1
|
Bacon SJ, Zhu Y, Ghosh P. Early spiral arteriole remodeling in the uterine-placental interface: A rat model. J Anat 2024; 244:1054-1066. [PMID: 38288680 PMCID: PMC11095304 DOI: 10.1111/joa.14019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 05/16/2024] Open
Abstract
The mammalian placenta's interface with the parent is a richly vascularized tissue whose development relies upon communication between many different cell types within the uterine microenvironment. The uterine blood vessels of the interface are reshaped during pregnancy into wide-bore, flaccid vessels that convey parental blood to the exchange region of the placenta. Invasive trophoblast as well as parental uterine macrophages and Natural Killer cells are involved in the stepwise remodeling of these vessels and their respective contributions to this crucial process are still being delineated. However, the earliest steps in arteriole remodeling are understudied as they are difficult to study in humans, and other species lack the deep trophoblast invasion that is so prominent a feature of placentation in humans. Here, we further characterize the rat, with deep hemochorial placentation akin to humans, as a model system in which to tease apart the earliest, relatively understudied events in spiral arteriole remodeling. We show that the rat uterine-placental interface increases in size and vascularity rapidly, before trophoblast invasion. The remodeling stages in the arterioles of the rat uterine-placental interface follow a sequence of anatomical changes similar to those in humans, and there are changes to the arterioles' muscular tunica media prior to the marked influx of immune cells. The rat is a tractable model in which to better understand the cell/cell interactions occurring in vivo in an intact tissue microenvironment over time.
Collapse
Affiliation(s)
- Sarah J Bacon
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts, USA
| | - Yuxi Zhu
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts, USA
| | - Priyanjali Ghosh
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts, USA
| |
Collapse
|
2
|
Li Q, Sharkey A, Sheridan M, Magistrati E, Arutyunyan A, Huhn O, Sancho-Serra C, Anderson H, McGovern N, Esposito L, Fernando R, Gardner L, Vento-Tormo R, Turco MY, Moffett A. Human uterine natural killer cells regulate differentiation of extravillous trophoblast early in pregnancy. Cell Stem Cell 2024; 31:181-195.e9. [PMID: 38237587 DOI: 10.1016/j.stem.2023.12.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 10/19/2023] [Accepted: 12/20/2023] [Indexed: 02/04/2024]
Abstract
In humans, balanced invasion of trophoblast cells into the uterine mucosa, the decidua, is critical for successful pregnancy. Evidence suggests that this process is regulated by uterine natural killer (uNK) cells, but how they influence reproductive outcomes is unclear. Here, we used our trophoblast organoids and primary tissue samples to determine how uNK cells affect placentation. By locating potential interaction axes between trophoblast and uNK cells using single-cell transcriptomics and in vitro modeling of these interactions in organoids, we identify a uNK cell-derived cytokine signal that promotes trophoblast differentiation at the late stage of the invasive pathway. Moreover, it affects transcriptional programs involved in regulating blood flow, nutrients, and inflammatory and adaptive immune responses, as well as gene signatures associated with disorders of pregnancy such as pre-eclampsia. Our findings suggest mechanisms on how optimal immunological interactions between uNK cells and trophoblast enhance reproductive success.
Collapse
Affiliation(s)
- Qian Li
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK.
| | - Andrew Sharkey
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Megan Sheridan
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Elisa Magistrati
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Anna Arutyunyan
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Oisin Huhn
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Carmen Sancho-Serra
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Holly Anderson
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Naomi McGovern
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Laura Esposito
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Ridma Fernando
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Lucy Gardner
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Roser Vento-Tormo
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome Sanger Institute, Cambridge CB10 1SA, UK.
| | | | - Ashley Moffett
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK.
| |
Collapse
|
3
|
Furukawa S, Tsuji N, Hayashi S, Kuroda Y, Kimura M, Kojima C, Takeuchi K. Effects of cyclophosphamide on rat placental development. J Toxicol Pathol 2023; 36:159-169. [PMID: 37577367 PMCID: PMC10412958 DOI: 10.1293/tox.2022-0144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 02/16/2023] [Indexed: 08/15/2023] Open
Abstract
We examined the morphological effects of cyclophosphamide (CPA) on placental development in pregnant rats. CPA was administered as a single dose to pregnant rats intraperitoneally at 0 mg/kg (the control group), 25 mg/kg on gestation day (GD) 12 (the CPA GD 12-treated group), and 25 mg/kg on GD 14 (the CPA GD 14-treated group). The fetal and placental weight decreased in the CPA-treated groups, complete fetal resorption from GD 17 onwards in the CPA GD 12-treated group, and external malformations in the CPA GD 14-treated group. Histopathologically, CPA induced apoptosis and/or cell proliferation inhibition in each part of the placenta. In the labyrinth zone, syncytiotrophoblasts were selectively reduced, resulting in a small placenta. In the basal zone, the number of spongiotrophoblasts was reduced, resulting in hypoplasia of glycogen cell islands. In addition, a small number of interstitial trophoblasts invaded the metrial gland from the basal zone on GD 15. The severity of these lesions was higher in the CPA GD 12-treated group than in the CPA GD 14-treated group. In the metrial gland, although the number of uterine natural killer cells was reduced, metrial gland development was not affected.
Collapse
Affiliation(s)
- Satoshi Furukawa
- Planning and Development Department, Nissan Chemical
Corporation, 2-5-1 Nihonbashi, Chuo-ku, Tokyo 103-6119, Japan
| | - Naho Tsuji
- Planning and Development, Agricultural Chemical Division,
Nissan Chemical Corporation, 2-5-1 Nihonbashi, Chuo-ku, Tokyo 103-6119, Japan
| | - Seigo Hayashi
- Biological Research Laboratories, Nissan Chemical
Corporation, 1470 Shiraoka, Shiraoka-shi, Saitama 349-0294, Japan
| | - Yusuke Kuroda
- Biological Research Laboratories, Nissan Chemical
Corporation, 1470 Shiraoka, Shiraoka-shi, Saitama 349-0294, Japan
| | - Masayuki Kimura
- Biological Research Laboratories, Nissan Chemical
Corporation, 1470 Shiraoka, Shiraoka-shi, Saitama 349-0294, Japan
| | - Chisato Kojima
- Biological Research Laboratories, Nissan Chemical
Corporation, 1470 Shiraoka, Shiraoka-shi, Saitama 349-0294, Japan
| | - Kazuya Takeuchi
- Biological Research Laboratories, Nissan Chemical
Corporation, 1470 Shiraoka, Shiraoka-shi, Saitama 349-0294, Japan
| |
Collapse
|
4
|
Cheng Y, Wang H, Shang J, Wang J, Yin J, Zhang J, Guo X, Wang S, Duan YG, Lee CL, Chiu PCN, Zhang J, Yeung WSB, Cao D, Yao Y. Transcriptomic analysis of mid-secretory endometrium reveals essential immune factors associated with pregnancy after single euploid blastocyst transfer. Am J Reprod Immunol 2023; 89:e13672. [PMID: 36542433 DOI: 10.1111/aji.13672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/30/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Implantation is a limiting factor for treatment success in assisted reproduction. Both embryonic and endometrial factors contribute to implantation. Embryonic factors have often been ignored in previous studies about the role of endometrium in implantation. In this study, we sought to identify the endometrial genes associated with negative pregnancy outcomes following the transfer of a single euploid blastocyst. METHODS Computational analyses of the transcriptomes of mid-secretory endometria from nine pregnant and seven non-pregnant patients in a cycle preceding the transfer of a single euploid blastocyst in a vitrified-warmed cycle were performed. RESULTS Principal component analysis of two reported endometrial receptivity gene sets showed close clustering of the pregnant and non-pregnant samples. Differential gene expression analysis and co-expression module analysis identified 131 genes associated with the pregnancy status. The endometrial signatures identified highlight the importance of immune and metabolic regulation in pregnancy outcome. Network analysis identified 20 hub genes that could predict pregnancy outcomes with 88.9% sensitivity and 85.7% specificity. Single-cell gene expression analysis highlighted the regulation of endometrial natural killer (NK) cells, T cells, and macrophages during embryo implantation. Immune cell abundance analysis supported the dysregulation of cytotoxic immune cells in the endometria of non-pregnant women. CONCLUSIONS We reported the first endometrial gene signature associated with pregnancy after elimination of embryo aneuploidy and highlighted the importance of the endometrial immune microenvironment and metabolic status in pregnancy outcomes.
Collapse
Affiliation(s)
- Yanfei Cheng
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.,Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hui Wang
- Department of Obstetrics and Gynecology, the First Medical Center of PLA General Hospital, Beijing, China
| | - Jin Shang
- Medical School of Chinese PLA, Beijing, China
| | - Jue Wang
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jingwen Yin
- Department of Obstetrics and Gynecology, Third Hospital, Peking University, Beijing, China
| | | | - Xinmeng Guo
- College of Medicine, Nankai University, Tianjin, China
| | - Sidong Wang
- Medical School of Chinese PLA, Beijing, China
| | - Yong-Gang Duan
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Cheuk-Lun Lee
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.,Department of Obstetrics and Gynaecology, The University of Hong Kong, Hong Kong S.A.R., China
| | - Philip C N Chiu
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.,Department of Obstetrics and Gynaecology, The University of Hong Kong, Hong Kong S.A.R., China
| | - Jian Zhang
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Shenzhen Key Laboratory of Metabolic Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - William S B Yeung
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.,Department of Obstetrics and Gynaecology, The University of Hong Kong, Hong Kong S.A.R., China
| | - Dandan Cao
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yuanqing Yao
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.,Department of Obstetrics and Gynecology, the First Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
5
|
Baines KJ, Klausner MS, Patterson VS, Renaud SJ. Interleukin-15 deficient rats have reduced osteopontin at the maternal-fetal interface. Front Cell Dev Biol 2023; 11:1079164. [PMID: 37152295 PMCID: PMC10157472 DOI: 10.3389/fcell.2023.1079164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 04/11/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction: Uterine Natural Killer (NK) cells are the predominant immune cells within the decidua during early pregnancy. These cells are thought to regulate aspects of decidualization and placental development, but their functions remain poorly characterized, especially in species with deeply invading trophoblasts such as humans and rats. Interleukin-15 (IL-15) is a cytokine required for NK cell development and survival. IL-15 mutant (IL15Δ/Δ) rats lack NK cells and exhibit altered placental development with precocious trophoblast invasion. In this study, we profiled gene expression differences between wild-type and IL15Δ/Δ implantation sites to reveal candidate factors produced by uterine NK cells that may regulate placentation and trophoblast invasion. Methods: Clariom S gene expression profiling was performed using implantation sites collected from pregnant wild-type and IL15Δ/Δ rats on gestational day 9.5. Levels and localization of perforin and osteopontin in implantation sites from wild-type and IL15Δ/Δ rats were further analyzed. The effect of osteopontin on the invasive capacity of rat trophoblasts was evaluated using Matrigel-based Transwell assays. Results: There were 257 genes differentially expressed between wild-type and IL15Δ/Δ implantation sites on gestational day 9.5, including decreased expression of various NK cell markers in IL15Δ/Δ rats, as well as Spp1, which encodes osteopontin. In wild-type rats, osteopontin was present within the decidua basalis and adjacent to the primitive placenta, and osteopontin colocalized with the NK cell marker perforin. Osteopontin was also detectable in uterine glands. Conversely, in IL15Δ/Δ rats, osteopontin and perforin were not readily detectable in the decidua despite robust osteopontin levels in uterine glands. Neutralization of osteopontin in media conditioned by cells isolated from the decidua decreased invasion of rat trophoblasts, suggesting that reduced levels of osteopontin are unlikely to account for the precocious trophoblast invasion in IL15Δ/Δ rats. Conclusion: Osteopontin is expressed by NK cells at the maternal-fetal interface in rats and may contribute to modulation of trophoblast invasion.
Collapse
Affiliation(s)
- Kelly J. Baines
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Michelle S. Klausner
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Violet S. Patterson
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Stephen J. Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
- Children’s Health Research Institute, Lawson Health Research Institute, London, ON, Canada
- *Correspondence: Stephen J. Renaud,
| |
Collapse
|
6
|
Hilton LR, Rätsep MT, VandenBroek MM, Jafri S, Laverty KJ, Mitchell M, Theilmann AL, Smart JA, Hawke LG, Moore SD, Renaud SJ, Soares MJ, Morrell NW, Ormiston ML. Impaired Interleukin-15 Signaling via BMPR2 Loss Drives Natural Killer Cell Deficiency and Pulmonary Hypertension. Hypertension 2022; 79:2493-2504. [PMID: 36043416 DOI: 10.1161/hypertensionaha.122.19178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 08/11/2022] [Indexed: 01/17/2023]
Abstract
BACKGROUND Natural killer (NK) cell impairment is a feature of pulmonary arterial hypertension (PAH) and contributes to vascular remodeling in animal models of disease. Although mutations in BMPR2, the gene encoding the BMP (bone morphogenetic protein) type-II receptor, are strongly associated with PAH, the contribution of BMPR2 loss to NK cell impairment remains unknown. We explored the impairment of IL (interleukin)-15 signaling, a central mediator of NK cell homeostasis, as both a downstream target of BMPR2 loss and a contributor to the pathogenesis of PAH. METHODS The expression, trafficking, and secretion of IL-15 and IL-15Rα (interleukin 15 α-type receptor) were assessed in human pulmonary artery endothelial cells, with or without BMPR2 silencing. NK cell development and IL-15/IL-15Rα levels were quantified in mice bearing a heterozygous knock-in of the R899X-BMPR2 mutation (bmpr2+/R899X). NK-deficient Il15-/- rats were exposed to the Sugen/hypoxia and monocrotaline models of PAH to assess the impact of impaired IL-15 signaling on disease severity. RESULTS BMPR2 loss reduced IL-15Rα surface presentation and secretion in human pulmonary artery endothelial cells via impaired trafficking through the trans-Golgi network. bmpr2+/R899X mice exhibited a decrease in NK cells, which was not attributable to impaired hematopoietic development but was instead associated with reduced IL-15/IL-15Rα levels in these animals. Il15-/- rats of both sexes exhibited enhanced disease severity in the Sugen/hypoxia model, with only male Il15-/- rats developing more severe PAH in response to monocrotaline. CONCLUSIONS This work identifies the loss of IL-15 signaling as a novel BMPR2-dependent contributor to NK cell impairment and pulmonary vascular disease.
Collapse
Affiliation(s)
- L Rhiannon Hilton
- Departments of Biomedical and Molecular Sciences, Medicine and Surgery, Queen's University, Kingston, Canada (L.B.H., M.T.R., M.M.V., K.J.L., M.M., A.L.T., J.A.S., L.G.H., M.L.O.)
| | - Matthew T Rätsep
- Departments of Biomedical and Molecular Sciences, Medicine and Surgery, Queen's University, Kingston, Canada (L.B.H., M.T.R., M.M.V., K.J.L., M.M., A.L.T., J.A.S., L.G.H., M.L.O.)
| | - M Martin VandenBroek
- Departments of Biomedical and Molecular Sciences, Medicine and Surgery, Queen's University, Kingston, Canada (L.B.H., M.T.R., M.M.V., K.J.L., M.M., A.L.T., J.A.S., L.G.H., M.L.O.)
| | - Salema Jafri
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom (S.J., S.D.M., N.W.M.)
| | - Kimberly J Laverty
- Departments of Biomedical and Molecular Sciences, Medicine and Surgery, Queen's University, Kingston, Canada (L.B.H., M.T.R., M.M.V., K.J.L., M.M., A.L.T., J.A.S., L.G.H., M.L.O.)
| | - Melissa Mitchell
- Departments of Biomedical and Molecular Sciences, Medicine and Surgery, Queen's University, Kingston, Canada (L.B.H., M.T.R., M.M.V., K.J.L., M.M., A.L.T., J.A.S., L.G.H., M.L.O.)
| | - Anne L Theilmann
- Departments of Biomedical and Molecular Sciences, Medicine and Surgery, Queen's University, Kingston, Canada (L.B.H., M.T.R., M.M.V., K.J.L., M.M., A.L.T., J.A.S., L.G.H., M.L.O.)
| | - James A Smart
- Departments of Biomedical and Molecular Sciences, Medicine and Surgery, Queen's University, Kingston, Canada (L.B.H., M.T.R., M.M.V., K.J.L., M.M., A.L.T., J.A.S., L.G.H., M.L.O.)
| | - Lindsey G Hawke
- Departments of Biomedical and Molecular Sciences, Medicine and Surgery, Queen's University, Kingston, Canada (L.B.H., M.T.R., M.M.V., K.J.L., M.M., A.L.T., J.A.S., L.G.H., M.L.O.)
| | - Stephen D Moore
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom (S.J., S.D.M., N.W.M.)
| | - Stephen J Renaud
- Department of Anatomy and Cell Biology, Western University, London, Canada (S.J.R.)
| | - Michael J Soares
- Departments of Pathology and Laboratory Medicine and Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City (M.J.S.)
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom (S.J., S.D.M., N.W.M.)
| | - Mark L Ormiston
- Departments of Biomedical and Molecular Sciences, Medicine and Surgery, Queen's University, Kingston, Canada (L.B.H., M.T.R., M.M.V., K.J.L., M.M., A.L.T., J.A.S., L.G.H., M.L.O.)
| |
Collapse
|
7
|
Abstract
The hemochorial placentation site is characterized by a dynamic interplay between trophoblast cells and maternal cells. These cells cooperate to establish an interface required for nutrient delivery to promote fetal growth. In the human, trophoblast cells penetrate deep into the uterus. This is not a consistent feature of hemochorial placentation and has hindered the establishment of suitable animal models. The rat represents an intriguing model for investigating hemochorial placentation with deep trophoblast cell invasion. In this study, we used single-cell RNA sequencing to characterize the transcriptome of the invasive trophoblast cell lineage, as well as other cell populations within the rat uterine-placental interface during early (gestation day [gd] 15.5) and late (gd 19.5) stages of intrauterine trophoblast cell invasion. We identified a robust set of transcripts that define invasive trophoblast cells, as well as transcripts that distinguished endothelial, smooth muscle, natural killer, and macrophage cells. Invasive trophoblast, immune, and endothelial cell populations exhibited distinct spatial relationships within the uterine-placental interface. Furthermore, the maturation stage of invasive trophoblast cell development could be determined by assessing gestation stage-dependent changes in transcript expression. Finally, and most importantly, expression of a prominent subset of rat invasive trophoblast cell transcripts is conserved in the invasive extravillous trophoblast cell lineage of the human placenta. These findings provide foundational data to identify and interrogate key conserved regulatory mechanisms essential for the development and function of an important compartment within the hemochorial placentation site that is essential for a healthy pregnancy.
Collapse
|
8
|
Haddad FL, Patel SV, Doornaert EE, De Oliveira C, Allman BL, Baines KJ, Renaud SJ, Schmid S. Interleukin 15 modulates the effects of poly I:C maternal immune activation on offspring behaviour. Brain Behav Immun Health 2022; 23:100473. [PMID: 35668725 PMCID: PMC9166394 DOI: 10.1016/j.bbih.2022.100473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 01/21/2023] Open
Abstract
Maternal infections during pregnancy are linked with an increased risk for disorders like Autism Spectrum Disorder and schizophrenia in the offspring. Although precise mechanisms are still unclear, clinical and preclinical evidence suggest a strong role for maternal immune activation (MIA) in the neurodevelopmental disruptions caused by maternal infection. Previously, studies using the Polyinosinic:Polycytidylic (Poly I:C) MIA preclinical model showed that cytokines like Interleukin 6 (Il6) are important mediators of MIA's effects. In this study, we hypothesized that Il15 may similarly act as a mediator of Poly I:C MIA, given its role in the antiviral immune response. To test this hypothesis, we induced Poly I:C MIA at gestational day 9.5 in wildtype (WT) and Il15−/− rat dams and tested their offspring in adolescence and adulthood. Poly I:C MIA and Il15 knockout produced both independent and synergistic effects on offspring behaviour. Poly I:C MIA decreased startle reactivity in adult WT offspring but resulted in increased adolescent anxiety and decreased adult locomotor activity in Il15−/− offspring. In addition, Poly I:C MIA led to genotype-independent effects on locomotor activity and prepulse inhibition. Finally, we showed that Il15−/− offspring exhibit distinct phenotypes that were unrelated to Poly I:C MIA including altered startle reactivity, locomotion and signal transduction in the auditory brainstem. Overall, our findings indicate that the lack of Il15 can leave offspring either more or less susceptible to Poly I:C MIA, depending on the phenotype in question. Future studies should examine the contribution of fetal versus maternal Il15 in MIA to determine the precise developmental mechanisms underlying these changes. Poly I:C MIA decreases startle reactivity in adult WT but not Il15−/− offspring. Il15−/− offspring exposed to Poly I:C MIA show altered PPI and open field exploration. Il15−/− rats exhibit distinct behavioural phenotypes independent from MIA.
Collapse
|
9
|
Marquardt RM, Ahn SH, Reske JJ, Chandler RL, Petroff MG, Kim TH, Jeong JW. Endometrial Epithelial ARID1A Is Required for Uterine Immune Homeostasis during Early Pregnancy. Int J Mol Sci 2022; 23:6067. [PMID: 35682747 PMCID: PMC9181301 DOI: 10.3390/ijms23116067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/13/2022] [Accepted: 05/24/2022] [Indexed: 12/14/2022] Open
Abstract
A growing body of work suggests epigenetic dysregulation contributes to endometriosis pathophysiology and female infertility. The chromatin remodeling complex subunit AT-rich interaction domain 1A (ARID1A) must be properly expressed to maintain normal uterine function. Endometrial epithelial ARID1A is indispensable for pregnancy establishment in mice through regulation of endometrial gland function; however, ARID1A expression is decreased in infertile women with endometriosis. We hypothesized that ARID1A performs critical operations in the endometrial epithelium necessary for fertility besides maintaining gland function. To identify alterations in uterine gene expression resulting from loss of epithelial ARID1A, we performed RNA-sequencing analysis on pre-implantation uteri from LtfiCre/+Arid1af/f and control mice. Differential expression analysis identified 4181 differentially expressed genes enriched for immune-related ingenuity canonical pathways including agranulocyte adhesion and diapedesis and natural killer cell signaling. RT-qPCR confirmed an increase in pro-inflammatory cytokine and macrophage-related gene expression but a decrease in natural killer cell signaling. Immunostaining confirmed a uterus-specific increase in macrophage infiltration. Flow cytometry delineated an increase in inflammatory macrophages and a decrease in uterine dendritic cells in LtfiCre/+Arid1af/f uteri. These findings demonstrate a role for endometrial epithelial ARID1A in suppressing inflammation and maintaining uterine immune homeostasis, which are required for successful pregnancy and gynecological health.
Collapse
Affiliation(s)
- Ryan M. Marquardt
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA; (R.M.M.); (J.J.R.); (R.L.C.); (T.H.K.)
- Cell and Molecular Biology Program, College of Natural Science, Michigan State University, East Lansing, MI 48824, USA;
| | - Soo Hyun Ahn
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA;
| | - Jake J. Reske
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA; (R.M.M.); (J.J.R.); (R.L.C.); (T.H.K.)
| | - Ronald L. Chandler
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA; (R.M.M.); (J.J.R.); (R.L.C.); (T.H.K.)
| | - Margaret G. Petroff
- Cell and Molecular Biology Program, College of Natural Science, Michigan State University, East Lansing, MI 48824, USA;
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA;
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Tae Hoon Kim
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA; (R.M.M.); (J.J.R.); (R.L.C.); (T.H.K.)
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA; (R.M.M.); (J.J.R.); (R.L.C.); (T.H.K.)
| |
Collapse
|
10
|
Shukla V, Soares MJ. Modeling Trophoblast Cell-Guided Uterine Spiral Artery Transformation in the Rat. Int J Mol Sci 2022; 23:ijms23062947. [PMID: 35328368 PMCID: PMC8950824 DOI: 10.3390/ijms23062947] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 12/20/2022] Open
Abstract
The rat possesses hemochorial placentation with deep intrauterine trophoblast cell invasion and trophoblast-guided uterine spiral artery remodeling, which resembles human placentation. Uterine spiral arteries are extensively remodeled to deliver sufficient supply of maternal blood and nutrients to the developing fetus. Inadequacies in these key processes negatively impact fetal growth and development. Recent innovations in genome editing combined with effective phenotyping strategies have provided new insights into placental development. Application of these research approaches has highlighted both conserved and species-specific features of hemochorial placentation. The review provides foundational information on rat hemochorial placental development and function during physiological and pathological states, especially as related to the invasive trophoblast cell-guided transformation of uterine spiral arteries. Our goal is to showcase the utility of the rat as a model for in vivo mechanistic investigations targeting regulatory events within the uterine-placental interface.
Collapse
Affiliation(s)
- Vinay Shukla
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Correspondence: (V.S.); (M.J.S.)
| | - Michael J. Soares
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy, Kansas City, MO 64108, USA
- Correspondence: (V.S.); (M.J.S.)
| |
Collapse
|
11
|
Hung TH, Hung JT, Wu CE, Huang Y, Lee CW, Yeh CT, Chung YH, Lo FY, Lai LC, Tung JK, Yu J, Yeh CN, Yu AL. Globo H Is a Promising Theranostic Marker for Intrahepatic Cholangiocarcinoma. Hepatol Commun 2022; 6:194-208. [PMID: 34558839 PMCID: PMC8710794 DOI: 10.1002/hep4.1800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 12/29/2022] Open
Abstract
Recent studies support the development of cancer therapeutics to target Globo H-ceramide, the most prevalent tumor-associated carbohydrate antigen in epithelial cancers. Herein, we evaluated the expression of Globo H and its prognostic significance in intrahepatic cholangiocarcinoma (ICC) and conducted preclinical studies to assess the antitumor activity of Globo H-specific antibody in thioacetamide (TAA)-induced ICC in rats. Globo H-ceramide in tumor specimens was detected by immunohistochemistry (IHC) and mass spectrometry. Antitumor efficacy of anti-Globo H mAbVK9 was evaluated in TAA-induced ICC in rat. Natural killer (NK) cells and their related genes were analyzed by IHC and quantitative real-time polymerase chain reaction. Data mining revealed that B3GALT5 and FUT2, the key enzymes for Globo H biosynthesis, were significantly up-regulated in human ICC. In addition, Globo H expression was detected in 41% (63 of 155) of ICC tumor specimens by IHC staining, and validated by mass spectrometric analysis of two IHC-positive tumors. Patients with Globo H positive tumors had significantly shorter relapse-free survival (RFS) and overall survival (P = 0.0003 and P = 0.002, respectively). Multivariable Cox regression analysis identified Globo H expression as an independent unfavorable predictor for RFS (hazard ratio: 1.66, 95% confidence interval: 1.08-2.36, P = 0.02) in ICC. Furthermore, gradual emergence of Globo H in liver tissues over 6 months in TAA-treated rats recapitulated the multistage progression of ICC in vivo. Importantly, administration of anti-Globo H mAbVK9 in rats bearing TAA-induced ICC significantly suppressed tumor growth with increased NK cells in the tumor microenvironment. Conclusion: Globo H is a theranostic marker in ICC.
Collapse
Affiliation(s)
- Tsai-Hsien Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Jung-Tung Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chiao-En Wu
- Department of Hematology-Oncology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Yenlin Huang
- Department of Anatomic Pathology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Chang Gung University, Taoyuan, Taiwan
| | - Chien-Wei Lee
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chau-Ting Yeh
- Liver Research Center, Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Hsiu Chung
- Department of Medical Research and Development, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Fei-Yun Lo
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Li-Chun Lai
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - John K Tung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - John Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
- Chang Gung University, Taoyuan, Taiwan
| | - Chun-Nan Yeh
- Department of Surgery and Liver Research Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Alice L Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Pediatrics, University of California in San Diego, San Diego, CA
- Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
12
|
Intersection of regulatory pathways controlling hemostasis and hemochorial placentation. Proc Natl Acad Sci U S A 2021; 118:2111267118. [PMID: 34876522 DOI: 10.1073/pnas.2111267118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2021] [Indexed: 11/18/2022] Open
Abstract
Hemochorial placentation is characterized by the development of trophoblast cells specialized to interact with the uterine vascular bed. We utilized trophoblast stem (TS) cell and mutant rat models to investigate regulatory mechanisms controlling trophoblast cell development. TS cell differentiation was characterized by acquisition of transcript signatures indicative of an endothelial cell-like phenotype, which was highlighted by the expression of anticoagulation factors including tissue factor pathway inhibitor (TFPI). TFPI localized to invasive endovascular trophoblast cells of the rat placentation site. Disruption of TFPI in rat TS cells interfered with development of the endothelial cell-like endovascular trophoblast cell phenotype. Similarly, TFPI was expressed in human invasive/extravillous trophoblast (EVT) cells situated within first-trimester human placental tissues and following differentiation of human TS cells. TFPI was required for human TS cell differentiation to EVT cells. We next investigated the physiological relevance of TFPI at the placentation site. Genome-edited global TFPI loss-of-function rat models revealed critical roles for TFPI in embryonic development, resulting in homogeneous midgestation lethality prohibiting analysis of the role of TFPI as a regulator of the late-gestation wave of intrauterine trophoblast cell invasion. In vivo trophoblast-specific TFPI knockdown was compatible with pregnancy but had profound effects at the uterine-placental interface, including restriction of the depth of intrauterine trophoblast cell invasion while leading to the accumulation of natural killer cells and increased fibrin deposition. Collectively, the experimentation implicates TFPI as a conserved regulator of invasive/EVT cell development, uterine spiral artery remodeling, and hemostasis at the maternal-fetal interface.
Collapse
|
13
|
Iqbal K, Pierce SH, Kozai K, Dhakal P, Scott RL, Roby KF, Vyhlidal CA, Soares MJ. Evaluation of Placentation and the Role of the Aryl Hydrocarbon Receptor Pathway in a Rat Model of Dioxin Exposure. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:117001. [PMID: 34747641 PMCID: PMC8574979 DOI: 10.1289/ehp9256] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
BACKGROUND Our environment is replete with chemicals that can affect embryonic and extraembryonic development. Dioxins, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), are compounds affecting development through the aryl hydrocarbon receptor (AHR). OBJECTIVES The purpose of this investigation was to examine the effects of TCDD exposure on pregnancy and placentation and to evaluate roles for AHR and cytochrome P450 1A1 (CYP1A1) in TCDD action. METHODS Actions of TCDD were examined in wild-type and genome-edited rat models. Placenta phenotyping was assessed using morphological, biochemical, and molecular analyses. RESULTS TCDD exposures were shown to result in placental adaptations and at higher doses, pregnancy termination. Deep intrauterine endovascular trophoblast cell invasion was a prominent placentation site adaptation to TCDD. TCDD-mediated placental adaptations were dependent upon maternal AHR signaling but not upon placental or fetal AHR signaling nor the presence of a prominent AHR target, CYP1A1. At the placentation site, TCDD activated AHR signaling within endothelial cells but not trophoblast cells. Immune and trophoblast cell behaviors at the uterine-placental interface were guided by the actions of TCDD on endothelial cells. DISCUSSION We identified an AHR regulatory pathway in rats activated by dioxin affecting uterine and trophoblast cell dynamics and the formation of the hemochorial placenta. https://doi.org/10.1289/EHP9256.
Collapse
Affiliation(s)
- Khursheed Iqbal
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Stephen H. Pierce
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Keisuke Kozai
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Pramod Dhakal
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Regan L. Scott
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Katherine F. Roby
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Anatomy and Cell Biology, KUMC, Kansas City, Kansas, USA
| | - Carrie A. Vyhlidal
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children’s Mercy Kansas City, Kansas City, Missouri
- Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas City, Missouri
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Michael J. Soares
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
- Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas City, Missouri
- Department of Obstetrics and Gynecology, KUMC, Kansas City, Kansas, USA
| |
Collapse
|
14
|
Dhakal P, Fitzgerald HC, Kelleher AM, Liu H, Spencer TE. Uterine glands impact embryo survival and stromal cell decidualization in mice. FASEB J 2021; 35:e21938. [PMID: 34547143 DOI: 10.1096/fj.202101170rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 11/11/2022]
Abstract
Uterine glands are essential for the establishment of pregnancy and have critical roles in endometrial receptivity to blastocyst implantation, stromal cell decidualization, and placentation. Uterine gland dysfunction is considered a major contributing factor to pregnancy loss, however our understanding of how glands impact embryo survival and stromal cell decidualization is incomplete. Forkhead box A2 (FOXA2) is expressed only in the glandular epithelium and regulates its development and function. Mice with a conditional deletion of FOXA2 in the uterus are infertile due to defective embryo implantation arising from a lack of leukemia inhibitory factor (LIF), a critical factor of uterine gland origin. Here, a glandless FOXA2-deficient mouse model, coupled with LIF repletion to rescue the implantation defect, was used to investigate the roles of uterine glands in embryo survival and decidualization. Studies found that embryo survival and decidualization were compromised in glandless FOXA2-deficient mice on gestational day 6.5, resulting in abrupt pregnancy loss by day 7.5. These findings strongly support the hypothesis that uterine glands secrete factors other than LIF that impact embryo survival and stromal cell decidualization for pregnancy success.
Collapse
Affiliation(s)
- Pramod Dhakal
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| | | | - Andrew M Kelleher
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA.,Division of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, Missouri, USA
| | - Hongyu Liu
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| | - Thomas E Spencer
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA.,Division of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
15
|
Kozai K, Iqbal K, Moreno-Irusta A, Scott RL, Simon ME, Dhakal P, Fields PE, Soares MJ. Protective role of IL33 signaling in negative pregnancy outcomes associated with lipopolysaccharide exposure. FASEB J 2021; 35:e21272. [PMID: 33423320 DOI: 10.1096/fj.202001782rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/15/2020] [Accepted: 11/30/2020] [Indexed: 01/21/2023]
Abstract
Interleukin 33 (IL33) signaling has been implicated in the establishment and maintenance of pregnancy and in pregnancy disorders. The goal of this project was to evaluate the role of IL33 signaling in rat pregnancy. The rat possesses hemochorial placentation with deep intrauterine trophoblast invasion; features also characteristic of human placentation. We generated and characterized a germline mutant rat model for IL33 using CRISPR/Cas9 genome editing. IL33 deficient rats exhibited deficits in lung responses to an inflammatory stimulus (Sephadex G-200) and to estrogen-induced uterine eosinophilia. Female rats deficient in IL33 were fertile and exhibited pregnancy outcomes (gestation length and litter size) similar to wild-type rats. Placental weight was adversely affected by the disruption of IL33 signaling. A difference in pregnancy-dependent adaptations to lipopolysaccharide (LPS) exposure was observed between wild-type and IL33 deficient pregnancies. Pregnancy in wild-type rats treated with LPS did not differ significantly from pregnancy in vehicle-treated wild-type rats. In contrast, LPS treatment decreased fetal survival rate, fetal and placental weights, and increased fetal growth restriction in IL33 deficient rats. In summary, a new rat model for investigating IL33 signaling has been established. IL33 signaling participates in the regulation of placental development and protection against LPS-induced fetal and placental growth restriction.
Collapse
Affiliation(s)
- Keisuke Kozai
- Institute for Reproduction and Perinatal Research, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas, KS, USA
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas, KS, USA
| | - Ayelen Moreno-Irusta
- Institute for Reproduction and Perinatal Research, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas, KS, USA
| | - Regan L Scott
- Institute for Reproduction and Perinatal Research, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas, KS, USA
| | - Mikaela E Simon
- Institute for Reproduction and Perinatal Research, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas, KS, USA
| | - Pramod Dhakal
- Institute for Reproduction and Perinatal Research, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas, KS, USA
| | - Patrick E Fields
- Institute for Reproduction and Perinatal Research, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas, KS, USA
| | - Michael J Soares
- Institute for Reproduction and Perinatal Research, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas, KS, USA.,Department of Pediatrics, University of Kansas Medical Center, Kansas, KS, USA.,Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas, KS, USA.,Center for Perinatal Research, Children's Mercy Research Institute, Children's Mercy, Kansas, MO, USA
| |
Collapse
|
16
|
Huhn O, Zhao X, Esposito L, Moffett A, Colucci F, Sharkey AM. How Do Uterine Natural Killer and Innate Lymphoid Cells Contribute to Successful Pregnancy? Front Immunol 2021; 12:607669. [PMID: 34234770 PMCID: PMC8256162 DOI: 10.3389/fimmu.2021.607669] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 05/10/2021] [Indexed: 12/19/2022] Open
Abstract
Innate lymphoid cells (ILCs) are the most abundant immune cells in the uterine mucosa both before and during pregnancy. Circumstantial evidence suggests they play important roles in regulating placental development but exactly how they contribute to the successful outcome of pregnancy is still unclear. Uterine ILCs (uILCs) include subsets of tissue-resident natural killer (NK) cells and ILCs, and until recently the phenotype and functions of uILCs were poorly defined. Determining the specific roles of each subset is intrinsically challenging because of the rapidly changing nature of the tissue both during the menstrual cycle and pregnancy. Single-cell RNA sequencing (scRNAseq) and high dimensional flow and mass cytometry approaches have recently been used to analyse uILC populations in the uterus in both humans and mice. This detailed characterisation has significantly changed our understanding of the heterogeneity within the uILC compartment. It will also enable key clinical questions to be addressed including whether specific uILC subsets are altered in infertility, miscarriage and pregnancy disorders such as foetal growth restriction and pre-eclampsia. Here, we summarise recent advances in our understanding of the phenotypic and functional diversity of uILCs in non-pregnant endometrium and first trimester decidua, and review how these cells may contribute to successful placental development.
Collapse
Affiliation(s)
- Oisín Huhn
- Department of Obstetrics and Gynaecology, National Institute for Health Research Cambridge, Biomedical Research Centre, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience University of Cambridge, Cambridge, United Kingdom
| | - Xiaohui Zhao
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience University of Cambridge, Cambridge, United Kingdom
| | - Laura Esposito
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience University of Cambridge, Cambridge, United Kingdom
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Ashley Moffett
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience University of Cambridge, Cambridge, United Kingdom
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Francesco Colucci
- Department of Obstetrics and Gynaecology, National Institute for Health Research Cambridge, Biomedical Research Centre, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience University of Cambridge, Cambridge, United Kingdom
| | - Andrew M. Sharkey
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience University of Cambridge, Cambridge, United Kingdom
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
17
|
Fournier SB, D'Errico JN, Stapleton PA. Uterine Vascular Control Preconception and During Pregnancy. Compr Physiol 2021; 11:1871-1893. [PMID: 34061977 DOI: 10.1002/cphy.c190015] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Successful pregnancy and reproduction are dependent on adequate uterine blood flow, placental perfusion, and vascular responsivity to fetal demands. The ability to support pregnancy centers on systemic adaptation and endometrial preparation through decidualization, embryonic implantation, trophoblast invasion, arterial/arteriolar reactivity, and vascular remodeling. These adaptations occur through responsiveness to endocrine signaling and local uteroplacental mediators. The purpose of this article is to highlight the current knowledge associated with vascular remodeling and responsivity during uterine preparation for and during pregnancy. We focus on maternal cardiovascular systemic and uterine modifications, endometrial decidualization, implantation and invasion, uterine and spiral artery remodeling, local uterine regulatory mechanisms, placentation, and pathological consequences of vascular dysfunction during pregnancy. © 2021 American Physiological Society. Compr Physiol 11:1-23, 2021.
Collapse
Affiliation(s)
- Sara B Fournier
- Environmental and Occupational Health Sciences Institute, Piscataway, New Jersey, USA
| | - Jeanine N D'Errico
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Phoebe A Stapleton
- Environmental and Occupational Health Sciences Institute, Piscataway, New Jersey, USA.,Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
18
|
Varberg KM, Soares MJ. Paradigms for investigating invasive trophoblast cell development and contributions to uterine spiral artery remodeling. Placenta 2021; 113:48-56. [PMID: 33985793 DOI: 10.1016/j.placenta.2021.04.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 04/16/2021] [Accepted: 04/18/2021] [Indexed: 12/21/2022]
Abstract
Uterine spiral arteries are extensively remodeled during placentation to ensure sufficient delivery of maternal blood to the developing fetus. Uterine spiral arterial remodeling is complex, as cells originating from both mother and developing conceptus interact at the maternal interface to regulate the extracellular matrix remodeling and vasculature restructuring necessary for successful placentation. Despite this complexity, one mechanism critical to spiral artery remodeling is trophoblast cell invasion into the maternal compartment. Invasive trophoblast cells include both interstitial and endovascular populations that exhibit spatiotemporal differences in uterine invasion, including proximity to uterine spiral arteries. Interstitial trophoblast cells invade the uterine parenchyma where they are interspersed among stromal cells. Endovascular trophoblast cells infiltrate uterine spiral arteries, replace endothelial cells, adopt a pseudo-endothelial cell phenotype, and engineer vessel remodeling. Impaired trophoblast cell invasion and, consequently, insufficient uterine spiral arterial remodeling can lead to the development of pregnancy disorders, such as preeclampsia, intrauterine growth restriction, and premature birth. This review provides insights into invasive trophoblast cells and their function during normal placentation as well as in settings of disease.
Collapse
Affiliation(s)
- Kaela M Varberg
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA; Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA.
| | - Michael J Soares
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA; Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA; Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Center for Perinatal Research, Children's Mercy Research Institute, Children's Mercy Kansas City, Missouri 64108, USA.
| |
Collapse
|
19
|
Chenouard V, Remy S, Tesson L, Ménoret S, Ouisse LH, Cherifi Y, Anegon I. Advances in Genome Editing and Application to the Generation of Genetically Modified Rat Models. Front Genet 2021; 12:615491. [PMID: 33959146 PMCID: PMC8093876 DOI: 10.3389/fgene.2021.615491] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
The rat has been extensively used as a small animal model. Many genetically engineered rat models have emerged in the last two decades, and the advent of gene-specific nucleases has accelerated their generation in recent years. This review covers the techniques and advances used to generate genetically engineered rat lines and their application to the development of rat models more broadly, such as conditional knockouts and reporter gene strains. In addition, genome-editing techniques that remain to be explored in the rat are discussed. The review also focuses more particularly on two areas in which extensive work has been done: human genetic diseases and immune system analysis. Models are thoroughly described in these two areas and highlight the competitive advantages of rat models over available corresponding mouse versions. The objective of this review is to provide a comprehensive description of the advantages and potential of rat models for addressing specific scientific questions and to characterize the best genome-engineering tools for developing new projects.
Collapse
Affiliation(s)
- Vanessa Chenouard
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
- genOway, Lyon, France
| | - Séverine Remy
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
| | - Laurent Tesson
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
| | - Séverine Ménoret
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
- CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, Nantes Université, Nantes, France
| | - Laure-Hélène Ouisse
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
| | | | - Ignacio Anegon
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
| |
Collapse
|
20
|
Marinić M, Mika K, Chigurupati S, Lynch VJ. Evolutionary transcriptomics implicates HAND2 in the origins of implantation and regulation of gestation length. eLife 2021; 10:61257. [PMID: 33522483 PMCID: PMC7943190 DOI: 10.7554/elife.61257] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 01/29/2021] [Indexed: 12/19/2022] Open
Abstract
The developmental origins and evolutionary histories of cell types, tissues, and organs contribute to the ways in which their dysfunction produces disease. In mammals, the nature, development and evolution of maternal-fetal interactions likely influence diseases of pregnancy. Here we show genes that evolved expression at the maternal-fetal interface in Eutherian mammals play essential roles in the evolution of pregnancy and are associated with immunological disorders and preterm birth. Among these genes is HAND2, a transcription factor that suppresses estrogen signaling, a Eutherian innovation allowing blastocyst implantation. We found dynamic HAND2 expression in the decidua throughout the menstrual cycle and pregnancy, gradually decreasing to a low at term. HAND2 regulates a distinct set of genes in endometrial stromal fibroblasts including IL15, a cytokine also exhibiting dynamic expression throughout the menstrual cycle and gestation, promoting migration of natural killer cells and extravillous cytotrophoblasts. We demonstrate that HAND2 promoter loops to an enhancer containing SNPs implicated in birth weight and gestation length regulation. Collectively, these data connect HAND2 expression at the maternal-fetal interface with evolution of implantation and gestational regulation, and preterm birth.
Collapse
Affiliation(s)
- Mirna Marinić
- Department of Human Genetics, University of Chicago, Chicago, United States
| | - Katelyn Mika
- Department of Human Genetics, University of Chicago, Chicago, United States
| | | | - Vincent J Lynch
- Department of Biological Sciences, University at Buffalo, Buffalo, United States
| |
Collapse
|
21
|
Iqbal K, Dhakal P, Pierce SH, Soares MJ. Catechol-O-methyltransferase and Pregnancy Outcome: an Appraisal in Rat. Reprod Sci 2021; 28:462-469. [PMID: 33048315 PMCID: PMC8082470 DOI: 10.1007/s43032-020-00348-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/05/2020] [Indexed: 01/19/2023]
Abstract
Catechol-O-methyltransferase (COMT) has been shown to be a key regulator of pregnancy outcomes in mouse, and its deficiency is causative in the development of a preeclampsia-like disease process. Preeclampsia is a human pregnancy disorder associated with failure of intrauterine trophoblast cell invasion and trophoblast-guided uterine spiral artery remodeling, which are not well-developed in mouse. The purpose of this study was to investigate COMT in rat, a species with deep intrauterine trophoblast invasion. To accomplish this task, we used clustered regularly interspaced short palindromic repeats/Cas9-mediated genome editing of the rat Comt gene. A Comt null rat model was established and its fertility characterized. Comt null male and female rats were viable and fertile. COMT deficiency did not significantly impact pregnancy outcomes, including litter size, placental and fetal weights, Mendelian and sex ratios, or pregnancy-dependent adaptations to hypoxia. Collectively, our findings indicate that pregnancy-associated phenotypic outcomes of COMT deficiency are not equivalent in mouse and rat. In rat, COMT is not required for a successful pregnancy outcome.
Collapse
Affiliation(s)
- Khursheed Iqbal
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas, USA.
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas, USA.
| | - Pramod Dhakal
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas, USA
- Department of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Stephen H Pierce
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas, USA
| | - Michael J Soares
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas, USA
- Department of Pediatrics, University of Kansas Medical Center, Kansas, USA
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas, USA
- Center for Perinatal Research, Children's Mercy Research Institute, Children's Mercy, Kansas, MO, USA
| |
Collapse
|
22
|
Sacks G, Finkelstein E. Natural killer cells and reproductive success. Am J Reprod Immunol 2020; 85:e13291. [DOI: 10.1111/aji.13291] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 06/15/2020] [Indexed: 12/30/2022] Open
Affiliation(s)
- Gavin Sacks
- IVFAustralia Sydney Australia
- University of New South Wales Sydney Australia
- St George hospital and Royal Hospital for Women Sydney Australia
| | | |
Collapse
|
23
|
Sliz A, Locker KCS, Lampe K, Godarova A, Plas DR, Janssen EM, Jones H, Herr AB, Hoebe K. Gab3 is required for IL-2- and IL-15-induced NK cell expansion and limits trophoblast invasion during pregnancy. Sci Immunol 2020; 4:4/38/eaav3866. [PMID: 31375526 DOI: 10.1126/sciimmunol.aav3866] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 07/03/2019] [Indexed: 12/12/2022]
Abstract
The scaffolding protein Grb2-associated binding protein 3 (Gab3) is a member of the Gab family, whose functions have remained elusive. Here, we identify Gab3 as a key determinant of peripheral NK cell expansion. Loss of Gab3 resulted in impaired IL-2 and IL-15-induced NK cell priming and expansion due to a selective impairment in MAPK signaling but not STAT5 signaling. In vivo, we found that Gab3 is required for recognition and elimination of "missing-self" and tumor targets. Unexpectedly, our studies also revealed that Gab3 plays an important role during pregnancy. Gab3-deficient mice exhibited impaired uterine NK cell expansion associated with abnormal spiral artery remodeling and increased trophoblast invasion in the decidua basalis. This coincided with stillbirth, retained placenta, maternal hemorrhage, and undelivered fetoplacental units at term. Thus, Gab3 is a key component required for cytokine-mediated NK cell priming and expansion that is essential for antitumor responses and limits trophoblast cell invasion during pregnancy.
Collapse
Affiliation(s)
- Anna Sliz
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA.,Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Kathryn C S Locker
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA.,Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Kristin Lampe
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA.,Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA
| | - Alzbeta Godarova
- Biomedical Informatics Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - David R Plas
- Vontz Center for Molecular Studies, University of Cincinnati, Cincinnati, OH 45267, USA
| | | | - Helen Jones
- Division of General Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Andrew B Herr
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA.,Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | | |
Collapse
|
24
|
Du M, Wang W, Huang L, Guan X, Lin W, Yao J, Li L. Natural killer cells in the pathogenesis of preeclampsia: a double-edged sword. J Matern Fetal Neonatal Med 2020; 35:1028-1035. [PMID: 32188324 DOI: 10.1080/14767058.2020.1740675] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Objective: To investigate the relationship between natural killer (NK) cells, extravillous trophoblast cells (EVTs) and vessel remodeling in early human pregnancy, and the association between NK cells and preeclampsia (PE) in late human pregnancy.Methods: Human decidual tissues from women with normal pregnancies were collected and examined for the relationship of NK cells with uterine vessel remodeling using immunohistochemistry. Percentages of peripheral blood NK (pNK) and decidual NK (dNK) cells and the levels of intracellular interferon (IFN)-γ, perforin and granzyme B in normal pregnancies, late-onset and early-onset PE were analyzed using flow cytometry. Cytolytic functions of dNK cells from normal and PE pregnancies were examined. Effects of conditioned medium (CM) of dNK cells from normal and PE pregnancies on first trimester trophoblast invasion and migration were tested.Results: In early pregnancy samples (9-13 weeks of gestation), we noted moderate vessel remodeling with abundant perivascular NK cells but a limited number of surrounding EVTs. The numbers of both human pNK cells and dNK cells and intracellular interferon (IFN)-γ, perforin and granzyme B production were significantly higher in PE compared with normal pregnancies at the time of delivery for both early- and late-onset disease. dNK cells from PE pregnancies not only killed first trimester trophoblasts but also inhibited their invasion and migration when compared to normal controls.Conclusion: Our results suggest that NK cells, in conjunction with EVTs, may play an important role in controlling uterine SA remodeling at the early stages of vessel remodeling, but they contribute to the pathogenesis of PE in late pregnancy.
Collapse
Affiliation(s)
- Mingyue Du
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Weijia Wang
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Lina Huang
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Xiuting Guan
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Wen Lin
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Jingxin Yao
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Liping Li
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
25
|
Utero-placental vascular remodeling during late gestation in Sprague-Dawley rats. Pregnancy Hypertens 2020; 20:36-43. [PMID: 32172168 DOI: 10.1016/j.preghy.2020.02.007] [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] [Received: 10/12/2019] [Revised: 02/14/2020] [Accepted: 02/28/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Remodeling of the uterine spiral arteries and blood vessels within the placenta allows delivery of nutrients to the growing utero-fetal-placental unit. While abnormal remodeling of these vessels is thought to play an important role in syndromes including intrauterine growth restriction and preeclampsia, there are a lack of studies that have quantified vascular remodeling in normal pregnant rats. Thus, the purpose of this study was to quantify time-dependent remodeling of the utero-placental vasculature during late gestation in normal pregnant rats. METHODS Timed-pregnant Sprague-Dawley rats were used. Gestational days of 14 and 19 were chosen because this when a large amount of fetal and placental growth occurs. A combined method of perfusion-casting and 3D micro-computed tomography were utilized to construct ex-vivo utero-placental vasculature images. RESULTS Significant spiral artery remodeling occurred between days 14 and 19. Vessel density shifted away from a distribution of smaller to larger diameters by day 19. Total spiral artery area and average diameter were increased by day 19. Moreover, branching and tortuosity of the spiral arteries were greater by day 19. In rodents, spiral arteries feed into the central canal vessels that funnel to sites of exchange. Canal vessel area and diameter were increased by day 19. CONCLUSIONS Our study supports a quantitative method to examine the placental vasculature showing that significant vascular remodeling occurs during late gestation in the utero-placental-fetal unit of the normal pregnant rat. This method may serve as a tool to investigate fundamental pathophysiological mechanisms underlying placental-related diseases in rat models.
Collapse
|
26
|
Sato Y. Endovascular trophoblast and spiral artery remodeling. Mol Cell Endocrinol 2020; 503:110699. [PMID: 31899258 DOI: 10.1016/j.mce.2019.110699] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/22/2019] [Accepted: 12/30/2019] [Indexed: 10/25/2022]
Abstract
Spiral artery remodeling, which is indispensable for successful pregnancy, is accomplished by endovascular trophoblasts that move upstream along the arterial wall, replace the endothelium, and disrupt the muscular lining. This review outlines the possible factors that could regulate endovascular trophoblast differentiation and invasion. First, high oxygen tension in the spiral artery could initiate endovascular trophoblast invasion. Second, activation of maternal decidual natural killer (dNK) cells could support perivascular invasion of interstitial trophoblasts and consequently could facilitate the endovascular trophoblast invasion. Third, maternal platelets trapped by the endovascular trophoblasts could enhance endovascular trophoblast invasion, which is in part mediated by chemokine CCL5 (C-C motif ligand 5) released from the activated platelets and chemokine receptor CCR1 (C-C chemokine receptor type 1) expressed specifically on the endovascular trophoblasts. The rat, in which trophoblast cells exhibit extensive interstitial and endovascular invasion, could be a suitable model animal for the study of human spiral artery remodeling. Apparently paradoxical results came from the rat study, i.e., exposure to hypoxia or depletion of dNK cells resulted in acceleration of the endovascular trophoblast invasion. This implies the presence of as-yet-undetermined regulator(s) whose effects on endovascular trophoblast invasion surpass the effects of surrounding oxygen tension or maternal dNK cells. In the future, clarification of the molecular differences between human interstitial and endovascular trophoblasts as well as establishment of the pregnant rat model exhibiting shallow endovascular trophoblast invasion and preeclamptic symptoms will contribute to elucidating the mechanism of spiral artery remodeling.
Collapse
Affiliation(s)
- Yukiyasu Sato
- Department of Obstetrics and Gynecology, Takamatsu Red Cross Hospital, 4-1-3 Banchou, Takamatsu, 760-0017, Japan.
| |
Collapse
|
27
|
Nandy D, Das S, Islam S, Ain R. Molecular regulation of vascular smooth muscle cell phenotype switching by trophoblast cells at the maternal-fetal interface. Placenta 2020; 93:64-73. [PMID: 32250741 DOI: 10.1016/j.placenta.2020.02.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/12/2020] [Accepted: 02/24/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Establishment of hemochorial placenta is associated with development and remodelling of uterine vasculature at the maternal fetal interface. This results in calibration of high resistance uterine arteries to flaccid low resistance vessels resulting in increased blood flow to the placenta and fetus in humans and rodents. Mechanisms underlying these remodelling events are poorly understood. In this report, we examine regulation of vascular remodelling using vascular smooth muscle cell (VSMC) phenotype switching as a primary parameter. METHODS Cellular dynamics was assessed by Immunofluorescence, qRT-PCR, western blotting in timed pregnant rat tissue. In vitro co-culture of trophoblast cells with vascular smooth muscle cells was used to understand regulation mechanism. RESULTS Analysis of cellular dynamics on days 13.5, 16.5 and 19.5 of gestation in the rat metrial gland, the entry point of uterine arteries, revealed that invasion of trophoblast cells preceded disappearance of VSMC α-SMA, a contractile state marker. Co-culture of VSMCs with trophoblast cells in vitro recapitulated trophoblast-induced de-differentiation of VSMCs in vivo. Interestingly, co-culturing with trophoblast cells activated PDGFRβ signalling in VSMCs, an effect mediated by secreted PDGF-BB from trophoblast cells. Trophoblast cells failed to elicit its effect on VSMC de-differentiation upon inhibition of PDGFRβ signalling using a selective inhibitor. Moreover, co-culturing with trophoblast cells also led to substantial increase in Akt activation and a modest increase in Erk phosphorylation in VSMCs and this effect was abolished by PDGFRβ inhibition. DISCUSSION Our results highlight that trophoblast cells direct VSMC phenotype switching and trophoblast derived PDGF-BB is one of the modulator.
Collapse
Affiliation(s)
- Debdyuti Nandy
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata, 700032, West Bengal, India
| | - Shreya Das
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata, 700032, West Bengal, India
| | - Safirul Islam
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata, 700032, West Bengal, India
| | - Rupasri Ain
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata, 700032, West Bengal, India; Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, West Bengal, India.
| |
Collapse
|
28
|
Sojka DK. Uterine Natural Killer Cell Heterogeneity: Lessons From Mouse Models. Front Immunol 2020; 11:290. [PMID: 32153593 PMCID: PMC7046796 DOI: 10.3389/fimmu.2020.00290] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 02/05/2020] [Indexed: 11/13/2022] Open
Abstract
Natural killer (NK) cells are the most abundant lymphocytes at the maternal-fetal interface. Epidemiological data implicate NK cells in human pregnancy outcomes. Discoveries using mouse NK cells have guided subsequent advances in human NK cell biology. However, it remains challenging to identify mouse and human uterine NK (uNK) cell function(s) because of the dynamic changes in the systemic-endocrinological and local uterine structural microenvironments during pregnancy. This review discusses functional similarities and differences between mouse and human NK cells at the maternal-fetal interface.
Collapse
Affiliation(s)
- Dorothy K Sojka
- Rheumatology Division, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
29
|
Meyer N, Zenclussen AC. Immune Cells in the Uterine Remodeling: Are They the Target of Endocrine Disrupting Chemicals? Front Immunol 2020; 11:246. [PMID: 32140155 PMCID: PMC7043066 DOI: 10.3389/fimmu.2020.00246] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 01/30/2020] [Indexed: 12/17/2022] Open
Abstract
Sufficient uterine remodeling is essential for fetal survival and development. Pathologies related to poor remodeling have a negative impact on maternal and fetal health even years after birth. Research of the last decades yielded excellent studies demonstrating the key role of immune cells in the remodeling processes. This review summarizes the current knowledge about the relevance of immune cells for uterine remodeling during pregnancy and further discusses immunomodulatory effects of man-made endocrine disrupting chemicals on immune cells.
Collapse
Affiliation(s)
- Nicole Meyer
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Ana Claudia Zenclussen
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
30
|
Goldman-Wohl D, Gamliel M, Mandelboim O, Yagel S. Learning from experience: cellular and molecular bases for improved outcome in subsequent pregnancies. Am J Obstet Gynecol 2019; 221:183-193. [PMID: 30802436 DOI: 10.1016/j.ajog.2019.02.037] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 02/07/2019] [Accepted: 02/18/2019] [Indexed: 12/23/2022]
Abstract
The frequencies of preeclampsia, fetal growth restriction, fetal demise, and low birthweight are lower in subsequent pregnancies. Enhanced maternal cardiovascular adaptation, shorter first and second stages of labor, and more robust lactation also have been observed in subsequent as compared with first pregnancies. We sought to investigate the cellular and molecular bases for better outcomes in subsequent pregnancies. Based on the knowledge that specialized immune cells at the maternal-fetal interface, decidual natural killer cells, promote development of the placental bed and conversion of the spiral arteries by secreting a myriad of angiogenic and growth factors, we asked whether decidual natural killer cells differ in subsequent as compared with first pregnancies. This idea stemmed from recent studies suggesting that natural killer cells, although part of the innate immune system, possess some features of adaptive immunity, including a certain type of immune cell memory, termed trained immunity. We found that decidual natural killer cells from parous women "remember pregnancy" and differ from decidual natural killer cells of primigravidae. Compared with the decidual natural killer cells of first pregnancy, these cells, that we termed pregnancy-trained decidual natural killer cells, express greater levels of the natural killer receptors NKG2C and leukocyte immunoglobulin-like receptor B1, which interact with ligands expressed on invasive trophoblasts. Furthermore, they secrete greater levels of several growth factors, including vascular endothelial growth factor α as well as interferon-γ, augmenting remodeling of the placental bed. We propose that this pregnancy-trained memory dwells in the epigenome, where memory of stimuli is known to persist even when the stimulus is no longer present. This epigenetic memory apparently resides in endometrial natural killer cells between pregnancies. We suggest that this trained memory, which we coined pregnancy-trained decidual natural killer cells, may be the missing link in the immune basis for enhanced subsequent pregnancy. Epigenetic memory (chromatin modification) also may afford a global explanation for additional findings of enhanced maternal cardiovascular adaptation, shorter first and second stages of labor, and more robust lactation. Understanding the molecular and cellular bases of improved outcomes of subsequent pregnancy may lead to the development of treatment modalities designed for women at high risk for pregnancy disorders originating at the maternal-fetal interface.
Collapse
Affiliation(s)
- Debra Goldman-Wohl
- Magda and Richard Hoffman Center for Human Placenta Research, Department of Obstetrics and Gynecology, Hebrew University Hadassah Medical Center, Jerusalem, Israel
| | - Moriya Gamliel
- The Concern Foundation Laboratories at the Lautenberg Centre for Immunology and Cancer Research, IMRIC, Faculty of Medicine, Hebrew University Hadassah Medical Center, Jerusalem, Israel
| | - Ofer Mandelboim
- The Concern Foundation Laboratories at the Lautenberg Centre for Immunology and Cancer Research, IMRIC, Faculty of Medicine, Hebrew University Hadassah Medical Center, Jerusalem, Israel
| | - Simcha Yagel
- Magda and Richard Hoffman Center for Human Placenta Research, Department of Obstetrics and Gynecology, Hebrew University Hadassah Medical Center, Jerusalem, Israel.
| |
Collapse
|
31
|
Soares MJ, Varberg KM, Iqbal K. Hemochorial placentation: development, function, and adaptations. Biol Reprod 2019; 99:196-211. [PMID: 29481584 DOI: 10.1093/biolre/ioy049] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/21/2018] [Indexed: 11/12/2022] Open
Abstract
Placentation is a reproductive adaptation that permits fetal growth and development within the protected confines of the female reproductive tract. Through this important role, the placenta also determines postnatal health and susceptibility to disease. The hemochorial placenta is a prominent feature in primate and rodent development. This manuscript provides an overview of the basics of hemochorial placental development and function, provides perspectives on major discoveries that have shaped placental research, and thoughts on strategies for future investigation.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas, USA and the Center for Perinatal Research, Children΄s Research Institute, Children΄s Mercy, Kansas City, Missouri, USA
| | - Kaela M Varberg
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
32
|
Yoshinaga K. A historical review of blastocyst implantation research. Biol Reprod 2018; 99:175-195. [PMID: 30010858 PMCID: PMC6279068 DOI: 10.1093/biolre/ioy093] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/17/2018] [Accepted: 04/23/2018] [Indexed: 02/07/2023] Open
Abstract
Research development on blastocyst implantation was reviewed in three sections: primate implantation, ungulate farm animal implantation, and the general process of blastocyst implantation in small rodents. Future research directions of this area are suggested.
Collapse
Affiliation(s)
- Koji Yoshinaga
- Fertility and Infertility Branch, Division of Extramural Research, NICHD, NIH,
Bethesda, Maryland, USA
| |
Collapse
|
33
|
Soares MJ, Iqbal K, Kozai K. Hypoxia and Placental Development. Birth Defects Res 2018; 109:1309-1329. [PMID: 29105383 DOI: 10.1002/bdr2.1135] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/04/2017] [Indexed: 12/17/2022]
Abstract
Hemochorial placentation is orchestrated through highly regulated temporal and spatial decisions governing the fate of trophoblast stem/progenitor cells. Trophoblast cell acquisition of specializations facilitating invasion and uterine spiral artery remodeling is a labile process, sensitive to the environment, and represents a process that is vulnerable to dysmorphogenesis in pathologic states. Hypoxia is a signal guiding placental development, and molecular mechanisms directing cellular adaptations to low oxygen tension are integral to trophoblast cell differentiation and placentation. Hypoxia can also be used as an experimental tool to investigate regulatory processes controlling hemochorial placentation. These developmental processes are conserved in mouse, rat, and human placentation. Consequently, elements of these developmental events can be modeled and hypotheses tested in trophoblast stem cells and in genetically manipulated rodents. Hypoxia is also a consequence of a failed placenta, yielding pathologies that can adversely affect maternal adjustments to pregnancy, fetal health, and susceptibility to adult disease. The capacity of the placenta for adaptation to environmental challenges highlights the importance of its plasticity in safeguarding a healthy pregnancy. Birth Defects Research 109:1309-1329, 2017.© 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas.,Fetal Health Research, Children's Research Institute, Children's Mercy, Kansas City, Missouri
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Keisuke Kozai
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|