51
|
Pereyra S, Sosa C, Bertoni B, Sapiro R. Transcriptomic analysis of fetal membranes reveals pathways involved in preterm birth. BMC Med Genomics 2019; 12:53. [PMID: 30935390 PMCID: PMC6444860 DOI: 10.1186/s12920-019-0498-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 03/10/2019] [Indexed: 12/21/2022] Open
Abstract
Background Preterm birth (PTB), defined as infant delivery before 37 weeks of completed gestation, results from the interaction of both genetic and environmental components and constitutes a complex multifactorial syndrome. Transcriptome analysis of PTB has proven challenging because of the multiple causes of PTB and the numerous maternal and fetal gestational tissues that must interact to facilitate parturition. The transcriptome of the chorioamnion membranes at the site of rupture in PTB and term fetuses may reflect the molecular pathways of preterm labor. Methods In this work, chorioamnion membranes from severe preterm and term fetuses were analyzed using RNA sequencing. Functional annotations and pathway analysis of differentially expressed genes were performed with the GAGE and GOSeq packages. A subset of differentially expressed genes in PTB was validated in a larger cohort using qRT-PCR and by comparing our results with genes and pathways previously reported in the literature. Results A total of 270 genes were differentially expressed (DE): 252 were upregulated and 18 were down-regulated in severe preterm births relative to term births. Inflammatory and immunological pathways were upregulated in PTB. Both types of pathways were previously suggested to lead to PTB. Pathways that were not previously reported in PTB, such as the hemopoietic pathway, appeared upregulated in preterm membranes. A group of 18 downregulated genes discriminated between term and severe preterm cases. These genes potentially characterize a severe preterm transcriptome pattern and therefore are candidate genes for understanding the syndrome. Some of the downregulated genes are involved in the nervous system, morphogenesis (WNT1, DLX5, PAPPA2) and ion channel complexes (KCNJ16, KCNB1), making them good candidates as biomarkers of PTB. Conclusions The identification of this DE gene pattern will help with the development of a multi-gene disease classifier. These markers were generated in an admixed South American population in which PTB has a high incidence. Since the genetic background may differentially impact different populations, it is necessary to include populations such as those from South America and Africa, which are usually excluded from high-throughput approaches. These classifiers should be compared to those in other populations to obtain a global landscape of PTB. Electronic supplementary material The online version of this article (10.1186/s12920-019-0498-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Silvana Pereyra
- Departamento de Genética, Facultad de Medicina, Universidad de la República, Av. General Flores 2125, C.P, 11800, Montevideo, Uruguay
| | - Claudio Sosa
- Clínica Ginecotologica "C", Centro Hospitalario Pereira Rossell, Facultad de Medicina, Universidad de la República, Bvar. General Artigas 1590, C:P.11600, Montevideo, Uruguay
| | - Bernardo Bertoni
- Departamento de Genética, Facultad de Medicina, Universidad de la República, Av. General Flores 2125, C.P, 11800, Montevideo, Uruguay
| | - Rossana Sapiro
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Av. General Flores 2125, C.P, 11800, Montevideo, Uruguay.
| |
Collapse
|
52
|
Arenas-Hernandez M, Romero R, Xu Y, Panaitescu B, Garcia-Flores V, Miller D, Ahn H, Done B, Hassan SS, Hsu CD, Tarca AL, Sanchez-Torres C, Gomez-Lopez N. Effector and Activated T Cells Induce Preterm Labor and Birth That Is Prevented by Treatment with Progesterone. THE JOURNAL OF IMMUNOLOGY 2019; 202:2585-2608. [PMID: 30918041 DOI: 10.4049/jimmunol.1801350] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 02/26/2019] [Indexed: 12/21/2022]
Abstract
Preterm labor commonly precedes preterm birth, the leading cause of perinatal morbidity and mortality worldwide. Most research has focused on establishing a causal link between innate immune activation and pathological inflammation leading to preterm labor and birth. However, the role of maternal effector/activated T cells in the pathogenesis of preterm labor/birth is poorly understood. In this study, we first demonstrated that effector memory and activated maternal T cells expressing granzyme B and perforin are enriched at the maternal-fetal interface (decidua) of women with spontaneous preterm labor. Next, using a murine model, we reported that prior to inducing preterm birth, in vivo T cell activation caused maternal hypothermia, bradycardia, systemic inflammation, cervical dilation, intra-amniotic inflammation, and fetal growth restriction, all of which are clinical signs associated with preterm labor. In vivo T cell activation also induced B cell cytokine responses, a proinflammatory macrophage polarization, and other inflammatory responses at the maternal-fetal interface and myometrium in the absence of an increased influx of neutrophils. Finally, we showed that treatment with progesterone can serve as a strategy to prevent preterm labor/birth and adverse neonatal outcomes by attenuating the proinflammatory responses at the maternal-fetal interface and cervix induced by T cell activation. Collectively, these findings provide mechanistic evidence showing that effector and activated T cells cause pathological inflammation at the maternal-fetal interface, in the mother, and in the fetus, inducing preterm labor and birth and adverse neonatal outcomes. Such adverse effects can be prevented by treatment with progesterone, a clinically approved strategy.
Collapse
Affiliation(s)
- Marcia Arenas-Hernandez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI 48201.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201.,Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, 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 48824.,Center for Molecular Obstetrics and Genetics, Wayne State University, Detroit, MI 48201
| | - Yi Xu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI 48201.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Bogdan Panaitescu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI 48201.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI 48201.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Derek Miller
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI 48201.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Hyunyoung Ahn
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Bogdan Done
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI 48201.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Sonia S Hassan
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI 48201.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201.,Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Chaur-Dong Hsu
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201.,Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201
| | - Adi L Tarca
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI 48201.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201.,Department of Computer Science, Wayne State University College of Engineering, Detroit, MI 48202; and
| | - Carmen Sanchez-Torres
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI 48201; .,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201.,Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI 48201
| |
Collapse
|
53
|
Flores-Espinosa P, Vega-Sánchez R, Mancilla-Herrera I, Bermejo-Martínez L, Preciado-Martínez E, Olmos-Ortiz A, Méndez I, Estrada-Gutiérrez G, Quesada-Reyna B, Helguera-Repetto C, Irles C, Zaga-Clavellina V. Prolactin selectively inhibits the LPS-induced chemokine secretion of human foetal membranes. J Matern Fetal Neonatal Med 2019; 33:4083-4089. [PMID: 30880514 DOI: 10.1080/14767058.2019.1596255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Background: Inflammation is a condition that jeopardizes the continuity of pregnancy because it increases the secretion of chemokines that favor the migration of leukocytes from maternal and fetal circulations to the cervix, placenta, and the chorioamniotic membranes. During pregnancy, the level of prolactin (PRL) in the amniotic fluid is high; there is evidence to suggest that PRL contributes to maintain a privileged immune environment in the amniotic cavity. We test the effect of prolactin on the secretion profile of chemokines in human fetal membranes.Methods: Nine fetal membranes collected from healthy nonlabouring cesarean deliveries at term. We placed whole membrane explants in a two-chamber culture system. Choriodecidua and amniotic chambers were pretreated with 250, 500, 1000, or 4000 ng/ml of PRL for 24 h, then choriodecidua was cotreated with 500 ng/ml of lipopolysaccharide (LPS) and PRL for 24 h. We used ELISA to measure secreted levels of four chemokines (RANTES, monocyte chemoattractant protein 1 (MCP-1), MIP-1α, and IL-8) in both amnion and choriodecidua regions.Results: In comparison with basal conditions, LPS treatment induced significantly higher secretion of RANTES, MCP-1, and MIP-1α, but not of IL-8. RANTES was mainly produced by choriodecidua and cotreatment with PRL significantly decreased its LPS-induced secretion. MCP-1 was primarily produced by the amnion and its secretion was only inhibited by 4000 ng/ml of PRL. Both membrane regions produced MIP-1α, which was significantly inhibited at 1000 and 4000 ng/ml PRL concentrations. IL-8 showed no significant changes regardless of PRL concentration.Conclusion: PRL inhibits the differential secretion of proinflammatory chemokines by human fetal membranes.
Collapse
Affiliation(s)
- P Flores-Espinosa
- Immunobiochemistry Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes (INPer IER), Mexico City, Mexico
| | - R Vega-Sánchez
- Nutrition and Bioprogramming Branch, INPer IER, Mexico City, Mexico
| | | | - L Bermejo-Martínez
- Immunobiochemistry Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes (INPer IER), Mexico City, Mexico
| | - E Preciado-Martínez
- Immunobiochemistry Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes (INPer IER), Mexico City, Mexico
| | - A Olmos-Ortiz
- Immunobiochemistry Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes (INPer IER), Mexico City, Mexico
| | - I Méndez
- Cellular and Molecular Neurobiology Branch, Instituto de Neurobiología, Querétaro, Mexico
| | - G Estrada-Gutiérrez
- Immunobiochemistry Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes (INPer IER), Mexico City, Mexico
| | - B Quesada-Reyna
- Gyneco-Obstetric Division, Hospital de Ginecología-Obstetricia No. 4, Mexico City, Mexico
| | - C Helguera-Repetto
- Immunobiochemistry Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes (INPer IER), Mexico City, Mexico
| | - C Irles
- Physiology and Cell Development Branch, INPer IER, Mexico City, Mexico
| | - V Zaga-Clavellina
- Immunobiochemistry Branch, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes (INPer IER), Mexico City, Mexico
| |
Collapse
|
54
|
Bonney EA, Johnson MR. The role of maternal T cell and macrophage activation in preterm birth: Cause or consequence? Placenta 2019; 79:53-61. [PMID: 30929747 DOI: 10.1016/j.placenta.2019.03.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 03/06/2019] [Indexed: 12/18/2022]
Abstract
The role of the immune system in term (TL) and preterm labor (PTL) is unknown. Despite the fact that globally, PTL remains the most important cause of childhood mortality. Infection, typically of the fetal membranes, termed chorioamnionitis, is the best-understood driver of PTL, but the mechanisms underpinning other causes, including idiopathic and stretch-induced PTL, are unclear, but may well involve activation of the maternal immune system. The final common pathway of placental dysfunction, fetal membrane rupture, cervical dilation and uterine contractions are highly complex processes. At term, choriodecidual rather than myometrial inflammation is thought to drive the onset of labor and similar findings are present in different types of PTL including idiopathic PTL. Although accumulated data has confirmed an association between the immune response and preterm birth, there is yet a need to understand if this response is an initiator or a consequence of tissue-level dysregulation. This review focuses on the potential role of macrophages and T cells in innate and adaptive immunity relevant to preterm birth in humans and animal models.
Collapse
Affiliation(s)
- Elizabeth A Bonney
- Department of Obstetrics, Gynecology and Reproductive Sciences University of Vermont, Larner College of Medicine, Burlington, VT, USA.
| | - Mark R Johnson
- Faculty of Medicine, Department of Surgery & Cancer, Imperial College, London, United Kingdom
| |
Collapse
|
55
|
Saliba J, Coutaud B, Solovieva V, Lu F, Blank V. Regulation of CXCL1 chemokine and CSF3 cytokine levels in myometrial cells by the MAFF transcription factor. J Cell Mol Med 2019; 23:2517-2525. [PMID: 30669188 PMCID: PMC6433675 DOI: 10.1111/jcmm.14136] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/01/2018] [Indexed: 12/16/2022] Open
Abstract
Cytokines play key roles in a variety of reproductive processes including normal parturition as well as preterm birth. Our previous data have shown that MAFF, a member of the MAF family of bZIP transcription factors, is rapidly induced by pro‐inflammatory cytokines in PHM1‐31 myometrial cells. We performed loss‐of‐function studies in PHM1‐31 cells to identify MAFF dependent genes. We showed that knockdown of MAFF significantly decreased CXCL1 chemokine and CSF3 cytokine transcript and protein levels. Using chromatin immunoprecipitation analyzes, we confirmed CXCL1 and CSF3 genes as direct MAFF targets. We also demonstrated that MAFF function in PHM1‐31 myometrial cells is able to control cytokine and matrix metalloproteinase gene expression in THP‐1 monocytic cells in a paracrine fashion. Our studies provide valuable insights into the MAFF dependent transcriptional network governing myometrial cell function. The data suggest a role of MAFF in parturition and/or infection‐induced preterm labour through modulation of inflammatory processes in the microenvironment.
Collapse
Affiliation(s)
- James Saliba
- Lady Davis Institute for Medical Research, Montreal, Quebec, Canada
| | - Baptiste Coutaud
- Lady Davis Institute for Medical Research, Montreal, Quebec, Canada
| | - Vera Solovieva
- Lady Davis Institute for Medical Research, Montreal, Quebec, Canada
| | - Fangshi Lu
- Lady Davis Institute for Medical Research, Montreal, Quebec, Canada
| | - Volker Blank
- Lady Davis Institute for Medical Research, Montreal, Quebec, Canada.,Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada.,Department of Physiology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
56
|
Wang B, Wang P, Parobchak N, Treff N, Tao X, Wang J, Rosen T. Integrated RNA-seq and ChIP-seq analysis reveals a feed-forward loop regulating H3K9ac and key labor drivers in human placenta. Placenta 2019; 76:40-50. [PMID: 30642670 DOI: 10.1016/j.placenta.2019.01.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/06/2019] [Accepted: 01/08/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Chromatin alterations are important mediators of gene expression changes. We have recently shown that activated non-canonical NF-κB signaling (RelB/p52) recruits histone acetyltransferase CBP and deacetylase HDAC1 to selectively acetylate H3K9 (H3K9ac) to induce expression of corticotropin-releasing hormone (CRH) and prostaglandin-endoperoxide synthase-2 (PTGS2) in the human placenta. Both of these genes play a role in initiating parturition in human pregnancy. METHODS We performed chromatin immunoprecipitation followed by gene sequencing (ChIP-seq) in primary term human cytotrophoblast (CTB) with use of antibodies to RelB, CBP, HDAC1 and H3K9ac. We further associated these chromatin alterations with gene expression changes from mid-trimester to term in CTB by RNA sequencing (RNA-seq). RESULTS We detected a genome-wide differential gene enrichment between mid-trimester and term human placenta. Pathway analysis identified that cytokine-cytokine receptor interaction, NF-κB, and TNF are the leading pathways enriched in term placenta and associated with these chromatin alterations. DISCUSSIONS Our analysis has provided the first-time characterization of the key players of human placental origin with molecular changes resulting from chromatin modifications, which could drive human labor.
Collapse
Affiliation(s)
- Bingbing Wang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
| | - Panwen Wang
- Department of Health Sciences Research, Center for Individualized Medicine, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Nataliya Parobchak
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Nathan Treff
- Genomic Prediction Inc, North Brunswick, NJ, 08902, USA
| | - Xin Tao
- Foundation for Embryonic Competence, Basking Ridge, NJ, 07920, USA
| | - Junwen Wang
- Department of Health Sciences Research, Center for Individualized Medicine, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Todd Rosen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
57
|
Ronzoni S, Steckle V, D'Souza R, Murphy KE, Lye S, Shynlova O. Cytokine Changes in Maternal Peripheral Blood Correlate With Time-to-Delivery in Pregnancies Complicated by Premature Prelabor Rupture of the Membranes. Reprod Sci 2018; 26:1266-1276. [PMID: 30541390 DOI: 10.1177/1933719118815590] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Premature prelabor rupture of the membranes (PPROM) causes one-third of preterm births worldwide and is most likely caused by subclinical intrauterine infection and/or inflammation. We proposed that women with systemic inflammation at the time of PPROM would have shorter latency. Peripheral blood samples were collected from 20 singleton pregnant women with PPROM between 23 ± 1 and 33 ± 6 weeks. The first sample was drawn within 48 hours of admission, followed by weekly blood draws until delivery. Pregnancies complicated with acute chorioamnionitis, preeclampsia, intrauterine growth restriction, obesity, substance abuse, and chronic maternal disease were excluded. Twenty uncomplicated, gestational age-matched pregnancies served as controls. Plasma concentration of 39 cytokines was measured longitudinally using Luminex assays to investigate their value as predictive biomarkers of latency. Women with PPROM exhibited significantly lower plasma concentration of interferon-γ-inducible protein 10-Chemokine (c-x-c motif; IP10/CXCL10), Chemokine (c-x-c motif) Ligand 9 (MIG/CXCL9), Platelet-derived growth factor BB (PDGFbb), and cutaneous T cell-attracting chemokine, also known as CCL27/CCL27 than controls at admission but significantly elevated interleukin (IL)1RA, tumor necrosis factor α, monocyte chemotactic protein-1/CCL2 at delivery compared to admission. Women with PPROM who delivered within 7 days had significantly lower plasma concentration of anti-inflammatory cytokine IL1RA than those with latency periods >7 days. The IL1RA and endotoxin activity in conjunction with clinical parameters results in excellent prediction of latency to delivery (area under the receiver-operating characteristic curve = 0.91). We concluded that higher levels of anti-inflammatory cytokines in women with PPROM were associated with increased latency until delivery, likely due to counterbalancing of proinflammatory load. When used in conjunction with other predictive characteristics of time until delivery, cytokines may further assist clinical decision-making and optimize pregnancy outcomes in women with PPROM.
Collapse
Affiliation(s)
- Stefania Ronzoni
- 1 Department of Obstetrics and Gynecology, Sunnybrook Health Science Centre, University of Toronto, Ontario, Canada
| | - Valerie Steckle
- 2 Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Toronto, Ontario, Canada.,3 Department of Physiology, University of Toronto, Ontario, Canada
| | - Rohan D'Souza
- 2 Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Toronto, Ontario, Canada.,4 Department of Obstetrics and Gynecology, University of Toronto, Ontario, Canada
| | - Kellie E Murphy
- 2 Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Toronto, Ontario, Canada.,4 Department of Obstetrics and Gynecology, University of Toronto, Ontario, Canada
| | - Stephen Lye
- 2 Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Toronto, Ontario, Canada.,3 Department of Physiology, University of Toronto, Ontario, Canada.,4 Department of Obstetrics and Gynecology, University of Toronto, Ontario, Canada
| | - Oksana Shynlova
- 2 Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Toronto, Ontario, Canada.,3 Department of Physiology, University of Toronto, Ontario, Canada.,4 Department of Obstetrics and Gynecology, University of Toronto, Ontario, Canada
| |
Collapse
|
58
|
Mizoguchi M, Ishida Y, Nosaka M, Kimura A, Kuninaka Y, Yahata T, Nanjo S, Toujima S, Minami S, Ino K, Mukaida N, Kondo T. Prevention of lipopolysaccharide-induced preterm labor by the lack of CX3CL1-CX3CR1 interaction in mice. PLoS One 2018; 13:e0207085. [PMID: 30399192 PMCID: PMC6219809 DOI: 10.1371/journal.pone.0207085] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/24/2018] [Indexed: 01/28/2023] Open
Abstract
Preterm labor (PTL) is the most common cause of neonatal death and long-term adverse outcome. The pharmacological agents for PTL prevention are palliative and frequently fail to prevent PTL and improve neonatal outcome. It is essential to fully understand the molecular mechanisms of PTL in order to develop novel therapeutic methods against PTL. Several lines of evidence indicate some chemokines are expressed in gestational tissues during labor or PTL. To reveal the pathophysiological roles of the CX3CL1-CX3CR1 axis in PTL, we performed present study using LPS-induced PTL mice model in CX3CR1-deficient (Cx3cr1-/-) mice. We indicated that PTL was suppressed in Cx3cr1-/- mice and immunoneutralization of CX3CL1 in WT mice. From immunohistochemical and the gene expression analyses, the CX3CL1-CX3CR1 axis has detrimental roles in PTL through intrauterine recruitment of macrophages and the enhancement of macrophage-derived inflammatory mediators. Thus, the CX3CL1-CX3CR1 axis may be a good molecular target for preventing PTL.
Collapse
Affiliation(s)
- Mika Mizoguchi
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama, Japan
| | - Yuko Ishida
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Mizuho Nosaka
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Akihiko Kimura
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yumi Kuninaka
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Tamaki Yahata
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama, Japan
| | - Sakiko Nanjo
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama, Japan
| | - Saori Toujima
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama, Japan
| | - Sawako Minami
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama, Japan
| | - Kazuhiko Ino
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
- * E-mail:
| |
Collapse
|
59
|
Lim W, Bae H, Bazer FW, Song G. Characterization of C-C motif chemokine ligand 4 in the porcine endometrium during the presence of the maternal-fetal interface. Dev Biol 2018; 441:146-158. [PMID: 30056935 DOI: 10.1016/j.ydbio.2018.06.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 06/27/2018] [Accepted: 06/29/2018] [Indexed: 11/27/2022]
Abstract
Chemokines and their receptors play a crucial role in embryo implantation at the maternal-fetal interface during pregnancy. In this study, we investigated the role of CCL4 in development of the porcine endometrium in the early gestational period. Porcine CCL4 showed high similarity with the human counterpart, and mRNA expression of CCL4 and its receptor (CCR5) was predominantly present in the endometrium during early pregnancy. Treatment with CCL4 increased proliferation of porcine uterine luminal epithelial (pLE) cells by activation of PI3K and MAPK signal transduction. In addition, CCL4 recovered the endoplasmic-reticulum stress-reduced proliferation and decreased the unfolded protein response in pLE cells. Besides, the lipopolysaccharide-activated NF-κB pathway was suppressed in response to CCL4 in pLE cells. Inhibition of CCR5 decreased the proliferation of pLE cells and activation of the PI3K and MAPK pathways by CCL4. Furthermore, CCL4 enhanced conceptus-maternal interactions between porcine trophectoderm (pTr) cells and pLE cells during early pregnancy by activating expression of migration and implantation-related genes. Collectively, the results suggest that CCL4 may improve successful implantation in early pregnancy in pigs.
Collapse
Affiliation(s)
- Whasun Lim
- Department of Biomedical Sciences, Catholic Kwandong University, Gangneung 25601, Republic of Korea
| | - Hyocheol Bae
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Fuller W Bazer
- Center for Animal Biotechnology and Genomics and Department of Animal Science, Texas A&M University, College Station 77843, TX, USA
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
60
|
Rinaldi SF, Makieva S, Saunders PT, Rossi AG, Norman JE. Immune cell and transcriptomic analysis of the human decidua in term and preterm parturition. Mol Hum Reprod 2018; 23:708-724. [PMID: 28962035 PMCID: PMC5909855 DOI: 10.1093/molehr/gax038] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 08/14/2017] [Indexed: 12/11/2022] Open
Abstract
STUDY QUESTION Is labour, both at term and preterm, associated with alterations in decidual lymphocyte densities and widespread changes to the decidual transcriptome? SUMMARY ANSWER The onset of parturition, both at term and preterm, is associated with widespread gene expression changes in the decidua, many of which are related to inflammatory signalling, but is not associated with changes in the number of any of the decidual lymphocyte populations examined. WHAT IS KNOWN ALREADY Given its location, directly at the maternal–foetal interface, the decidua is likely to play a pivotal role in the onset of parturition, however, the molecular events occurring in the decidua in association with the onset of labour, both at term and preterm, remain relatively poorly defined. Using flow cytometry and microarray analysis, the present study aimed to investigate changes to the immune cell milieu of the decidua in association with the onset of parturition and define the decidual gene signature associated with term and preterm labour (PTL). STUDY DESIGN, SIZE, DURATION This study used decidual samples collected from 36 women across four clinical groups: term (38–42 weeks of gestation) not in labour, TNL; term in labour, TL; preterm (<35 weeks of gestation)not in labour, PTNL; and preterm in labour, PTL. PARTICIPANTS/MATERIALS, SETTING, METHODS Decidual lymphocytes were isolated from fresh decidual tissue collected from women in each of our four patient groups and stained with a panel of antibodies (CD45, CD3, CD19, CD56, CD4, CD8 and TCRVα24-Jα18) to investigate lymphocyte populations present in the decidua (TNL, n = 8; TL, n = 7; PTNL, n = 5; PTL, n = 5). RNA was extracted from decidual tissue and subjected to Illumina HT-12v4.0 BeadChip expression microarrays (TNL, n = 11; TL, n = 8; PTNL, n = 7; PTL, n = 10). Quantitative real-time PCR (qRT-PCR) was used to validate the microarray results. MAIN RESULTS AND THE ROLE OF CHANCE The relative proportions of decidual lymphocytes (T cells, NK cells, B cells and invariant natural killer (iNKT) cells) were unaffected by either gestation or labour status. However, we found elevated expression of the non-classical MHC-protein, CD1D, in PTL decidua samples (P < 0.05), suggesting the potential for increased activation of decidual invariant NKT (iNKT) cells in PTL. Both term and PTL were associated with widespread gene expression changes, particularly related to inflammatory signalling. Up-regulation of candidate genes in TL (IL-6, PTGS2, ATF3, IER3 and TNFAIP3) and PTL (CXCL8, MARCO, LILRA3 and PLAU) were confirmed by qRT-PCR analysis. LARGE SCALE DATA Microarray data are available at www.ebi.ac.uk/arrayexpress under accession number E-MTAB-5353. LIMITATIONS REASONS FOR CAUTION Whilst no changes in lymphocyte number were observed across our patient samples, we did not investigate the activation state of any of the immune cell sub-populations examined, therefore, it is possible that the function of these cells may be altered in association with labour onset. Additionally, the results of our transcriptomic analyses are descriptive and at this stage, we cannot prove direct causal link with the up-regulation of any of the genes examined and the onset of either term or PTL. WIDER IMPLICATIONS OF THE FINDINGS Our findings demonstrate that the onset of parturition is associated with widespread changes to the decidual transcriptome, and there are distinct gene expression changes associated with term and PTL. We confirmed that an inflammatory signature is present within the decidua, and we also report the up-regulation of several genes involved in regulating the inflammatory response. The identification of genes involved in regulating the inflammatory response may provide novel molecular targets for the development of new, more effective therapies for the prevention of preterm birth (PTB). Such targets are urgently required. STUDY FUNDING AND COMPETING INTEREST(S) This work was supported by Medical Research Council (grant number MR/L002657/1) and Tommy's, the baby charity. Jane Norman has had research grants from the charity Tommy's and from the National Institute for Health Research on PTB during the lifetime of this project. Jane Norman also sits on a data monitoring committee for GSK for a study on PTB prevention and her institution receives financial recompense for this. The other authors do not have any conflicts of interest to declare.
Collapse
Affiliation(s)
- S F Rinaldi
- MRC Centre for Reproductive Health and Tommy's Centre for Maternal and Fetal Health, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - S Makieva
- MRC Centre for Reproductive Health and Tommy's Centre for Maternal and Fetal Health, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - P T Saunders
- MRC Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - A G Rossi
- MRC Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - J E Norman
- MRC Centre for Reproductive Health and Tommy's Centre for Maternal and Fetal Health, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| |
Collapse
|
61
|
Amabebe E, Chapman DR, Stern VL, Stafford G, Anumba DOC. Mid-gestational changes in cervicovaginal fluid cytokine levels in asymptomatic pregnant women are predictive markers of inflammation-associated spontaneous preterm birth. J Reprod Immunol 2018; 126:1-10. [PMID: 29367099 PMCID: PMC5886036 DOI: 10.1016/j.jri.2018.01.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 11/10/2017] [Accepted: 01/05/2018] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Perturbation of the choriodecidual space before the onset of spontaneous preterm birth (sPTB) could lead to a concomitant rise in both cervicovaginal fluid (CVF) cytokine and fetal fibronectin (FFN), and assessing the concentrations of both markers could improve the prediction of sPTB (delivery before 37 completed weeks of gestation). Therefore, we prospectively determined mid-trimester changes in CVF cytokine and FFN concentrations, and their predictive capacity for sPTB in asymptomatic pregnant women. STUDY DESIGN CVF collected at 20+0-22+6 weeks (n = 47: Preterm-delivered = 22, Term-delivered = 25) and 26+0-28+6 weeks (n = 50: Preterm-delivered = 17, Term-delivered = 33) from 63 asymptomatic pregnant women at risk of sPTB were examined. Cytokine and FFN concentrations were determined by multiplexed bead-based immunoassay and 10Q Rapid analysis (Hologic, MA, USA) respectively. The 20+0-22+6/26+0-28+6 weeks ratios of cytokines and FFN concentrations were compared between preterm- and term-delivered women using Receiver Operating Characteristics curves to predict sPTB. Also, bacterial 16S rDNA from 64 samples (20+0-22+6 weeks n = 36, 26+0-28+6 weeks n = 28) was amplified by polymerase chain reaction to determine associations between vaginal microflora, cytokine and FFN concentrations. RESULTS Changes in RANTES and IL-1β concentrations between 20+0-22+6 and 26+0-28+6 weeks, expressed as a ratios, were predictive of sPTB, RANTES (AUC = 0.82, CI = 0.62-0.94) more so than IL-1β (AUC = 0.71, CI = 0.53-0.85) and FFN (not predictive). Combining these markers (AUC = 0.83, CI = 0.63-0.95) showed similar predictive capacity as RANTES alone. FFN concentrations at 26+0-28+6 weeks correlated with IL-1β (r = 0.4, P = 0.002) and RANTES (r = 0.3, P = 0.03). In addition, there was increased prevalence of vaginal anaerobes including Bacteroides, Fusobacterium and Mobiluncus between gestational time points in women who experienced sPTB compared to the term women (P = 0.0006). CONCLUSIONS CVF RANTES and IL-1β in mid-trimester of pregnancy correlate with quantitative FFN. The levels of CVF RANTES and IL-1β decline significantly in women who deliver at term unlike women who deliver preterm. This observation suggests that sPTB may be characterised by sustained choriodecidual inflammation and may have clinical value in serial screening for sPTB if confirmed by larger studies.
Collapse
Affiliation(s)
- Emmanuel Amabebe
- Academic Unit of Reproductive and Developmental Medicine, The University of Sheffield, Sheffield, UK
| | - David R Chapman
- Academic Unit of Reproductive and Developmental Medicine, The University of Sheffield, Sheffield, UK
| | - Victoria L Stern
- Academic Unit of Reproductive and Developmental Medicine, The University of Sheffield, Sheffield, UK
| | - Graham Stafford
- Integrated BioSciences Group, School of Clinical Dentistry, The University of Sheffield, Sheffield, UK
| | - Dilly O C Anumba
- Academic Unit of Reproductive and Developmental Medicine, The University of Sheffield, Sheffield, UK.
| |
Collapse
|
62
|
Presicce P, Park CW, Senthamaraikannan P, Bhattacharyya S, Jackson C, Kong F, Rueda CM, DeFranco E, Miller LA, Hildeman DA, Salomonis N, Chougnet CA, Jobe AH, Kallapur SG. IL-1 signaling mediates intrauterine inflammation and chorio-decidua neutrophil recruitment and activation. JCI Insight 2018; 3:98306. [PMID: 29563340 DOI: 10.1172/jci.insight.98306] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 02/13/2018] [Indexed: 12/31/2022] Open
Abstract
Neutrophil infiltration of the chorioamnion-decidua tissue at the maternal-fetal interface (chorioamnionitis) is a leading cause of prematurity, fetal inflammation, and perinatal mortality. We induced chorioamnionitis in preterm rhesus macaques by intraamniotic injection of LPS. Here, we show that, during chorioamnionitis, the amnion upregulated phospho-IRAK1-expressed neutrophil chemoattractants CXCL8 and CSF3 in an IL-1-dependent manner. IL-1R blockade decreased chorio-decidua neutrophil accumulation, neutrophil activation, and IL-6 and prostaglandin E2 concentrations in the amniotic fluid. Neutrophils accumulating in the chorio-decidua had increased survival mediated by BCL2A1, and IL-1R blockade also decreased BCL2A1+ chorio-decidua neutrophils. Readouts for inflammation in a cohort of women with preterm delivery and chorioamnionitis were similar to findings in the rhesus macaques. IL-1 is a potential therapeutic target for chorioamnionitis and associated morbidities.
Collapse
Affiliation(s)
| | | | | | | | - Courtney Jackson
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | - Cesar M Rueda
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Emily DeFranco
- Department of Obstetrics/Gynecology, Maternal-Fetal Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Lisa A Miller
- California National Primate Research Center, Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, UCD, Davis, California, USA
| | - David A Hildeman
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Nathan Salomonis
- Division of Biomedical informatics, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio, USA
| | - Claire A Chougnet
- Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | |
Collapse
|
63
|
Miller D, Romero R, Unkel R, Xu Y, Vadillo-Ortega F, Hassan SS, Gomez-Lopez N. CD71+ erythroid cells from neonates born to women with preterm labor regulate cytokine and cellular responses. J Leukoc Biol 2018; 103:761-775. [PMID: 29389020 DOI: 10.1002/jlb.5a0717-291rrr] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 12/08/2017] [Accepted: 12/14/2017] [Indexed: 12/21/2022] Open
Abstract
Neonatal CD71+ erythroid cells are thought to have immunosuppressive functions. Recently, we demonstrated that CD71+ erythroid cells from neonates born to women who underwent spontaneous preterm labor (PTL) are reduced to levels similar to those of term neonates; yet, their functional properties are unknown. Herein, we investigated the functionality of CD71+ erythroid cells from neonates born to women who underwent spontaneous preterm or term labor. CD71+ erythroid cells from neonates born to women who underwent PTL displayed a similar mRNA profile to that of those from term neonates. The direct contact between preterm or term neonatal CD71+ erythroid cells and maternal mononuclear immune cells, but not soluble products from these cells, induced the release of proinflammatory cytokines and a reduction in the release of TGF-β. Moreover, PTL-derived neonatal CD71+ erythroid cells (1) modestly altered CD8+ T cell activation; (2) inhibited conventional CD4+ and CD8+ T-cell expansion; (3) suppressed the expansion of CD8+ regulatory T cells; (4) regulated cytokine responses mounted by myeloid cells in the presence of a microbial product; and (5) indirectly modulated T-cell cytokine responses. In conclusion, neonatal CD71+ erythroid cells regulate neonatal T-cell and myeloid responses and their direct contact with maternal mononuclear cells induces a proinflammatory response. These findings provide insight into the biology of neonatal CD71+ erythroid cells during the physiologic and pathologic processes of labor.
Collapse
Affiliation(s)
- Derek Miller
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, 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, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, 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
| | - Ronald Unkel
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - 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, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Felipe Vadillo-Ortega
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Facultad de Medicina, UNAM, Branch at Instituto Nacional de Medicina Genomica, Mexico City, Mexico
| | - Sonia S Hassan
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, 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
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, 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
| |
Collapse
|
64
|
do Imperio GE, Bloise E, Javam M, Lye P, Constantinof A, Dunk C, Dos Reis FM, Lye SJ, Gibb W, Ortiga-Carvalho TM, Matthews SG. Chorioamnionitis Induces a Specific Signature of Placental ABC Transporters Associated with an Increase of miR-331-5p in the Human Preterm Placenta. Cell Physiol Biochem 2018; 45:591-604. [PMID: 29402780 PMCID: PMC7202864 DOI: 10.1159/000487100] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/10/2017] [Indexed: 12/16/2022] Open
Abstract
Background/Aims The ATP-binding cassette (ABC) transporters mediate drug biodisposition and immunological responses in the placental barrier. In vitro infective challenges alter expression of specific placental ABC transporters. We hypothesized that chorioamnionitis induces a distinct pattern of ABC transporter expression. Methods Gene expression of 50 ABC transporters was assessed using TaqMan® Human ABC Transporter Array, in preterm human placentas without (PTD; n=6) or with histological chorioamnionitis (PTDC; n=6). Validation was performed using qPCR, immunohistochemistry and Western blot. MicroRNAs known to regulate P-glycoprotein (P-gp) were examined by qPCR. Results Up-regulation of ABCB9, ABCC2 and ABCF2 mRNA was detected in chorioamnionitis (p<0.05), whereas placental ABCB1 (P-gp; p=0.051) and ABCG2 (breast cancer resistance protein-BCRP) mRNA levels (p=0.055) approached near significant up-regulation. In most cases, the magnitude of the effect significantly correlated to the severity of inflammation. Upon validation, increased placental ABCB1 and ABCG2 mRNA levels (p<0.05) were observed. At the level of immunohistochemistry, while BCRP was increased (p<0.05), P-gp staining intensity was significantly decreased (p<0.05) in PTDC. miR-331-5p, involved in P-gp suppression, was upregulated in PTDC (p<0.01) and correlated to the grade of chorioamnionitis (p<0.01). Conclusions Alterations in the expression of ABC transporters will likely lead to modified transport of clinically relevant compounds at the inflamed placenta. A better understanding of the potential role of these transporters in the events surrounding PTD may also enable new strategies to be developed for prevention and treatment of PTD.
Collapse
Affiliation(s)
- Guinever Eustaquio do Imperio
- Departments of Physiology, Toronto, Ontario, Canada.,Laboratory of Translational Endocrinology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Enrrico Bloise
- Laboratory of Translational Endocrinology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Departments of Morphology, Belo Horizonte, Brazil
| | - Mohsen Javam
- Departments of Physiology, Toronto, Ontario, Canada
| | | | | | - Caroline Dunk
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | | | - Stephen James Lye
- Departments of Physiology, Toronto, Ontario, Canada.,Obstetrics and Gynecology, Toronto, Ontario, Canada.,Medicine, University of Toronto, Toronto, Ontario, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - William Gibb
- Departments of Obstetrics & Gynecology and Department of Cellular & Molecular Medicine, University of Ottawa, Toronto, Ontario, Canada
| | - Tania M Ortiga-Carvalho
- Laboratory of Translational Endocrinology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Stephen Giles Matthews
- Departments of Physiology, Toronto, Ontario, Canada.,Obstetrics and Gynecology, Toronto, Ontario, Canada.,Medicine, University of Toronto, Toronto, Ontario, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| |
Collapse
|
65
|
Choriodecidual leukocytes display a unique gene expression signature in spontaneous labor at term. Genes Immun 2018; 20:56-68. [PMID: 29362510 PMCID: PMC6358585 DOI: 10.1038/s41435-017-0010-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/01/2017] [Accepted: 11/16/2017] [Indexed: 12/11/2022]
Abstract
Prior to and during the process of human labor, maternal circulating leukocytes infiltrate the maternal-fetal interface (choriodecidua) and become activated resembling choriodecidual leukocytes. Since, there is no evidence comparing maternal circulating and choriodecidual leukocytes, herein, we characterized their transcriptome and explored the biological processes enriched in choriodecidual leukocytes. From women undergoing spontaneous term labor we isolated circulating and choriodecidual leukocytes, performed microarray analysis (n = 5) and qRT-PCR validation (n = 9) and interaction network analysis with up-regulated genes. We found 270 genes up-regulated and only 17 genes down-regulated in choriodecidual leukocytes compared to maternal circulating leukocytes. The most up-regulated genes were CCL18, GPNMB, SEPP1, FN1, RNASE1, SPP1, C1QC, and PLTP. The biological processes enriched in choriodecidual leukocytes were cell migration and regulation of immune response, chemotaxis, and humoral immune responses. Our results show striking differences between the transcriptome of choriodecidual and maternal circulating leukocytes. Choriodecidual leukocytes are enriched in immune mediators implicated in the spontaneous process of labor at term.
Collapse
|
66
|
Cappelletti M, Lawson MJ, Chan CC, Wilburn AN, Divanovic S. Differential outcomes of TLR2 engagement in inflammation-induced preterm birth. J Leukoc Biol 2017; 103:535-543. [PMID: 29345344 PMCID: PMC6084304 DOI: 10.1002/jlb.3ma0717-274rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/17/2017] [Accepted: 10/19/2017] [Indexed: 01/06/2023] Open
Abstract
Preterm birth (PTB) is the leading cause of neonatal mortality worldwide. Infection and inflammation are considered main causes of PTB. Among multiple pathogens, Gram‐positive bacteria are commonly linked with induction of PTB. Although activation of innate immune responses, via TLR2 engagement, by Gram‐positive bacteria is a likely cause, whether induction of PTB depends on the potency of specific microbial components to induce Toll‐like receptor (TLR)2‐driven inflammation has not been elucidated. Here, we show that TLR2 activation by synthetic lipopeptides, Pam2Cys, and Pam3Cys specifically, variably influenced inflammation and subsequent induction of PTB. Pam2Cys challenge, compared to Pam3Cys, induced PTB and promoted significantly higher expression of inflammatory cytokines, specifically IL‐6 and IFN‐β, both in vivo and in vitro. Notably, antibody‐mediated neutralization of IL‐6 or genetic deletion of type I IFN receptor (IFNAR) was sufficient to protect from Pam2Cys‐driven PTB and to temper excessive proinflammatory cytokine production. Conversely, IFN‐β or IL‐6 was not sufficient to promote induction of PTB by Pam3Cys. In summary, our data implies a divergent function of TLR2‐activating lipopeptides in the magnitude and type of ligand‐driven inflammatory vigor in induction of PTB.
Collapse
Affiliation(s)
- Monica Cappelletti
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Matthew J Lawson
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Molecular, Cellular and Biochemical Pharmacology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Calvin C Chan
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Adrienne N Wilburn
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Senad Divanovic
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
67
|
Vistnes M, Tapia G, Mårild K, Midttun Ø, Ueland PM, Viken MK, Magnus P, Berg JP, Gillespie KM, Skrivarhaug T, Njølstad PR, Joner G, Størdal K, Stene LC. Plasma immunological markers in pregnancy and cord blood: A possible link between macrophage chemo-attractants and risk of childhood type 1 diabetes. Am J Reprod Immunol 2017; 79. [PMID: 29266506 DOI: 10.1111/aji.12802] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 11/24/2017] [Indexed: 12/18/2022] Open
Abstract
PROBLEM Previous studies have suggested that immune perturbations during pregnancy can affect offspring type 1 diabetes (T1D) risk. We aimed to identify immunological markers that could predict offspring T1D or that were linked to T1D risk factors. METHOD OF STUDY We quantified selected circulating immunological markers in mid-pregnancy (interleukin [IL]-1β, IL-1ra, IL-2Rα, IL-2, -4, -5, -6, -10, -12p70, 13, -17A, GM-CSF, IFN-γ, CXCL10, CCL 2, CCL3, CCL4, TNF) and cord blood plasma (neopterin and kynurenine/tryptophan ratio) in a case-control study with 175 mother/child T1D cases (median age 5.8, range 0.7-13.0 years) and 552 controls. RESULTS Pre-pregnancy obesity was positively associated with CCL4, CXCL10, kynurenine/tryptophan ratio and neopterin (P < .01). The established T1D SNPs rs1159465 (near IL2RA) and rs75352297 (near CCR2 and CCR3) were positively associated with IL-2Rα and CCL4, respectively (P < .01). There was a borderline association of CCL4 and offspring T1D risk, independent of maternal obesity and genotype. When grouping the immunological markers, there was a borderline association (P = .05) with M1 phenotype and no association between M2-, Th1-, Th2- or Th17 phenotypes and offspring T1D risk. CONCLUSION Increased mid-pregnancy CCL4 levels showed borderline associations with increased offspring T1D risk, which may indicate a link between environmental factors in pregnancy and offspring T1D risk.
Collapse
Affiliation(s)
- Maria Vistnes
- Department of Internal Medicine, Diakonhjemmet Hospital, Oslo, Norway.,Institute for Experimental Medical Research, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - German Tapia
- Norwegian Institute of Public Health, Oslo, Norway
| | - Karl Mårild
- Norwegian Institute of Public Health, Oslo, Norway.,Barbara Davis Center, University of Colorado, Aurora, CO, USA
| | | | - Per M Ueland
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Laboratory of Clinical Biochemistry, Haukeland University Hospital, Bergen, Norway
| | - Marte K Viken
- Department of Immunology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Per Magnus
- Norwegian Institute of Public Health, Oslo, Norway
| | - Jens P Berg
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kathleen M Gillespie
- Diabetes and Metabolism, School of Clinical Sciences, Southmead Hospital, University of Bristol, Bristol, UK
| | - Torild Skrivarhaug
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Pål R Njølstad
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Pediatrics and Adolescent Medicine, Haukeland University Hospital, Bergen, Norway
| | - Geir Joner
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Ketil Størdal
- Norwegian Institute of Public Health, Oslo, Norway.,Pediatric Department, Østfold Hospital Trust, Grålum, Norway
| | - Lars C Stene
- Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
68
|
Makinson R, Lloyd K, Rayasam A, McKee S, Brown A, Barila G, Grissom N, George R, Marini M, Fabry Z, Elovitz M, Reyes TM. Intrauterine inflammation induces sex-specific effects on neuroinflammation, white matter, and behavior. Brain Behav Immun 2017; 66:277-288. [PMID: 28739513 PMCID: PMC6916731 DOI: 10.1016/j.bbi.2017.07.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 07/14/2017] [Accepted: 07/20/2017] [Indexed: 02/06/2023] Open
Abstract
Exposure to inflammation during pregnancy has been linked to adverse neurodevelopmental consequences for the offspring. One common route through which a developing fetus is exposed to inflammation is with intrauterine inflammation. To that end, we utilized an animal model of intrauterine inflammation (IUI; intrauterine lipopolysaccharide (LPS) administration, 50µg, E15) to assess placental and fetal brain inflammatory responses, white matter integrity, anxiety-related behaviors (elevated zero maze, light dark box, open field), microglial counts, and the CNS cytokine response to an acute injection of LPS in both males and females. These studies revealed that for multiple endpoints (fetal brain cytokine levels, cytokine response to adult LPS challenge) male IUI offspring were uniquely affected by intrauterine inflammation, while for other endpoints (behavior, microglial number) both sexes were similarly affected. These data advance our understanding of sex-specific effects of early life exposure to inflammation in a translationally- relevant model.
Collapse
Affiliation(s)
- Ryan Makinson
- University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Kelsey Lloyd
- University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Aditya Rayasam
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison
| | - Sarah McKee
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA
| | - Amy Brown
- Maternal and Child Health Research Center, Department of OBGYN, University of Pennsylvania, Philadelphia, PA
| | - Guillermo Barila
- Maternal and Child Health Research Center, Department of OBGYN, University of Pennsylvania, Philadelphia, PA
| | - Nicola Grissom
- University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Robert George
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA
| | - Matt Marini
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison
| | - Michal Elovitz
- Maternal and Child Health Research Center, Department of OBGYN, University of Pennsylvania, Philadelphia, PA
| | - Teresa M. Reyes
- University of Cincinnati, College of Medicine, Cincinnati, OH
| |
Collapse
|
69
|
Orlovic M, Tomic V, Vukojevic K, Hudic I, Mandic V, Azinovic I, Soldo D, Kajic M, Soljic V. Decreased expression of MMP-9 in CD8 + cells in placenta with severe preeclampsia. Biotech Histochem 2017; 92:288-296. [PMID: 28498052 DOI: 10.1080/10520295.2017.1309069] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
We compared the number of CD4-positive (CD4+) and CD8-positive (CD8+) cells in severe and non-severe preeclampsia (PE), and in normal pregnancy. We also evaluated the expression of matrix metalloproteinase 9 (MMP-9) in CD4+ and CD8+ cells. Immunohistochemistry for CD4+ and CD8+ was performed on the decidua basalis of 15 severe and 13 non-severe PE women and compared to decidual tissue of 19 normal pregnancies (control group). Co-expression of MMP-9 with CD8+ and CD4+ cells was determined by double immunofluorescence staining. The median number of CD8+ cells/mm2 was significantly lower for the severe PE group than for the normal pregnancy group, as was the number of CD4+ cells and MMP-9+CD8+ cells. No statistical difference was found between the non-severe PE group and the normal pregnancy group. The significant decrease of CD4+, CD8+ and MMP-9+CD8+ cells at the fetal-maternal interface only in the severe PE group suggests that immunological disorders play a role in the pathophysiology of severe PE.
Collapse
Affiliation(s)
- M Orlovic
- a Department of Gynecology , University Hospital in Mostar
| | - V Tomic
- a Department of Gynecology , University Hospital in Mostar.,b Faculty of Health Studies , University of Mostar
| | - K Vukojevic
- c Laboratory of Morphology, Department of Histology and Embryology , School of Medicine, University of Mostar , Mostar , Bosnia and Herzegovina.,d Laboratory for Early Human Development, Department of Anatomy , Histology and Embryology, School of Medicine, University of Split , Split , Croatia
| | - I Hudic
- e Clinic of Gynecology and Obstetrics, University Clinical Center , Tuzla
| | - V Mandic
- a Department of Gynecology , University Hospital in Mostar
| | - I Azinovic
- c Laboratory of Morphology, Department of Histology and Embryology , School of Medicine, University of Mostar , Mostar , Bosnia and Herzegovina
| | - D Soldo
- a Department of Gynecology , University Hospital in Mostar
| | - M Kajic
- a Department of Gynecology , University Hospital in Mostar
| | - V Soljic
- c Laboratory of Morphology, Department of Histology and Embryology , School of Medicine, University of Mostar , Mostar , Bosnia and Herzegovina.,f Department of Pathology , Cytology and Forensic Medicine, University Hospital in Mostar , Mostar , Bosnia and Herzegovina
| |
Collapse
|
70
|
Gomez-Lopez N, Romero R, Xu Y, Garcia-Flores V, Leng Y, Panaitescu B, Miller D, Abrahams VM, Hassan SS. Inflammasome assembly in the chorioamniotic membranes during spontaneous labor at term. Am J Reprod Immunol 2017; 77:10.1111/aji.12648. [PMID: 28233423 PMCID: PMC5429868 DOI: 10.1111/aji.12648] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 01/17/2017] [Indexed: 12/22/2022] Open
Abstract
PROBLEM Inflammasome activation requires two steps: priming and assembly of the multimeric complex. The second step includes assembly of the sensor molecule and adaptor protein ASC (an apoptosis-associated speck-like protein containing a CARD), which results in ASC speck formation and the recruitment of caspase (CASP)-1. Herein, we investigated whether there is inflammasome assembly in the chorioamniotic membranes and choriodecidual leukocytes from women who underwent spontaneous labor at term. METHOD OF STUDY Using in situ proximity ligation assays, ASC/CASP-1 complexes were determined in the chorioamniotic membranes from women who delivered at term without labor or underwent spontaneous labor at term with or without acute histologic chorioamnionitis (n=10-11 each). Also, ASC speck formation was determined by flow cytometry in the choriodecidual leukocytes isolated from women who delivered at term with or without spontaneous labor (n=9-12 each). RESULTS (i) ASC/CASP-1 complexes were detected in the chorioamniotic membranes; (ii) ASC/CASP-1 complexes were greater in the chorioamniotic membranes from women who underwent spontaneous labor at term than in those without labor; (iii) ASC/CASP-1 complexes were even more abundant in the chorioamniotic membranes from women who underwent spontaneous labor at term with acute histologic chorioamnionitis than in those without this placental lesion; (iv) ASC speck formation was detected in the choriodecidual leukocytes; and (v) ASC speck formation was greater in the choriodecidual leukocytes isolated from women who underwent spontaneous labor at term than in those without labor. CONCLUSION There is inflammasome assembly in the chorioamniotic membranes and choriodecidual leukocytes during spontaneous labor at term.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - 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, NICHD/NIH/DHHS, 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
| | - Yi Xu
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
| | - Valeria Garcia-Flores
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
| | - Yaozhu Leng
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
| | - Bogdan Panaitescu
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
| | - Derek Miller
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
| | - Vikki M. Abrahams
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sonia S. Hassan
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
| |
Collapse
|
71
|
Choudhury RH, Dunk CE, Lye SJ, Aplin JD, Harris LK, Jones RL. Extravillous Trophoblast and Endothelial Cell Crosstalk Mediates Leukocyte Infiltration to the Early Remodeling Decidual Spiral Arteriole Wall. THE JOURNAL OF IMMUNOLOGY 2017; 198:4115-4128. [PMID: 28396316 DOI: 10.4049/jimmunol.1601175] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 03/14/2017] [Indexed: 12/16/2022]
Abstract
Decidual spiral arteriole (SpA) remodeling is essential to ensure optimal uteroplacental blood flow during human pregnancy, yet very little is known about the regulatory mechanisms. Uterine decidual NK (dNK) cells and macrophages infiltrate the SpAs and are proposed to initiate remodeling before colonization by extravillous trophoblasts (EVTs); however, the trigger for their infiltration is unknown. Using human first trimester placenta, decidua, primary dNK cells, and macrophages, we tested the hypothesis that EVTs activate SpA endothelial cells to secrete chemokines that have the potential to recruit maternal immune cells into SpAs. Gene array, real-time PCR, and ELISA analyses showed that treatment of endothelial cells with EVT conditioned medium significantly increased production of two chemokines, CCL14 and CXCL6. CCL14 induced chemotaxis of both dNK cells and decidual macrophages, whereas CXCL6 also induced dNK cell migration. Analysis of the decidua basalis from early pregnancy demonstrated expression of CCL14 and CXCL6 by endothelial cells in remodeling SpAs, and their cognate receptors are present in both dNK cells and macrophages. Neutralization studies identified IL-6 and CXCL8 as factors secreted by EVTs that induce endothelial cell CCL14 and CXCL6 expression. This study has identified intricate crosstalk between EVTs, SpA cells, and decidual immune cells that governs their recruitment to SpAs in the early stages of remodeling and has identified potential key candidate factors involved. This provides a new understanding of the interactions between maternal and fetal cells during early placentation and highlights novel avenues for research to understand defective SpA remodeling and consequent pregnancy pathology.
Collapse
Affiliation(s)
- Ruhul H Choudhury
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9WL, United Kingdom; .,Academic Health Science Centre, St. Mary's Hospital, Manchester M13 9WL, United Kingdom
| | - Caroline E Dunk
- Research Centre for Women's and Infants' Health, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5T 3H7, Canada; and
| | - Stephen J Lye
- Research Centre for Women's and Infants' Health, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5T 3H7, Canada; and
| | - John D Aplin
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9WL, United Kingdom.,Academic Health Science Centre, St. Mary's Hospital, Manchester M13 9WL, United Kingdom
| | - Lynda K Harris
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9WL, United Kingdom.,Academic Health Science Centre, St. Mary's Hospital, Manchester M13 9WL, United Kingdom.,Manchester Pharmacy School, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Rebecca L Jones
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9WL, United Kingdom.,Academic Health Science Centre, St. Mary's Hospital, Manchester M13 9WL, United Kingdom
| |
Collapse
|
72
|
Cappelletti M, Presicce P, Lawson MJ, Chaturvedi V, Stankiewicz TE, Vanoni S, Harley IT, McAlees JW, Giles DA, Moreno-Fernandez ME, Rueda CM, Senthamaraikannan P, Sun X, Karns R, Hoebe K, Janssen EM, Karp CL, Hildeman DA, Hogan SP, Kallapur SG, Chougnet CA, Way SS, Divanovic S. Type I interferons regulate susceptibility to inflammation-induced preterm birth. JCI Insight 2017; 2:e91288. [PMID: 28289719 DOI: 10.1172/jci.insight.91288] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Preterm birth (PTB) is a leading worldwide cause of morbidity and mortality in infants. Maternal inflammation induced by microbial infection is a critical predisposing factor for PTB. However, biological processes associated with competency of pathogens, including viruses, to induce PTB or sensitize for secondary bacterial infection-driven PTB are unknown. We show that pathogen/pathogen-associated molecular pattern-driven activation of type I IFN/IFN receptor (IFNAR) was sufficient to prime for systemic and uterine proinflammatory chemokine and cytokine production and induction of PTB. Similarly, treatment with recombinant type I IFNs recapitulated such effects by exacerbating proinflammatory cytokine production and reducing the dose of secondary inflammatory challenge required for induction of PTB. Inflammatory challenge-driven induction of PTB was eliminated by defects in type I IFN, TLR, or IL-6 responsiveness, whereas the sequence of type I IFN sensing by IFNAR on hematopoietic cells was essential for regulation of proinflammatory cytokine production. Importantly, we also show that type I IFN priming effects are conserved from mice to nonhuman primates and humans, and expression of both type I IFNs and proinflammatory cytokines is upregulated in human PTB. Thus, activation of the type I IFN/IFNAR axis in pregnancy primes for inflammation-driven PTB and provides an actionable biomarker and therapeutic target for mitigating PTB risk.
Collapse
Affiliation(s)
| | - Pietro Presicce
- Division of Neonatology/Pulmonary Biology, Cincinnati Children's Hospital Research Foundation
| | - Matthew J Lawson
- Division of Immunobiology.,Molecular, Cellular and Biochemical Pharmacology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | | - Simone Vanoni
- Division of Allergy and Immunology, Cincinnati Children's Hospital Research Foundation, and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | | - Daniel A Giles
- Division of Immunobiology.,Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | | | | | | - Rebekah Karns
- Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Research Foundation, and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | | | | | | - Simon P Hogan
- Division of Allergy and Immunology, Cincinnati Children's Hospital Research Foundation, and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Suhas G Kallapur
- Division of Neonatology/Pulmonary Biology, Cincinnati Children's Hospital Research Foundation
| | | | | | | |
Collapse
|
73
|
El-Azzamy H, Balogh A, Romero R, Xu Y, LaJeunesse C, Plazyo O, Xu Z, Price TG, Dong Z, Tarca AL, Papp Z, Hassan SS, Chaiworapongsa T, Kim CJ, Gomez-Lopez N, Than NG. Characteristic Changes in Decidual Gene Expression Signature in Spontaneous Term Parturition. J Pathol Transl Med 2017; 51:264-283. [PMID: 28226203 PMCID: PMC5445200 DOI: 10.4132/jptm.2016.12.20] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 12/03/2016] [Accepted: 12/20/2016] [Indexed: 11/29/2022] Open
Abstract
Background The decidua has been implicated in the “terminal pathway” of human term parturition, which is characterized by the activation of pro-inflammatory pathways in gestational tissues. However, the transcriptomic changes in the decidua leading to terminal pathway activation have not been systematically explored. This study aimed to compare the decidual expression of developmental signaling and inflammation-related genes before and after spontaneous term labor in order to reveal their involvement in this process. Methods Chorioamniotic membranes were obtained from normal pregnant women who delivered at term with spontaneous labor (TIL, n = 14) or without labor (TNL, n = 15). Decidual cells were isolated from snap-frozen chorioamniotic membranes with laser microdissection. The expression of 46 genes involved in decidual development, sex steroid and prostaglandin signaling, as well as pro- and anti-inflammatory pathways, was analyzed using high-throughput quantitative real-time polymerase chain reaction (qRT-PCR). Chorioamniotic membrane sections were immunostained and then semi-quantified for five proteins, and immunoassays for three chemokines were performed on maternal plasma samples. Results The genes with the highest expression in the decidua at term gestation included insulin-like growth factor-binding protein 1 (IGFBP1), galectin-1 (LGALS1), and progestogen-associated endometrial protein (PAEP); the expression of estrogen receptor 1 (ESR1), homeobox A11 (HOXA11), interleukin 1β (IL1B), IL8, progesterone receptor membrane component 2 (PGRMC2), and prostaglandin E synthase (PTGES) was higher in TIL than in TNL cases; the expression of chemokine C-C motif ligand 2 (CCL2), CCL5, LGALS1, LGALS3, and PAEP was lower in TIL than in TNL cases; immunostaining confirmed qRT-PCR data for IL-8, CCL2, galectin-1, galectin-3, and PAEP; and no correlations between the decidual gene expression and the maternal plasma protein concentrations of CCL2, CCL5, and IL-8 were found. Conclusions Our data suggests that with the initiation of parturition, the decidual expression of anti-inflammatory mediators decreases, while the expression of pro-inflammatory mediators and steroid receptors increases. This shift may affect downstream signaling pathways that can lead to parturition.
Collapse
Affiliation(s)
- Haidy El-Azzamy
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
| | - Andrea Balogh
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA.,Department of Immunology, Eotvos Lorand University, Budapest, Hungary
| | - Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA.,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Yi Xu
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
| | | | - Olesya Plazyo
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
| | - Zhonghui Xu
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
| | - Theodore G Price
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
| | - Zhong Dong
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA
| | - Adi L Tarca
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University, School of Medicine, Detroit, MI, USA
| | - Zoltan Papp
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
| | - Sonia S Hassan
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University, School of Medicine, Detroit, MI, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University, School of Medicine, Detroit, MI, USA
| | - Chong Jai Kim
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA.,Department of Pathology, Wayne State University, School of Medicine, Detroit, MI, USA.,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University, School of Medicine, Detroit, MI, USA
| | - Nandor Gabor Than
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University, School of Medicine, Detroit, MI, USA.,Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary.,Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary.,First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| |
Collapse
|
74
|
Polettini J, Cobo T, Kacerovsky M, Vinturache AE, Laudanski P, Peelen MJCS, Helmer H, Lamont RF, Takeda J, Lapointe J, Torloni MR, Zhong N, Menon R. Biomarkers of spontaneous preterm birth: a systematic review of studies using multiplex analysis. J Perinat Med 2017; 45:71-84. [PMID: 27514075 DOI: 10.1515/jpm-2016-0097] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/22/2016] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Despite decades of research on risk indicators of spontaneous preterm birth (PTB), reliable biomarkers are still not available to screen or diagnose high-risk pregnancies. Several biomarkers in maternal and fetal compartments have been mechanistically linked to PTB, but none of them are reliable predictors of pregnancy outcome. This systematic review was conducted to synthesize the knowledge on PTB biomarkers identified using multiplex analysis. MATERIALS AND METHODS Three electronic databases (PubMed, EMBASE and Web of Science) were searched for studies in any language reporting the use of multiplex assays for maternal biomarkers associated with PTB published from January 2005 to March 2014. RESULTS Retrieved citations (3631) were screened, and relevant studies (33) were selected for full-text reading. Ten studies were included in the review. Forty-two PTB-related proteins were reported, and RANTES and IL-10 (three studies) followed by MIP-1β, GM-CSF, Eotaxin, and TNF-RI (two studies) were reported more than once in maternal serum. However, results could not be combined due to heterogeneity in type of sample, study population, assay, and analysis methods. CONCLUSION By this systematic review, we conclude that multiplex assays are a potential technological advancement for identifying biomarkers of PTB, although no single or combination of biomarkers could be identified to predict PTB risk.
Collapse
|
75
|
Abstract
BACKGROUND Runs of homozygosity (ROH) are consecutive homozygous genotypes, which may result from population inbreeding or consanguineous marriages. ROH enhance the expression of recessive traits. METHODS We mapped ROH in a case control study of women delivering at term compared with women delivering at or before 34 wk gestation. Gene sets known to be important in risk of preterm birth were examined for their overlap with identified ROH segments. RESULTS While we found no evidence of increased burden of ROH or copy number variations in mothers delivering at or before 34 wk compared with term, we identified 424 genome-wide 50 kb segments with significant difference in abundance of overlapping ROH segments in cases vs. controls, P < 0.05. These regions overlap 199 known genes. We found preterm birth associated genes (CXCR4, MYLK, PAK1) and genes shown to have an evolutionary link to preterm (CXCR4, PPP3CB, C6orf57, DUSP13, and SLC25A45) with significant differences in abundance of overlapping ROH blocks in cases vs. controls, P < 0.001. CONCLUSION We conclude, while we found no significant burden of ROH, we did identify genomic regions with significantly greater abundance of ROH blocks in women delivering preterm that overlapped genes known to be involved in preterm birth.
Collapse
|
76
|
Sivarajasingam SP, Imami N, Johnson MR. Myometrial cytokines and their role in the onset of labour. J Endocrinol 2016; 231:R101-R119. [PMID: 27647860 DOI: 10.1530/joe-16-0157] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 09/19/2016] [Indexed: 12/12/2022]
Abstract
Human labour is an inflammatory event, physiologically driven by an interaction between hormonal and mechanical factors and pathologically associated with infection, bleeding and excessive uterine stretch. The initiation and communicators of inflammation is still not completely understood; however, a key role for cytokines has been implicated. We summarise the current understanding of the nature and role of cytokines, chemokines and hormones and their involvement in signalling within the myometrium particularly during labour.
Collapse
Affiliation(s)
- S P Sivarajasingam
- Department of Surgery and CancerImperial College London, Chelsea and Westminster Hospital, London, UK
| | - N Imami
- Department of MedicineImperial College London, London, UK
| | - M R Johnson
- Department of Surgery and CancerImperial College London, Chelsea and Westminster Hospital, London, UK
| |
Collapse
|
77
|
Bergeron J, Gerges N, Guiraut C, Grbic D, Allard MJ, Fortier LC, Vaillancourt C, Sébire G. Activation of the IL-1β/CXCL1/MMP-10 axis in chorioamnionitis induced by inactivated Group B Streptococcus. Placenta 2016; 47:116-123. [PMID: 27780533 DOI: 10.1016/j.placenta.2016.09.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/07/2016] [Accepted: 09/25/2016] [Indexed: 11/19/2022]
Abstract
Infection or inflammation during pregnancy is known to lead to maternal immune activation triggering a fetal inflammatory response syndrome associated with deleterious effects, such as brain injury and neurodevelopmental disabilities. Group B Streptococcus (GBS) - one of the most common bacterium colonizing pregnant women - can be responsible for chorioamnionitis. Given that interleukin (IL)-1β has a major role in anti-GBS host defense, we hypothesized that IL-1β-driven innate immune response is implicated in GBS-induced chorioamnionitis. Using a rat model of GBS-induced chorioamnionitis, this study showed that inflammatory response to this pathogen was associated with maternal and placental IL-1β hyper expression. Following placental chemokine (C-X-C motif) ligand 1 (CXCL1) production, polymorphonuclear leukocytes (PMN) placental infiltration started at 24 h post-GBS exposure, and MMP-10 was released within these placentas. At 72 h, PMN infiltration extended to membranes and to membranes' arteries. This was associated with IL-1β release within the fetus blood at 72 h. Such a GBS-associated inflammatory cascade might be deleterious for fetal organs. These results pave the way toward targeted placento-protective anti-inflammatory strategies against GBS-induced chorioamnionitis.
Collapse
Affiliation(s)
- Julie Bergeron
- Département de pédiatrie, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Québec, J1H 5N4, Canada.
| | - Noha Gerges
- Departments of Pediatrics and Neurology and Neurosurgery, McGill University, 1001, Décarie boulevard, room EM0.3211, Montreal, Québec, H4A 3J1, Canada.
| | - Clémence Guiraut
- Departments of Pediatrics and Neurology and Neurosurgery, McGill University, 1001, Décarie boulevard, room EM0.3211, Montreal, Québec, H4A 3J1, Canada.
| | - Djordje Grbic
- Département de pédiatrie, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Québec, J1H 5N4, Canada.
| | - Marie-Julie Allard
- Departments of Pediatrics and Neurology and Neurosurgery, McGill University, 1001, Décarie boulevard, room EM0.3211, Montreal, Québec, H4A 3J1, Canada
| | - Louis-Charles Fortier
- Département de Microbiologie, Université de Sherbrooke, 3201 rue Jean Mignault, Sherbrooke, Quebec, J1E 4K8, Canada.
| | - Cathy Vaillancourt
- INRS- Centre Institut Armand Frappier, 531 boulevard des Prairies, Laval, Quebec, H7V 1B7, Canada.
| | - Guillaume Sébire
- Département de pédiatrie, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Québec, J1H 5N4, Canada; Departments of Pediatrics and Neurology and Neurosurgery, McGill University, 1001, Décarie boulevard, room EM0.3211, Montreal, Québec, H4A 3J1, Canada.
| |
Collapse
|
78
|
Inflammatory gene networks in term human decidual cells define a potential signature for cytokine-mediated parturition. Am J Obstet Gynecol 2016; 214:284.e1-284.e47. [PMID: 26348374 DOI: 10.1016/j.ajog.2015.08.075] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 08/17/2015] [Accepted: 08/31/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND Inflammation is a proximate mediator of preterm birth and fetal injury. During inflammation several microRNAs (22 nucleotide noncoding ribonucleic acid (RNA) molecules) are up-regulated in response to cytokines such as interleukin-1β. MicroRNAs, in most cases, fine-tune gene expression, including both up-regulation and down-regulation of their target genes. However, the role of pro- and antiinflammatory microRNAs in this process is poorly understood. OBJECTIVE The principal goal of the work was to examine the inflammatory genomic profile of human decidual cells challenged with a proinflammatory cytokine known to be present in the setting of preterm parturition. We determined the coding (messenger RNA) and noncoding (microRNA) sequences to construct a network of interacting genes during inflammation using an in vitro model of decidual stromal cells. STUDY DESIGN The effects of interleukin-1β exposure on mature microRNA expression were tested in human decidual cell cultures using the multiplexed NanoString platform, whereas the global inflammatory transcriptional response was measured using oligonucleotide microarrays. Differential expression of select transcripts was confirmed by quantitative real time-polymerase chain reaction. Bioinformatics tools were used to infer transcription factor activation and regulatory interactions. RESULTS Interleukin-1β elicited up- and down-regulation of 350 and 78 nonredundant transcripts (false discovery rate < 0.1), respectively, including induction of numerous cytokines, chemokines, and other inflammatory mediators. Whereas this transcriptional response included marked changes in several microRNA gene loci, the pool of fully processed, mature microRNA was comparatively stable following a cytokine challenge. Of a total of 6 mature microRNAs identified as being differentially expressed by NanoString profiling, 2 (miR-146a and miR-155) were validated by quantitative real time-polymerase chain reaction. Using complementary bioinformatics approaches, activation of several inflammatory transcription factors could be inferred downstream of interleukin-1β based on the overall transcriptional response. Further analysis revealed that miR-146a and miR-155 both target genes involved in inflammatory signaling, including Toll-like receptor and mitogen-activated protein kinase pathways. CONCLUSION Stimulation of decidual cells with interleukin-1β alters the expression of microRNAs that function to temper proinflammatory signaling. In this setting, some microRNAs may be involved in tissue-level inflammation during the bulk of gestation and assist in pregnancy maintenance.
Collapse
|
79
|
Seminal Plasma Promotes Lesion Development in a Xenograft Model of Endometriosis. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 185:1409-22. [PMID: 25907757 DOI: 10.1016/j.ajpath.2015.01.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 12/24/2014] [Accepted: 01/20/2015] [Indexed: 12/13/2022]
Abstract
The factors that predispose one-tenth of reproductive-aged women to endometriosis are poorly understood. We determined that genetic deficiency in transforming growth factor β1 impairs endometriosis-like lesion growth in mice. Given that seminal plasma is an abundant source of transforming growth factor β, we evaluated the effect of exposure to seminal plasma on the growth of endometrial lesions. Human endometrial explants were exposed to seminal plasma or to control medium before transfer to Prkdc(scid)-mutant (severe combined immunodeficient) mice. Xenografts exposed to seminal plasma showed an eightfold increase in volume and a 4.3-fold increase in weight after 14 days. These increases were associated with increased proliferation of endometrial epithelial cells and enhanced survival and proliferation of human stromal cells compared with those in control lesions, in which human stromal cell persistence was negligible. Although the distribution of macrophages was altered, their number and activation status did not change in response to seminal plasma. Seminal plasma stimulated the production of a variety of cytokines in endometrial tissue, including growth-regulated oncogene, granulocyte macrophage colony-stimulating factor, and IL-1β. These data suggest that seminal plasma enhances the formation of endometriosis-like lesion via a direct effect on endometrial cell survival and proliferation, rather than via macrophage-mediated mechanisms. These findings raise the possibility that endometrial exposure to seminal plasma could contribute to endometriotic disease progression in women.
Collapse
|
80
|
Karjalainen MK, Ojaniemi M, Haapalainen AM, Mahlman M, Salminen A, Huusko JM, Määttä TA, Kaukola T, Anttonen J, Ulvila J, Haataja R, Teramo K, Kingsmore SF, Palotie A, Muglia LJ, Rämet M, Hallman M. CXCR3 Polymorphism and Expression Associate with Spontaneous Preterm Birth. THE JOURNAL OF IMMUNOLOGY 2015. [PMID: 26209629 DOI: 10.4049/jimmunol.1501174] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Spontaneous preterm birth (SPTB) is a major factor associating with deaths and with lowered quality of life in humans. Environmental and genetic factors influence the susceptibility. Previously, by analyzing families with recurrent SPTB in linkage analysis, we identified a linkage peak close to the gene encoding CXCR3. Present objectives were to investigate the association of CXCR3 with SPTB in Finnish mothers (n = 443) and infants (n = 747), to analyze CXCR3 expression levels in human placenta and levels of its ligands in umbilical cord blood, and to verify the influence of Cxcr3 on SPTB-associating cytokines in mice. We detected an association between an intronic CXCR3 polymorphism, rs2280964, and SPTB in infants from families with recurrent preterm births (p = 0.009 versus term controls, odds ratio 0.52, 95% confidence interval 0.32-0.86). The minor allele was protective and undertransmitted to SPTB infants (p = 0.007). In the placenta and fetal membranes, the rs2280964 major allele homozygotes had higher expression levels than minor allele homozygotes; decidual trophoblasts showed strong CXCR3 immunoreactivity. Expression was higher in SPTB placentas compared with those from elective deliveries. Concentration of a CXCR3 ligand, CXCL9, was increased in cord blood from SPTB, and the protective rs2280964 allele was associated with low CXCL9. In CXCR3-deficient mice (Mus musculus), SPTB-associating cytokines were not acutely increased in amniotic fluid after preterm birth-inducing dose of maternal LPS. Our results indicate that CXCR3 contributes to SPTB. Activation of CXCR3 signaling may disturb the maternal-fetal tolerance, and this may promote labor.
Collapse
Affiliation(s)
- Minna K Karjalainen
- PEDEGO Research Center and Medical Research Center Oulu, University of Oulu, 90014 Oulu, Finland; Department of Children and Adolescents, Oulu University Hospital, 90029 Oulu, Finland;
| | - Marja Ojaniemi
- PEDEGO Research Center and Medical Research Center Oulu, University of Oulu, 90014 Oulu, Finland; Department of Children and Adolescents, Oulu University Hospital, 90029 Oulu, Finland
| | - Antti M Haapalainen
- PEDEGO Research Center and Medical Research Center Oulu, University of Oulu, 90014 Oulu, Finland; Department of Children and Adolescents, Oulu University Hospital, 90029 Oulu, Finland
| | - Mari Mahlman
- PEDEGO Research Center and Medical Research Center Oulu, University of Oulu, 90014 Oulu, Finland; Department of Children and Adolescents, Oulu University Hospital, 90029 Oulu, Finland
| | - Annamari Salminen
- PEDEGO Research Center and Medical Research Center Oulu, University of Oulu, 90014 Oulu, Finland; Department of Children and Adolescents, Oulu University Hospital, 90029 Oulu, Finland
| | - Johanna M Huusko
- PEDEGO Research Center and Medical Research Center Oulu, University of Oulu, 90014 Oulu, Finland; Department of Children and Adolescents, Oulu University Hospital, 90029 Oulu, Finland
| | - Tomi A Määttä
- PEDEGO Research Center and Medical Research Center Oulu, University of Oulu, 90014 Oulu, Finland; Department of Children and Adolescents, Oulu University Hospital, 90029 Oulu, Finland
| | - Tuula Kaukola
- PEDEGO Research Center and Medical Research Center Oulu, University of Oulu, 90014 Oulu, Finland; Department of Children and Adolescents, Oulu University Hospital, 90029 Oulu, Finland
| | - Julia Anttonen
- PEDEGO Research Center and Medical Research Center Oulu, University of Oulu, 90014 Oulu, Finland; Department of Children and Adolescents, Oulu University Hospital, 90029 Oulu, Finland
| | - Johanna Ulvila
- PEDEGO Research Center and Medical Research Center Oulu, University of Oulu, 90014 Oulu, Finland; Department of Children and Adolescents, Oulu University Hospital, 90029 Oulu, Finland
| | - Ritva Haataja
- Biocenter Oulu, University of Oulu, 90014 Oulu, Finland
| | - Kari Teramo
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, Finland
| | | | - Aarno Palotie
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142; Institute for Molecular Medicine Finland, University of Helsinki, 00014 Helsinki, Finland; Psychiatric and Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114; Department of Neurology, Massachusetts General Hospital, Boston, MA 02114
| | - Louis J Muglia
- Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Mika Rämet
- PEDEGO Research Center and Medical Research Center Oulu, University of Oulu, 90014 Oulu, Finland; Department of Children and Adolescents, Oulu University Hospital, 90029 Oulu, Finland; BioMediTech, University of Tampere, 33014 Tampere, Finland; and Department of Pediatrics, Tampere University Hospital, 33521 Tampere, Finland
| | - Mikko Hallman
- PEDEGO Research Center and Medical Research Center Oulu, University of Oulu, 90014 Oulu, Finland; Department of Children and Adolescents, Oulu University Hospital, 90029 Oulu, Finland
| |
Collapse
|
81
|
Rajagopal SP, Hutchinson JL, Dorward DA, Rossi AG, Norman JE. Crosstalk between monocytes and myometrial smooth muscle in culture generates synergistic pro-inflammatory cytokine production and enhances myocyte contraction, with effects opposed by progesterone. Mol Hum Reprod 2015; 21:672-86. [PMID: 26002969 PMCID: PMC4518137 DOI: 10.1093/molehr/gav027] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 05/19/2015] [Indexed: 11/14/2022] Open
Abstract
Both term and preterm parturition are characterized by an influx of macrophages and neutrophils into the myometrium and cervix, with co-incident increased peripheral blood monocyte activation. Infection and inflammation are strongly implicated in the pathology of preterm labour (PTL), with progesterone considered a promising candidate for its prevention or treatment. In this study, we investigated the effect of monocytes on myometrial smooth muscle cell inflammatory cytokine production both alone and in response to LPS, a TLR4 agonist used to trigger PTL in vivo. We also investigated the effect of monocytes on myocyte contraction. Monocytes, isolated from peripheral blood samples from term pregnant women, were cultured alone, or co-cultured with PHM1-41 myometrial smooth muscle cells, for 24 h. In a third set of experiments, PHM1-41 myocytes were cultured for 24 h in isolation. Cytokine secretion was determined by ELISA or multiplex assays. Co-culture of monocytes and myocytes led to synergistic secretion of pro-inflammatory cytokines and chemokines including IL-6, IL-8 and MCP-1, with the secretion being further enhanced by LPS (100 ng/ml). The synergistic secretion of IL-6 and IL-8 from co-cultures was mediated in part by direct cell–cell contact, and by TNF. Conditioned media from co-cultures stimulated contraction of PHM1-41 myocytes, and the effect was inhibited by progesterone. Both progesterone and IL-10 inhibited LPS-stimulated IL-6 and IL-8 secretion from co-cultures, while progesterone also inhibited chemokine secretion. These data suggest that monocytes infiltrating the myometrium at labour participate in crosstalk that potentiates pro-inflammatory cytokine secretion, an effect that is enhanced by LPS, and can augment myocyte contraction. These effects are all partially inhibited by progesterone.
Collapse
Affiliation(s)
- S P Rajagopal
- MRC Centre for Reproductive Health, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - J L Hutchinson
- MRC Centre for Reproductive Health, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - D A Dorward
- MRC Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - A G Rossi
- MRC Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - J E Norman
- MRC Centre for Reproductive Health, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
82
|
Xu Y, Plazyo O, Romero R, Hassan SS, Gomez-Lopez N. Isolation of Leukocytes from the Human Maternal-fetal Interface. J Vis Exp 2015:e52863. [PMID: 26067211 DOI: 10.3791/52863] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pregnancy is characterized by the infiltration of leukocytes in the reproductive tissues and at the maternal-fetal interface (decidua basalis and decidua parietalis). This interface is the anatomical site of contact between maternal and fetal tissues; therefore, it is an immunological site of action during pregnancy. Infiltrating leukocytes at the maternal-fetal interface play a central role in implantation, pregnancy maintenance, and timing of delivery. Therefore, phenotypic and functional characterizations of these leukocytes will provide insight into the mechanisms that lead to pregnancy disorders. Several protocols have been described in order to isolate infiltrating leukocytes from the decidua basalis and decidua parietalis; however, the lack of consistency in the reagents, enzymes, and times of incubation makes it difficult to compare these results. Described herein is a novel approach that combines the use of gentle mechanical and enzymatic dissociation techniques to preserve the viability and integrity of extracellular and intracellular markers in leukocytes isolated from the human tissues at the maternal-fetal interface. Aside from immunophenotyping, cell culture, and cell sorting, the future applications of this protocol are numerous and varied. Following this protocol, the isolated leukocytes can be used to determine DNA methylation, expression of target genes, in vitro leukocyte functionality (i.e., phagocytosis, cytotoxicity, T-cell proliferation, and plasticity, etc.), and the production of reactive oxygen species at the maternal-fetal interface. Additionally, using the described protocol, this laboratory has been able to describe new and rare leukocytes at the maternal-fetal interface.
Collapse
Affiliation(s)
- Yi Xu
- Perinatology Research Branch, NICHD/NIH/DHHS
| | | | - Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS; Department of Obstetrics and Gynecology, University of Michigan; Department of Epidemiology and Biostatistics, Michigan State University; Department of Molecular Obstetrics and Genetics, Wayne State University
| | - Sonia S Hassan
- Perinatology Research Branch, NICHD/NIH/DHHS; Department of Obstetrics and Gynecology, Wayne State University School of Medicine
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, NICHD/NIH/DHHS; Department of Obstetrics and Gynecology, Wayne State University School of Medicine; Department of Immunology and Microbiology, Wayne State University School of Medicine;
| |
Collapse
|
83
|
Yang S, Reid G, Challis JRG, Kim SO, Gloor GB, Bocking AD. Is there a role for probiotics in the prevention of preterm birth? Front Immunol 2015; 6:62. [PMID: 25741339 PMCID: PMC4330906 DOI: 10.3389/fimmu.2015.00062] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 02/01/2015] [Indexed: 01/12/2023] Open
Abstract
Preterm birth (PTB) continues to be a global health challenge. An over-production of inflammatory cytokines and chemokines, as well as an altered maternal vaginal microbiome has been implicated in the pathogenesis of inflammation/infection-associated PTB. Lactobacillus represents the dominant species in the vagina of most healthy pregnant women. The depletion of Lactobacillus in women with bacterial vaginosis (BV) has been associated with an increased risk of PTB. It remains unknown at what point an aberrant vaginal microbiome composition specifically induces the cascade leading to PTB. The ability of oral or vaginal lactobacilli probiotics to reduce BV occurrence and/or dampen inflammation is being considered as a means to prevent PTB. Certain anti-inflammatory properties of lactobacilli suggest potential mechanisms. To date, clinical studies have not been powered with sufficiently high rates of PTB, but overall, there is merit in examining this promising area of clinical science.
Collapse
Affiliation(s)
- Siwen Yang
- Department of Physiology, Obstetrics and Gynecology, University of Toronto , Toronto, ON , Canada ; Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital , Toronto, ON , Canada
| | - Gregor Reid
- Department of Microbiology and Immunology, Western University , London, ON , Canada ; Lawson Health Research Institute , London, ON , Canada
| | - John R G Challis
- Department of Physiology, Obstetrics and Gynecology, University of Toronto , Toronto, ON , Canada ; Department of Obstetrics and Gynecology, The University of Western Australia , Perth, WA , Australia
| | - Sung O Kim
- Department of Microbiology and Immunology, Western University , London, ON , Canada ; Lawson Health Research Institute , London, ON , Canada
| | - Gregory B Gloor
- Department of Biochemistry, Western University , London, ON , Canada
| | - Alan D Bocking
- Department of Physiology, Obstetrics and Gynecology, University of Toronto , Toronto, ON , Canada ; Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital , Toronto, ON , Canada
| |
Collapse
|
84
|
Presicce P, Senthamaraikannan P, Alvarez M, Rueda CM, Cappelletti M, Miller LA, Jobe AH, Chougnet CA, Kallapur SG. Neutrophil recruitment and activation in decidua with intra-amniotic IL-1beta in the preterm rhesus macaque. Biol Reprod 2015; 92:56. [PMID: 25537373 PMCID: PMC4342792 DOI: 10.1095/biolreprod.114.124420] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 09/26/2014] [Accepted: 12/22/2014] [Indexed: 11/01/2022] Open
Abstract
Chorioamnionitis, an infection/inflammation of the fetomaternal membranes, is frequently associated with preterm delivery. The mechanisms of inflammation in chorioamnionitis are poorly understood. We hypothesized that neutrophils recruited to the decidua would be the major producers of proinflammatory cytokines. We injected intra-amniotic (IA) interleukin 1beta (IL-1beta) at ∼80% gestation in rhesus macaque monkeys, Macaca mulatta, delivered the fetuses surgically 24 h or 72 h after IA injections, and investigated the role of immune cells in the chorion-amnion decidua. IA IL-1beta induced a robust infiltration of neutrophils and significant increases of proinflammatory cytokines in the chorioamnion decidua at 24 h after exposure, with a subsequent decrease at 72 h. Neutrophils in the decidua were the major source of tumor necrosis factor alpha (TNFalpha) and IL-8. Interestingly, IA IL-1beta also induced a significant increase in anti-inflammatory indoleamine 2,3-dioxygenase (IDO) expression in the decidua neutrophils. The frequency of regulatory T cells (Tregs) and FOXP3 mRNA expression in the decidua did not change after IA IL-1beta injection. Collectively, our data demonstrate that in this model of sterile chorioamnionitis, the decidua neutrophils cause the inflammation in the gestational tissues but may also act as regulators to dampen the inflammation. These results help to understand the contribution of neutrophils to the pathogenesis of chorioamnionitis-induced preterm labor.
Collapse
Affiliation(s)
- Pietro Presicce
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | | | - Manuel Alvarez
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Cesar M Rueda
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Monica Cappelletti
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Lisa A Miller
- California National Primate Research Center, University of California Davis, Davis, California Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California
| | - Alan H Jobe
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Claire A Chougnet
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Suhas G Kallapur
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
85
|
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: 15.0] [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
|
86
|
Gomez-Lopez N, Tong WC, Arenas-Hernandez M, Tanaka S, Hajar O, Olson DM, Taggart MJ, Mitchell BF. Chemotactic activity of gestational tissues through late pregnancy, term labor, and RU486-induced preterm labor in Guinea pigs. Am J Reprod Immunol 2014; 73:341-52. [PMID: 25329235 DOI: 10.1111/aji.12333] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 09/23/2014] [Indexed: 01/22/2023] Open
Abstract
PROBLEM Is increased leukocyte chemotactic activity (CA) from gestational tissues necessary for term or preterm labor in guinea pigs? METHOD OF STUDY Tissue extracts were prepared from pregnant guinea pig decidua-myometrium, cervix, fetal membranes (amniochorion), and placenta during early third trimester (n = 8), term not in labor (TNL, n = 5), and term spontaneous labor (TL, n = 6), RU486-induced preterm labor (PTL, n = 6), or controls (cPTL, n = 5). Leukocyte CA was assessed using a modified Boyden chamber assay. Extract chemokine and maternal progesterone concentrations were quantified by enzyme immunoassay. RESULTS Only the extracts from amniochorion demonstrated increased CA through late gestation and labor. In contrast, CA was decreased in extracts from amniochorion and cervix from animals after RU486-induced PTL. Maternal progesterone concentrations remained high in all groups. CONCLUSION Leukocyte CA of intrauterine tissues is increased in term spontaneous labor. However, RU486-induced preterm labor occurs in the absence of increased CA.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Department of Obstetrics and Gynecology, Perinatology Research Branch, Wayne State University, Detroit, MI, USA
| | | | | | | | | | | | | | | |
Collapse
|
87
|
Stephen GL, Lui S, Hamilton SA, Tower CL, Harris LK, Stevens A, Jones RL. Transcriptomic profiling of human choriodecidua during term labor: inflammation as a key driver of labor. Am J Reprod Immunol 2014; 73:36-55. [PMID: 25283845 DOI: 10.1111/aji.12328] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 09/05/2014] [Indexed: 01/10/2023] Open
Abstract
PROBLEM Inflammation is a driver of labor in myometrium and cervix; however, the involvement of decidua is poorly defined. We have reported decidual leukocyte infiltration prior to and during labor; the regulators of these inflammatory processes are unknown. METHOD OF STUDY Choriodecidua RNA obtained after term labor or elective cesarean delivery was applied to Affymetrix GeneChips. Pathway analysis and gene validation were performed. RESULTS Extensive inflammatory activation was identified in choriodecidua following labor, predominantly upregulation of genes regulating leukocyte trafficking and cytokine signalling. Genes governing cell fate, tissue remodelling, and translation were also altered. Upregulation of candidate genes (ICAM1, CXCR4, CD44, TLR4, SOCS3, BCL2A, and IDO) was confirmed. NFκB, STAT1&3, HMGB1, and miRNA-21, miRNA-46, miRNA-141, and miRNA-200 were predicted upstream regulators. CONCLUSION This study confirms inflammatory processes are major players in labor events in choriodecidua, as in other gestational tissues. Suppressing uterine inflammation is likely to be critical for arresting premature labor.
Collapse
Affiliation(s)
- Gillian L Stephen
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK; St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | | | | | | | | | | | | |
Collapse
|
88
|
Enhanced natural killer-cell and T-cell responses to influenza A virus during pregnancy. Proc Natl Acad Sci U S A 2014; 111:14506-11. [PMID: 25246558 DOI: 10.1073/pnas.1416569111] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Pregnant women experience increased morbidity and mortality after influenza infection, for reasons that are not understood. Although some data suggest that natural killer (NK)- and T-cell responses are suppressed during pregnancy, influenza-specific responses have not been previously evaluated. Thus, we analyzed the responses of women that were pregnant (n = 21) versus those that were not (n = 29) immediately before inactivated influenza vaccination (IIV), 7 d after vaccination, and 6 wk postpartum. Expression of CD107a (a marker of cytolysis) and production of IFN-γ and macrophage inflammatory protein (MIP) 1β were assessed by flow cytometry. Pregnant women had a significantly increased percentage of NK cells producing a MIP-1β response to pH1N1 virus compared with nonpregnant women pre-IIV [median, 6.66 vs. 0.90% (P = 0.0149)] and 7 d post-IIV [median, 11.23 vs. 2.81% (P = 0.004)], indicating a heightened chemokine response in pregnant women that was further enhanced by the vaccination. Pregnant women also exhibited significantly increased T-cell production of MIP-1β and polyfunctionality in NK and T cells to pH1N1 virus pre- and post-IIV. NK- and T-cell polyfunctionality was also enhanced in pregnant women in response to the H3N2 viral strain. In contrast, pregnant women had significantly reduced NK- and T-cell responses to phorbol 12-myristate 13-acetate and ionomycin. This type of stimulation led to the conclusion that NK- and T-cell responses during pregnancy are suppressed, but clearly this conclusion is not correct relative to the more biologically relevant assays described here. Robust cellular immune responses to influenza during pregnancy could drive pulmonary inflammation, explaining increased morbidity and mortality.
Collapse
|
89
|
Du MR, Wang SC, Li DJ. The integrative roles of chemokines at the maternal-fetal interface in early pregnancy. Cell Mol Immunol 2014; 11:438-48. [PMID: 25109684 DOI: 10.1038/cmi.2014.68] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 06/29/2014] [Accepted: 07/01/2014] [Indexed: 12/14/2022] Open
Abstract
Embryos express paternal antigens that are foreign to the mother, but the mother provides a special immune milieu at the fetal-maternal interface to permit rather than reject the embryo growth in the uterus until parturition by establishing precise crosstalk between the mother and the fetus. There are unanswered questions in the maintenance of pregnancy, including the poorly understood phenomenon of maternal tolerance to the allogeneic conceptus, and the remarkable biological roles of placental trophoblasts that invade the uterine wall. Chemokines are multifunctional molecules initially described as having a role in leukocyte trafficking and later found to participate in developmental processes such as differentiation and directed migration. It is increasingly evident that the gestational uterine microenvironment is characterized, at least in part, by the differential expression and secretion of chemokines that induce selective trafficking of leukocyte subsets to the maternal-fetal interface and regulate multiple events that are closely associated with normal pregnancy. Here, we review the expression and function of chemokines and their receptors at the maternal-fetal interface, with a special focus on chemokine as a key component in trophoblast invasiveness and placental angiogenesis, recruitment and instruction of immune cells so as to form a fetus-supporting milieu during pregnancy. The chemokine network is also involved in pregnancy complications.
Collapse
|
90
|
Tower CL, Lui S, Jones RL. Are chemokines therapeutic targets for the prevention of preterm labor? Immunotherapy 2014; 6:901-4. [DOI: 10.2217/imt.14.70] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Clare L Tower
- Maternal &Fetal Health Research Centre, Faculty of Medical & Human Sciences, Institute of Human Development, University of Manchester, Manchester, M13 9WL, UK
- Manchester Academic Health Science Centre, St Mary's Hospital, Central Manchester University Hospital NHS Foundation Trust, Manchester, M13 9WL, UK
| | - Sylvia Lui
- Maternal &Fetal Health Research Centre, Faculty of Medical & Human Sciences, Institute of Human Development, University of Manchester, Manchester, M13 9WL, UK
- Manchester Academic Health Science Centre, St Mary's Hospital, Central Manchester University Hospital NHS Foundation Trust, Manchester, M13 9WL, UK
| | - Rebecca L Jones
- Maternal &Fetal Health Research Centre, Faculty of Medical & Human Sciences, Institute of Human Development, University of Manchester, Manchester, M13 9WL, UK
- Manchester Academic Health Science Centre, St Mary's Hospital, Central Manchester University Hospital NHS Foundation Trust, Manchester, M13 9WL, UK
| |
Collapse
|
91
|
Yang S, Li W, Challis JRG, Reid G, Kim SO, Bocking AD. Probiotic Lactobacillus rhamnosus GR-1 supernatant prevents lipopolysaccharide-induced preterm birth and reduces inflammation in pregnant CD-1 mice. Am J Obstet Gynecol 2014; 211:44.e1-44.e12. [PMID: 24486224 DOI: 10.1016/j.ajog.2014.01.029] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/20/2013] [Accepted: 01/21/2014] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The objective of this study was to determine the effect of probiotic Lactobacillus rhamnosus GR-1 supernatant (GR-1 SN) on lipopolysaccharide-induced preterm birth (PTB) and outputs of cytokines, chemokines, and progesterone in pregnant CD-1 mice. STUDY DESIGN We compared PTB rates after intrauterine injection of lipopolysaccharide with and without previous GR-1 SN treatment. Cytokines and chemokines in the maternal plasma, myometrium, placenta, and amniotic fluid were examined with multiplex assay, and circulating maternal progesterone was measured with enzyme-linked immunoassay. Statistical significance was assessed with 2-tailed 1-way analysis of variance or analysis of variance on ranks. Fetal sex ratios in mice that delivered preterm were compared with those that delivered at term after lipopolysaccharide and GR-1 SN treatments. RESULTS GR-1 SN reduced lipopolysaccharide-induced PTB by 43%. GR-1 SN significantly decreased the lipopolysaccharide-induced production of interleukin (IL)-1β, -6, and -12p40, tumor necrosis factor-α, CCL4, and CCL5 in maternal plasma; IL-6, -12p70, -17, and -13 and tumor necrosis factor-α in myometrium; IL-6, -12p70, and -17 in placenta; and IL-6, tumor necrosis factor-α, CCL3, and CCL4 in amniotic fluid. Maternal plasma progesterone was reduced significantly after lipopolysaccharide injection with and without GR-1 SN pretreatment. There was no difference in fetal sex ratios between mice that delivered preterm and those that did not after lipopolysaccharide and GR-1 SN treatments. CONCLUSION The supernatant of probiotic L rhamnosus GR-1 attenuated lipopolysaccharide-induced inflammation and PTB in vivo. GR-1 SN may confer therapeutic benefits in the prevention of infection-associated PTB by controlling systemic and intrauterine inflammation.
Collapse
Affiliation(s)
- Siwen Yang
- Departments of Physiology and Obstetrics and Gynecology, University of Toronto Faculty of Medicine, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Wei Li
- Departments of Physiology and Obstetrics and Gynecology, University of Toronto Faculty of Medicine, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - John R G Challis
- Departments of Physiology and Obstetrics and Gynecology, University of Toronto Faculty of Medicine, Toronto, ON, Canada; Faculty of Health Sciences, Simon Fraser University, Vancouver, BC, Canada
| | - Gregor Reid
- Department of Microbiology and Immunology, Western University Faculty of Medicine, London, ON, Canada; Lawson Health Research Institute, London, ON, Canada
| | - Sung O Kim
- Department of Microbiology and Immunology, Western University Faculty of Medicine, London, ON, Canada
| | - Alan D Bocking
- Departments of Physiology and Obstetrics and Gynecology, University of Toronto Faculty of Medicine, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.
| |
Collapse
|
92
|
Faust K, Göpel W, Moser K, Temole G, Bartels M, Wieg C, Tröger B, Herting E, Härtel C. Differential expression of antimicrobial polypeptides in cord blood samples of preterm and term infants. Acta Paediatr 2014; 103:e143-7. [PMID: 24387008 DOI: 10.1111/apa.12544] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 12/09/2013] [Accepted: 12/16/2013] [Indexed: 10/25/2022]
Abstract
AIM To determine levels of antimicrobial polypeptides (AMP) in cord blood of term and preterm neonates and to investigate influencing factors. METHODS In a single-centre study, n = 139 preterm infants and n = 36 term infants were included. AMP levels were analysed in supernatants of whole cord blood cultures with a standardised concentration of 5 × 10(6) white blood cells/mL via enzyme-linked immunosorbent assay (ELISA). RESULTS Lactoferrin, human neutrophil peptides (HNP) 1-3 and bacterial permeability-increasing protein (BPI) expression in cord blood of preterm infants were influenced by the cause of preterm delivery, that is increased levels in infants with clinical amniotic infection. AMP levels also weakly correlated with white blood cell and neutrophil count at birth. In the whole cohort, no association between gestational age or birthweight with AMP levels was found. In the subgroup of infants without clinical amniotic infection (n = 77 preterm infants, n = 36 healthy term infants), we noted a weak correlation between gestational age and lactoferrin, calprotectin and HNP1-3 levels. In addition to that, we observed higher levels of lactoferrin and HNP1-3 in large-for-gestational-age infants. CONCLUSION Our study confirms that several factors influence cord blood AMP levels which underlines the difficulties of using AMP levels as biomarkers of immunological response.
Collapse
Affiliation(s)
- Kirstin Faust
- Department of Paediatrics; University of Lübeck; Lübeck Germany
| | - Wolfgang Göpel
- Department of Paediatrics; University of Lübeck; Lübeck Germany
| | | | | | - Maren Bartels
- Department of Paediatrics; University of Lübeck; Lübeck Germany
| | | | - Birte Tröger
- Department of Paediatrics; University of Lübeck; Lübeck Germany
| | - Egbert Herting
- Department of Paediatrics; University of Lübeck; Lübeck Germany
| | | |
Collapse
|
93
|
Rinaldi SF, Catalano RD, Wade J, Rossi AG, Norman JE. Decidual neutrophil infiltration is not required for preterm birth in a mouse model of infection-induced preterm labor. THE JOURNAL OF IMMUNOLOGY 2014; 192:2315-25. [PMID: 24501200 PMCID: PMC3932811 DOI: 10.4049/jimmunol.1302891] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Parturition is associated with a leukocyte influx into the intrauterine tissues; however, the exact role these leukocytes play in the onset of labor remains unclear. Neutrophil infiltration of the uteroplacental tissues has been particularly associated with infection-associated preterm labor (PTL) in both women and mouse models. In this study, we investigated the role of neutrophils in a mouse model of infection-induced PTL. Intrauterine administration of LPS on day 17 of gestation resulted in a 7-fold increase in the number of decidual neutrophils compared with control mice receiving PBS (p < 0.01; n = 8–11). We hypothesized that neutrophil influx is necessary for PTL and that neutrophil depletion would abolish preterm birth. To test this hypothesis, mice were depleted of neutrophils by treatment with anti–Gr-1, anti–Ly-6G, or the appropriate IgG control Ab on day 16 of gestation prior to LPS on day 17 (n = 6–7). Successful neutrophil depletion was confirmed by flow cytometry and immunohistochemistry. Neutrophil depletion with Gr-1 resulted in reduced uterine and placental Il-1β expression (p < 0.05). Neutrophil depletion with Ly-6G reduced uterine Il-1β and Tnf-α expression (p < 0.05). However, neutrophil depletion with either Ab did not delay LPS-induced preterm birth. Collectively, these data show that decidual neutrophil infiltration is not essential for the induction of infection-induced PTL in the mouse, but that neutrophils contribute to the LPS-induced inflammatory response of the uteroplacental tissues.
Collapse
Affiliation(s)
- Sara F Rinaldi
- Medical Research Council Centre for Reproductive Health, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | | | | | | | | |
Collapse
|
94
|
Bardou M, Hadi T, Mace G, Pesant M, Debermont J, Barrichon M, Wendremaire M, Laurent N, Sagot P, Lirussi F. Systemic increase in human maternal circulating CD14+CD16- MCP-1+ monocytes as a marker of labor. Am J Obstet Gynecol 2014; 210:70.e1-9. [PMID: 23994222 DOI: 10.1016/j.ajog.2013.08.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/27/2013] [Accepted: 08/27/2013] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To study the influence of pregnancy and labor on the proportion and level of activation of monocyte subpopulations in human pregnancy. STUDY DESIGN Peripheral blood samples were obtained from healthy nonpregnant women (n = 6); women in the third-trimester of healthy pregnancies (n = 18) and women with preterm premature rupture of membranes (n = 46), just before delivery for the last 2 groups. Monocyte subpopulations were characterized by flow cytometry using CD14, CD16, and activation level using macrophage chemoattractant protein-1 (MCP-1) and CCR2 antibodies. RESULTS The relative proportion of each monocyte subset in nonpregnant women was similar to that in women with healthy or complicated pregnancies. However, pregnancy was associated with a significant decrease in MCP-1 expressing monocytes (79.5% ± 19.8% vs 9.3% ± 6.8% and 11.9% ± 8.3% for nonpregnant, healthy pregnancy, and preterm premature rupture of membranes (respectively, P < .05). Spontaneous labor was associated with a return to nonpregnant values for the proportion of MCP-1 expressing monocytes in both normal (74.4% ± 16.9) and preterm premature rupture of membranes pregnancy (68.4% ± 35.6), irrespective of the mode of delivery (vaginal or cesarean section). This was not observed in women who delivered without spontaneous labor onset. CCR-2 (MCP-1 receptor) expression was not modified in monocytes at the time of labor, but was significantly increased in granulocytes (3646 ± 1080 vs 7338 ± 2718 for nonlaboring and laboring preterm premature rupture of membranes, respectively, P < .05) CONCLUSION: In light of previous reports of a role for MCP-1 in labor, our results suggest the downregulation of activation levels of monocytes, via MCP-1 expression might be involved in maternofetal immune tolerance. Monocyte reactivation might be associated with labor.
Collapse
Affiliation(s)
- Marc Bardou
- Centre d'Investigations Cliniques Plurithématique 803 (INSERM CIC-P 803), Dijon, France; Center for Lipid Research, INSERM (Institut National de la Santé et de la Recherche Médicale), Dijon, France; CHU de Dijon, Dijon, France
| | - Tarik Hadi
- Centre d'Investigations Cliniques Plurithématique 803 (INSERM CIC-P 803), Dijon, France; Center for Lipid Research, INSERM (Institut National de la Santé et de la Recherche Médicale), Dijon, France; CHU de Dijon, Dijon, France
| | | | - Matthieu Pesant
- Humanitas Clinical and Research Center, Leukocyte Biology Laboratory, University of Milan, Milan, Italy
| | - Julie Debermont
- Centre d'Investigations Cliniques Plurithématique 803 (INSERM CIC-P 803), Dijon, France
| | - Marina Barrichon
- Center for Lipid Research, INSERM (Institut National de la Santé et de la Recherche Médicale), Dijon, France; CHU de Dijon, Dijon, France
| | - Maeva Wendremaire
- Center for Lipid Research, INSERM (Institut National de la Santé et de la Recherche Médicale), Dijon, France; CHU de Dijon, Dijon, France; Laboratoire de Pharmacologie-Toxicologie, Dijon, France
| | | | - Paul Sagot
- CHU de Dijon, Dijon, France; Service de Gynécologie et d'Obstétrique, Dijon, France
| | - Frédéric Lirussi
- Center for Lipid Research, INSERM (Institut National de la Santé et de la Recherche Médicale), Dijon, France; CHU de Dijon, Dijon, France; Laboratoire de Pharmacologie-Toxicologie, Dijon, France
| |
Collapse
|
95
|
Goddard LM, Ton AN, Org T, Mikkola HKA, Iruela-Arispe ML. Selective suppression of endothelial cytokine production by progesterone receptor. Vascul Pharmacol 2013; 59:36-43. [PMID: 23747964 DOI: 10.1016/j.vph.2013.06.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 06/01/2013] [Indexed: 01/01/2023]
Abstract
Steroid hormones are well-recognized suppressors of the inflammatory response, however, their cell- and tissue-specific effects in the regulation of inflammation are far less understood, particularly for the sex-related steroids. To determine the contribution of progesterone in the endothelium, we have characterized and validated an in vitro culture system in which human umbilical vein endothelial cells constitutively express human progesterone receptor (PR). Using next generation RNA-sequencing, we identified a selective group of cytokines that are suppressed by progesterone both under physiological conditions and during pathological activation by lipopolysaccharide. In particular, IL-6, IL-8, CXCL2/3, and CXCL1 were found to be direct targets of PR, as determined by ChIP-sequencing. Regulation of these cytokines by progesterone was also confirmed by bead-based multiplex cytokine assays and quantitative PCR. These findings provide a novel role for PR in the direct regulation of cytokine levels secreted by the endothelium. They also suggest that progesterone-PR signaling in the endothelium directly impacts leukocyte trafficking in PR-expressing tissues.
Collapse
Affiliation(s)
- Lauren M Goddard
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | |
Collapse
|