1
|
Katsumata S, Ota K, Takahashi T, Mitsui J, Uchida N, Hiraoka K, Komiya A, Kawai K. Successful pregnancy and live birth despite discontinuation of hormone replacement therapy at 3 weeks and 5 days of gestation following vitrified-warmed embryo transfer under a hormone replacement therapy cycle: a case report and literature review. BMC Pregnancy Childbirth 2024; 24:831. [PMID: 39707226 DOI: 10.1186/s12884-024-07059-w] [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: 12/04/2023] [Accepted: 12/10/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND In freeze-thawed embryo transfer (FET) cycles, hormone replacement treatment (HRT) is crucial for implantation and pregnancy maintenance. HRT typically continues until the 10th week of pregnancy owing to a luteoplacental shift, although a definitive HRT regimen remains undetermined. We present the case of a woman who underwent FET during an HRT cycle and ceased HRT after a negative pregnancy test at 3 weeks and 5 days, who went on to deliver a healthy baby. CASE PRESENTATION A 30-year-old Japanese woman with primary infertility was scheduled for FET. Estrogen supplementation was initiated as part of the HRT cycle for endometrial preparation. After achieving an endometrial thickness of 8 mm, progesterone supplementation was commenced, and the transfer of a 4BB blastocyst occurred 5 days after initiating progesterone treatment. At a gestational age of 3 weeks and 5 days, her serum human chorionic gonadotropin (hCG) level was only 8.3 mIU/mL, leading to discontinuation of HRT due to the absence of pregnancy. However, at 6 weeks and 1 day, her serum hCG levels significantly rose to 9359 mIU/mL, prompting the resumption of HRT. Ultrasonography confirmed the presence of a gestational sac and cardiac activity in the uterus, and HRT was continued until the 10th week. Ultimately, she delivered a healthy female neonate vaginally, weighing 2601 g at 40 weeks and 6 days. CONCLUSIONS Progesterone supplementation is customary in FET with HRT cycles, although it has raised the possibility that there is demonstrating the potential for an ongoing pregnancy and resulting in a healthy baby under no progesterone replacement prior to the luteoplacental shift in this case. The duration and dosage of progesterone in luteal support for FET with HRT warrant further investigation.
Collapse
Affiliation(s)
| | - Kuniaki Ota
- Kameda IVF Clinic Makuhari, Makuhari, 261-8501, Japan
- Fukushima Medical Center for Children and Women, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
- Department of Obstetrics and Gynecology, Tokyo Rosai Hospital, Tokyo, 143-0013, Japan
| | - Toshifumi Takahashi
- Fukushima Medical Center for Children and Women, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan.
| | | | - Nozomi Uchida
- Kameda IVF Clinic Makuhari, Makuhari, 261-8501, Japan
| | | | - Akira Komiya
- Kameda IVF Clinic Makuhari, Makuhari, 261-8501, Japan
| | | |
Collapse
|
2
|
Hou L, Liang X, Zeng L, Wang Q, Chen Z. Conventional and modern markers of pregnancy of unknown location: Update and narrative review. Int J Gynaecol Obstet 2024; 167:957-967. [PMID: 39022869 DOI: 10.1002/ijgo.15807] [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: 03/27/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024]
Abstract
Pregnancy of unknown location (PUL) is a temporary pathologic or physiologic phenomenon of early pregnancy that requires follow up to determine the final pregnancy outcome. Evidence indicated that PUL patients suffer a remarkably higher rate of adverse pregnancy outcomes, represented by ectopic gestation and early pregnancy loss, than the general population. In the past few decades, discussion about PUL has never stopped, and a variety of markers have been widely investigated for the early and accurate evaluation of PUL, including serum biomarkers, ultrasound imaging features, multivariate analysis, and the diagnosis of ectopic pregnancy based on risk stratification. So far, machine learning (ML) methods represented by M4 and M6 logistic regression have gained a level of recognition and are continually improving. Nevertheless, the heterogeneity of PUL markers, mainly caused by the limited sample size, the differences in population and technical maturity, etc., have hampered the management of PUL. With the advancement of multidisciplinary integration and cutting-edge technologies (e.g. artificial intelligence, prediction model development, and telemedicine), novel markers, and strategies for the management of PUL are expected to be developed. In this review, we summarize both conventional and novel markers (represented by artificial intelligence) for PUL assessment and management, investigate their advancements, limitations and challenges, and propose insights on future research direction and clinical application.
Collapse
Affiliation(s)
- Likang Hou
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- The First Affiliated Hospital, Medical Imaging Center, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiaowen Liang
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, Department of Medical Imaging, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Lingqing Zeng
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- The First Affiliated Hospital, Medical Imaging Center, Hengyang Medical School, University of South China, Hengyang, China
| | - Qian Wang
- The First Affiliated Hospital, Center for Reproductive Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Zhiyi Chen
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, China
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, Department of Medical Imaging, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| |
Collapse
|
3
|
Dias Da Silva I, Wuidar V, Zielonka M, Pequeux C. Unraveling the Dynamics of Estrogen and Progesterone Signaling in the Endometrium: An Overview. Cells 2024; 13:1236. [PMID: 39120268 PMCID: PMC11312103 DOI: 10.3390/cells13151236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/25/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
The endometrium is crucial for the perpetuation of human species. It is a complex and dynamic tissue lining the inner wall of the uterus, regulated throughout a woman's life based on estrogen and progesterone fluctuations. During each menstrual cycle, this multicellular tissue undergoes cyclical changes, including regeneration, differentiation in order to allow egg implantation and embryo development, or shedding of the functional layer in the absence of pregnancy. The biology of the endometrium relies on paracrine interactions between epithelial and stromal cells involving complex signaling pathways that are modulated by the variations of estrogen and progesterone levels across the menstrual cycle. Understanding the complexity of estrogen and progesterone receptor signaling will help elucidate the mechanisms underlying normal reproductive physiology and provide fundamental knowledge contributing to a better understanding of the consequences of hormonal imbalances on gynecological conditions and tumorigenesis. In this narrative review, we delve into the physiology of the endometrium, encompassing the complex signaling pathways of estrogen and progesterone.
Collapse
Grants
- J.0165.24, 7.6529.23, J.0153.22, 7.4580.21F, 7.6518.21, J.0131.19 Fund for Scientific Research
- FSR-F-2023-FM, FSR-F-2022-FM, FSR-F-2021-FM, FSR-F-M-19/6761 University of Liège
- 2020, 2021, 2022 Fondation Léon Fredericq
Collapse
Affiliation(s)
| | | | | | - Christel Pequeux
- Tumors and Development, Estrogen-Sensitive Tissues and Cancer Team, GIGA-Cancer, Laboratory of Biology, University of Liège, 4000 Liège, Belgium; (I.D.D.S.); (V.W.); (M.Z.)
| |
Collapse
|
4
|
Nayeri T, Sarvi S, Daryani A. Effective factors in the pathogenesis of Toxoplasmagondii. Heliyon 2024; 10:e31558. [PMID: 38818168 PMCID: PMC11137575 DOI: 10.1016/j.heliyon.2024.e31558] [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: 01/03/2023] [Revised: 05/11/2024] [Accepted: 05/17/2024] [Indexed: 06/01/2024] Open
Abstract
Toxoplasma gondii (T. gondii) is a cosmopolitan protozoan parasite in humans and animals. It infects about 30 % of the human population worldwide and causes potentially fatal diseases in immunocompromised hosts and neonates. For this study, five English-language databases (ScienceDirect, ProQuest, Web of Science, PubMed, and Scopus) and the internet search engine Google Scholar were searched. This review was accomplished to draw a global perspective of what is known about the pathogenesis of T. gondii and various factors affecting it. Virulence and immune responses can influence the mechanisms of parasite pathogenesis and these factors are in turn influenced by other factors. In addition to the host's genetic background, the type of Toxoplasma strain, the routes of transmission of infection, the number of passages, and different phases of parasite life affect virulence. The identification of virulence factors of the parasite could provide promising insights into the pathogenesis of this parasite. The results of this study can be an incentive to conduct more intensive research to design and develop new anti-Toxoplasma agents (drugs and vaccines) to treat or prevent this infection. In addition, further studies are needed to better understand the key agents in the pathogenesis of T. gondii.
Collapse
Affiliation(s)
- Tooran Nayeri
- Infectious and Tropical Diseases Research Center, Dezful University of Medical Sciences, Dezful, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahabeddin Sarvi
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahmad Daryani
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
5
|
Dawid M, Pich K, Mlyczyńska E, Respekta-Długosz N, Wachowska D, Greggio A, Szkraba O, Kurowska P, Rak A. Adipokines in pregnancy. Adv Clin Chem 2024; 121:172-269. [PMID: 38797542 DOI: 10.1016/bs.acc.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Reproductive success consists of a sequential events chronology, starting with the ovum fertilization, implantation of the embryo, placentation, and cellular processes like proliferation, apoptosis, angiogenesis, endocrinology, or metabolic changes, which taken together finally conduct the birth of healthy offspring. Currently, many factors are known that affect the regulation and proper maintenance of pregnancy in humans, domestic animals, or rodents. Among the determinants of reproductive success should be distinguished: the maternal microenvironment, genes, and proteins as well as numerous pregnancy hormones that regulate the most important processes and ensure organism homeostasis. It is well known that white adipose tissue, as the largest endocrine gland in our body, participates in the synthesis and secretion of numerous hormones belonging to the adipokine family, which also may regulate the course of pregnancy. Unfortunately, overweight and obesity lead to the expansion of adipose tissue in the body, and its excess in both women and animals contributes to changes in the synthesis and release of adipokines, which in turn translates into dramatic changes during pregnancy, including those taking place in the organ that is crucial for the proper progress of pregnancy, i.e. the placenta. In this chapter, we are summarizing the current knowledge about levels of adipokines and their role in the placenta, taking into account the physiological and pathological conditions of pregnancy, e.g. gestational diabetes mellitus, preeclampsia, or intrauterine growth restriction in humans, domestic animals, and rodents.
Collapse
Affiliation(s)
- Monika Dawid
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Karolina Pich
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Ewa Mlyczyńska
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Natalia Respekta-Długosz
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Dominka Wachowska
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Aleksandra Greggio
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Oliwia Szkraba
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Patrycja Kurowska
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Agnieszka Rak
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland.
| |
Collapse
|
6
|
Xu Y, Liu X, Zeng W, Zhu Y, Dong J, Wu F, Chen C, Sharma S, Lin Y. DOCK1 insufficiency disrupts trophoblast function and pregnancy outcomes via DUSP4-ERK pathway. Life Sci Alliance 2024; 7:e202302247. [PMID: 37967942 PMCID: PMC10651491 DOI: 10.26508/lsa.202302247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/17/2023] Open
Abstract
Abnormal trophoblast function is associated with diseases such as recurrent spontaneous abortion, pre-eclampsia, and preterm birth, and endangers maternal and fetal health. However, the underlying regulatory mechanisms remain unclear. In this study, we found DOCK1 expression is decreased in the placental villi of patients with recurrent spontaneous abortion, and that its expression determined the invasive properties of extravillous trophoblasts (EVTs), highlighting a previously unknown role of DOCK1 in regulating EVT function. Furthermore, DOCK1 deficiency disturbed the ubiquitinated degradation of DUSP4, leading to its accumulation. This caused inactivation of the ERK signaling pathway, resulting in inadequate EVT migration and invasion. DOCK1 was implicated in regulating the ubiquitin levels of DUSP4, possibly by modulating the E3 ligase enzyme HUWE1. The results of our in vivo experiments confirmed that the DOCK1 inhibitor TBOPP caused miscarriage in mice by inactivating the DUSP4/ERK pathway. Collectively, our results revealed the crucial role of DOCK1 in the regulation of EVT function via the DUSP4-ERK pathway and a basis for the development of novel treatments for adverse pregnancy outcomes caused by trophoblast dysfunction.
Collapse
Affiliation(s)
- Yichi Xu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaorui Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weihong Zeng
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yueyue Zhu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Junpeng Dong
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fan Wu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cailian Chen
- Department of Automation, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of System Control and Information Processing, Ministry of Education of China, Shanghai, China
| | - Surendra Sharma
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Yi Lin
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Matsuo S, Kotani T, Tano S, Ushida T, Imai K, Nakamura T, Osuka S, Goto M, Osawa M, Asada Y, Kajiyama H. Risk factors for non-previa placenta accreta spectrum in pregnancies conceived through frozen embryo transfer during a hormone replacement cycle in Japan. Reprod Med Biol 2024; 23:e12592. [PMID: 39050787 PMCID: PMC11266119 DOI: 10.1002/rmb2.12592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/27/2024] [Accepted: 06/07/2024] [Indexed: 07/27/2024] Open
Abstract
Purpose Non-previa placenta accreta spectrum (PAS) is associated with assisted reproductive technology (ART), particularly frozen embryo transfer during hormone replacement therapy (HRC-FET). We especially aimed to evaluate the prevalence and risk factors for non-previa PAS in HRC-FET pregnancies. Methods Overall, 279 women who conceived through ART at three ART facilities and delivered at a single center were included in this retrospective study. Data regarding endometrial thickness at embryo transfer, previous histories, and type of embryo transfer-HRC-FET, frozen embryo transfer during a natural ovulatory cycle (NC-FET), and fresh embryo transfer (Fresh-ET)-were collected. Univariable logistic regression analyses were conducted. Results The prevalence of non-previa PAS was 27/192 (14.1%) in the HRC-FET group and 0 (0.0%) in both the NC-FET and Fresh-ET groups. Significantly high odds ratio [95% confidence interval] of non-previa PAS was associated with a history of artificial abortion (6.45 [1.98-21.02]), endometrial thickness <8.0 mm (6.11 [1.06-35.12]), resolved low-lying placenta (5.73 [2.13-15.41]), multiparity (2.90 [1.26-6.69]), polycystic ovarian syndrome (2.62 [1.02-6.71]), and subchorionic hematoma (2.49 [1.03-6.04]). Conclusions A history of artificial abortion, endometrial thickness <8.0 mm, and resolved low-lying placenta may help in antenatal detection of a high-risk population of non-previa PAS in HRC-FET pregnancies.
Collapse
Affiliation(s)
- Seiko Matsuo
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Tomomi Kotani
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
- Division of Reproduction and Perinatology, Center for Maternal‐Neonatal CareNagoya University HospitalNagoyaJapan
| | - Sho Tano
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Takafumi Ushida
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
- Division of Reproduction and Perinatology, Center for Maternal‐Neonatal CareNagoya University HospitalNagoyaJapan
| | - Kenji Imai
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Tomoko Nakamura
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
- Division of Reproduction and Perinatology, Center for Maternal‐Neonatal CareNagoya University HospitalNagoyaJapan
| | - Satoko Osuka
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Maki Goto
- Department of Obstetrics and GynecologyOkazaki City HospitalOkazakiJapan
| | | | | | - Hiroaki Kajiyama
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
8
|
Kazemian A, Tavares Pereira M, Aslan S, Payan-Carreira R, Reichler IM, Agaoglu RA, Kowalewski MP. Membrane-bound progesterone receptors in the canine uterus and placenta; possible targets in the maintenance of pregnancy. Theriogenology 2023; 210:68-83. [PMID: 37480804 DOI: 10.1016/j.theriogenology.2023.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/04/2023] [Accepted: 07/04/2023] [Indexed: 07/24/2023]
Abstract
To date, the biological functions of P4 within the canine placenta have been attributed to maternal stroma-derived decidual cells as the only placental cells expressing the nuclear P4 receptor (PGR). However, P4 can also exert its effects via membrane-bound receptors. To test the hypothesis that membrane-bound P4 receptors are involved in regulating placental function in the dog, the expression of mPRα, -β, -γ, PGRMC1 and -2 was investigated in the uterine and placental compartments derived from different stages of pregnancy and from prepartum luteolysis. Further, to assess the PGR signaling-mediated effects upon membrane P4 receptors in canine decidual cells, in vitro decidualized dog uterine stromal (DUS) cells were treated with type II antigestagens (aglepristone or mifepristone). The expression of all membrane P4 receptors was detectable in reproductive tissues and in DUS cells. The main findings indicate their distinguishable placental spatio-temporal distribution; PGRMC2 was predominantly found in decidual cells, PGRMC1 was strong in maternal endothelial compartments, and syncytiotrophoblast showed abundant levels of mPRα and mPRβ. In vitro decidualization was associated with increased expression of PGRMC1 and -2, while their protein levels were diminished by antigestagen treatment. The involvement of membrane-bound P4 signaling in the regulation of canine placental function is implied, with P4 effects being directly exerted through maternal and fetal cellular compartments. The indirect effects of PGR might involve the modulation of membrane-bound receptors availability in decidual cells, implying a self-regulatory loop of P4 in regulating the availability of its own receptors in the canine placenta.
Collapse
Affiliation(s)
- Ali Kazemian
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.
| | - Miguel Tavares Pereira
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.
| | - Selim Aslan
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Near East University, Nicosia, Cyprus.
| | - Rita Payan-Carreira
- School of Science and Technology, Department of Veterinary Medicine, University of Évora, Évora, Portugal.
| | - Iris M Reichler
- Clinic of Reproductive Medicine, Vetsuisse Faculty, University of Zurich (UZH), Zurich, Switzerland.
| | - Reha A Agaoglu
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Turkey.
| | - Mariusz P Kowalewski
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland; Center for Clinical Studies (ZKS), Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
9
|
Çinar M, Tokmak A, Kuru Pekcan M, Sarsu M, Üstün Y, Özakşit G. Does mid-luteal progesterone predict pregnancy in intrauterine insemination cycles following sequential clomiphene citrate and gonadotropin treatment? Medicine (Baltimore) 2023; 102:e34754. [PMID: 37657005 PMCID: PMC10476853 DOI: 10.1097/md.0000000000034754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/24/2023] [Indexed: 09/03/2023] Open
Abstract
This study aimed to determine whether serum mid-luteal progesterone (MLP) levels measured in the current treatment cycles of infertile women undergoing controlled ovarian hyperstimulation and intrauterine insemination following the sequential use of clomiphene citrate and gonadotropin may predict pregnancy. A total of 107 consecutive anovulatory women were included in this prospective cohort study. Patients with other causes of infertility were also excluded from the study. None of the patients received progesterone treatment for luteal phase support. The data recorded for each woman included age, body mass index, infertility type and duration, basal hormone levels, and previous and current cycle characteristics with MLP levels. Ovulation was confirmed using MLP and sonographic evaluation in all patients. An MLP level of > 3 ng/mL was regarded as a sign of ovulation. After treatment, the patients were divided into 2 groups according to the presence or absence of pregnancy, and the obtained data were compared between the groups. There were no significant differences in age, body mass index, or basal hormone levels between the 2 groups (all P > .05). However, the duration of infertility was significantly shorter in the pregnancy group (P = .003). The anovulation rate in this cohort was 18.7% (n = 20). A total of 15 (14%) were examined. MLP levels were 25.1 ± 13.8 ng/mL and 18.3 ± 14.5 ng/mL in the pregnant and nonpregnant groups, respectively (P:.089). Based on the receiver operating characteristic curve analysis, it was determined that there was no predictive value of the mid-luteal phase progesterone level for pregnancy in patients in whom ovulation was detected. Mid-luteal serum progesterone levels did not predict pregnancy in infertile women who underwent controlled ovarian hyperstimulation with sequential clomiphene citrate plus gonadotropin treatment and intrauterine insemination.
Collapse
Affiliation(s)
- Mehmet Çinar
- Zekai Tahir Burak Women’s Health Research and Education Hospital, Obstetrics and Gynecology, Ankara, Turkey
| | - Aytekin Tokmak
- Zekai Tahir Burak Women’s Health Research and Education Hospital, Obstetrics and Gynecology, Ankara, Turkey
| | - Meryem Kuru Pekcan
- Zekai Tahir Burak Women’s Health Research and Education Hospital, Obstetrics and Gynecology, Ankara, Turkey
| | - Mustafa Sarsu
- Zekai Tahir Burak Women’s Health Research and Education Hospital, Obstetrics and Gynecology, Ankara, Turkey
| | - Yaprak Üstün
- Zekai Tahir Burak Women’s Health Research and Education Hospital, Obstetrics and Gynecology, Ankara, Turkey
| | - Gülnur Özakşit
- Zekai Tahir Burak Women’s Health Research and Education Hospital, Obstetrics and Gynecology, Ankara, Turkey
| |
Collapse
|
10
|
Earhart AP, Karasseva NG, Storey KM, Olthoff B, Sarker MB, Laffey KG, Lange MJ, Rector RS, Schulz LC, Gil D, Neuhauser CM, Schrum AG. Lower female survival from an opportunistic infection reveals progesterone-driven sex bias in trained immunity. Cell Rep 2023; 42:113007. [PMID: 37590139 PMCID: PMC10528383 DOI: 10.1016/j.celrep.2023.113007] [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: 01/19/2023] [Revised: 06/08/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023] Open
Abstract
Immune responses differ between females and males, although such sex-based variance is incompletely understood. Observing that bacteremia of the opportunistic pathogen Burkholderia gladioli caused many more deaths of female than male mice bearing genetic deficiencies in adaptive immunity, we determined that this was associated with sex bias in the innate immune memory response called trained immunity. Female attenuation of trained immunity varies with estrous cycle stage and correlates with serum progesterone, a hormone that decreases glycolytic capacity and recall cytokine secretion induced by antigen non-specific stimuli. Progesterone receptor antagonism rescues female trained immune responses and survival from controlled B. gladioli infection to magnitudes similar to those of males. These data demonstrate progesterone-dependent sex bias in trained immunity where attenuation of female responses is associated with survival outcomes from opportunistic infection.
Collapse
Affiliation(s)
- Alexander P Earhart
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Natalia G Karasseva
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Kathryn M Storey
- Division of Biological Sciences, College of Arts & Science, University of Missouri, Columbia, MO 65212, USA
| | - Benjamin Olthoff
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Md Bodruzzaman Sarker
- Division of Animal Sciences, College of Agriculture, Food & Natural Resources, University of Missouri, Columbia, MO 65212, USA
| | - Kimberly G Laffey
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Margaret J Lange
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - R Scott Rector
- Department of Nutrition and Exercise Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, USA; Department of Medicine, School of Medicine, University of Missouri, Columbia, MO 65212, USA; Research Service, Harry S. Truman Memorial VA Hospital, University of Missouri, Columbia, MO 65212, USA; NextGen Precision Health Institute, University of Missouri, Columbia, MO 65212, USA
| | - Laura C Schulz
- Department of Obstetrics, Gynecology, & Women's Health, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Diana Gil
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA; NextGen Precision Health Institute, University of Missouri, Columbia, MO 65212, USA; Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65212, USA; Department of Chemical & Biomedical Engineering, College of Engineering, University of Missouri, Columbia, MO 65212, USA
| | - Claudia M Neuhauser
- Division of Research, Department of Mathematics, University of Houston, Houston, TX 77204, USA
| | - Adam G Schrum
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA; Division of Animal Sciences, College of Agriculture, Food & Natural Resources, University of Missouri, Columbia, MO 65212, USA; NextGen Precision Health Institute, University of Missouri, Columbia, MO 65212, USA; Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65212, USA; Department of Chemical & Biomedical Engineering, College of Engineering, University of Missouri, Columbia, MO 65212, USA.
| |
Collapse
|
11
|
Stevens Brentjens LBPM, Obukhova D, den Hartog JE, Delvoux B, Koskivuori J, Auriola S, Häkkinen MR, Bui BN, van Hoogenhuijze NE, Mackens S, Mol F, de Bruin JP, Besselink D, Teklenburg G, Kukushkina V, Salumets A, Broekmans FJM, van Golde RJT, Esteki MZ, Romano A. An integrative analysis of endometrial steroid metabolism and transcriptome in relation to endometrial receptivity in in vitro fertilization patients. F&S SCIENCE 2023; 4:219-228. [PMID: 37142054 DOI: 10.1016/j.xfss.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/27/2023] [Accepted: 04/24/2023] [Indexed: 05/06/2023]
Abstract
OBJECTIVE To study the relationship between the steroid concentration in the endometrium, in serum, and the gene expression level of steroid-metabolizing enzymes in the context of endometrial receptivity in in vitro fertilization (IVF) patients. DESIGN Case-control study of 40 IVF patients recruited in the SCRaTCH study (NTR5342), a randomized controlled trial investigating pregnancy outcome after "endometrial scratching." Endometrial biopsies and serum were obtained from patients with a first failed IVF cycle randomized to the endometrial scratch in the midluteal phase of the natural cycle before the next fresh embryo transfer during the second IVF cycle. SETTING University hopsital. PATIENTS Twenty women with clinical pregnancy were compared with 20 women who did not conceive after fresh embryo transfer. Cases and controls were matched for primary vs. secondary infertility, embryo quality, and age. INTERVENTION None. MAIN OUTCOME MEASURE(S) Steroid concentrations in endometrial tissue homogenates and serum were measured with liquid chromatography-mass spectrometry. The endometrial transcriptome was profiled by RNA-sequencing, followed by principal component analysis and differential expression analysis. False discovery rate-adjusted and log-fold change >|0.5| were selected as the threshold for differentially expressed genes. RESULT(S) Estrogen levels were comparable in both serum (n = 16) and endometrium (n = 40). Androgens and 17-hydroxyprogesterone were higher in serum than that in endometrium. Although steroid levels did not vary between pregnant and nonpregnant groups, subgroup analysis of primary women with infertility showed a significantly lower estrone concentration and estrone:androstenedione ratio in serum of the pregnant group (n = 5) compared with the nonpregnant group (n = 2). Expression of 34 out of 46 genes encoding the enzymes controlling the local steroid metabolism was detected, and estrogen receptor β gene was differentially expressed between pregnant and nonpregnant women. When only the primary infertile group was considered, 28 genes were differentially expressed between pregnant and nonpregnant women, including HSD11B2, that catalyzes the conversion of cortisol into cortisone. CONCLUSION(S) Steroidomic and transcriptomic analyses show that steroid concentrations are regulated by the local metabolism in the endometrium. Although no differences were found in endometrial steroid concentration in the pregnant and nonpregnant IVF patients, primary women with infertility showed deviations in steroid levels and gene expression, indicating that a more homogeneous patient group is required to uncover the exact role of steroid metabolism in endometrial receptivity. CLINICAL TRIAL REGISTRATION NUMBER The study was registered in the Dutch trial registry (www.trialregister.nl), registration number NL5193/NTR5342, available at https://trialsearch.who.int/Trial2.aspx?TrialID=NTR6687. The date of registration is July 31, 2015. The first enrollment is on January 1, 2016.
Collapse
Affiliation(s)
- Linda B P M Stevens Brentjens
- Department of Obstetrics and Gynecology, Maastricht University Medical Center+, Maastricht, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands.
| | - Darina Obukhova
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands; Department of Clinical Genetics, Maastricht University, Maastricht, the Netherlands
| | - Janneke E den Hartog
- Department of Obstetrics and Gynecology, Maastricht University Medical Center+, Maastricht, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Bert Delvoux
- Department of Obstetrics and Gynecology, Maastricht University Medical Center+, Maastricht, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | | | - Seppo Auriola
- University of Eastern Finland, School of Pharmacy, Kuopio, Finland
| | - Merja R Häkkinen
- Department of Health Security, Finnish Institute for Health and Welfare (THL), Kuopio, Finland
| | - Bich N Bui
- Department of Gynecology and Reproductive Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Nienke E van Hoogenhuijze
- Department of Gynecology and Reproductive Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Shari Mackens
- Center for Reproductive Medicine, UZ Brussel, Jette, Belgium
| | - Femke Mol
- Center for Reproductive Medicine, Reproduction and Development, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Jan Peter de Bruin
- Department of Obstetrics and Gynecology, Jeroen Bosch Hospital-Hertogenbosch, the Netherlands
| | - Dagmar Besselink
- Department of Obstetrics and Gynecology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Gijs Teklenburg
- Isala Fertility Clinic, Isala Hospital, Zwolle, the Netherlands
| | | | - Andres Salumets
- Competence Centre on Health Technologies, Tartu, Estonia; Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia; Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Frank J M Broekmans
- Department of Gynecology and Reproductive Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Ron J T van Golde
- Department of Obstetrics and Gynecology, Maastricht University Medical Center+, Maastricht, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Masoud Zamani Esteki
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands; Department of Clinical Genetics, Maastricht University, Maastricht, the Netherlands
| | - Andrea Romano
- Department of Obstetrics and Gynecology, Maastricht University Medical Center+, Maastricht, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
12
|
Liang C, Chung HF, Dobson AJ, Cade JE, Greenwood DC, Hayashi K, Hardy R, Kuh D, Schouw YTVD, Sandin S, Weiderpass E, Mishra GD. Is there a link between infertility, miscarriage, stillbirth, and premature or early menopause? Results from pooled analyses of 9 cohort studies. Am J Obstet Gynecol 2023; 229:47.e1-47.e9. [PMID: 37059411 DOI: 10.1016/j.ajog.2023.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/14/2023] [Accepted: 04/02/2023] [Indexed: 04/16/2023]
Abstract
BACKGROUND Some reproductive factors (such as age at menarche and parity) have been shown to be associated with age at natural menopause, but there has been little quantitative analysis of the association between infertility, miscarriage, stillbirth, and premature (<40 years) or early menopause (40-44 years). In addition, it has been unknown whether the association differs between Asian and non-Asian women, although the age at natural menopause is younger among Asian women. OBJECTIVE This study aimed to investigate the association of infertility, miscarriage, and stillbirth with age at natural menopause, and whether the association differed by race (Asian and non-Asian). STUDY DESIGN This was a pooled individual participant data analysis from 9 observational studies contributing to the InterLACE consortium. Naturally postmenopausal women with data on at least 1 of the reproductive factors (ie, infertility, miscarriage, and stillbirth), age at menopause, and confounders (ie, race, education level, age at menarche, body mass index, and smoking status) were included. A multinomial logistic regression model was used to estimate relative risk ratios and 95% confidence intervals for the association of infertility, miscarriage, and stillbirth with premature or early menopause, adjusting for confounders. Between-study difference and within-study correlation were taken into account by including study as a fixed effect and indicating study as a cluster variable. We also examined the association with number of miscarriages (0, 1, 2, ≥3) and stillbirths (0, 1, ≥2), and tested whether the strength of association differed between Asian and non-Asian women. RESULTS A total of 303,594 postmenopausal women were included. Their median age at natural menopause was 50.0 years (interquartile range, 47.0-52.0). The percentages of women with premature and early menopause were 2.1% and 8.4%, respectively. The relative risk ratios (95% confidence intervals) of premature and early menopause were 2.72 (1.77-4.17) and 1.42 (1.15-1.74) for women with infertility; 1.31 (1.08-1.59) and 1.37 (1.14-1.65) for women with recurrent miscarriages; and 1.54 (1.52-1.56) and 1.39 (1.35-1.43) for women with recurrent stillbirths. Asian women with infertility, recurrent miscarriages (≥3), or recurrent stillbirths (≥2) had higher risk of premature and early menopause compared with non-Asian women with the same reproductive history. CONCLUSION Histories of infertility and recurrent miscarriages and stillbirths were associated with higher risk of premature and early menopause, and the associations differed by race, with stronger associations for Asian women with such reproductive history.
Collapse
|
13
|
Oltman J, Balachander S, Mol BW, Woolner AMF. Have we overlooked the role of mifepristone for the medical management of tubal ectopic pregnancy? Hum Reprod 2023:7193344. [PMID: 37295950 PMCID: PMC10391312 DOI: 10.1093/humrep/dead116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/12/2023] [Indexed: 06/12/2023] Open
Abstract
Ectopic pregnancy is a risk of both spontaneous and assisted reproduction pregnancies. The majority of ectopic pregnancies abnormally implant within a fallopian tube (extrauterine pregnancies). In haemodynamically stable women, medical or expectant treatment can be offered. Currently accepted medical treatment is using a drug called methotrexate. However, methotrexate has potential adverse effects, and a significant proportion of women will still require emergency surgery (up to 30%) to remove the ectopic pregnancy. Mifepristone (RU-486) has anti-progesterone effects and has a role in managing intrauterine pregnancy loss and termination of pregnancy. On reviewing the literature and given progesterone's pivotal role in sustaining pregnancy, we propose that we may have overlooked the role of mifepristone in the medical management of tubal ectopic pregnancy in haemodynamically stable women.
Collapse
Affiliation(s)
- Julia Oltman
- Aberdeen Centre for Women's Health Research, Institute of Applied Health Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Sanjana Balachander
- School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Ben W Mol
- Aberdeen Centre for Women's Health Research, Institute of Applied Health Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
- Monash Medical Centre Clayton, Obstetrics and Gynaecology, Monash Medical Centre, Clayton, Victoria, Australia
- Monash University, Clayton, Victoria, Australia
| | - Andrea M F Woolner
- Aberdeen Centre for Women's Health Research, Institute of Applied Health Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
14
|
Dingle K, Kassem OM, Azizieh F, AbdulHussain G, Raghupathy R. Quantitative analyses of cytokine profiles reveal hormone-mediated modulation of cytokine profiles in recurrent spontaneous miscarriage. Cytokine 2023; 164:156160. [PMID: 36804258 DOI: 10.1016/j.cyto.2023.156160] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/21/2023]
Abstract
PURPOSE Cytokines play important roles in pregnancy complications. Some hormones such as estrogen, progesterone, and dydrogesterone have been shown to alter cytokine profiles. Understanding how cytokine profiles are affected by these hormones is therefore an important step towards immunomodulatory therapies for pregnancy complications. We analyse previously published data on the effects of estrogen, progesterone, and dydrogesterone on cytokine balances in women having recurrent spontaneous miscarriages. MATERIALS AND METHODS Levels of eight cytokines (IFN-γ, IL-2, IL-6, IL-10, IL-13, IL-17, IL-23, TNF-α) from n = 22 women presenting unexplained recurrent spontaneous miscarriages were studied. Cytokine values were recorded after in vitro exposure of peripheral blood cells to estrogen, progesterone, and dydrogesterone. We expand on earlier analysis of the dataset by employing different statistical techniques including effect sizes for individual cytokine values, a more powerful statistical test, and adjusting p-values for multiple comparisons. We employ multivariate analysis methods, including to determine the relative magnitude of the effects of the hormone therapies on cytokines. A new statistical method is introduced based on pairwise distances able to accommodate complex relations in cytokine profiles. RESULTS We report several statistically significant differences in individual cytokine values between the control group and each hormone treated group, with estrogen affecting the fewest cytokines, and progesterone and dydrogesterone both affecting seven out of eight cytokines. Exposure to estrogen produces no large effects sizes however, while IFN-γ and IL-17 show large effect sizes for both progesterone and dydrogesterone, among other cytokines. Our new method for identifying which collections (i.e. subsets) of cytokines best distinguish contrasting groups identifies IFN-γ, IL-10 and IL-23 as especially noteworthy for both progesterone and dydrogesterone treatments. CONCLUSIONS While some statistically significant differences in cytokine levels after exposure to estrogen are found, these have small effect sizes and are unlikely to be clinically relevant. Progesterone and dydrogesterone both induce statistically significant and large effect-size differences in cytokine levels, hence therapy with these two progestogens is more likely to be clinically relevant. Univariate and multivariate methods for identifying cytokine importances provide insight into which groups of cytokines are most affected and in what ways by therapies.
Collapse
Affiliation(s)
- Kamaludin Dingle
- Centre for Applied Mathematics and Bioinformatics, Department of Mathematics and Natural Sciences, Gulf University for Science and Technology, Hawally, Kuwait; Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, CA, USA.
| | - Osama M Kassem
- Centre for Applied Mathematics and Bioinformatics, Department of Mathematics and Natural Sciences, Gulf University for Science and Technology, Hawally, Kuwait
| | - Fawaz Azizieh
- Centre for Applied Mathematics and Bioinformatics, Department of Mathematics and Natural Sciences, Gulf University for Science and Technology, Hawally, Kuwait
| | | | - Raj Raghupathy
- Department of Microbiology, Faculty of Medicine, Kuwait University, Kuwait
| |
Collapse
|
15
|
Chromogranin A: An Endocrine Factor of Pregnancy. Int J Mol Sci 2023; 24:ijms24054986. [PMID: 36902417 PMCID: PMC10002927 DOI: 10.3390/ijms24054986] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/26/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Pregnancy is a state of physiological and hormonal changes. One of the endocrine factors involved in these processes is chromogranin A, an acidic protein produced, among others, by the placenta. Although it has been previously linked to pregnancy, no existing articles have ever managed to clarify the role of this protein regarding this subject. Therefore, the aim of the present study is to gather knowledge of chromogranin A's function with reference to gestation and parturition, clarify elusive information, and, most importantly, to formulate hypotheses for the future studies to verify.
Collapse
|
16
|
Meyyazhagan A, Kuchi Bhotla H, Pappuswamy M, Tsibizova V, Al Qasem M, Di Renzo GC. Cytokine see-saw across pregnancy, its related complexities and consequences. Int J Gynaecol Obstet 2023; 160:516-525. [PMID: 35810391 DOI: 10.1002/ijgo.14333] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/17/2022] [Accepted: 07/07/2022] [Indexed: 01/20/2023]
Abstract
During pregnancy, a woman's immune system adapts to the changing hormonal concentrations, causing immunologic transition. These immunologic changes are required for a full-term pregnancy, preserving the fetus' innate and adaptive immunity. Preterm labor, miscarriage, gestational diabetes mellitus, and pre-eclampsia are all caused by abnormal cytokine expression during pregnancy and childbirth. A disruption in the cytokine balance can lead to autoimmune diseases or microbiologic infections, or to autoimmune illness remission during pregnancy with postpartum recurrence. The cytokine treatments are essential and damaging to the developing fetus. The current review summarizes the known research on cytokine changes during pregnancy and their possible consequences for pregnant women. Studies suggest that customizing medication for each woman and her progesterone levels should be based on the cytokine profile of each pregnant woman. Immune cells and chemicals play an important function in development of the placenta and embryo. During pregnancy, T cells divide and move, and a careful balance between proinflammatory and anti-inflammatory cytokines is necessary. The present review focuses on the mother's endurance in generating fetal cells and the immunologic mechanism involved.
Collapse
Affiliation(s)
- Arun Meyyazhagan
- Center for Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy.,PREIS International and European School of Perinatal, Neonatal and Reproductive Medicine, Florence, Italy
| | - Haripriya Kuchi Bhotla
- Department of Life Sciences, CHRIST (Deemed to be University), Bengaluru, Karnataka, India
| | - Manikantan Pappuswamy
- Department of Life Sciences, CHRIST (Deemed to be University), Bengaluru, Karnataka, India
| | - Valentina Tsibizova
- PREIS International and European School of Perinatal, Neonatal and Reproductive Medicine, Florence, Italy.,Institute of Perinatology and Pediatrics, Almazov National Medical Research Center, Saint-Petersburg, Russia
| | - Malek Al Qasem
- PREIS International and European School of Perinatal, Neonatal and Reproductive Medicine, Florence, Italy.,Department of Obstetrics and Gynecology, Faculty of Medicine, Mutah University, Al-Karak, Jordan
| | - Gian Carlo Di Renzo
- Center for Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy.,PREIS International and European School of Perinatal, Neonatal and Reproductive Medicine, Florence, Italy.,Department of Obstetrics and Gynecology, I.M. Sechenov First State University of Moscow, Moscow, Russia
| |
Collapse
|
17
|
Alawadhi M, Kilarkaje N, Mouihate A, Al-Bader MD. Role of progesterone on dexamethasone-induced alterations in placental vascularization and progesterone receptors in rats†. Biol Reprod 2023; 108:133-149. [PMID: 36322157 DOI: 10.1093/biolre/ioac192] [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: 04/27/2022] [Revised: 07/04/2022] [Accepted: 10/18/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Intrauterine growth restriction (IUGR) is manifested by lower maternal progesterone levels, smaller placental size, and decreased placental vascularity indicated by lower expression of vascular endothelial growth factor (VEGF). Studies showed that progesterone increases angiogenesis and induces VEGF expression in different tissues. Therefore, the aim of the present study is to evaluate the effect of progesterone on placental vascular bed and VEGF expression and the modulation of nuclear and membranous progesterone receptors (PR) in dexamethasone-induced rat IUGR model. METHODS Pregnant Sprague-Dawley rats were allocated into four groups and given intraperitoneal injections of either saline, dexamethasone, dexamethasone, and progesterone or progesterone. Injections started on gestation day (DG) 15 and lasted until the days of euthanization (19 and 21 DG). Enzyme-linked immunosorbent assay was used to evaluate plasma progesterone levels. Real-time PCR and western blotting were used to evaluate gene and protein expressions of VEGF, and PR in labyrinth and basal placental zones. Immunohistochemistry was used to locate VEGF and different PRs in placental cells. Immunofluorescence was used to monitor the expression of blood vessel marker (αSMA). RESULTS Dexamethasone decreased the vascular bed fraction and the expression of VEGF in both placental zones. Progesterone co-treatment with dexamethasone prevented this reduction. Nuclear and membrane PRs showed tissue-specific expression in different placental zones and responded differently to both dexamethasone and progesterone. CONCLUSIONS Progesterone treatment improves the outcomes in IUGR pregnancy. Progesterone alleviated DEX-induced IUGR probably by promoting placental VEGF and angiogenesis.
Collapse
Affiliation(s)
- Mariam Alawadhi
- Department of Physiology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Narayana Kilarkaje
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Abdeslam Mouihate
- Department of Physiology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Maie D Al-Bader
- Department of Physiology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
18
|
Liu H, Yu L, Ding Y, Peng M, Deng Y. Progesterone Enhances the Invasion of Trophoblast Cells by Activating PI3K/AKT Signaling Pathway to Prevent Preeclampsia. Cell Transplant 2023; 32:9636897221145682. [PMID: 36593749 PMCID: PMC9830574 DOI: 10.1177/09636897221145682] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
We aimed to explore whether the effect of progesterone on preeclampsia via the PI3K/AKT signaling pathway. First, we studied the role of progesterone in preeclampsia patients and HTR-8/Svneo cells by adding progesterone. Then PI3K inhibitor LY294002 was added. The effects of progesterone on preeclampsia were also studied in animals by constructing a preeclampsia rat model. CCK-8 and Transwell assay were applied to measure cell viability and invasion ability. ELISA was performed to measure progesterone, MMP-2, MMP-9, pro-inflammatory factors TNF-α, IL-1β, and anti-inflammatory factors IL-4, IL-10, and IL-13 levels. HE staining was used to detect the pathological changes in uterine spiral artery. Western blot was performed to detect Cyclin D1, PCNA, MMP-2, MMP-9, inflammatory factors TNF-α, IL-1β, IL-4, IL-10, IL-13, and PI3K/AKT signaling pathway related proteins AKT, p-AKT, PI3K, and p-PI3K expressions. Progesterone could reduce blood pressure and urine protein in pregnant women with preeclampsia. TNF-α and IL-1β levels were decreased, but IL-4, IL-10, IL-13, cyclin D1, and PCNA levels were increased in pregnant women with preeclampsia after using progesterone. After the use of progesterone, the symptoms of the PE model group were improved. Among them, the lumen of the placental uterine spiral artery was enlarged, and the fibrinoid necrosis of the uterine wall and acute atherosclerotic lesions were relieved. In addition, progesterone promoted HTR-8/Svneo cells proliferation and invasion. However, high expression of MMP-2, MMP-9, p-AKT, and p-PI3K in Normal and preeclampsia groups caused by progesterone was weakened after adding LY294002, indicating that progesterone could activate PI3K/AKT signaling pathway to regulate HTR-8/Svneo cells. Progesterone decreased urine protein and blood pressure of preeclampsia rats in a concentration-dependent manner. Moreover, progesterone activated the PI3K/AKT signaling pathway and inhibited the inflammatory response in preeclampsia rats.
Collapse
Affiliation(s)
- Hongyu Liu
- Department of Obstetrics and Gynaecology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ling Yu
- Department of Obstetrics and Gynaecology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yiling Ding
- Department of Obstetrics and Gynaecology, The Second Xiangya Hospital of Central South University, Changsha, China,Yiling Ding, Department of Obstetrics and Gynaecology, The Second Xiangya Hospital of Central South University, Renmin Middle Road 139, Changsha 410005, Hunan, China.
| | - Mei Peng
- Department of Obstetrics and Gynaecology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yali Deng
- Department of Obstetrics and Gynaecology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
19
|
Shi L, Meng F, Wang S, Hu Z, Li J, Tian F, Wang H, Zhu Y, Wang Y, Ge RS, Li H. Effects of phenolic compounds on 3β-hydroxysteroid dehydrogenase activity in human and rat placenta: Screening, mode of action, and docking analysis. J Steroid Biochem Mol Biol 2023; 225:106202. [PMID: 36241036 DOI: 10.1016/j.jsbmb.2022.106202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 11/07/2022]
Abstract
Human 3β-hydroxysteroid dehydrogenase type I (HSD3B1) and rat type IV (HSD3B4) in placentas catalyze the conversion of pregnenolone to progesterone, which plays a key role in maintaining pregnancy. Many phenolic compounds potentially inhibit HSD3B in placentas as endocrine disruptors. In this study, the effects of 16 phenolic compounds on the activity of human HSD3B1 and rat HSD3B4 were determined and the structure-activity relationship was compared. HSD3B1 in human placental microsomes and HSD3B4 in rat placental microsomes were used to measure their activities and pregnenolone and NAD+ were used as substrates. Of the 16 phenolic compounds, 4-nonylphenol, pentabromophenol, and 2-bromophenol resulted in residual human HSD3B1 activity lower than 50 % and 4-nonylphenol and pentabromophenol resulted in residual rat HSD3B4 activity lower than 50 %. 4-Nonylphenol, pentabromophenol, and 2-bromophenol were mixed inhibitors of human HSD3B1, with Ki values of 2.31, 3.58 and 4.86 µM, respectively, while 4-nonylphenol and pentabromophenol were noncompetitive inhibitors of rat HSD3B4 with Ki values of 20.86 and 141.8 µM. Molecular docking showed that 4-nonylphenol, pentabromophenol, and 2-bromophenol docked to the active sites of human HSD3B1 and rat HSD3B4, and the shift of residue S125 in human HSD3B1 to T125 in rat HSD3B4 could explain the species-dependent difference in their inhibitory potency and mode of action. This study demonstrates that 4-nonylphenol, pentabromophenol, and 2-bromophenol are mixed inhibitors of human placental HSD3B1, while 4-nonylphenol and pentabromophenol are noncompetitive inhibitors of rat HSD3B4, possibly blocking the placental steroidogenesis.
Collapse
Affiliation(s)
- Lei Shi
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Fangyan Meng
- Department of Anaesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Shaowei Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zhiyan Hu
- Department of Anaesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jingjing Li
- Department of Anaesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Fuhong Tian
- Department of Anaesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Haixing Wang
- Department of Anaesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yang Zhu
- Department of Anaesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yiyan Wang
- Department of Anaesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Ren-Shan Ge
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Department of Anaesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325000, Zhejiang Province, China.
| | - Huitao Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325000, Zhejiang Province, China.
| |
Collapse
|
20
|
Li WN, Dickson MJ, DeMayo FJ, Wu SP. The role of progesterone receptor isoforms in the myometrium. J Steroid Biochem Mol Biol 2022; 224:106160. [PMID: 35931328 PMCID: PMC9895129 DOI: 10.1016/j.jsbmb.2022.106160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/15/2022] [Accepted: 08/01/2022] [Indexed: 02/08/2023]
Abstract
Myometrial contraction is stringently controlled throughout pregnancy and parturition. Progesterone signaling, effecting through the progesterone receptor (PR), is pivotal in modulating uterine activity. Evidence has shown that two major PR isoforms, PR-A and PR-B, have distinct activities on gene regulation, and the ratio between these isoforms determines the contractility of the myometrium at different gestational stages. Herein, we focus on the regulation of PR activity in the myometrium, especially the differential actions of the two PR isoforms, which maintain uterine quiescence during pregnancy and regulate the switch to a contractile state at the onset of labor. To demonstrate the PR regulatory network and its mechanisms of actions on myometrial activity, we summarized the findings into three parts: Regulation of PR Expression and Isoform Levels, Progesterone Receptor Interacting Factors, and Biological Processes Regulated by Myometrial Progesterone Receptor Isoforms. Recent genomic and epigenomic data, from human specimens and mouse models, are recruited to support the existing knowledge and offer new insights and future directions in myometrial biology.
Collapse
Affiliation(s)
- Wan-Ning Li
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Mackenzie J Dickson
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Francesco J DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| | - San-Pin Wu
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
21
|
Low-Dose Tacrolimus Promotes the Migration and Invasion and Nitric Oxide Production in the Human-Derived First Trimester Extravillous Trophoblast Cells In Vitro. Int J Mol Sci 2022; 23:ijms23158426. [PMID: 35955565 PMCID: PMC9369346 DOI: 10.3390/ijms23158426] [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: 08/16/2021] [Revised: 07/12/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
Placentation is one of the most important determinants for a successful pregnancy, and this is dependent on the process of trophoblast migration and invasion. Progesterone receptors (PGR) are critical effectors of progesterone (P4) signaling that is required for trophoblast migration and invasion conducive to a successful gestation. In immune complicated pregnancies, evidence has shown that abnormal placentation occurs because of aberrant expression of PGR. Therapeutic intervention with tacrolimus (FK506) was able to restore PGR expression and improve pregnancy outcomes in immune-complicated gestations; however, the exact mode of action of tacrolimus in assisting placentation is not clear. Here, we attempt to uncover the mode of action of tacrolimus by examining its effects on trophoblast invasion and migration in the human-derived extravillous trophoblast (EVT) cell line, the HTR-8/SVneo cells. Using a variety of functional assays, we demonstrated that low-dose tacrolimus (10 ng/mL) was sufficient to significantly (p < 0.001) stimulate the migration and invasion of the HTR-8/SVneo cells, inducing their cytosolic/nuclear progesterone receptor expression and activation, and modulating their Nitric Oxide (NO) production. Moreover, tacrolimus abrogated the suppressive effect of the NOS inhibitor Nω- Nitro-L-Arginine Methyl Ester (L-NAME) on these vital processes critically involved in the establishment of human pregnancy. Collectively, our data suggest an immune-independent mode of action of tacrolimus in positively influencing placentation in complicated gestations, at least in part, through promoting the migration and invasion of the first trimester extravillous trophoblast cells by modulating their NO production and activating their cytosolic/nuclear progesterone-receptors. To our knowledge, this is the first report to show that the mode of action of tacrolimus as a monotherapy for implantation failure is plausibly PGR-dependent.
Collapse
|
22
|
Louwen F, Kreis NN, Ritter A, Friemel A, Solbach C, Yuan J. BCL6, a key oncogene, in the placenta, pre-eclampsia and endometriosis. Hum Reprod Update 2022; 28:890-909. [PMID: 35640966 PMCID: PMC9629482 DOI: 10.1093/humupd/dmac027] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/02/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The key oncogene B-cell lymphoma 6 (BCL6) drives malignant progression by promoting proliferation, overriding DNA damage checkpoints and blocking cell terminal differentiation. However, its functions in the placenta and the endometrium remain to be defined. OBJECTIVE AND RATIONALE Recent studies provide evidence that BCL6 may play various roles in the human placenta and the endometrium. Deregulated BCL6 might be related to the pathogenesis of pre-eclampsia (PE) as well as endometriosis. In this narrative review, we aimed to summarize the current knowledge regarding the pathophysiological role of BCL6 in these two reproductive organs, discuss related molecular mechanisms, and underline associated research perspectives. SEARCH METHODS We conducted a comprehensive literature search using PubMed for human, animal and cellular studies published until October 2021 in the following areas: BCL6 in the placenta, in PE and in endometriosis, in combination with its functions in proliferation, fusion, migration, invasion, differentiation, stem/progenitor cell maintenance and lineage commitment. OUTCOMES The data demonstrate that BCL6 is important in cell proliferation, survival, differentiation, migration and invasion of trophoblastic cells. BCL6 may have critical roles in stem/progenitor cell survival and differentiation in the placenta and the endometrium. BCL6 is aberrantly upregulated in pre-eclamptic placentas and endometriotic lesions through various mechanisms, including changes in gene transcription and mRNA translation as well as post-transcriptional/translational modifications. Importantly, increased endometrial BCL6 is considered to be a non-invasive diagnostic marker for endometriosis and a predictor for poor outcomes of IVF. These data highlight that BCL6 is crucial for placental development and endometrium homeostasis, and its upregulation is associated with the pathogenesis of PE, endometriosis and infertility. WIDER IMPLICATIONS The lesson learned from studies of the key oncogene BCL6 reinforces the notion that numerous signaling pathways and regulators are shared by tumors and reproductive organs. Their alteration may promote the progression of malignancies as well as the development of gestational and reproductive disorders.
Collapse
Affiliation(s)
- Frank Louwen
- Division of Obstetrics and Prenatal Medicine, Department of Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Nina-Naomi Kreis
- Division of Obstetrics and Prenatal Medicine, Department of Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Andreas Ritter
- Division of Obstetrics and Prenatal Medicine, Department of Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Alexandra Friemel
- Division of Obstetrics and Prenatal Medicine, Department of Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Christine Solbach
- Division of Obstetrics and Prenatal Medicine, Department of Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Juping Yuan
- Division of Obstetrics and Prenatal Medicine, Department of Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| |
Collapse
|
23
|
Aikaterini B, Sophia Z, Fanourios M, Panagiotis D, Timur G, Antonios M. Aging, a modulator of human endometrial stromal cell proliferation and decidualization. A role for implantation? Reprod Biomed Online 2022; 45:202-210. [DOI: 10.1016/j.rbmo.2022.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/24/2022] [Accepted: 03/30/2022] [Indexed: 01/09/2023]
|
24
|
Park JY, Mani S, Clair G, Olson HM, Paurus VL, Ansong CK, Blundell C, Young R, Kanter J, Gordon S, Yi AY, Mainigi M, Huh DD. A microphysiological model of human trophoblast invasion during implantation. Nat Commun 2022; 13:1252. [PMID: 35292627 PMCID: PMC8924260 DOI: 10.1038/s41467-022-28663-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 02/01/2022] [Indexed: 12/12/2022] Open
Abstract
Successful establishment of pregnancy requires adhesion of an embryo to the endometrium and subsequent invasion into the maternal tissue. Abnormalities in this critical process of implantation and placentation lead to many pregnancy complications. Here we present a microenigneered system to model a complex sequence of orchestrated multicellular events that plays an essential role in early pregnancy. Our implantation-on-a-chip is capable of reconstructing the three-dimensional structural organization of the maternal-fetal interface to model the invasion of specialized fetal extravillous trophoblasts into the maternal uterus. Using primary human cells isolated from clinical specimens, we demonstrate in vivo-like directional migration of extravillous trophoblasts towards a microengineered maternal vessel and their interactions with the endothelium necessary for vascular remodeling. Through parametric variation of the cellular microenvironment and proteomic analysis of microengineered tissues, we show the important role of decidualized stromal cells as a regulator of extravillous trophoblast migration. Furthermore, our study reveals previously unknown effects of pre-implantation maternal immune cells on extravillous trophoblast invasion. This work represents a significant advance in our ability to model early human pregnancy, and may enable the development of advanced in vitro platforms for basic and clinical research of human reproduction.
Collapse
Affiliation(s)
- Ju Young Park
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Sneha Mani
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Geremy Clair
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Heather M Olson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Vanessa L Paurus
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Charles K Ansong
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Cassidy Blundell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel Young
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica Kanter
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Scott Gordon
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alex Y Yi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Monica Mainigi
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Dan Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
25
|
The effect of progesterone administration on the expression of metastasis tumor antigens (MTA1 and MTA3) in placentas of normal and dexamethasone-treated rats. Mol Biol Rep 2022; 49:1935-1943. [PMID: 35037193 DOI: 10.1007/s11033-021-07005-5] [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: 05/05/2021] [Accepted: 11/23/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Dexamethasone (DEX) induces intrauterine growth restriction (IUGR) in pregnant rats. IUGR can occur due to apoptosis of trophoblasts, which is believed to be inhibited by progesterone (P4). A group of genes called MTAs play a role in proliferation and apoptosis. MTA1 upregulates trophoblasts proliferation and differentiation, while MTA3 downregulates proliferation and induces apoptosis. Hence, we hypothesized that during IUGR, placental MTA1 decreases and MTA3 increases and this is reversed by P4 treatment. METHODS Pregnant Sprague-Dawley rats were divided into 4 groups based on daily intraperitoneal injections: control (C, saline), DEX (DEX, 0.2 mg/kg/day), DEX and P4 (DEX + P4, DEX: 0.2 mg/kg/day, P4: 5 mg/kg/day) and P4-treated (P4, 5 mg/kg/day) groups. Injections were started on 15 dg until the day of dissection (19 or 21 dg). Gene and protein expressions of MTA1 and MTA3 were studied in the labyrinth (LZ) and basal (BZ) zones using real-time PCR and Western blotting, respectively. RESULTS DEX treatment induced 18% reduction in fetal body weight (p < 0.001) and 30% reduction in placental weight (p < 0.01). Maternal P4 level was also significantly lower in DEX treated groups (p < 0.05). MTA1 expression was decreased in the LZ (gene, p < 0.001) and BZ (protein p < 0.01), while MTA3 protein expression was upregulated in the LZ with DEX treatment (p < 0.001). These changes were reversed with P4 treatment. CONCLUSION The findings of the present study indicate that DEX induces IUGR through changing the expression of placental MTA1 and MTA3 antigens and P4 improved pregnancy outcome by preventing the changes in MTAs expression.
Collapse
|
26
|
Nagy B, Szekeres-Barthó J, Kovács GL, Sulyok E, Farkas B, Várnagy Á, Vértes V, Kovács K, Bódis J. Key to Life: Physiological Role and Clinical Implications of Progesterone. Int J Mol Sci 2021; 22:11039. [PMID: 34681696 PMCID: PMC8538505 DOI: 10.3390/ijms222011039] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/10/2021] [Accepted: 10/11/2021] [Indexed: 02/06/2023] Open
Abstract
The most recent studies of progesterone research provide remarkable insights into the physiological role and clinical importance of this hormone. Although the name progesterone itself means "promoting gestation", this steroid hormone is far more than a gestational agent. Progesterone is recognized as a key physiological component of not only the menstrual cycle and pregnancy but also as an essential steroidogenic precursor of other gonadal and non-gonadal hormones such as aldosterone, cortisol, estradiol, and testosterone. Based on current findings, progesterone and novel progesterone-based drugs have many important functions, including contraception, treatment of dysfunctional uterine bleeding, immune response, and prevention of cancer. Considering the above, reproduction and life are not possible without progesterone; thus, a better understanding of this essential molecule could enable safe and effective use of this hormone in many clinical conditions.
Collapse
Affiliation(s)
- Bernadett Nagy
- National Laboratory for Human Reproduction, University of Pécs, 7624 Pécs, Hungary; (J.S.-B.); (G.L.K.); (E.S.); (B.F.); (Á.V.); (V.V.); (K.K.); (J.B.)
- Department of Obstetrics and Gynecology, Medical School, University of Pécs, 7624 Pécs, Hungary
- MTA-PTE Human Reproduction Scientific Research Group, University of Pécs, 7624 Pécs, Hungary
| | - Júlia Szekeres-Barthó
- National Laboratory for Human Reproduction, University of Pécs, 7624 Pécs, Hungary; (J.S.-B.); (G.L.K.); (E.S.); (B.F.); (Á.V.); (V.V.); (K.K.); (J.B.)
- MTA-PTE Human Reproduction Scientific Research Group, University of Pécs, 7624 Pécs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pécs, Hungary
- Department of Medical Biology, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Gábor L. Kovács
- National Laboratory for Human Reproduction, University of Pécs, 7624 Pécs, Hungary; (J.S.-B.); (G.L.K.); (E.S.); (B.F.); (Á.V.); (V.V.); (K.K.); (J.B.)
- Szentágothai Research Center, University of Pécs, 7624 Pécs, Hungary
- Department of Laboratory Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Endre Sulyok
- National Laboratory for Human Reproduction, University of Pécs, 7624 Pécs, Hungary; (J.S.-B.); (G.L.K.); (E.S.); (B.F.); (Á.V.); (V.V.); (K.K.); (J.B.)
- Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pécs, 7624 Pécs, Hungary
| | - Bálint Farkas
- National Laboratory for Human Reproduction, University of Pécs, 7624 Pécs, Hungary; (J.S.-B.); (G.L.K.); (E.S.); (B.F.); (Á.V.); (V.V.); (K.K.); (J.B.)
- Department of Obstetrics and Gynecology, Medical School, University of Pécs, 7624 Pécs, Hungary
- MTA-PTE Human Reproduction Scientific Research Group, University of Pécs, 7624 Pécs, Hungary
| | - Ákos Várnagy
- National Laboratory for Human Reproduction, University of Pécs, 7624 Pécs, Hungary; (J.S.-B.); (G.L.K.); (E.S.); (B.F.); (Á.V.); (V.V.); (K.K.); (J.B.)
- Department of Obstetrics and Gynecology, Medical School, University of Pécs, 7624 Pécs, Hungary
- MTA-PTE Human Reproduction Scientific Research Group, University of Pécs, 7624 Pécs, Hungary
| | - Viola Vértes
- National Laboratory for Human Reproduction, University of Pécs, 7624 Pécs, Hungary; (J.S.-B.); (G.L.K.); (E.S.); (B.F.); (Á.V.); (V.V.); (K.K.); (J.B.)
- Department of Obstetrics and Gynecology, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Kálmán Kovács
- National Laboratory for Human Reproduction, University of Pécs, 7624 Pécs, Hungary; (J.S.-B.); (G.L.K.); (E.S.); (B.F.); (Á.V.); (V.V.); (K.K.); (J.B.)
- Department of Obstetrics and Gynecology, Medical School, University of Pécs, 7624 Pécs, Hungary
- MTA-PTE Human Reproduction Scientific Research Group, University of Pécs, 7624 Pécs, Hungary
| | - József Bódis
- National Laboratory for Human Reproduction, University of Pécs, 7624 Pécs, Hungary; (J.S.-B.); (G.L.K.); (E.S.); (B.F.); (Á.V.); (V.V.); (K.K.); (J.B.)
- Department of Obstetrics and Gynecology, Medical School, University of Pécs, 7624 Pécs, Hungary
- MTA-PTE Human Reproduction Scientific Research Group, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
27
|
Labarta E, Mariani G, Rodríguez-Varela C, Bosch E. Individualized luteal phase support normalizes live birth rate in women with low progesterone levels on the day of embryo transfer in artificial endometrial preparation cycles. Fertil Steril 2021; 117:96-103. [PMID: 34548167 DOI: 10.1016/j.fertnstert.2021.08.040] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To analyze the impact on live birth rates (LBRs) of the individualized luteal phase support (termed iLPS) in patients with low serum progesterone (P) levels compared with patients without iLPS. DESIGN Retrospective cohort study, December 1, 2018, to May 30, 2019. SETTING Private medical center. PATIENT(S) A total of 2,275 patients checked for serum P on the day of blastocyst transfer were analyzed. During the study period, 1,299 patients showed serum P levels of ≥9.2 ng/mL, whereas 550 showed serum P levels of <9.2 ng/mL and received iLPS. Additionally, a historical group of 426 patients with serum P levels of <9.2 ng/mL but no iLPS were used for comparison. Eligible patients were aged ≤50 years with adequate endometrium morphology after receiving estrogens. Luteal phase support was provided with micronized vaginal P (MVP) to all women. Patients with personalized initiation of exogenous P according to the endometrial receptivity assay test, polyps, fibroids distorting the cavity, or hydrosalpinx were not included in the analysis. INTERVENTION(S) As routine practice since December 2018, patients with low serum P levels received an iLPS with a daily injection of 25 mg of subcutaneous P from the day of embryo transfer (ET) in addition to standard LPS (400 mg of MVP twice a day). MAIN OUTCOME MEASURE(S) Live birth rate. RESULT(S) The LBR was 44.9% in the iLPS cases vs. 45.0% in patients with normal serum P levels (crude odds ratio [OR], 1.0; 95% confidence interval [CI], 0.82-1.22). By regression analysis, low serum P levels did not affect the LBR after adjusting for possible confounders (age, oocyte origin, fresh vs. frozen, day of ET, embryo quality, number of embryos transferred) (adjusted OR, 0.99; 95% CI, 0.79-1.25). Similarly, no differences were observed in other pregnancy outcomes between groups. The LBR was significantly higher in the group of patients who received additional subcutaneous P (iLPS) compared with the historical group with low serum P levels and no iLPS (44.9% vs. 37.3%; OR, 1.37; 95% CI, 1.06-1.78). In the overall population, patients showing P levels of <9.2 ng/mL on the day of ET were slightly younger and had higher body mass index and lower estradiol and P levels during the proliferative phase compared with patients with P levels of ≥9.2 ng/mL. No differences were observed with regard to the time in between the last dose of MVP and the serum P determination. After a multivariable logistic regression analysis, only body mass index and estradiol levels in the proliferative phase reminded statistically significant. Significant differences in the LBR were observed between patients with serum P levels of <9.2 ng/mL without iLPS and patients with serum P levels of ≥9.2 ng/mL when using either own or donated oocytes. CONCLUSION(S) Individualized LPS for patients with low serum P levels produces LBRs similar to those of patients with adequate serum P levels.
Collapse
Affiliation(s)
- Elena Labarta
- IVIRMA Valencia, Valencia, Spain; IVI Foundation, Valencia, Spain.
| | - Giulia Mariani
- IVIRMA Valencia, Valencia, Spain; IVIRMA Roma, Roma, Italy
| | | | - Ernesto Bosch
- IVIRMA Valencia, Valencia, Spain; IVI Foundation, Valencia, Spain
| |
Collapse
|
28
|
Sugianto NA, Heistermann M, Newman C, Macdonald DW, Buesching CD. Alternative reproductive strategies provide a flexible mechanism for assuring mating success in the European badgers (Meles meles): An investigation from hormonal measures. Gen Comp Endocrinol 2021; 310:113823. [PMID: 34044013 DOI: 10.1016/j.ygcen.2021.113823] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/19/2021] [Accepted: 05/21/2021] [Indexed: 10/21/2022]
Abstract
Selection-pressures differ with population density, but few studies investigate how this can affect reproductive physiology. European badger (Meles meles) density varies from solitary to group-living across their range, with reported mating periods throughout the entire year to specific seasonal periods. Badger reproduction is evolutionarily distinct, interrupting the direct progression from conception to gestation with delayed implantation (DI), allowing for superfecundation (SF). To establish the tactical mating flexibility afforded by DI*SF, we used cross-sectional population-level seasonal variation of circulating sex-steroids for 97 females from a high-density population. Oestradiol was highest in spring among non-parous females, then lower in summer, and remained low during following seasons, suggesting that the mating period was restricted to just spring. Oestrone was consistently higher than oestradiol; it was elevated in spring, lowest during summer, peaked in autumn, and remained elevated for pregnant females in winter. This suggests that oestrone sustains pre-implanted blastocysts throughout DI. Progesterone was low throughout, except during winter pregnancy, associated with implantation and luteal development. In contrast to multiple mating periods reported by lower-density studies, our oestradiol data suggest that, at high-density, females exhibit only one mating period (congruent with testosterone patterns in males studied previously in this same population). While additional mating periods during DI enhance fertility assurance at low-density, at high-density, we propose that when coitus is frequent, fertilisation is assured, precluding the need for further cycles and associated mating risks. This endocrinologically flexible DI*SF mating strategy likely represents a form of balancing selection, allowing badgers to succeed at a range of regional densities.
Collapse
Affiliation(s)
- Nadine Adrianna Sugianto
- Wildlife Conservation Research Unit, Department of Zoology, University of Oxford, Oxford, UK; School of Biosciences, University of Birmingham, Birmingham, UK.
| | - Michael Heistermann
- Endocrinology Laboratory, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany
| | - Chris Newman
- Wildlife Conservation Research Unit, Department of Zoology, University of Oxford, Oxford, UK; Cook's Lake Farming Forestry and Wildlife Inc (Ecological Consultancy), Queens County, Nova Scotia, Canada
| | - David W Macdonald
- Wildlife Conservation Research Unit, Department of Zoology, University of Oxford, Oxford, UK
| | - Christina D Buesching
- Cook's Lake Farming Forestry and Wildlife Inc (Ecological Consultancy), Queens County, Nova Scotia, Canada; Department of Biology, Irving K. Barber Faculty of Sciences, The University of British Columbia, Kelowna, British Columbia, Canada
| |
Collapse
|
29
|
Chae SA, Son JS, Du M. Prenatal exercise in fetal development: a placental perspective. FEBS J 2021; 289:3058-3071. [PMID: 34449982 DOI: 10.1111/febs.16173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/09/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
Abstract
Maternal obesity (MO) and gestational diabetes mellitus (GDM) are common in Western societies, which impair fetal development and predispose offspring to metabolic dysfunction. Placenta is the organ linking the mother to her fetus, and MO suppresses the development of vascular system and expression of nutrient transporters in placenta, thereby affecting fetal development. For maintaining its proper physiological function, placenta is energy demanding, which is met through extensive oxidative phosphorylation. However, the oxidative capacity of placenta is suppressed due to MO and GDM. Recently, several studies showed that physical activity during pregnancy enhances oxidative metabolism and improves placental function, which might be partially mediated by exerkines, referring to cytokines elicited by exercise. In addition, as an endocrine organ, placenta secretes cytokines, termed placentokines, including apelin, superoxide dismutase 3, irisin, and adiponectin, which mediate fetal development and maternal metabolism. Possible molecular mechanisms linking maternal exercise and placentokines to placental and fetal development are further discussed. As an emerging field, up to now, available studies are limited, mostly conducted in rodents. Given the epidemics of obesity and metabolic disorders, as well as the prevalence of maternal sedentary lifestyle, the effects of exercise of pregnant women on placental function and placentokine secretion, as well as their impacts on fetal development, need to be further examined.
Collapse
Affiliation(s)
- Song Ah Chae
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Jun Seok Son
- Laboratory of Perinatal Kinesioepigenetics, Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Min Du
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| |
Collapse
|
30
|
Whynott RM, Summers KM, Jakubiak M, Van Voorhis BJ, Mejia RB. The effect of weight and body mass index on serum progesterone values and live birth rate in cryopreserved in vitro fertilization cycles. F S Rep 2021; 2:195-200. [PMID: 34278354 PMCID: PMC8267385 DOI: 10.1016/j.xfre.2021.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 11/17/2022] Open
Abstract
Objective To determine if weight or body mass index (BMI) affects the serum progesterone level at the time of the pregnancy test in cryopreserved blastocyst transfer cycles and to determine if those serum progesterone levels affect live births. Design Retrospective cohort study. Setting US academic medical center. Patient(s) Six hundred thirty-three patients undergoing their first cryopreserved embryo transfer cycle. Intervention(s) None. Main Outcome Measure(s) The primary outcome was the serum progesterone level on the day of the pregnancy test by patient weight and BMI. Our secondary analysis assessed the serum progesterone effect on live birth rate (LBR) in a clinic where progesterone supplementation was increased if the progesterone level was <15 ng/mL on the day of the pregnancy test. Results(s) There was a strong negative correlation between serum progesterone level and both BMI and weight, with BMI accounting for 27% and weight accounting for 29% of the variance in progesterone level. Serum progesterone level on the day of the pregnancy test was <15 ng/mL in 3% of women weighing <68 kg compared with 29% of women weighing ≥90.7 kg. Among women weighing ≥90.7 kg, live birth occurred in 47% whose serum progesterone level was <15 ng/mL on the day of the pregnancy test compared with 49% in those with serum progesterone level of 15–19 ng/mL and 44% in those with serum progesterone level of ≥20 ng/mL. Conclusion(s) Body weight was a significant factor in serum progesterone level at the time of the pregnancy test, with nearly 30% of patients weighing ≥90.7 kg having serum progesterone level of <15 ng/mL, a value associated with lower LBRs in prior studies. However, we found no effect of low progesterone levels on LBR after cryopreserved embryo transfer cycles in a clinic where progesterone dosing was increased if serum progesterone levels were <15 ng/mL.
Collapse
Affiliation(s)
- Rachel M Whynott
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Karen M Summers
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Margurite Jakubiak
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Bradley J Van Voorhis
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Rachel B Mejia
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
31
|
Barbanti C, Centini G, Lazzeri L, Habib N, Labanca L, Zupi E, Afors K, Starace AC. Adenomyosis and infertility: the role of the junctional zone. Gynecol Endocrinol 2021; 37:577-583. [PMID: 33587014 DOI: 10.1080/09513590.2021.1878131] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
OBJECTIVE Adenomyosis is a benign uterine disorder characterized by the invasion of the endometrium within the myometrium, starting from the junctional zone (JZ), the inner hormone dependent layer of the myometrium that plays an important role in sperm transport, implantation and placentation. The resulting histological abnormalities and functional defects may represent the pathogenic substrate for infertility and pregnancy complications. The objective of this paper is to review the literature to evaluate the correlation between inner myometrium alterations and infertility and to assess the role of JZ in the origin of adverse obstetric outcomes of both spontaneous and in vitro fertilization (IVF) pregnancies. METHODS we searched Pubmed for all original and review articles in the English language from January1962 until December 2019, using the MeSH terms of 'adenomyosis', 'junctional zone', combined with 'infertility', 'obstetrical outcomes', 'spontaneous conception', 'in vitro fertilization' and 'classification'. The review was divided into three sections to assess this pathogenic correlation, evaluating also the importance of classification of the disease. RESULTS AND CONCLUSIONS Absent or incomplete remodeling of the JZ can affect uterine peristalsis, alter vascular plasticity of the spiral arteries and activate inflammatory pathways, all related to adverse obstetric outcomes. Despite these observations, there is still limited evidence whether adenomyosis is a cause of infertility. However, it is reasonable to screen patients for adenomyosis, to consider pregnant women with diffuse adenomyosis at high risk of adverse obstetric outcomes, and to evaluate the importance of a noninvasive validated classification in the management of women with adenomyosis.
Collapse
Affiliation(s)
- Chiara Barbanti
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Gabriele Centini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Lucia Lazzeri
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Nassir Habib
- Department of Obstetrics and Gynaecology, Beaujon Hospital-University of Paris, Paris, France
| | - Luca Labanca
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Errico Zupi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Karolina Afors
- Department of Obstetrics and Gynaecology, Whittington Hospital, London, UK
| | | |
Collapse
|
32
|
Luo X, Yang R, Bai Y, Li L, Lin N, Sun L, Liu J, Wu Z. Binding of microRNA-135a (miR-135a) to homeobox protein A10 ( HOXA10) mRNA in a high-progesterone environment modulates the embryonic implantation factors beta3-integrin (ITGβ3) and empty spiracles homeobox-2 (EMX2). ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:662. [PMID: 33987360 PMCID: PMC8106024 DOI: 10.21037/atm-21-596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background Patients with elevated circulating progesterone concentrations on the day of the human chorionic gonadotropin (hCG) trigger had relatively low implantation rates during assisted reproductive treatments. In this study, we assess the hypothesis that different concentrations of progesterone regulate the expression of homeobox protein A10 (HOXA10) and its downstream genes through miRNA-135a. Methods MicroRNA-135a (miR-135a), HOXA10, beta3-integrin (ITGβ3), and empty spiracles homeobox-2 (EMX2) expression levels in endometrial tissues from patients with elevated progesterone were measured. To determine the threshold of progesterone level which can impair implantation, Ishikawa cells were used to determine the expression of the aforementioned 4 genes after exposure to 5 graded concentrations of progesterone. The dual-luciferase reporter assay was used to verify whether miR-135a regulated the expression of HOXA10. Furthermore, the effects of HOXA10 on the expression of key endometrial receptivity genes ITGβ3 and EMX2 were confirmed. Results High progesterone levels promoted miR-135a expression in vivo, and miR-135a bound to the 3'-untranslated region (3'-UTR) of HOXA10 mRNA to inhibit HOXA10 expression. Reduction of HOXA10 promoted EMX2 expression and inhibited ITG-3 production. Progesterone promoted the expression of HOXA10 in vitro at low concentrations. However, when the concentration was greater than 10−7 ng/mL, progesterone inhibited HOXA10 by promoting miR-135a expression, thereby altering the expression of related genes and affecting endometrial receptivity. Conclusions In vitro, the trend in miR-135a expression (which first decreased and then increased) was in direct contrast to that of HOXA10 expression (which first increased and then decreased) as progesterone levels increased. The key factors regulating endometrial receptivity included ITGβ3 and EMX2, which were confirmed to be regulated by HOXA10. High progesterone levels affected miR-135a expression, and miR-135a inhibited HOXA10 expression, thereby affecting endometrial receptivity.
Collapse
Affiliation(s)
- Xi Luo
- Faculty of Life science and Technology, Kunming University of Science and Technology, Kunming, China.,Medical School, Kunming University of Science and Technology, Kunming, China.,Department of Reproductive Medicine, the First People's Hospital of Yunnan Province, Kunming, China.,Reproductive Medical Center of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Renxiang Yang
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Yun Bai
- Department of Reproductive Medicine, the First People's Hospital of Yunnan Province, Kunming, China.,Reproductive Medical Center of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Lei Li
- Department of Reproductive Medicine, the First People's Hospital of Yunnan Province, Kunming, China.,Reproductive Medical Center of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Na Lin
- Department of Reproductive Medicine, the First People's Hospital of Yunnan Province, Kunming, China.,Reproductive Medical Center of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Lan Sun
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Jianjun Liu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Research Center of Biomedical Engineering, Kunming Medical University, Kunming, China
| | - Ze Wu
- Department of Reproductive Medicine, the First People's Hospital of Yunnan Province, Kunming, China.,Reproductive Medical Center of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
33
|
Song Y, Zhou F, Tan X, Liu X, Ding J, Zhang C, Li F, Zhu W, Ma W, Hu R, Zhang M. Bushen Huoxue recipe attenuates early pregnancy loss via activating endometrial COX2-PGE2 angiogenic signaling in mice. BMC Complement Med Ther 2021; 21:36. [PMID: 33446182 PMCID: PMC7809844 DOI: 10.1186/s12906-021-03201-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 01/04/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND During the fresh cycles of in vitro fertilization and embryo transfer, a disturbance in the reproductive endocrine environment following controlled ovarian hyperstimulation (COH) is closely related to compromised endometrial receptivity. This is a major disadvantage for women during pregnancy. Based on the theory of traditional Chinese medicine, Bushen Huoxue recipe (BSHXR) has been indicated to facilitate embryo implantation. METHODS The COH model (Kunming breed) was induced by injecting mice with pregnant mare serum gonadotrophin (0.4 IU/g) and human chorionic gonadotropin (1 IU/g), followed by treatment with BSHXR at three different concentrations (5.7, 11.4, and 22.8 g/kg), Bushen recipe (BSR) (5.7 g/kg), and Huoxue recipe (HXR) (5.7 g/kg). After successful mating, the pregnancy rate and implantation sites were examined on embryo day 8 (ED8), and the weight ratio of endometrium was calculated on ED4 midnight. Serum estrogen, progesterone, and endometrial PGE2 levels were measured using enzyme-linked immunosorbent assay. The endometrial microvasculature was evaluated using CD31 immunostaining. The protein and mRNA levels of the angiogenic factors in the endometrium were evaluated using western blot, immunohistochemistry, and polymerase chain reaction. RESULTS In the COH group, the pregnancy rate and implantation sites were significantly decreased, and abnormal serum hormone levels and impaired endometrial vascular development were observed. After BSHXR treatment, the supraphysiological serum progesterone level in COH mice was restored to normalcy. Moreover, the abnormal expression of the endometrial pro-angiogenic factors, including HIF1α, COX2-PGE2 pathway, and the down-stream factors, namely, MMP2, MMP9, TIMP2, and FGF2 after subjecting mice to COH was significantly improved after BSHXR treatment. CONCLUSION BSHXR could improve embryo implantation by regulating hormonal balance and modulating endometrial angiogenesis in mice, without inducing any side effects in normal pregnancy.
Collapse
Affiliation(s)
- Yufan Song
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Fanru Zhou
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Xiujuan Tan
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Xia Liu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Jiahui Ding
- Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Chu Zhang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Fan Li
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Wenxin Zhu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Wenwen Ma
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Runan Hu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Mingmin Zhang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
34
|
Ullah R, Naz A, Akram HS, Ullah Z, Tariq M, Mithani A, Faisal A. Transcriptomic analysis reveals differential gene expression, alternative splicing, and novel exons during mouse trophoblast stem cell differentiation. Stem Cell Res Ther 2020; 11:342. [PMID: 32762732 PMCID: PMC7409654 DOI: 10.1186/s13287-020-01848-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 07/15/2020] [Accepted: 07/22/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Differentiation of mouse trophoblast stem cells (TSCs) to trophoblast giant cells (TGCs) has been widely used as a model system to study placental development and function. While several differentially expressed genes, including regulators of TSC differentiation, have been identified, a comprehensive analysis of the global expression of genes and splice variants in the two cell types has not been reported. RESULTS Here, we report ~ 7800 differentially expressed genes in TGCs compared to TSCs which include regulators of the cell cycle, apoptosis, cytoskeleton, cell mobility, embryo implantation, metabolism, and various signaling pathways. We show that several mitotic proteins, including Aurora A kinase, were downregulated in TGCs and that the activity of Aurora A kinase is required for the maintenance of TSCs. We also identify hitherto undiscovered, cell-type specific alternative splicing events in 31 genes in the two cell types. Finally, we also report 19 novel exons in 12 genes which are expressed in both TSCs and TGCs. CONCLUSIONS Overall, our results uncover several potential regulators of TSC differentiation and TGC function, thereby providing a valuable resource for developmental and molecular biologists interested in the study of stem cell differentiation and embryonic development.
Collapse
Affiliation(s)
- Rahim Ullah
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Ambreen Naz
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Hafiza Sara Akram
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Zakir Ullah
- Virginia Commonwealth University, Richmond, USA
| | - Muhammad Tariq
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Aziz Mithani
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan.
| | - Amir Faisal
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan.
| |
Collapse
|
35
|
Bobetsis YA, Graziani F, Gürsoy M, Madianos PN. Periodontal disease and adverse pregnancy outcomes. Periodontol 2000 2020; 83:154-174. [PMID: 32385871 DOI: 10.1111/prd.12294] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Periodontal diseases are considered not only to affect tooth-supporting tissues but also to have a cause-and-effect relationship with various systemic diseases and conditions, such as adverse pregnancy outcomes. Mechanistic studies provide strong evidence that periodontal pathogens can translocate from infected periodontium to the feto-placental unit and initiate a metastatic infection. However, the extent and mechanisms by which metastatic inflammation and injury contribute to adverse pregnancy outcomes still remain unclear. The presence of oral bacteria in the placenta of women with term gestation further complicates our understanding of the biology behind the role of periodontal pathogens in pregnancy outcomes. Epidemiological studies demonstrate many methodological inconsistencies and flaws that render comparisons difficult and conclusions insecure. Therefore, despite the fact that a number of prospective studies show a positive association between periodontal diseases and various adverse pregnancy outcomes, the evidence behind it is still weak. Future well-designed explanatory studies are necessary to verify this relationship and, if present, determine its magnitude. The majority of high-quality randomized controlled trials reveal that nonsurgical periodontal therapy during the second trimester of gestation does not improve pregnancy outcomes. From a biological standpoint, this can be partially explained by the fact that therapy rendered at the fourth to sixth months of pregnancy is too late to prevent placental colonization by periodontal pathogens and consequently incapable of affecting pathogen-induced injury at the feto-placental unit. Thus, interventions during the preconception period may be more meaningful. With the increase in our understanding on the potential association between periodontal disease and adverse pregnancy outcomes, it is clear that dental practitioners should provide periodontal treatment to pregnant women that is safe for both the mother and the unborn child. Although there is not enough evidence that the anti-infective therapy alters pregnancy outcomes, it improves health-promoting behavior and periodontal condition, which in turn advance general health and risk factor control.
Collapse
Affiliation(s)
- Yiorgos A Bobetsis
- Department of Periodontology, National and Kapodistrian University of Athens, Athens, Greece
| | - Filippo Graziani
- Department of Surgery, Medical, Molecular, and Critical Area Pathology, University of Pisa, Pisa, Italy
| | - Mervi Gürsoy
- Department of Periodontology, University of Turku, Turku, Finland
| | - Phoebus N Madianos
- Department of Periodontology, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
36
|
Quintero-Ronderos P, Laissue P. Genetic Variants Contributing to Early Recurrent Pregnancy Loss Etiology Identified by Sequencing Approaches. Reprod Sci 2020; 27:1541-1552. [PMID: 32430708 DOI: 10.1007/s43032-020-00187-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recurrent pregnancy loss (RPL) affects up to 5% of couples. It is believed that genetic factors contribute to the disease's etiology and pathophysiology. Hundreds of genes represent coherent RPL candidates due to mammalian implantation's inherent complexity. Sanger sequencing (direct sequencing) of candidate genes has identified potential RPL causative genes (and variants), including those regulating embryo implantation and pregnancy maintenance. Although this approach is a reliable technique, the simultaneous analysis of large genomic regions is challenging. Next-generation sequencing (NGS) technology has thus emerged as a useful alternative for determining genetic variants and transcriptomic disturbances contributing to monogenic and polygenic diseases pathogenesis. However, interpreting results remains challenging as NGS experiments provide an enormous amount of complex data. The molecular aspects of specific diseases must be fully understood for accurate interpretation of NGS data. This review was thus aimed at describing (for the first time) the most relevant studies involving Sanger and NGS sequencing, leading to the description of variants related to RPL pathogenesis. Successful RPL-related NGS initiatives (including RNAseq-based studies) and future challenges are discussed. We consider that the information given here should be useful for clinicians, scientists, and students to enable a better understanding of RPL etiology. It may also provide a basis for the development of diagnostic/prognostic approaches contributing toward translational medicine.
Collapse
Affiliation(s)
- Paula Quintero-Ronderos
- Center For Research in Genetics and Genomics (CIGGUR), GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 N° 63C-69, Bogotá, 1100100, Colombia
| | - Paul Laissue
- Center For Research in Genetics and Genomics (CIGGUR), GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 N° 63C-69, Bogotá, 1100100, Colombia.
| |
Collapse
|
37
|
Aleksenko L, Quaye IK. Pregnancy-induced Cardiovascular Pathologies: Importance of Structural Components and Lipids. Am J Med Sci 2020; 360:447-466. [PMID: 32540145 DOI: 10.1016/j.amjms.2020.05.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 03/09/2020] [Accepted: 05/07/2020] [Indexed: 01/22/2023]
Abstract
Pregnancy leads to adaptations for maternal and fetal energy needs. The cardiovascular system bears the brunt of the adaptations as the heart and vessels enable nutrient supply to maternal organs facilitated by the placenta to the fetus. The components of the cardiovascular system are critical in the balance between maternal homeostatic and fetus driven homeorhetic regulation. Since lipids intersect maternal cardiovascular function and fetal needs with growth and in stress, factors affecting lipid deposition and mobilization impact risk outcomes. Here, the cardiovascular components and functional derangements associated with cardiovascular pathology in pregnancy, vis-à-vis lipid deposition, mobilization and maternal and/or cardiac and fetal energy needs are detailed. Most reports on the components and associated pathology in pregnancy, are on derangements affecting the extracellular matrix and epicardial fat, followed by the endothelium, vascular smooth muscle, pericytes and myocytes. Targeted studies on all cardiovascular components and pathological outcomes in pregnancy will enhance targeted interventions.
Collapse
Affiliation(s)
- Larysa Aleksenko
- Division of Obstetrics and Gynecology, Department of Clinical Sciences, Lund University, Lund, Sweden.
| | - Isaac K Quaye
- Regent University College of Science and Technology, Accra, Ghana
| |
Collapse
|
38
|
Fractalkine Regulates HEC-1A/JEG-3 Interaction by Influencing the Expression of Implantation-Related Genes in an In Vitro Co-Culture Model. Int J Mol Sci 2020; 21:ijms21093175. [PMID: 32365902 PMCID: PMC7246682 DOI: 10.3390/ijms21093175] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/24/2020] [Accepted: 04/29/2020] [Indexed: 12/15/2022] Open
Abstract
Embryo implantation is a complex process regulated by a network of biological molecules. Recently, it has been described that fractalkine (CX3CL1, FKN) might have an important role in the feto-maternal interaction during gestation since the trophoblast cells express fractalkine receptor (CX3CR1) and the endometrium cells secrete fractalkine. CX3CR1 controls three major signalling pathways, PLC-PKC pathway, PI3K/AKT/NFκB pathway and Ras-mitogen-activated protein kinases (MAPK) pathways regulating proliferation, growth, migration and apoptosis. In this study, we focused on the molecular mechanisms of FKN treatment influencing the expression of implantation-related genes in trophoblast cells (JEG-3) both in mono-and in co-culture models. Our results reveal that FKN acted in a concentration and time dependent manner on JEG-3 cells. FKN seemed to operate as a positive regulator of implantation via changing the action of progesterone receptor (PR), activin receptor and bone morphogenetic protein receptor (BMPR). FKN modified also the expression of matrix metalloproteinase 2 and 9 controlling invasion. The presence of HEC-1A endometrial cells in the co-culture contributed to the effect of fractalkine on JEG-3 cells regulating implantation. The results suggest that FKN may contribute to the successful attachment and implantation of embryo.
Collapse
|
39
|
In-vitro investigation of calcitonin associated effects on the trophoblastic cells. Acta Histochem 2020; 122:151510. [PMID: 32024606 DOI: 10.1016/j.acthis.2020.151510] [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: 10/21/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 11/21/2022]
Abstract
Calcitonin is expressed in the epithelium of endometrium, and modulates zonula adherens junctions which are composed of cadherin-catenins complex during the implantation window. Trophoblastic cells which have complex interaction with the epithelial cells of endometrium during implantation were demonstrated to have calcitonin receptors. Mechanism of action of calcitonin on trophoblastic cells has not yet been elucidated. Therefore, it was aimed to determine the effects of calcitonin on the expressions of β-catenin and phospho-β-catenin in a dose depended manner under the influence of progesterone and estrogen hormones (P + E) by using JAR cell line through the immunocytochemical and Western blot analyses. Moreover, adherens junctions (AJs) were ultrastructurally investigated to assess the involvement of cadherin-catenin complex in accordance with the changes in the specified parameters. Immunocytochemical analysis showed that only 10 nM calcitonin treated group had increased expression of membranous β-catenin compared to the control group, while there was decreased expression of β-catenin in the nucleus of all the experimental groups. Cytoplasmic expressions of the phospho-β-catenin decreased in all experimental groups compared to the control group while the decrease in the nuclear expression was remarkable in the groups treated with P + E, and P + E + 250 nM calcitonin. Western blot analysis showed that total β-catenin and phospho-β-catenin expressions were not significantly different. Ultrastructural analysis showed that increase in the number of AJs was noticeable in the group treated with 10 nM calcitonin. Overall, the localization and expression levels of β-catenin and phospho-β-catenin suggest that calcitonin could show its effects through the non-canonical pathway in the trophoblastic cells.
Collapse
|
40
|
Yaw AM, Duong TV, Nguyen D, Hoffmann HM. Circadian rhythms in the mouse reproductive axis during the estrous cycle and pregnancy. J Neurosci Res 2020; 99:294-308. [PMID: 32128870 DOI: 10.1002/jnr.24606] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/17/2020] [Accepted: 02/12/2020] [Indexed: 12/26/2022]
Abstract
Molecular and behavioral timekeeping is regulated by the circadian system which includes the brain's suprachiasmatic nucleus (SCN) that translates environmental light information into neuronal and endocrine signals aligning peripheral tissue rhythms to the time of day. Despite the critical role of circadian rhythms in fertility, it remains unexplored how circadian rhythms change within reproductive tissues during pregnancy. To determine how estrous cycle and pregnancy impact phase relationships of reproductive tissues, we used PER2::Luciferase (PER2::LUC) circadian reporter mice and determined the time of day of PER2::LUC peak (phase) in the SCN, pituitary, uterus, and ovary. The relationships between reproductive tissue PER2::LUC phases changed throughout the estrous cycle and late pregnancy and were accompanied by changes to PER2::LUC period in the SCN, uterus, and ovary. To determine if the phase relationship adaptations were driven by sex steroids, we asked if progesterone, a hormone involved in estrous cyclicity and pregnancy, could regulate Per2-luciferase expression. Using an in vitro transfection assay, we found that progesterone increased Per2-luciferase expression in immortalized SCN (SCN2.2) and arcuate nucleus (KTAR) cells. In addition, progesterone shortened PER2::LUC period in ex vivo uterine tissue recordings collected during pregnancy. As progesterone dramatically increases during pregnancy, we evaluated wheel-running patterns in PER2::LUC mice. We confirmed that activity levels decrease during pregnancy and found that activity onset was delayed. Although SCN, but not arcuate nucleus, PER2::LUC period changed during late pregnancy, onset of locomotor activity did not correlate with SCN or arcuate nucleus PER2::LUC period.
Collapse
Affiliation(s)
- Alexandra M Yaw
- Department of Animal Science and the Reproductive and Developmental Science Program, Michigan State University, East Lansing, MI, USA
| | - Thu V Duong
- Department of Animal Science and the Reproductive and Developmental Science Program, Michigan State University, East Lansing, MI, USA
| | - Duong Nguyen
- Department of Animal Science and the Reproductive and Developmental Science Program, Michigan State University, East Lansing, MI, USA
| | - Hanne M Hoffmann
- Department of Animal Science and the Reproductive and Developmental Science Program, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
41
|
Pier B, Crellin C, Katre A, Conner MG, Novak L, Young SL, Arend R. Large, Non-Cavity Distorting Intramural Leiomyomas Decrease Leukemia Inhibitory Factor in the Secretory Phase Endometrium. Reprod Sci 2020; 27:569-574. [PMID: 32046421 PMCID: PMC7539805 DOI: 10.1007/s43032-019-00056-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 06/07/2019] [Indexed: 12/11/2022]
Abstract
Despite mounting evidence that large intramural leiomyomas decrease fecundity during in vitro fertilization cycles, few studies have demonstrated a mechanism for this impact. We hypothesize that large intramural leiomyomas (IM) decrease the expression of endometrial implantation factors during the window of implantation. We prospectively recruited sub-fertile patients with IM 3 cm or greater in size planning myomectomy and performed endometrial biopsies the day of planned myomectomy (n = 9). Preoperative screening demonstrated no intercavitary lesions. Control endometrial samples were obtained from young, normally menstruating women free of uterine leiomyomas (n = 8). Endometrial samples were obtained in the mid-secretory phase (average cycle day for control patients and intramural leiomyoma patients were 24.5 and 21.3, respectively). Expression of implantation markers HOXA10, leukemia inhibitory factor (LIF), ER-α, and PR was compared using quantitative immunohistochemistry. Standard descriptive statistics were used to compare H-scores between the cohorts. Patients with intramural leiomyomas were found to have decreased LIF compared to controls (p value < 0.001). Expressions of HOXA10 and PR were no different between cohorts; however, ER-α showed a trend toward increased expression in the fibroid cohort (p value 0.07). LIF is downregulated in the endometrium of patients with large IM. This study is among the first to show decreased LIF expression in patients with uterine leiomyomas. We hypothesize that this difference from previously published work is due to sampling the endometrium at the height of LIF expression. Further work is needed to show if LIF downregulation is corrected with leiomyoma resection.
Collapse
Affiliation(s)
- Bruce Pier
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Tripler Army Medical Center, Jarrett White Road, Honolulu, Hawaii 96859 USA
| | - Christopher Crellin
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Tripler Army Medical Center, Jarrett White Road, Honolulu, Hawaii 96859 USA
| | - Ashwini Katre
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Michael G. Conner
- Department of Pathology, University of Alabama at Birmingham, Birmingham, USA
| | - Lea Novak
- Department of Pathology, University of Alabama at Birmingham, Birmingham, USA
| | - Steven L Young
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Rebecca Arend
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
| |
Collapse
|
42
|
Brito C, M Silva T, M Castro M, Wyrwas W, Oliveira B, M Fonseca B, Oliveira P, W Roberts C, Teixeira N, Borges M. Toxoplasma gondii infection reduces serum progesterone levels and adverse effects at the maternal-foetal interface. Parasite Immunol 2019; 42:e12690. [PMID: 31802508 DOI: 10.1111/pim.12690] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 11/29/2022]
Abstract
AIMS Pregnant BALB/c mice infected with a Toxoplasma gondii type II strain were used to determine how pregnancy interferes with the development of maternal immunity to T gondii and how infection disrupts pregnancy and foetal development. METHODS Maternal and foetal parasite loads were assessed by amplification of T gondii SAG1 using qPCR. Adverse effects of infection were evaluated on foetal-placental development by quantification of implantation units undergoing resorption and by histopathological analyses. Serum progesterone levels were quantified by immunoassay. The effect of T gondii infection on maternal immunity was determined by assessing the cellular composition of spleens by flow cytometry. RESULTS Infected pregnant mice exhibited clinical signs of infection, inflammation and necrosis at the maternal-foetal interface and decreased serum progesterone levels. In infected mice, there was a clear effect of pregnancy and infection on macrophage cell numbers. However, no differences in the parasite load were detected between non-pregnant and pregnant mice. CONCLUSIONS Maternal T gondii infection induced adverse effects at the maternal-foetal interface. Alterations were found in immune spleen cells, dependent on the day of pregnancy, relative to nonpregnant animals. The results obtained suggest a pregnancy-dependent mechanism during T gondii infection able to interfere with macrophage numbers.
Collapse
Affiliation(s)
- Carina Brito
- Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Tânia M Silva
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Maria M Castro
- Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Weronika Wyrwas
- Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Bárbara Oliveira
- Rodent Animal Facility, Institute of Biomedical Sciences of Abel Salazar-University of Porto, Porto, Portugal
| | - Bruno M Fonseca
- UCIBIO/REQUIMTE, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Pedro Oliveira
- EPIUnit, ISPUP, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Craig W Roberts
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Natércia Teixeira
- UCIBIO/REQUIMTE, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Margarida Borges
- UCIBIO/REQUIMTE, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| |
Collapse
|
43
|
Isocitrate dehydrogenase type 2 (IDH2) is part of a multiprotein complex for placental steroidogenesis. Placenta 2019; 87:30-37. [PMID: 31542634 DOI: 10.1016/j.placenta.2019.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/09/2019] [Accepted: 09/16/2019] [Indexed: 11/20/2022]
Abstract
BACKGROUND Human syncytiotrophoblast mitochondria require the activity of the isocitrate dehydrogenase type 2 (IDH2) to obtain reduced coenzymes for progesterone (P4) synthesis. Data from the literature indicate that mitochondrial steroidogenic contact sites transform efficiently cholesterol into P4. In this research, we identified the IDH2 as a member of the steroidogenic contact site and analyzed the steroidogenic role of its activity. METHOD Human syncytiotrophoblast mitochondria were isolated by differential centrifugation, and steroidogenic contact sites were obtained by osmotic shock and sucrose gradient ultracentrifugation. In-gel native activity assay, mass spectroscopy, and western blot were used to identify the association of proteins and their activities. P4 was determined by immunofluorescence. RESULTS The IDH2 was mainly identified in steroidogenic contact sites, and its activity was associated with a complex of proteins with an apparent molecular mass of ~590 kDa. Mass spectroscopy showed many groups of proteins with several metabolic functions, including steroidogenesis and ATP synthesis. The IDH2 activity was coupled to P4 synthesis since in the presence of Ca2+ or Na2SeO3, inhibitors of the IDH2, the P4 production decreased. CONCLUSIONS The human syncytiotrophoblast mitochondria build contact sites for steroidogenesis. The IDH2, a non-membrane protein, supplies the NADPH required for the synthesis of P4 in a complex (steroidosome) that associate the proteins required to transform efficiently cholesterol into P4, which is necessary in pregnancy to maintain the relationship between mother and fetus. GENERAL SIGNIFICANCE The IDH2 is proposed as a check point in the regulation of placental steroidogenesis.
Collapse
|
44
|
Characterization of Oestrus Cycles in Namibian Swakara and Damara Sheep through Determination of Circannual Plasma Progesterone Levels. J Vet Med 2019; 2019:5320718. [PMID: 31380447 PMCID: PMC6657608 DOI: 10.1155/2019/5320718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/08/2019] [Accepted: 07/01/2019] [Indexed: 11/17/2022] Open
Abstract
A year-long prospective study characterized the seasonality of oestrus cycles in primiparous, nonpregnant Swakara (n=8) and Damara (n=5) ewes through surveillance of plasma progesterone (P4) levels. During this period, Swakara and Damara groups evidently averaged 23 oestrus cycles with an average length of 17 days. Damara ewes showed greater mean peak plasma P4 levels (11.4±0.16ng/ml) than Swakara ewes (5.4±0.11ng/ml) (P<0.05). Oestrus cycles in Damara ewes showed relatively uniform plasma P4 peaks throughout the year ranging from 10.6±0.16 to 12.6±0.24ng/ml. In Swakara ewes, P4 peaks were highest in the autumn oestrus cycles (from 7.1±0.16 to 7.5±0.11ng/ml), rapidly declining through winter to 2.2±0.08ng/ml by midspring and then rapidly increasing to 4.9±0.37ng/ml at the commencement of summer, followed by a gradual increase from 5.7± to 7.1±ng/ml by the start of autumn. The annual mean area under the curve temporal progesterone measurements (AUCPM) in Damara ewes (115.9±18.6ng⁎day/ml) was greater than that in Swakara ewes (58.6±25.3ng⁎day/ml) (p<0.05). For Swakara ewes, the mean AUCPM in summer and autumn cycles (68.2±14.7 and 79.5±10.0ng⁎day/ml, respectively) were greater than those in spring and winter cycles (28.7±12.3 and 55.0±27.3ng⁎day/ml), respectively (P<0.05). There was no seasonal variation in the exposure of the Damara ewes to P4 in between seasons (P>0.05), though, however, the Damara ewes had greater P4 levels than the Swakara ewes (P<0.05). Progesterone profiles showed that Swakara ewes possessed 'residual' seasonality, whereas the Damara ewes were no longer seasonal. The implications of this disparity in the seasonal exposure of Swakara and Damara ewes to luteal P4 on fertility warrant further investigation.
Collapse
|
45
|
Olvera-Sánchez S, Esparza-Perusquía M, Flores-Herrera O, Urban-Sosa VA, Martínez F. Aspectos generales del transporte de colesterol en la esteroidogénesis de la placenta humana. TIP REVISTA ESPECIALIZADA EN CIENCIAS QUÍMICO-BIOLÓGICAS 2019. [DOI: 10.22201/fesz.23958723e.2019.0.180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
La placenta humana requiere de colesterol para sintetizar la progesterona que mantiene la relación entre el feto y la madre, lo que le permite concluir de manera exitosa el embarazo. La placenta incorpora el colesterol principalmente a través de las lipoproteínas de baja densidad (LDL) que se obtienen del torrente circulatorio materno por un mecanismo de endocitosis. A los endosomas que se generan en este proceso se les unen varias proteínas conformando los endosomas tardíos, que degradan las LDL y liberan el colesterol a las mitocondrias del sinciciotrofoblasto que lo transforman en pregnenolona y posteriormente en progesterona. Las proteínas de fusión de membranas denominados complejos SNARE participan en la liberación del colesterol en sitios de contacto específicos en donde se localizan las proteínas mitocondriales responsables de la esteroidogénesis.
Collapse
|
46
|
In Vitro Effect of Estradiol and Progesterone on Ovine Amniotic Epithelial Cells. Stem Cells Int 2019; 2019:8034578. [PMID: 31049069 PMCID: PMC6458847 DOI: 10.1155/2019/8034578] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 01/30/2019] [Accepted: 02/05/2019] [Indexed: 01/26/2023] Open
Abstract
Amniotic epithelial cells (AECs), an emerging source of extrafoetal stem cells, have recently attracted attention for their great regenerative potential. Since AEC amplifications are accompanied by the loss of their native epithelial phenotype and by the progressive reduction of relevant biological properties, the issue to be addressed is the development of effective culture protocols. In this context, recently, it has been demonstrated that progesterone (P4) supplementation during ovine AEC (oAEC) expansion could prevent the undesirable epithelial-mesenchymal transition (EMT). In contrast, there is no information to date on the role of the other pregnancy steroids in culture. With this aim, the present study has been designed to clarify the impact of estradiol (E2), alone or in combination with P4 (12.5 μM and 25 μM), during oAEC amplification. Steroid supplementations were assessed by testing oAEC proliferation, stemness, EMT, and osteogenic or chondrogenic plasticity. The results indicated that EMT can be prevented exclusively in the presence of high doses of P4, while it occurred rapidly in cells exposed to E2 as denoted by protein (cytokeratin-8 and alpha-SMA) and gene expression (vimentin and snail) profiles. Moreover, steroid exposure was able to influence highly oAEC plasticity. Particularly, P4-treated cells displayed a precommitment towards osteogenic lineage, confirmed by the upregulation of OCN, RUNX2, and the greater deposition of calcium nodules. Conversely, P4 exposure inhibited oAEC chondrogenic differentiation, which was induced in E2-treated cells as confirmed by the upregulation of chondrogenesis-related genes (SOX9, ACAN, and COL2A1) and by the accumulation of Alcian blue-positive extracellular matrix. Simultaneously, E2-treated cells remained unresponsive to osteogenic inductive stimuli. In conclusion, media supplementation with high doses of steroids may be adopted to modulate phenotype and plasticity during oAEC amplification. Relevantly, the osteo or chondro steroid-induced precommitment may open unprecedented cell-based therapies to face the unsolved orthopaedic issues related to osteochondral regeneration.
Collapse
|
47
|
Quintero-Ronderos P, Laissue P. Genetic Variants Contributing to Early Recurrent Pregnancy Loss Etiology Identified by Sequencing Approaches. Reprod Sci 2019:1933719119831769. [PMID: 30879428 DOI: 10.1177/1933719119831769] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recurrent pregnancy loss (RPL) affects up to 5% of couples. It is believed that genetic factors contribute to the disease's etiology and pathophysiology. Hundreds of genes represent coherent RPL candidates due to mammalian implantation's inherent complexity. Sanger sequencing (direct sequencing) of candidate genes has identified potential RPL causative genes (and variants), including those regulating embryo implantation and pregnancy maintenance. Although this approach is a reliable technique, the simultaneous analysis of large genomic regions is challenging. Next-generation sequencing (NGS) technology has thus emerged as a useful alternative for determining genetic variants and transcriptomic disturbances contributing to monogenic and polygenic diseases pathogenesis. However, interpreting results remains challenging as NGS experiments provide an enormous amount of complex data. The molecular aspects of specific diseases must be fully understood for accurate interpretation of NGS data. This review was thus aimed at describing (for the first time) the most relevant studies involving Sanger and NGS sequencing, leading to the description of variants related to RPL pathogenesis. Successful RPL-related NGS initiatives (including RNAseq-based studies) and future challenges are discussed. We consider that the information given here should be useful for clinicians, scientists, and students to enable a better understanding of RPL etiology. It may also provide a basis for the development of diagnostic/prognostic approaches contributing toward translational medicine.
Collapse
Affiliation(s)
- Paula Quintero-Ronderos
- 1 Center For Research in Genetics and Genomics (CIGGUR), GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Paul Laissue
- 1 Center For Research in Genetics and Genomics (CIGGUR), GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
48
|
Kuo CY, Shevchuk M, Opfermann J, Guo T, Santoro M, Fisher JP, Kim PCW. Trophoblast-endothelium signaling involves angiogenesis and apoptosis in a dynamic bioprinted placenta model. Biotechnol Bioeng 2019; 116:181-192. [PMID: 30298908 PMCID: PMC6289739 DOI: 10.1002/bit.26850] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 09/26/2018] [Accepted: 10/04/2018] [Indexed: 01/01/2023]
Abstract
Trophoblast invasion and remodeling of the maternal spiral arteries are required for pregnancy success. Aberrant endothelium-trophoblast crosstalk may lead to preeclampsia, a pregnancy complication that has serious effects on both the mother and the baby. However, our understanding of the mechanisms involved in this pathology remains elementary because the current in vitro models cannot describe trophoblast-endothelium interactions under dynamic culture. In this study, we developed a dynamic three-dimensional (3D) placenta model by bioprinting trophoblasts and an endothelialized lumen in a perfusion bioreactor. We found the 3D printed perfusion bioreactor system significantly augmented responses of endothelial cells by encouraging network formations and expressions of angiogenic markers, cluster of differentiation 31 (CD31), matrix metalloproteinase-2 (MMP2), matrix metalloproteinase-9 (MMP9), and vascular endothelial growth factor A (VEGFA). Bioprinting favored colocalization of trophoblasts with endothelial cells, similar to in vivo observations. Additional analysis revealed that trophoblasts reduced the angiogenic responses by reducing network formation and motility rates while inducing apoptosis of endothelial cells. Moreover, the presence of endothelial cells appeared to inhibit trophoblast invasion rates. These results clearly demonstrated the utility and potential of bioprinting and perfusion bioreactor system to model trophoblast-endothelium interactions in vitro. Our bioprinted placenta model represents a crucial step to develop advanced research approach that will expand our understanding and treatment options of preeclampsia and other pregnancy-related pathologies.
Collapse
Affiliation(s)
- Che-Ying Kuo
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Mariya Shevchuk
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Justin Opfermann
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
| | - Ting Guo
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Marco Santoro
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Peter CW Kim
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
- School of Medicine and Health Sciences, The George Washington University, Washington, DC
| |
Collapse
|
49
|
Chen W, Chen J, Xu M, Zhong Z, Zhang Q, Yang W, Huang G. Electroacupuncture facilitates implantation by enhancing endometrial angiogenesis in a rat model of ovarian hyperstimulation. Biol Reprod 2019; 100:268-280. [PMID: 30084973 PMCID: PMC6335210 DOI: 10.1093/biolre/ioy176] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 07/06/2018] [Accepted: 07/31/2018] [Indexed: 01/31/2023] Open
Abstract
Controlled ovarian hyperstimulation (COH) impairs the synchronized development of endometrium and embryo, resulting in the failure of embryo implantation. Here, we investigated what effects electroacupuncture had on embryo implantation in COH rats. Female rats were randomly assigned to four groups: normal (N), model (M), electroacupuncture (EA), and electroacupuncture pretreatment (PEA). Rats in groups M, EA, PEA were injected with pregnant mare serum gonadotropin (PMSG) and human chorionic gonadotropin to establish the COH model. Rats in group EA received electroacupuncture treatment from the PMSG injection day to the 3rd day of pregnancy (D3), while those in group PEA received electroacupuncture treatment for 3 days before the PMSG day and continuing to D3. Furthermore, another 30 female rats who received the same treatment as the rats in group PEA were injected with siVEGFR2 into uterine lumen. The endometrial microvascular density (MVD) and the expression levels of vascular endothelial growth factor-A, angiopoietin-1, and fibroblast growth factor-2 were significantly lower in groups M than in groups N and PEA. The percentage of dolichos biflorus agglutinin positive uterine natural killer cells in groups N, EA and PEA was higher than that in group M. After the siVEGFR2 injection, the protein expression levels of vascular endothelial growth factor receptor 2 (VEGFR2), PI3K, p-AKT and p-ERK, the embryo number and the MVD were significantly reduced. In conclusion, electroacupuncture can facilitate embryo implantation in COH rats by activating the VEGFR2/PI3K/AKT and VEGFR2/ERK signaling pathways which have a positive relationship with endometrial angiogenesis.
Collapse
Affiliation(s)
- Wei Chen
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jie Chen
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Menghao Xu
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhiyan Zhong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qing Zhang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Yang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guangying Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
50
|
Borges M, Magalhães Silva T, Brito C, Teixeira N, Roberts CW. How does toxoplasmosis affect the maternal-foetal immune interface and pregnancy? Parasite Immunol 2018; 41:e12606. [PMID: 30471137 DOI: 10.1111/pim.12606] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 11/13/2018] [Accepted: 11/20/2018] [Indexed: 12/11/2022]
Abstract
Toxoplasma gondii is a zoonotic parasite which, depending on the geographical location, can infect between 10% and 90% of humans. Infection during pregnancy may result in congenital toxoplasmosis. The effects on the foetus vary depending on the stage of gestation in which primary maternal infection arises. A large body of research has focused on understanding immune response to toxoplasmosis, although few studies have addressed how it is affected by pregnancy or the pathological consequences of infection at the maternal-foetal interface. There is a lack of knowledge about how maternal immune cells, specifically macrophages, are modulated during infection and the resulting consequences for parasite control and pathology. Herein, we discuss the potential of T. gondii infection to affect the maternal-foetal interface and the potential of pregnancy to disrupt maternal immunity to T. gondii infection.
Collapse
Affiliation(s)
- Margarida Borges
- UCIBIO/REQUIMTE, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Tânia Magalhães Silva
- Instituto de Biologia Molecular e Celular (IBMC), University of Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Carina Brito
- UCIBIO/REQUIMTE, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Natércia Teixeira
- UCIBIO/REQUIMTE, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Craig W Roberts
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|