-inflammatory effect in association with IL-10.
Collapse
Affiliation(s)
- Tamara Stampalija
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD and Detroit, MI 48201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Stampalija T, Chaiworapongsa T, Romero R, Chaemsaithong P, Korzeniewski SJ, Schwartz AG, Ferrazzi EM, Dong Z, Hassan SS. Maternal plasma concentrations of sST2 and angiogenic/anti-angiogenic factors in preeclampsia. J Matern Fetal Neonatal Med 2013; 26:1359-70. [PMID: 23488689 DOI: 10.3109/14767058.2013.784256] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Angiogenic/anti-angiogenic factors have emerged as one of the promising biomarkers for the prediction of preeclampsia. Since not all patients with preeclampsia can be identified by these analytes, the search for additional biomarkers continues. The soluble form of ST2 (sST2), a protein capable of binding to interleukin (IL)-33 and thus contributing to a Th1-biased immune response, has been reported to be elevated in maternal plasma of women with preeclampsia. The aims of this study were to examine: (1) differences in maternal plasma concentrations of sST2 and IL-33 between women diagnosed with preeclampsia and those having uncomplicated pregnancies; (2) the relationship between sST2, umbilical and uterine artery Doppler velocimetry, and the severity of preeclampsia; and (3) the performance of sST2 and angiogenic/anti-angiogenic factors in identifying patients with preeclampsia at the time of diagnosis. METHODS This cross-sectional study included women with preeclampsia (n = 106) and women with an uncomplicated pregnancy (n = 131). Plasma concentrations of sST2, IL-33, soluble vascular endothelial growth factor receptor (sVEGFR)-1, soluble endoglin (sEng) and placental growth factor (PlGF) were determined by enzyme linked immune sorbent assay. Area under the receiver operating characteristic curve (AUC) for the identification of preeclampsia was examined for each analyte. RESULTS (1) Patients with preeclampsia had a higher mean plasma concentrations of sST2 than those with an uncomplicated pregnancy (p < 0.0001), while no significant difference in the mean plasma concentration of IL-33 between the two groups was observed; (2) the magnitude of this difference was greater in early-onset, compared to late-onset disease, and in severe compared to mild preeclampsia; (3) sST2 plasma concentrations did not correlate with the results of uterine or umbilical artery Doppler velocimetry (p = 0.7 and p = 1, respectively) among women with preeclampsia; (4) sST2 correlated positively with plasma concentrations of sVEGFR1-1 and sEng (Spearman's Rho = 0.72 and 0.63; each p < 0.0001), and negatively with PlGF (Spearman's Rho = -0.56, p < 0.0001); and (5) while the AUC achieved by sST2 and angiogenic/anti-angiogenic factors in identifying women with preeclampsia at the time of diagnosis were non-significantly different prior to term (<37 weeks of gestation), thereafter the AUC achieved by sST2 was significantly less than that achieved by angiogenic/anti-angiogenic factors. CONCLUSIONS Preeclampsia is associated with increased maternal plasma concentrations of sST2. The findings that sST2 concentrations do not correlate with uterine or umbilical artery Doppler velocimetry in women with preeclampsia suggest that elevated maternal plasma sST2 concentrations in preeclampsia are not related to the increased impedance to flow in the utero-placental circulation. The performance of sST2 in identifying preeclampsia at the time of diagnosis prior to 37 weeks of gestation was comparable to that of angiogenic/anti-angiogenic factors. It remains to be elucidated if an elevation of maternal plasma sST2 concentrations in pregnancy is specific to preeclampsia.
Collapse
|