1
|
Clarke GS, Vincent AD, Ladyman SR, Gatford KL, Page AJ. Circadian patterns of behaviour change during pregnancy in mice. J Physiol 2024; 602:6531-6552. [PMID: 38477893 PMCID: PMC11607885 DOI: 10.1113/jp285553] [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: 08/24/2023] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Food intake and activity adapt during pregnancy to meet the increased energy demands. In comparison to non-pregnant females, pregnant mice consume more food, eating larger meals during the light phase, and reduce physical activity. How pregnancy changes the circadian timing of behaviour was less clear. We therefore randomised female C57BL/6J mice to mating for study until early (n = 10), mid- (n = 10) or late pregnancy (n = 11) or as age-matched, non-pregnant controls (n = 12). Mice were housed individually in Promethion cages with a 12 h light-12 h dark cycle [lights on at 07.00 h, Zeitgeber (ZT)0] for behavioural analysis. Food intake between ZT10 and ZT11 was greater in pregnant than non-pregnant mice on days 6.5-12.5 and 12.5-17.5. In mice that exhibited a peak in the last 4 h of the light phase (ZT8-ZT12), peaks were delayed by 1.6 h in the pregnant compared with the non-pregnant group. Food intake immediately after dark-phase onset (ZT13-ZT14) was greater in the pregnant than non-pregnant group during days 12.5-17.5. Water intake patterns corresponded to food intake. From days 0.5-6.5 onwards, the pregnant group moved less during the dark phase, with decreased probability of being awake, in comparison to the non-pregnant group. The onset of dark-phase activity, peaks in activity, and wakefulness were all delayed during pregnancy. In conclusion, increased food intake during pregnancy reflects increased amplitude of eating behaviour, without longer duration. Decreases in activity also contribute to positive energy balance in pregnancy, with delays to all measured behaviours evident from mid-pregnancy onwards. KEY POINTS: Circadian rhythms synchronise daily behaviours including eating, drinking and sleep, but how these change in pregnancy is unclear. Food intake increased, with delays in peaks of food intake behaviour late in the light phase from days 6.5 to 12.5 of pregnancy, in comparison to the non-pregnant group. The onset of activity after lights off (dark phase) was delayed in pregnant compared with non-pregnant mice. Activity decreased by ∼70% in the pregnant group, particularly in the dark (active) phase, with delays in peaks of wakefulness evident from days 0.5-6.5 of pregnancy onwards. These behavioural changes contribute to positive energy balance during pregnancy. Delays in circadian behaviours during mouse pregnancy were time period and pregnancy stage specific, implying different regulatory mechanisms.
Collapse
Affiliation(s)
- Georgia S. Clarke
- School of BiomedicineUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Nutrition, Diabetes & Gut Health, Lifelong Health ThemeSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Robinson Research InstituteUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Andrew D. Vincent
- Freemasons Centre for Male Health & Wellbeing, Adelaide Medical SchoolThe University of AdelaideAdelaideSouth AustraliaAustralia
| | - Sharon R. Ladyman
- Centre for Neuroendocrinology, School of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
- Department of AnatomySchool of Biomedical SciencesDunedinNew Zealand
| | - Kathryn L. Gatford
- School of BiomedicineUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Nutrition, Diabetes & Gut Health, Lifelong Health ThemeSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Robinson Research InstituteUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Amanda J. Page
- School of BiomedicineUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Nutrition, Diabetes & Gut Health, Lifelong Health ThemeSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
| |
Collapse
|
2
|
Hu Q, Gui Y, Cao C, Xie J, Tang H. Single-cell sequencing reveals transcriptional dynamics regulated by ERα in mouse ovaries. PLoS One 2024; 19:e0313867. [PMID: 39570927 PMCID: PMC11581351 DOI: 10.1371/journal.pone.0313867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/31/2024] [Indexed: 11/24/2024] Open
Abstract
CONTEXT Estrogen receptor α (ERα) is a key regulator of reproductive function, particularly in ovarian development and function, yet the specifics of its role at the molecular level remain unclear. AIMS The study aims to elucidate the molecular mechanisms of ERα-regulated transcriptional dynamics in ovarian cells using ERα knockout (αERKO) mice created via CRISPR/Cas9. METHODS Single-cell RNA sequencing (scRNA-seq) was used to compare transcriptomes from individual ovarian cells in both wild type and αERKO mice. Bioinformatics analyses identified distinct cell populations and their transcriptional profiles post ERα deletion. KEY RESULTS Distinct oocyte and granulosa cell populations were identified, with ERα deletion disrupting the regulation of genes linked to ovarian infertility, the ovulation cycle, and steroidogenesis. Greb1 expression in granulosa cells was found to be ERα-dependent. CONCLUSIONS ERα deletion significantly alters the transcriptional landscape of ovarian cells, affecting genes and pathways central to ovarian function and the ovulation process. IMPLICATIONS The findings provide an in-depth, single-cell view of ERα's role in the reproductive system, offering insights that may lead to novel treatments for ovarian disorders.
Collapse
Affiliation(s)
- Qicai Hu
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
- Institute of Obstetrics and Gynecology, Shenzhen PKU-HKUST Medical Center, Shenzhen, P. R. China
- Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecologic Diseases, Shenzhen, P. R. China
| | - Yiqian Gui
- Institute Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Congcong Cao
- Guangdong and Shenzhen Key Laboratory of Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, China
| | - Jun Xie
- Guangdong and Shenzhen Key Laboratory of Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, China
| | - Huiru Tang
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
- Cheerland Watson Precision Medicine Co. LTD, Shenzhen, China
| |
Collapse
|
3
|
Hamburg-Shields E, Mesiano S. The hormonal control of parturition. Physiol Rev 2024; 104:1121-1145. [PMID: 38329421 PMCID: PMC11380996 DOI: 10.1152/physrev.00019.2023] [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: 04/20/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/09/2024] Open
Abstract
Parturition is a complex physiological process that must occur in a reliable manner and at an appropriate gestation stage to ensure a healthy newborn and mother. To this end, hormones that affect the function of the gravid uterus, especially progesterone (P4), 17β-estradiol (E2), oxytocin (OT), and prostaglandins (PGs), play pivotal roles. P4 via the nuclear P4 receptor (PR) promotes uterine quiescence and for most of pregnancy exerts a dominant block to labor. Loss of the P4 block to parturition in association with a gain in prolabor actions of E2 are key transitions in the hormonal cascade leading to parturition. P4 withdrawal can occur through various mechanisms depending on species and physiological context. Parturition in most species involves inflammation within the uterine tissues and especially at the maternal-fetal interface. Local PGs and other inflammatory mediators may initiate parturition by inducing P4 withdrawal. Withdrawal of the P4 block is coordinated with increased E2 actions to enhance uterotonic signals mediated by OT and PGs to promote uterine contractions, cervix softening, and membrane rupture, i.e., labor. This review examines recent advances in research to understand the hormonal control of parturition, with focus on the roles of P4, E2, PGs, OT, inflammatory cytokines, and placental peptide hormones together with evolutionary biology of and implications for clinical management of human parturition.
Collapse
Affiliation(s)
- Emily Hamburg-Shields
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, Ohio, United States
- Department of Obstetrics and Gynecology, University Hospitals of Cleveland, Cleveland, Ohio, United States
| | - Sam Mesiano
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, Ohio, United States
- Department of Obstetrics and Gynecology, University Hospitals of Cleveland, Cleveland, Ohio, United States
| |
Collapse
|
4
|
Kwak YT, Montalbano AP, Kelleher AM, Colon-Caraballo M, Kraus WL, Mahendroo M, Mendelson CR. Decline in corepressor CNOT1 in the pregnant myometrium near term impairs progesterone receptor function and increases contractile gene expression. J Biol Chem 2024; 300:107484. [PMID: 38897566 PMCID: PMC11301068 DOI: 10.1016/j.jbc.2024.107484] [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: 05/04/2023] [Revised: 05/18/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Progesterone (P4), acting via its nuclear receptor (PR), is critical for pregnancy maintenance by suppressing proinflammatory and contraction-associated protein (CAP)/contractile genes in the myometrium. P4/PR partially exerts these effects by tethering to NF-κB bound to their promot-ers, thereby decreasing NF-κB transcriptional activity. However, the underlying mechanisms whereby P4/PR interaction blocks proinflammatory and CAP gene expression are not fully understood. Herein, we characterized CCR-NOT transcription complex subunit 1 (CNOT1) as a corepressor that also interacts within the same chromatin complex as PR-B. In mouse myome-trium increased expression of CAP genes Oxtr and Cx43 at term coincided with a marked decline in expression and binding of CNOT1 to NF-κB-response elements within the Oxtr and Cx43 promoters. Increased CAP gene expression was accompanied by a pronounced decrease in enrichment of repressive histone marks and increase in enrichment of active histone marks to this genomic region. These changes in histone modification were associated with changes in expression of corresponding histone modifying enzymes. Myometrial tissues from P4-treated 18.5 dpc pregnant mice manifested increased Cnot1 expression at 18.5 dpc, compared to vehicle-treated controls. P4 treatment of PR-expressing hTERT-HM cells enhanced CNOT1 expression and its recruitment to PR bound NF-κB-response elements within the CX43 and OXTR promoters. Furthermore, knockdown of CNOT1 significantly increased expression of contractile genes. These novel findings suggest that decreased expression and DNA-binding of the P4/PR-regulated transcriptional corepressor CNOT1 near term and associated changes in histone modifications at the OXTR and CX43 promoters contribute to the induction of myometrial contractility leading to parturition.
Collapse
Affiliation(s)
- Youn-Tae Kwak
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Alina P Montalbano
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Andrew M Kelleher
- Department of Obstetrics & Gynecology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Laboratory of Signaling and Gene Regulation, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Cecil H. and Ida Green Center for Reproductive Biology Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Department of Obstetrics, Gynecology, and Women's Health, University of Missouri, Columbia, Missouri, USA
| | - Mariano Colon-Caraballo
- Department of Obstetrics & Gynecology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Cecil H. and Ida Green Center for Reproductive Biology Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - W Lee Kraus
- Laboratory of Signaling and Gene Regulation, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Cecil H. and Ida Green Center for Reproductive Biology Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Mala Mahendroo
- Department of Obstetrics & Gynecology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Cecil H. and Ida Green Center for Reproductive Biology Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Carole R Mendelson
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Department of Obstetrics & Gynecology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Cecil H. and Ida Green Center for Reproductive Biology Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; North Texas March of Dimes Birth Defects Center, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
5
|
DeTomaso A, Kim H, Shauh J, Adulla A, Zigo S, Ghoul M, Presicce P, Kallapur SG, Goodman W, Tilburgs T, Way SS, Hackney D, Moore J, Mesiano S. Progesterone inactivation in decidual stromal cells: A mechanism for inflammation-induced parturition. Proc Natl Acad Sci U S A 2024; 121:e2400601121. [PMID: 38861608 PMCID: PMC11194587 DOI: 10.1073/pnas.2400601121] [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: 01/12/2024] [Accepted: 04/25/2024] [Indexed: 06/13/2024] Open
Abstract
The process of human parturition involves inflammation at the interface where fetal chorion trophoblast cells interact with maternal decidual stromal (DS) cells and maternal immune cells in the decidua (endometrium of pregnancy). This study tested the hypothesis that inflammation at the chorion-decidua interface (CDI) induces labor by negating the capacity for progesterone (P4) to block labor and that this is mediated by inactivation of P4 in DS cells by aldo-keto reductase family 1 member C1 (AKR1C1). In human, Rhesus macaque, and mouse CDI, AKR1C1 expression increased in association with term and preterm labor. In a human DS cell line and in explant cultures of term human fetal membranes containing the CDI, the prolabor inflammatory cytokine, interleukin-1ß (IL-1ß), and media conditioned by LPS-stimulated macrophages increased AKR1C1 expression and coordinately reduced nuclear P4 levels and P4 responsiveness. Loss of P4 responsiveness was overcome by inhibition of AKR1C1 activity, inhibition of AKR1C1 expression, and bypassing AKR1C1 activity with a P4 analog that is not metabolized by AKR1C1. Increased P4 activity in response to AKR1C1 inhibition was prevented by the P4 receptor antagonist RU486. Pharmacologic inhibition of AKR1C1 activity prevented parturition in a mouse model of inflammation-induced preterm parturition. The data suggest that inflammatory stimuli at the CDI drive labor by inducing AKR1C1-mediated P4 inactivation in DS cells and that inhibiting and/or bypassing of AKR1C1-mediated P4 inactivation is a plausible therapeutic strategy to mitigate the risk of inflammation-associated preterm birth.
Collapse
Affiliation(s)
- Angela DeTomaso
- Department of Pathology, Case Western Reserve University, Cleveland, OH44106
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
| | - Hyeyon Kim
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
| | - Jacqueline Shauh
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
| | - Anika Adulla
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
| | - Sarah Zigo
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
| | - Maya Ghoul
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
| | - Pietro Presicce
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Suhas G. Kallapur
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA90095
| | - Wendy Goodman
- Department of Pathology, Case Western Reserve University, Cleveland, OH44106
| | - Tamara Tilburgs
- Cincinnati Children’s Hospital, Center for Inflammation and Tolerance, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH45229
| | - Sing-Sing Way
- Cincinnati Children’s Hospital, Center for Inflammation and Tolerance, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH45229
| | - David Hackney
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
- Department of Obstetrics and Gynecology, University Hospitals, Cleveland, OH44106
| | - John Moore
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH44106
| | - Sam Mesiano
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH44106
- Department of Obstetrics and Gynecology, University Hospitals, Cleveland, OH44106
| |
Collapse
|
6
|
Shynlova O, Nadeem L, Lye S. Progesterone control of myometrial contractility. J Steroid Biochem Mol Biol 2023; 234:106397. [PMID: 37683774 DOI: 10.1016/j.jsbmb.2023.106397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/01/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023]
Abstract
During pregnancy, the primary function of the uterus is to be quiescent and not contract, which allows the growing fetus to develop and mature. A uterine muscle layer, myometrium, is composed of smooth muscle cells (SMCs). Before the onset of labor contractions, the uterine SMCs experience a complex biochemical and molecular transformation involving the expression of contraction-associated proteins. Labor is initiated when genes in SMCs are activated in response to a combination of hormonal, inflammatory and mechanical signals. In this review, we provide an overview of molecular mechanisms regulating the process of parturition in humans, focusing on the hormonal control of the myometrium, particularly the steroid hormone progesterone. The primary reason for discussing the regulation of myometrial contractility by progesterone is the importance of the clinical problem of preterm birth. It is thought that the hormonal mechanisms regulating premature uterine contractions represent an untimely triggering of the normal events occurring during term parturition. Yet, our knowledge of the complex and redundant hormonal pathways controlling uterine contractile activity leading to delivery of the neonate remains incomplete. Finally, we introduce recent animal studies using a novel class of drugs, Selective Progesterone Receptor Modulators, targeting progesterone signaling to prevent premature myometrial contractions.
Collapse
Affiliation(s)
- Oksana Shynlova
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto M5G 1X5, Canada; Department of Physiology, University of Toronto, M5S 1A1, Canada; Department of Obstetrics & Gynecology, University of Toronto, M5S 1A1, Canada.
| | - Lubna Nadeem
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto M5G 1X5, Canada
| | - Stephen Lye
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto M5G 1X5, Canada; Department of Physiology, University of Toronto, M5S 1A1, Canada; Department of Obstetrics & Gynecology, University of Toronto, M5S 1A1, Canada
| |
Collapse
|
7
|
Kennedy EA, Jurado KA. IL-33 is alarmin the uterus for labor. Immunity 2023; 56:467-469. [PMID: 36921571 DOI: 10.1016/j.immuni.2023.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Timely labor is critical for both infant and maternal health, yet the mechanisms underlying the initiation of childbirth remain unclear. In this issue of Immunity, Siewiera et al. demonstrate a vital role for innate type 2 immune responses in controlling uterus-intrinsic onset of labor in mice.1.
Collapse
Affiliation(s)
- Elizabeth A Kennedy
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Kellie Ann Jurado
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Paul M, Zakar T, Phung J, Gregson A, Barreda AP, Butler TA, Walker FR, Pennell C, Smith R, Paul JW. 20α-Hydroxysteroid Dehydrogenase Expression in the Human Myometrium at Term and Preterm Birth: Relationships to Fetal Sex and Maternal Body Mass Index. Reprod Sci 2023:10.1007/s43032-023-01183-2. [PMID: 36765000 DOI: 10.1007/s43032-023-01183-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 01/27/2023] [Indexed: 02/12/2023]
Abstract
The mechanism by which human labor is initiated in the presence of elevated circulating progesterone levels remains unknown. Recent evidence indicates that the progesterone-metabolizing enzyme, 20α-hydroxysteroid dehydrogenase (20α-HSD), encoded by the gene AKR1C1, may contribute to functional progesterone withdrawal. We found that AKR1C1 expression significantly increased with labor onset in term myometrium, but not in preterm myometrium. Among preterm laboring deliveries, clinically diagnosed chorioamnionitis was associated with significantly elevated AKR1C1 expression. AKR1C1 expression positively correlated with BMI before labor and negatively correlated with BMI during labor. Analysis by fetal sex showed that AKR1C1 expression was significantly higher in women who delivered male babies compared to women who delivered female babies at term, but not preterm. Further, in pregnancies where the fetus was female, AKR1C1 expression positively correlated with the mother's age and BMI at the time of delivery. In conclusion, the increase in myometrial AKR1C1 expression with term labor is consistent with 20α-HSD playing a role in local progesterone metabolism to promote birth. Interestingly, this role appears to be specific to term pregnancies where the fetus is male. Upregulated AKR1C1 expression in the myometrium at preterm in-labor with clinical chorioamnionitis suggests that increased 20α-HSD activity is a mechanism through which inflammation drives progesterone withdrawal in preterm labor. The link between AKR1C1 expression and maternal BMI may provide insight into why maternal obesity is often associated with dysfunctional labor. Higher myometrial AKR1C1 expression in male pregnancies may indicate fetal sex-related differences in the mechanisms that precipitate labor onset at term.
Collapse
Affiliation(s)
- Marina Paul
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
- Centre for Rehab Innovations, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Tamas Zakar
- Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Centre, New Lambton Heights, NSW, 2305, Australia
- John Hunter Hospital, New Lambton Heights, NSW, 2305, Australia
| | - Jason Phung
- Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Centre, New Lambton Heights, NSW, 2305, Australia
- John Hunter Hospital, New Lambton Heights, NSW, 2305, Australia
| | - Amy Gregson
- Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Centre, New Lambton Heights, NSW, 2305, Australia
| | - Anna Paredes Barreda
- Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Centre, New Lambton Heights, NSW, 2305, Australia
| | - Trent A Butler
- John Hunter Hospital, New Lambton Heights, NSW, 2305, Australia
| | - Frederick R Walker
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
- Centre for Rehab Innovations, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Craig Pennell
- Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Centre, New Lambton Heights, NSW, 2305, Australia
- John Hunter Hospital, New Lambton Heights, NSW, 2305, Australia
| | - Roger Smith
- Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia
- Mothers and Babies Research Centre, New Lambton Heights, NSW, 2305, Australia
- John Hunter Hospital, New Lambton Heights, NSW, 2305, Australia
| | - Jonathan W Paul
- Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia.
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, 2308, Australia.
- Mothers and Babies Research Centre, New Lambton Heights, NSW, 2305, Australia.
| |
Collapse
|
9
|
YOMOGITA H, ITO H, HASHIMOTO K, KUDO A, FUKUSHIMA T, ENDO T, HIRATE Y, AKIMOTO Y, KOMADA M, KANAI Y, MIYASAKA N, KANAI-AZUMA M. A possible function of Nik-related kinase in the labyrinth layer of delayed delivery mouse placentas. J Reprod Dev 2023; 69:32-40. [PMID: 36567126 PMCID: PMC9939280 DOI: 10.1262/jrd.2022-120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In mice and humans, Nik-related protein kinase (Nrk) is an X-linked gene that encodes a serine/threonine kinase belonging to GCK group 4. Nrk knockout (Nrk KO) mice exhibit delayed delivery, possibly due to defective communication between the Nrk KO conceptus and its mother. However, the mechanism of delayed labor remains largely unknown. Here, we found that in pregnant mothers with the Nrk KO conceptus, the serum progesterone (P4) and placental lactogen (PL-2) concentrations in late pregnancy were higher than those in the wild type. Moreover, we demonstrated that Nrk is expressed in trophoblast giant cells (TGCs) and syncytiotrophoblast-2 (SynT-2) in the labyrinth layer of the mouse placenta. In the human placenta, NRK is also expressed in Syn-T in villi. Both human Syn-T and mouse TGCs of the labyrinth layer are present within fetal tissues that are in direct contact with the maternal blood. The labyrinth layer of the Nrk KO conceptus was gigantic, with enlarged cytoplasm and Golgi bodies in the TGCs. To investigate the function of Nrk in the labyrinth layer, a differentially expressed gene (DEG) analysis was performed. The DEG analysis revealed that labor-promoting factors, such as prostaglandins, were decreased, and pregnancy-maintaining factors, such as the prolactin family and P4 receptor, were increased. These findings suggest that the Nrk KO mice exhibit delayed delivery owing to high P4 concentrations caused by the hypersecretion of pregnancy-maintaining factors, such as PL-2, from the placenta.
Collapse
Affiliation(s)
- Hiroshi YOMOGITA
- Department of Perinatal and Women’s Medicine, Tokyo Medical and Dental University, Tokyo 113-8510, Japan,Center for Experimental Animals, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Hikaru ITO
- Center for Experimental Animals, Tokyo Medical and Dental University, Tokyo 113-8510, Japan,Research Facility Center for Science and Technology, Kagawa University, Kagawa 761-0793, Japan
| | - Kento HASHIMOTO
- Center for Experimental Animals, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Akihiko KUDO
- Department of Microscopic Anatomy, Kyorin University School of Medicine, Tokyo 181-8611, Japan
| | - Toshiaki FUKUSHIMA
- Cell Biology Center, Tokyo Institute of Technology, Kanagawa 226-8503, Japan
| | - Tsutomu ENDO
- Center for Experimental Animals, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Yoshikazu HIRATE
- Center for Experimental Animals, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Yoshihiro AKIMOTO
- Department of Microscopic Anatomy, Kyorin University School of Medicine, Tokyo 181-8611, Japan
| | - Masayuki KOMADA
- Cell Biology Center, Tokyo Institute of Technology, Kanagawa 226-8503, Japan
| | - Yoshiakira KANAI
- Department of Veterinary Anatomy, University of Tokyo, Tokyo 113-8657, Japan
| | - Naoyuki MIYASAKA
- Department of Perinatal and Women’s Medicine, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Masami KANAI-AZUMA
- Center for Experimental Animals, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| |
Collapse
|
10
|
Detlefsen AJ, Paulukinas RD, Penning TM. Germline Mutations in Steroid Metabolizing Enzymes: A Focus on Steroid Transforming Aldo-Keto Reductases. Int J Mol Sci 2023; 24:1873. [PMID: 36768194 PMCID: PMC9915212 DOI: 10.3390/ijms24031873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Steroid hormones synchronize a variety of functions throughout all stages of life. Importantly, steroid hormone-transforming enzymes are ultimately responsible for the regulation of these potent signaling molecules. Germline mutations that cause dysfunction in these enzymes cause a variety of endocrine disorders. Mutations in SRD5A2, HSD17B3, and HSD3B2 genes that lead to disordered sexual development, salt wasting, and other severe disorders provide a glimpse of the impacts of mutations in steroid hormone transforming enzymes. In a departure from these established examples, this review examines disease-associated germline coding mutations in steroid-transforming members of the human aldo-keto reductase (AKR) superfamily. We consider two main categories of missense mutations: those resulting from nonsynonymous single nucleotide polymorphisms (nsSNPs) and cases resulting from familial inherited base pair substitutions. We found mutations in human AKR1C genes that disrupt androgen metabolism, which can affect male sexual development and exacerbate prostate cancer and polycystic ovary syndrome (PCOS). Others may be disease causal in the AKR1D1 gene that is responsible for bile acid deficiency. However, given the extensive roles of AKRs in steroid metabolism, we predict that with expanding publicly available data and analysis tools, there is still much to be uncovered regarding germline AKR mutations in disease.
Collapse
Affiliation(s)
- Andrea J. Detlefsen
- Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ryan D. Paulukinas
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Trevor M. Penning
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
11
|
An In Vivo Screening Model for Investigation of Pathophysiology of Human Implantation Failure. Biomolecules 2022; 13:biom13010079. [PMID: 36671464 PMCID: PMC9856033 DOI: 10.3390/biom13010079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/14/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
To improve current infertility treatments, it is important to understand the pathophysiology of implantation failure. However, many molecules are involved in the normal biological process of implantation and the roles of each molecule and the molecular mechanism are not fully understood. This review highlights the hemagglutinating virus of Japan (HVJ; Sendai virus) envelope (HVJ-E) vector, which uses inactivated viral particles as a local and transient gene transfer system to the murine uterus during the implantation period in order to investigate the molecular mechanism of implantation. In vivo screening in mice using the HVJ-E vector system suggests that signal transducer and activator of transcription-3 (Stat-3) could be a diagnostic and therapeutic target for women with a history of implantation failure. The HVJ-E vector system hardly induces complete defects in genes; however, it not only suppresses but also transiently overexpresses some genes in the murine uterus. These features may be useful in investigating the pathophysiology of implantation failure in women.
Collapse
|
12
|
The effect of pregnancy on meibomian gland, tear film, cornea and anterior segment parameters. Photodiagnosis Photodyn Ther 2022; 40:103070. [PMID: 35987462 DOI: 10.1016/j.pdpdt.2022.103070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 12/14/2022]
Abstract
AIM In this prospective study, we aimed to examine the effect of physiological and pathological changes that occur during pregnancy in regard to Meibomian Gland (MG) structure, tear film, cornea and anterior segment parameters. METHODS The following groups were compared: 49 eyes of 49 pregnant women at 16-20 weeks of pregnancy (P16 Group), 46 eyes of 46 pregnant women at 32-36 weeks of pregnancy (P32 Group) and 51 eyes of 51 participants who were not pregnant (P0 Group). The groups were compared in terms of the first break-up time (NIF-BUT) and average break-up time (NIAvg-BUT) values. Non-contact meibography and MG loss rates were also compared. RESULTS The groups were found to be compatible in terms of age (P=0.052). The mean NIF-BUT values in the P16, P32 and P0 groups were 4.7 ±2.7, 6 ±3 and 6.7 ±3.1 seconds, respectively (P=0.055). The mean MG loss rates for the upper lid in the P16, P32 and P0 groups were 35.3%±12.6, 33.4%±11.4 and 15.5%±5.4, respectively. The loss rates for the lower lid in the P16, P32 and P0 groups were found to be 40.5%±18.6, 40.5%± 14.4 and 20.1%±8.1, respectively (P=0.000, p=0.000). The mean anterior chamber value (ACV) was found in the P16, P32 and P0 groups with 160.8 ±31.8, 150.9 ±26.5 and 165.9 ±26.5 µm3, respectively (P=0.035). CONCLUSION MG loss was found to be higher in pregnant groups compared to the non-pregnant groups. We found minimal instability in the tear film of the pregnant groups. We believe that pregnant women should be followed closely in terms of ocular surface diseases.
Collapse
|
13
|
Yomogita H, Miyasaka N, Kanai-Azuma M. A Review of Delayed Delivery Models and the Analysis Method in Mice. J Dev Biol 2022; 10:jdb10020020. [PMID: 35645296 PMCID: PMC9149829 DOI: 10.3390/jdb10020020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 11/16/2022] Open
Abstract
In humans, the incidence of post-term delivery is 1–10%. Post-term delivery significantly increases the risk of cesarean section or neonatal intensive care unit (NICU) admission. Despite these serious challenges, the cause of prolonged delivery remains unclear. Several common factors of delayed parturition between mice and humans will help elucidate the mechanisms of pregnancy and labor. At present, gene modification techniques are rapidly developing; however, there are limited reviews available describing the mouse phenotype analysis as a human model for post-term delivery. We classified the delayed-labor mice into nine types according to their causes. In mice, progesterone (P₄) maintains pregnancy, and the most common cause of delayed labor is luteolysis failure. Other contributing factors include humoral molecules in the fetus/placenta, uterine contractile dysfunction, poor cervical ripening, and delayed implantation. The etiology of delayed parturition is overexpression of the pregnancy maintenance mechanism or suppression of the labor induction mechanism. Here, we describe how to investigated their causes using mouse genetic analysis. In addition, we generated a list to identify the causes. Our review will help understand the findings obtained using the mouse model, providing a foundation for conducting more systematic research on delayed delivery.
Collapse
Affiliation(s)
- Hiroshi Yomogita
- Department of Perinatal and Women’s Medicine, Tokyo Medical and Dental University, Tokyo 113-8510, Japan; (H.Y.); (N.M.)
- Center for Experimental Animals, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Naoyuki Miyasaka
- Department of Perinatal and Women’s Medicine, Tokyo Medical and Dental University, Tokyo 113-8510, Japan; (H.Y.); (N.M.)
| | - Masami Kanai-Azuma
- Center for Experimental Animals, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
- Correspondence: ; Tel.: +813-3813-6111
| |
Collapse
|
14
|
Progesterone Receptor Signaling in the Uterus Is Essential for Pregnancy Success. Cells 2022; 11:cells11091474. [PMID: 35563781 PMCID: PMC9104461 DOI: 10.3390/cells11091474] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 12/25/2022] Open
Abstract
The uterus plays an essential role in the reproductive health of women and controls critical processes such as embryo implantation, placental development, parturition, and menstruation. Progesterone receptor (PR) regulates key aspects of the reproductive function of several mammalian species by directing the transcriptional program in response to progesterone (P4). P4/PR signaling controls endometrial receptivity and decidualization during early pregnancy and is critical for the establishment and outcome of a successful pregnancy. PR is also essential throughout gestation and during labor, and it exerts critical roles in the myometrium, mainly by the specialized function of its two isoforms, progesterone receptor A (PR-A) and progesterone receptor B (PR-B), which display distinct and separate roles as regulators of transcription. This review summarizes recent studies related to the roles of PR function in the decidua and myometrial tissues. We discuss how PR acquired key features in placental mammals that resulted in a highly specialized and dynamic role in the decidua. We also summarize recent literature that evaluates the myometrial PR-A/PR-B ratio at parturition and discuss the efficacy of current treatment options for preterm birth.
Collapse
|
15
|
Overview of human 20 alpha-hydroxysteroid dehydrogenase (AKR1C1): Functions, regulation, and structural insights of inhibitors. Chem Biol Interact 2021; 351:109746. [PMID: 34780792 DOI: 10.1016/j.cbi.2021.109746] [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: 08/31/2021] [Revised: 10/28/2021] [Accepted: 11/10/2021] [Indexed: 11/22/2022]
Abstract
Human aldo-keto reductase family 1C1 (AKR1C1) is an important enzyme involved in human hormone metabolism, which is mainly responsible for the metabolism of progesterone in the human body. AKR1C1 is highly expressed and has an important relationship with the occurrence and development of various diseases, especially some cancers related to hormone metabolism. Nowadays, many inhibitors against AKR1C1 have been discovered, including some synthetic compounds and natural products, which have certain inhibitory activity against AKR1C1 at the target level. Here we briefly reviewed the physiological and pathological functions of AKR1C1 and the relationship with the disease, and then summarized the development of AKR1C1 inhibitors, elucidated the interaction between inhibitors and AKR1C1 through molecular docking results and existing co-crystal structures. Finally, we discussed the design ideals of selective AKR1C1 inhibitors from the perspective of AKR1C1 structure, discussed the prospects of AKR1C1 in the treatment of human diseases in terms of biomarkers, pre-receptor regulation and single nucleotide polymorphisms, aiming to provide new ideas for drug research targeting AKR1C1.
Collapse
|
16
|
Gene Expression Profiling of Skeletal Muscles. Genes (Basel) 2021; 12:genes12111718. [PMID: 34828324 PMCID: PMC8621074 DOI: 10.3390/genes12111718] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/21/2021] [Accepted: 10/27/2021] [Indexed: 12/13/2022] Open
Abstract
Next-generation sequencing provides an opportunity for an in-depth biocomputational analysis to identify gene expression patterns between soleus and tibialis anterior, two well-characterized skeletal muscles, and analyze their gene expression profiling. RNA read counts were analyzed for differential gene expression using the R package edgeR. Differentially expressed genes were filtered using a false discovery rate of less than 0.05 c, a fold-change value of more than twenty, and an association with overrepresented pathways based on the Reactome pathway over-representation analysis tool. Most of the differentially expressed genes associated with soleus are coded for components of lipid metabolism and unique contractile elements. Differentially expressed genes associated with tibialis anterior encoded mostly for glucose and glycogen metabolic pathway regulatory enzymes and calcium-sensitive contractile components. These gene expression distinctions partly explain the genetic basis for skeletal muscle specialization, and they may help to explain skeletal muscle susceptibility to disease and drugs and further refine tissue engineering approaches.
Collapse
|
17
|
Tomac J, Mazor M, Lisnić B, Golemac M, Kveštak D, Bralić M, Bilić Zulle L, Brinkmann MM, Dölken L, Reinert LS, Paludan SR, Krmpotić A, Jonjić S, Juranić Lisnić V. Viral infection of the ovaries compromises pregnancy and reveals innate immune mechanisms protecting fertility. Immunity 2021; 54:1478-1493.e6. [PMID: 34015257 DOI: 10.1016/j.immuni.2021.04.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 11/29/2020] [Accepted: 04/16/2021] [Indexed: 12/16/2022]
Abstract
Viral infections during pregnancy are a considerable cause of adverse outcomes and birth defects, and the underlying mechanisms are poorly understood. Among those, cytomegalovirus (CMV) infection stands out as the most common intrauterine infection in humans, putatively causing early pregnancy loss. We employed murine CMV as a model to study the consequences of viral infection on pregnancy outcome and fertility maintenance. Even though pregnant mice successfully controlled CMV infection, we observed highly selective, strong infection of corpus luteum (CL) cells in their ovaries. High infection densities indicated complete failure of immune control in CL cells, resulting in progesterone insufficiency and pregnancy loss. An abundance of gap junctions, absence of vasculature, strong type I interferon (IFN) responses, and interaction of innate immune cells fully protected the ovarian follicles from viral infection. Our work provides fundamental insights into the effect of CMV infection on pregnancy loss and mechanisms protecting fertility.
Collapse
Affiliation(s)
- Jelena Tomac
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia
| | - Marija Mazor
- Center for Proteomics, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia
| | - Berislav Lisnić
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia; Center for Proteomics, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia
| | - Mijo Golemac
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia
| | - Daria Kveštak
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia
| | - Marina Bralić
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia
| | - Lidija Bilić Zulle
- Clinical Hospital Rijeka, University of Rijeka, B. Branchetta 20, 51000 Rijeka, Croatia
| | - Melanie M Brinkmann
- Viral Immune Modulation Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; Institute of Genetics, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Lars Dölken
- Institute for Virology and Immunobiology, Julius-Maximilians-Universität Würzburg, 97080 Würzburg, Germany; Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Centre for Infection Research, 97080 Würzburg, Germany
| | - Line S Reinert
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Soren R Paludan
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Astrid Krmpotić
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia
| | - Stipan Jonjić
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia; Center for Proteomics, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia.
| | - Vanda Juranić Lisnić
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia; Center for Proteomics, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia.
| |
Collapse
|
18
|
Nadeem L, Balendran R, Dorogin A, Mesiano S, Shynlova O, Lye SJ. Pro-inflammatory signals induce 20α-HSD expression in myometrial cells: A key mechanism for local progesterone withdrawal. J Cell Mol Med 2021; 25:6773-6785. [PMID: 34114342 PMCID: PMC8278114 DOI: 10.1111/jcmm.16681] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 01/03/2023] Open
Abstract
Metabolism of progesterone (P4) by the enzyme 20α hydroxysteroid dehydrogenase (20α‐HSD) in myometrial cells is postulated to be a mechanism for P4 withdrawal, which occurs concomitant to uterine inflammation (physiologic or infection‐induced) and associated activation of transcription factors: NF‐кB and AP‐1, common to term and preterm labour. We found that 20α‐HSD protein is significantly increased in human myometrium during term labour, and in mouse uterus during term and preterm labour. Treatment of human myometrial cells with the pro‐inflammatory mediators, lipopolysaccharide (LPS, mimicking infection) and 12‐O‐tetradecanoylphorbol‐13‐acetate (TPA, mimicking inflammation), induced 20α‐HSD gene expression and increased 20α‐HSD protein abundance. LPS treatment decreased P4 release into the culture medium and resulted in up‐regulation of GJA1 in the hTERT‐HM cells. The NF‐кB /AP‐1 transcription factors mediated effects of LPS and TPA on 20α‐HSD gene transcription. Both pro‐inflammatory stimuli induced 20α‐HSD promoter activity in LPS/TPA‐treated cells which was significantly attenuated by inhibition of NF‐кB (JSH: 20 µM) or AP‐1 signalling (T5224: 10 µM). Deletion of NF‐кB consensus sites abrogated LPS‐mediated promoter induction, while removal of AP‐1 sites reversed the TPA‐mediated induction of 20α‐HSD promoter. We conclude that inflammatory stimuli (physiologic or pathologic) that activate NF‐кB or AP‐1 induce 20α‐HSD transcription and subsequent local P4 withdrawal resulting in up‐regulation of GJA1 and activation of myometrium that precedes labour.
Collapse
Affiliation(s)
- Lubna Nadeem
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Rathesh Balendran
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Anna Dorogin
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Sam Mesiano
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Oksana Shynlova
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics & Gynecology, University of Toronto, Toronto, ON, Canada
| | - Stephen J Lye
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics & Gynecology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
19
|
Corneal Biomechanical Changes in Third Trimester of Pregnancy. ACTA ACUST UNITED AC 2021; 57:medicina57060600. [PMID: 34200925 PMCID: PMC8230443 DOI: 10.3390/medicina57060600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 12/22/2022]
Abstract
Background and Objectives: There is a clear evidence that pregnancy is associated with high production of sex hormones. During the first, second and third trimester of pregnancy, blood hormones levels increase gradually. Cells with affinity for sex hormones have been identified in different ocular tissues, such as: lid, lacrimal gland, meibomian gland, bulbar and palpebral conjunctivae, cornea, iris, ciliary body, lens, retina (retinal pigment epithelium) and choroid. This is why pregnancy is associated with changes at ocular level, involving anterior and posterior segments. Several clinical trials have been made trying to highlight changes in corneal biomechanics during pregnancy. By conducting this review, we want to evaluate both the changes in parameters that define corneal biomechanics and intraocular pressure values in pregnant. Materials and Methods: Following a systematic search in the literature related mainly to changes in corneal biomechanics during pregnancy, focusing on the paper published in the last decade, we included in a meta-analysis the cumulative results of three prospective comparative studies. Results: Important changes in corneal biomechanics (corneal hysteresis and corneal resistance factor) parameters were observed in women in the third trimester of pregnancy, but these variations were not statistically significant. Also, a decrease in intraocular pressure was mentioned in these women, but only the corneal compensation intraocular pressure showed a decrease with statistical significance. Conclusions: A decrease in corneal compensatory intraocular pressure was observed in pregnant women in the third trimester of pregnancy, but without other statistically significant changes resulting from the analysis of the other three parameters (corneal hysteresis, corneal resistance factor and Goldmann-correlated intraocular pressure).
Collapse
|
20
|
Khader N, Shchuka VM, Shynlova O, Mitchell JA. Transcriptional control of parturition: insights from gene regulation studies in the myometrium. Mol Hum Reprod 2021; 27:gaab024. [PMID: 33823545 PMCID: PMC8126590 DOI: 10.1093/molehr/gaab024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/09/2021] [Indexed: 12/19/2022] Open
Abstract
The onset of labour is a culmination of a series of highly coordinated and preparatory physiological events that take place throughout the gestational period. In order to produce the associated contractions needed for foetal delivery, smooth muscle cells in the muscular layer of the uterus (i.e. myometrium) undergo a transition from quiescent to contractile phenotypes. Here, we present the current understanding of the roles transcription factors play in critical labour-associated gene expression changes as part of the molecular mechanistic basis for this transition. Consideration is given to both transcription factors that have been well-studied in a myometrial context, i.e. activator protein 1, progesterone receptors, oestrogen receptors, and nuclear factor kappa B, as well as additional transcription factors whose gestational event-driving contributions have been demonstrated more recently. These transcription factors may form pregnancy- and labour-associated transcriptional regulatory networks in the myometrium to modulate the timing of labour onset. A more thorough understanding of the transcription factor-mediated, labour-promoting regulatory pathways holds promise for the development of new therapeutic treatments that can be used for the prevention of preterm labour in at-risk women.
Collapse
Affiliation(s)
- Nawrah Khader
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Virlana M Shchuka
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Oksana Shynlova
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Obstetrics & Gynaecology, University of Toronto, ON, Canada
| | - Jennifer A Mitchell
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
21
|
Verma P, Hassan MI, Singh A, Singh IK. Design and development of novel inhibitors of aldo-ketoreductase 1C1 as potential lead molecules in treatment of breast cancer. Mol Cell Biochem 2021; 476:2975-2987. [PMID: 33770316 DOI: 10.1007/s11010-021-04134-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/06/2021] [Indexed: 11/29/2022]
Abstract
Aldo-keto reductase 1C1 (AKR1C1) is a hydroxysteroid dehydrogenase, known to inactivate the biologically active progesterone into its corresponding 20 α-hydroxyprogesterone. Increased expression of the AKR1C1 gene in oncogenesis is linked with resistance to various anticancer agents and hence it is considered as an emerging drug target for the design and developing the novel anticancer drugs. We have performed QSAR pharmacophore modeling for AKR1C1 inhibitors followed by a virtual screening of ~ 59,000 compounds present at the Maybridge database. The screened compounds were refined using drug-like filters of Lipinski rule, ADMET plot, molecular docking and scoring and subsequently top 20 hits were selected. Selected compounds were subjected to the in vitro for AKR1C1 inhibition assay and best seven compounds bearing excellent binding affinity to the AKR1C1 were finally selected. The identified compounds may be exploited in hit-to-lead development and may also prove as an interventional strategy in preventing a pre-term birth due to declining levels of progesterone.
Collapse
Affiliation(s)
- Priyanka Verma
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, 110019, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Archana Singh
- Department of Botany, Hansraj College, University of Delhi, New Delhi, 110007, India.
| | - Indrakant K Singh
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, 110019, India. .,Deshbandhu College, University of Delhi, New Delhi, 110019, India.
| |
Collapse
|
22
|
Decidual cell FKBP51-progesterone receptor binding mediates maternal stress-induced preterm birth. Proc Natl Acad Sci U S A 2021; 118:2010282118. [PMID: 33836562 PMCID: PMC7980401 DOI: 10.1073/pnas.2010282118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Depression and posttraumatic stress disorder increase the risk of idiopathic preterm birth (iPTB); however, the exact molecular mechanism is unknown. Depression and stress-related disorders are linked to increased FK506-binding protein 51 (FKBP51) expression levels in the brain and/or FKBP5 gene polymorphisms. Fkbp5-deficient (Fkbp5 -/-) mice resist stress-induced depressive and anxiety-like behaviors. FKBP51 binding to progesterone (P4) receptors (PRs) inhibits PR function. Moreover, reduced PR activity and/or expression stimulates human labor. We report enhanced in situ FKBP51 expression and increased nuclear FKBP51-PR binding in decidual cells of women with iPTB versus gestational age-matched controls. In Fkbp5 +/+ mice, maternal restraint stress did not accelerate systemic P4 withdrawal but increased Fkbp5, decreased PR, and elevated AKR1C18 expression in uteri at E17.25 followed by reduced P4 levels and increased oxytocin receptor (Oxtr) expression at 18.25 in uteri resulting in PTB. These changes correlate with inhibition of uterine PR function by maternal stress-induced FKBP51. In contrast, Fkbp5 -/- mice exhibit prolonged gestation and are completely resistant to maternal stress-induced PTB and labor-inducing uterine changes detected in stressed Fkbp5 +/+ mice. Collectively, these results uncover a functional P4 withdrawal mechanism mediated by maternal stress-induced enhanced uterine FKBP51 expression and FKPB51-PR binding, resulting in iPTB.
Collapse
|
23
|
Luo Y, Qiao X, Ma Y, Deng H, Xu CC, Xu L. Irisin deletion induces a decrease in growth and fertility in mice. Reprod Biol Endocrinol 2021; 19:22. [PMID: 33581723 PMCID: PMC7881587 DOI: 10.1186/s12958-021-00702-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 01/28/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Irisin, which is cleaved from fibronectin type III domain-containing protein 5 (Fndc5), plays an important role in energy homeostasis. The link between energy metabolism and reproduction is well known. However, the biological actions of irisin in reproduction remain largely unexplored. METHODS In this study, we generated Fndc5 gene mutation to create irisin deficient mice. Female wild-type (WT) and Fndc5 mutant mice were fed with standard chow for 48 weeks. Firstly, the survival rate, body weight and fertility were described in mice. Secondly, the levels of steroid hormones in serum were measured by ELISA, and the estrus cycle and the appearance of follicles were determined by vaginal smears and ovarian continuous sections. Thirdly, mRNA-sequencing analysis was used to compare gene expression between the ovaries of Fndc5 mutant mice and those of WT mice. Finally, the effects of exogenous irisin on steroid hormone production was investigated in KGN cells. RESULTS The mice lacking irisin presented increased mortality, reduced body weight and poor fertility. Analysis of sex hormones showed decreased levels of estradiol, follicle-stimulating hormone and luteinizing hormone, and elevated progesterone levels in Fndc5 mutant mice. Irisin deficiency in mice was associated with irregular estrus, reduced ratio of antral follicles. The expressions of Akr1c18, Mamld1, and Cyp19a1, which are involved in the synthesis of steroid hormones, were reduced in the ovaries of mutant mice. Exogenous irisin could promote the expression of Akr1c18, Mamld1, and Cyp19a1 in KGN cells, stimulating estradiol production and inhibiting progesterone secretion. CONCLUSIONS Irisin deficiency was related to disordered endocrinology metabolism in mice. The irisin deficient mice showed poor growth and development, and decreased fertility. Irisin likely have effects on the expressions of Akr1c18, Mamld1 and Cyp19a1 in ovary, regulating the steroid hormone production. This study provides novel insights into the potential role of irisin in mammalian growth and reproduction.
Collapse
Affiliation(s)
- Yunyao Luo
- Reproductive Endocrinology and Regulation Laboratory West China Second University Hospital, Sichuan University, #20 Section 3, Ren Min Nan Road, Chengdu, 610041, Sichuan, China
- The Joint Laboratory for Reproductive Medicine of Sichuan University-The Chinese University of Hong Kong, Chengdu, People's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) Ministry of Education, Chengdu, People's Republic of China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiaoyong Qiao
- Reproductive Endocrinology and Regulation Laboratory West China Second University Hospital, Sichuan University, #20 Section 3, Ren Min Nan Road, Chengdu, 610041, Sichuan, China
- The Joint Laboratory for Reproductive Medicine of Sichuan University-The Chinese University of Hong Kong, Chengdu, People's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) Ministry of Education, Chengdu, People's Republic of China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yaxian Ma
- Reproductive Endocrinology and Regulation Laboratory West China Second University Hospital, Sichuan University, #20 Section 3, Ren Min Nan Road, Chengdu, 610041, Sichuan, China
- The Joint Laboratory for Reproductive Medicine of Sichuan University-The Chinese University of Hong Kong, Chengdu, People's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) Ministry of Education, Chengdu, People's Republic of China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hongxia Deng
- Reproductive Endocrinology and Regulation Laboratory West China Second University Hospital, Sichuan University, #20 Section 3, Ren Min Nan Road, Chengdu, 610041, Sichuan, China
- The Joint Laboratory for Reproductive Medicine of Sichuan University-The Chinese University of Hong Kong, Chengdu, People's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) Ministry of Education, Chengdu, People's Republic of China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Charles C Xu
- College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Liangzhi Xu
- Reproductive Endocrinology and Regulation Laboratory West China Second University Hospital, Sichuan University, #20 Section 3, Ren Min Nan Road, Chengdu, 610041, Sichuan, China.
- The Joint Laboratory for Reproductive Medicine of Sichuan University-The Chinese University of Hong Kong, Chengdu, People's Republic of China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) Ministry of Education, Chengdu, People's Republic of China.
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
24
|
Abstract
Term labour is a state of physiological inflammation orchestrated by multiple uterine tissues (both fetal and maternal). This physiological inflammation preceding and accompanying labour onset is characterized by an increase in cytokine and chemokine secretion by the fetal membranes, as well as uterine tissues (i.e., decidua and myometrium). Pro-inflammatory cytokines and chemokines activate circulating maternal peripheral leukocytes as well as the uterine vascular endothelium to permit leukocyte infiltration into the uterus. This inflammatory milieu, in the absence of infection, is required for the initiation of labour as the uterine-infiltrated leukocytes secrete matrix metalloproteinases to induce fetal membrane rupture and cervical ripening as well as various labour mediators, which promote contractions of the myometrium. Myometrial activation at term and the onset of labour contractions are directly related to the changes in the ovarian/placental hormone progesterone and its downstream mediators (i.e., the progesterone receptors, PRA/B), which are also critical for maintenance of pregnancy. Our recent data provides direct evidence in support of local and functional P4 withdrawal in the uterine muscle (myometrium) via the activator protein-1 (AP-1) mediated pathway. This review outlines known mechanisms regulating activation of human labour, including progesterone and cytokine signaling. Understanding of the molecular mechanism of myometrial activation and labour onset could facilitate the development of new therapeutics for high-risk pregnant women to prevent premature uterine activation and preterm birth.
Collapse
Affiliation(s)
- Oksana Shynlova
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada; Departments of Physiology and University of Toronto, Ontario, Canada; Obstetrics & Gynecology, University of Toronto, Ontario, Canada.
| | - Lubna Nadeem
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Jianhong Zhang
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Caroline Dunk
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Stephen Lye
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada; Departments of Physiology and University of Toronto, Ontario, Canada; Obstetrics & Gynecology, University of Toronto, Ontario, Canada
| |
Collapse
|
25
|
Green ES, Arck PC. Pathogenesis of preterm birth: bidirectional inflammation in mother and fetus. Semin Immunopathol 2020; 42:413-429. [PMID: 32894326 PMCID: PMC7508962 DOI: 10.1007/s00281-020-00807-y] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/14/2020] [Indexed: 12/18/2022]
Abstract
Preterm birth (PTB) complicates 5–18% of pregnancies globally and is a leading cause of maternal and fetal morbidity and mortality. Most PTB is spontaneous and idiopathic, with largely undefined causes. To increase understanding of PTB, much research in recent years has focused on using animal models to recapitulate the pathophysiology of PTB. Dysfunctions of maternal immune adaptations have been implicated in a range of pregnancy pathologies, including PTB. A wealth of evidence arising from mouse models as well as human studies is now available to support that PTB results from a breakdown in fetal-maternal tolerance, along with excessive, premature inflammation. In this review, we examine the current knowledge of the bidirectional communication between fetal and maternal systems and its role in the immunopathogenesis of PTB. These recent insights significantly advance our understanding of the pathogenesis of PTB, which is essential to ultimately designing more effective strategies for early prediction and subsequent prevention of PTB.
Collapse
Affiliation(s)
- Ella Shana Green
- Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251, Hamburg, Germany
| | - Petra Clara Arck
- Department of Obstetrics and Fetal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20251, Hamburg, Germany.
| |
Collapse
|
26
|
Lee-Thacker S, Jeon H, Choi Y, Taniuchi I, Takarada T, Yoneda Y, Ko C, Jo M. Core Binding Factors are essential for ovulation, luteinization, and female fertility in mice. Sci Rep 2020; 10:9921. [PMID: 32555437 PMCID: PMC7303197 DOI: 10.1038/s41598-020-64257-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 03/12/2020] [Indexed: 12/17/2022] Open
Abstract
Core Binding Factors (CBFs) are a small group of heterodimeric transcription factor complexes composed of DNA binding proteins, RUNXs, and a non-DNA binding protein, CBFB. The LH surge increases the expression of Runx1 and Runx2 in ovulatory follicles, while Cbfb is constitutively expressed. To investigate the physiological significance of CBFs, we generated a conditional mutant mouse model in which granulosa cell expression of Runx2 and Cbfb was deleted by the Esr2Cre. Female Cbfbflox/flox;Esr2cre/+;Runx2flox/flox mice were infertile; follicles developed to the preovulatory follicle stage but failed to ovulate. RNA-seq analysis of mutant mouse ovaries collected at 11 h post-hCG unveiled numerous CBFs-downstream genes that are associated with inflammation, matrix remodeling, wnt signaling, and steroid metabolism. Mutant mice also failed to develop corpora lutea, as evident by the lack of luteal marker gene expression, marked reduction of vascularization, and excessive apoptotic staining in unruptured poorly luteinized follicles, consistent with dramatic reduction of progesterone by 24 h after hCG administration. The present study provides in vivo evidence that CBFs act as essential transcriptional regulators of both ovulation and luteinization by regulating the expression of key genes that are involved in inflammation, matrix remodeling, cell differentiation, vascularization, and steroid metabolisms in mice.
Collapse
Affiliation(s)
- Somang Lee-Thacker
- Department of Obstetrics and Gynecology, Chandler Medical Center, 800 Rose Street, University of Kentucky, Lexington, KY, 40536-0298, USA
| | - Hayce Jeon
- Department of Obstetrics and Gynecology, Chandler Medical Center, 800 Rose Street, University of Kentucky, Lexington, KY, 40536-0298, USA
| | - Yohan Choi
- Department of Obstetrics and Gynecology, Chandler Medical Center, 800 Rose Street, University of Kentucky, Lexington, KY, 40536-0298, USA
| | - Ichiro Taniuchi
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Takeshi Takarada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Yukio Yoneda
- Section of Prophylactic Pharmacology, Kanazawa University, Venture Business Laboratory 402, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - CheMyong Ko
- Department of Comparative Biosciences, College of Veterinary Medicine, 2001 South Lincoln Avenue, University of Illinois at Urbana-Champaign, Urbana, Illinois, 61802, USA
| | - Misung Jo
- Department of Obstetrics and Gynecology, Chandler Medical Center, 800 Rose Street, University of Kentucky, Lexington, KY, 40536-0298, USA.
| |
Collapse
|
27
|
Amano T, Ripperger JA, Albrecht U. Changing the light schedule in late pregnancy alters birth timing in mice. Theriogenology 2020; 154:212-222. [PMID: 32650187 DOI: 10.1016/j.theriogenology.2020.05.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/11/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022]
Abstract
In rats, birth timing is affected by changes in the light schedule until the middle of the pregnancy period. This phenomenon can be used to control birth timing in the animal industry and/or clinical fields. However, changes in the light schedule until the middle of the pregnancy period can damage the fetus by affecting the development of the major organs. Thus, we compared birth timing in mice kept under a 12-h light/12-h darkness schedule (L/D) throughout pregnancy with that of mice kept under a light schedule that changed from L/D to constant light (L/L) or constant darkness (D/D) from day 17.5 of pregnancy, the latter phase of the pregnancy period. On average, the pregnancy period was longer in D/D mice (19.9 days) than L/L or L/D mice (19.5 and 19.3 days, respectively, P < 0.05), confirming that light schedule affects birth timing. The average number of newborns was the same in L/L, L/D, and D/D mice (7.5, 7.8, and 7.9, respectively), but the average newborn weight of L/L mice (1.3 g) was lower than that of L/D and D/D mice (both 1.4 g, P < 0.05), indicating that constant light has detrimental effects on fetus growth. However, the percentage of dead newborns was the same between L/L, L/D, and D/D mice (11.1, 10.6, and 3.6%, respectively). The serum progesterone level on day 18.5 of pregnancy in L/D mice was 42.8 ng/ml, lower (P < 0.05) than that of D/D mice (65.3 ng/ml), suggesting that light schedule affects luteolysis. The average pregnancy period of mice lacking a circadian clock kept under D/D conditions from day 17.5 of pregnancy (KO D/D) (20.3 days) was delayed compared with wild-type (WT) D/D mice (P < 0.05). However, the average number of newborns, percentage of births with dead pups, and weight per newborn of KO D/D mice (7.6, 3.6%, and 1.4 g, respectively) were the same as WT mice kept under D/D conditions. A direct effect of the circadian clock on the mechanism(s) regulating birth timing was questionable, as the lighter average weight per KO fetus (0.6 g) versus WT fetus (0.7 g) on day 17.5 of pregnancy might have caused the delay in birth. The range of birth timing in KO D/D mice was the same as that of WT D/D mice, indicating that the circadian clock does not concentrate births at one time.
Collapse
Affiliation(s)
- Tomoko Amano
- College of Agriculture, Food and Environment Sciences, Department of Sustainable Agriculture, Laboratory of Animal Genetics, Rakuno Gakuen University, 582 Midorimachi Bunkyodai, Ebetsu, Hokkaido, 069-8501, Japan.
| | - Jürgen A Ripperger
- Department of Biology/Unit of Biochemistry, Faculty of Sciences, University of Fribourg, Chemin du Musée 5, CH-1700, Fribourg, Switzerland
| | - Urs Albrecht
- Department of Biology/Unit of Biochemistry, Faculty of Sciences, University of Fribourg, Chemin du Musée 5, CH-1700, Fribourg, Switzerland
| |
Collapse
|
28
|
Ransick A, Lindström NO, Liu J, Zhu Q, Guo JJ, Alvarado GF, Kim AD, Black HG, Kim J, McMahon AP. Single-Cell Profiling Reveals Sex, Lineage, and Regional Diversity in the Mouse Kidney. Dev Cell 2020; 51:399-413.e7. [PMID: 31689386 DOI: 10.1016/j.devcel.2019.10.005] [Citation(s) in RCA: 243] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/26/2019] [Accepted: 10/04/2019] [Indexed: 01/13/2023]
Abstract
Chronic kidney disease affects 10% of the population with notable differences in ethnic and sex-related susceptibility to kidney injury and disease. Kidney dysfunction leads to significant morbidity and mortality and chronic disease in other organ systems. A mouse-organ-centered understanding underlies rapid progress in human disease modeling and cellular approaches to repair damaged systems. To enhance an understanding of the mammalian kidney, we combined anatomy-guided single-cell RNA sequencing of the adult male and female mouse kidney with in situ expression studies and cell lineage tracing. These studies reveal cell diversity and marked sex differences, distinct organization and cell composition of nephrons dependent on the time of nephron specification, and lineage convergence, in which contiguous functionally related cell types are specified from nephron and collecting system progenitor populations. A searchable database, Kidney Cell Explorer (https://cello.shinyapps.io/kidneycellexplorer/), enables gene-cell relationships to be viewed in the anatomical framework of the kidney.
Collapse
Affiliation(s)
- Andrew Ransick
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Nils O Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Jing Liu
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Qin Zhu
- Graduate Program in Genomics and Computational Biology, Biomedical Graduate Studies, University of Pennsylvania, 160 BRB II/III - 421 Curie Blvd, Philadelphia, PA 19104-6064, USA
| | - Jin-Jin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Gregory F Alvarado
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Albert D Kim
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Hannah G Black
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Junhyong Kim
- Department of Biology, University of Pennsylvania, 415 S. University Ave, Philadelphia, PA 19104, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
29
|
The Interaction of lncRNA XLOC-2222497, AKR1C1, and Progesterone in Porcine Endometrium and Pregnancy. Int J Mol Sci 2020; 21:ijms21093232. [PMID: 32370225 DOI: 10.3390/ijms21093232] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/26/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022] Open
Abstract
The endometrium is an important tissue for pregnancy and plays an important role in reproduction. In this study, high-throughput transcriptome sequencing was performed in endometrium samples of Meishan and Yorkshire pigs on days 18 and 32 of pregnancy. Aldo-keto reductase family 1 member C1 (AKR1C1) was found to be a differentially expressed gene, and was identified by quantitative real-time PCR (qRT-PCR) and Western blot. Immunohistochemistry results revealed the cellular localization of the AKR1C1 protein in the endometrium. Luciferase activity assay demonstrated that the AKR1C1 core promoter region was located in the region from -706 to -564, containing two nuclear factor erythroid 2-related factor 2 (NRF2) binding sites (antioxidant response elements, AREs). XLOC-2222497 was identified as a nuclear long non-coding RNA (lncRNA) highly expressed in the endometrium. XLOC-2222497 overexpression and knockdown have an effect on the expression of AKR1C1. Endocrinologic measurement showed the difference in progesterone levels between Meishan and Yorkshire pigs. Progesterone treatment upregulated AKR1C1 and XLOC-2222497 expression in porcine endometrial epithelial cells. In conclusion, transcriptome analysis revealed differentially expressed transcripts during the early pregnancy process. Further experiments demonstrated the interaction of XLOC-2222497/AKR1C1/progesterone in the endometrium and provided new potential targets for pregnancy maintenance and its control.
Collapse
|
30
|
Timaxian C, Raymond-Letron I, Bouclier C, Gulliver L, Le Corre L, Chébli K, Guillou A, Mollard P, Balabanian K, Lazennec G. The health status alters the pituitary function and reproduction of mice in a Cxcr2-dependent manner. Life Sci Alliance 2020; 3:3/3/e201900599. [PMID: 32041848 PMCID: PMC7010316 DOI: 10.26508/lsa.201900599] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/18/2022] Open
Abstract
This study explores the effects of microbiota on reproductive function of Cxcr2 knockout animals. Cxcr2 is involved in the control of pituitary action and the subsequent development of mammary gland, uterus and ovary. Microbiota and chronic infections can affect not only immune status, but also the overall physiology of animals. Here, we report that chronic infections dramatically modify the phenotype of Cxcr2 KO mice, impairing in particular, their reproduction ability. We show that exposure of Cxcr2 KO females to multiple types of chronic infections prevents their ability to cycle, reduces the development of the mammary gland and alters the morphology of the uterus due to an impairment of ovary function. Mammary gland and ovary transplantation demonstrated that the hormonal contexture was playing a crucial role in this phenomenon. This was further evidenced by alterations to circulating levels of sex steroid and pituitary hormones. By analyzing at the molecular level the mechanisms of pituitary dysfunction, we showed that in the absence of Cxcr2, bystander infections affect leukocyte migration, adhesion, and function, as well as ion transport, synaptic function behavior, and reproduction pathways. Taken together, these data reveal that a chemokine receptor plays a direct role in pituitary function and reproduction in the context of chronic infections.
Collapse
Affiliation(s)
- Colin Timaxian
- Centre National de la Recherche Scientifique (CNRS), SYS2DIAG-ALCEDIAG, Cap Delta, Montpellier, France.,CNRS, Groupement de Recherche 3697 "Microenvironment of Tumor Niches," Micronit, France
| | - Isabelle Raymond-Letron
- Department of Histopathology, National Veterinary School of Toulouse, France and Platform of Experimental and Compared Histopathology, STROMALab, Unité de recherche mixte (UMR) Université Paul Sabatier/CNRS 5223, Etablissement français du sang, Institut national de la santé et de la recherche médicale (Inserm) U1031, Toulouse, France
| | - Céline Bouclier
- Centre National de la Recherche Scientifique (CNRS), SYS2DIAG-ALCEDIAG, Cap Delta, Montpellier, France
| | | | - Ludovic Le Corre
- Nutrition et Toxicologie Alimentaire (NUTOX) Laboratory - INSERM Lipides, Nutrition, Cancer UMR 1231 - AgrosupDijon, Dijon, France
| | - Karim Chébli
- Equipe Metazoan Messenger RNAs Metabolism, Montpellier, France
| | - Anne Guillou
- Institut de Génomique Fonctionnelle, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Patrice Mollard
- Institut de Génomique Fonctionnelle, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Karl Balabanian
- CNRS, Groupement de Recherche 3697 "Microenvironment of Tumor Niches," Micronit, France.,Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160, Paris, France
| | - Gwendal Lazennec
- Centre National de la Recherche Scientifique (CNRS), SYS2DIAG-ALCEDIAG, Cap Delta, Montpellier, France .,CNRS, Groupement de Recherche 3697 "Microenvironment of Tumor Niches," Micronit, France
| |
Collapse
|
31
|
Myometrial activation: Novel concepts underlying labor. Placenta 2020; 92:28-36. [PMID: 32056784 DOI: 10.1016/j.placenta.2020.02.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/21/2022]
Abstract
Term labour is a state of physiological inflammation orchestrated by multiple uterine tissues (both fetal and maternal). This physiological inflammation preceding and accompanying labour onset is characterized by an increase in cytokine and chemokine secretion by the fetal membranes, as well as uterine tissues (i.e., decidua and myometrium). Pro-inflammatory cytokines and chemokines activate circulating maternal peripheral leukocytes as well as the uterine vascular endothelium to permit leukocyte infiltration into the uterus. This inflammatory milieu, in the absence of infection, is required for the initiation of labour as the uterine-infiltrated leukocytes secrete matrix metalloproteinases to induce fetal membrane rupture and cervical ripening as well as various labour mediators, which promote contractions of the myometrium. Myometrial activation at term and the onset of labour contractions are directly related to the changes in the ovarian/placental hormone progesterone and its downstream mediators (i.e., the progesterone receptors, PRA/B), which are also critical for maintenance of pregnancy. Our recent data provides direct evidence in support of local and functional P4 withdrawal in the uterine muscle (myometrium) via the activator protein-1 (AP-1) mediated pathway. This review outlines known mechanisms regulating activation of human labour, including progesterone and cytokine signaling. Understanding of the molecular mechanism of myometrial activation and labour onset could facilitate the development of new therapeutics for high-risk pregnant women to prevent premature uterine activation and preterm birth.
Collapse
|
32
|
Solano ME, Arck PC. Steroids, Pregnancy and Fetal Development. Front Immunol 2020; 10:3017. [PMID: 32038609 PMCID: PMC6987319 DOI: 10.3389/fimmu.2019.03017] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 12/09/2019] [Indexed: 12/21/2022] Open
Abstract
Maternal glucocorticoids critically rise during pregnancy reaching up to a 20-fold increase of mid-pregnancy concentrations. Concurrently, another steroid hormone, progesterone, increases. Progesterone, which shows structural similarities to glucocorticoids, can bind the intracellular glucocorticoid receptor, although with lower affinity. Progesterone is essential for the establishment and continuation of pregnancy and it is generally acknowledged to promote maternal immune tolerance to fetal alloantigens through a wealth of immunomodulatory mechanisms. Despite the potent immunomodulatory capacity of glucocorticoids, little is known about their role during pregnancy. Here we aim to compare general aspects of glucocorticoids and progesterone during pregnancy, including shared common steroidogenic pathways, plasma transporters, regulatory pathways, expression of receptors, and mechanisms of action in immune cells. It was recently acknowledged that progesterone receptors are not ubiquitously expressed on immune cells and that pivotal features of progesterone induced- maternal immune adaptations to pregnancy are mediated via the glucocorticoid receptor, including e.g., T regulatory cells expansion. We hypothesize that a tight equilibrium between progesterone and glucocorticoids is critically required and recapitulate evidence supporting that their disequilibrium underlie pregnancy complications. Such a disequilibrium can occur, e.g., after maternal stress perception, which triggers the release of glucocorticoids and impair progesterone secretion, resulting in intrauterine inflammation. These endocrine misbalance might be interconnected, as increase in glucocorticoid synthesis, e.g., upon stress, may occur in detriment of progesterone steroidogenesis, by depleting the common precursor pregnenolone. Abundant literature supports that progesterone deficiency underlies pregnancy complications in which immune tolerance is challenged. In these settings, it is largely yet undefined if and how glucocorticoids are affected. However, although progesterone immunomodulation during pregnancy appear to be chiefly mediated glucocorticoid receptors, excess glucocorticoids cannot compensate by progesterone deficiency, indicating that additional und still undercover mechanisms are at play.
Collapse
Affiliation(s)
- Maria Emilia Solano
- Department for Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Petra Clara Arck
- Department for Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
33
|
Shehroz M, Aslam M, Ali Khan M, Aiman S, Gul Afridi S, Khan A. The In Silico Characterization of a Salicylic Acid Analogue Coding Gene Clusters in Selected Pseudomonas fluorescens Strains. IRANIAN JOURNAL OF BIOTECHNOLOGY 2019; 17:e2250. [PMID: 32671125 PMCID: PMC7357695 DOI: 10.30498/ijb.2019.95299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND The microbial genome sequences provide solid in silico framework for interpretation of their drug-like chemical scaffolds biosynthetic potentials. Pseudomonas fluorescens strains are metabolically versatile and producing therapeutically important natural products. OBJECTIVES The key objective of the present study was to mine the publically available data of P. fluorescens strains genomes for putative drug-like metabolites identification. MATERIALS AND METHODS We implemented the computational biology resources of AntiSMASH and BAGEL3 for the secondary metabolites prediction from P. fluorescens strains genome sequences. The predicted secondary metabolites were evaluated using drug discovery chemoinformatics resources, like Drugbank database search and molecular docking inspection. RESULTS The analyses unveiled a wide array of chemical scaffolds biosynthesis in different P. fluorescens strains. Subsequently, the drug-like potential evaluation of these metabolites identified few strains, including P. fluorescens PT14, P. fluorescens A5O6, and P. fluorescens FW300-N2E3 that harbor the biosynthetic gene clusters for salicylic acid-like metabolite biosynthesis. The molecular docking inspection of this metabolite against human cyclooxygenase and aldo-keto reductase targets revealed its feasible inhibitory potentials like other salicylate compounds. CONCLUSION The computational biology and drug discovery analyses identified different gene clusters in P. fluorescens genomes coding for salicylic acid-like chemotypes biosynthesis. These gene clusters may worthy to target through metabolic engineering for the massive production of salicylates-like chemical scaffolds from microbial resources.
Collapse
Affiliation(s)
- Muhammad Shehroz
- Department of Biochemistry, Abdul Wali Khan University Mardan, Khyber Pakhtunkhwa, Pakistan
| | - Muneeba Aslam
- Department of Biochemistry, Abdul Wali Khan University Mardan, Khyber Pakhtunkhwa, Pakistan
| | - Munazza Ali Khan
- Department of Biochemistry, Abdul Wali Khan University Mardan, Khyber Pakhtunkhwa, Pakistan
| | - Sara Aiman
- Department of Biochemistry, Abdul Wali Khan University Mardan, Khyber Pakhtunkhwa, Pakistan
| | - Sahib Gul Afridi
- Department of Biochemistry, Abdul Wali Khan University Mardan, Pakistan
| | - Asifullah Khan
- Department of Biochemistry, Abdul Wali Khan University Mardan, Pakistan
| |
Collapse
|
34
|
Yoshida M, Takayanagi Y, Ichino-Yamashita A, Sato K, Sugimoto Y, Kimura T, Nishimori K. Functional Hierarchy of Uterotonics Required for Successful Parturition in Mice. Endocrinology 2019; 160:2800-2810. [PMID: 31517984 PMCID: PMC6887699 DOI: 10.1210/en.2019-00499] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 09/09/2019] [Indexed: 11/23/2022]
Abstract
Parturition is an essential process in placental mammals for giving birth to offspring. However, the molecular machineries of parturition are not fully understood. We investigated whether oxytocin plays a crucial role in the progress of parturition in cooperation with the prostaglandin F2α (PGF2α) receptor. We first examined alterations in the expression of uterine contraction-associated genes in uteri of oxytocin receptor-deficient mice (Oxtr-/-) during parturition. We found that induction of cyclooxygenase (COX)-2 and connexin 43 expression was impaired in Oxtr-/-, whereas that of PGF2α receptor expression was not. We next generated mice with double knockout of genes for the oxytocin receptor/oxytocin and PGF2α receptor (Oxtr-/-;Ptgfr-/- and Oxt-/-;Ptgfr-/-) and evaluated their parturition with Oxtr-/-, Oxt-/-, Ptgfr-/-, and wild-type mice. In Oxtr-/-;Ptgfr-/- and Oxt-/-;Ptgfr-/-, pregnancy rates were similar to those of other genotypes. However, normal parturition was not observed in Oxtr-/-;Ptgfr-/- or Oxt-/-;Ptgfr-/- because of persistent progesterone from the corpus luteum, as observed in Ptgfr-/-. We administered RU486, a progesterone antagonist, to Ptgfr-/-, Oxtr-/-;Ptgfr-/-, and Oxt-/-;Ptgfr-/- on gestation day 19. These mice were able to deliver a living first pup and the parturition onset was similar to that in Ptgfr-/-. Meanwhile, unlike Ptgfr-/-, ∼75% of Oxtr-/-;Ptgfr-/- and Oxt-/-;Ptgfr-/- administered RU486 remained in labor at 24 hours after the onset of parturition. All of the pups that experienced prolonged labor died. We thus revealed that the oxytocin receptor is an upstream regulator of COX-2 and connexin 43 in the uterus during parturition and that both oxytocin/oxytocin receptor and PGF2α receptor are major components for successful parturition.
Collapse
Affiliation(s)
- Masahide Yoshida
- Laboratory of Molecular Biology, Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University, Sendai-shi, Miyagi-ken, Japan
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke-shi, Tochigi-ken, Japan
| | - Yuki Takayanagi
- Laboratory of Molecular Biology, Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University, Sendai-shi, Miyagi-ken, Japan
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke-shi, Tochigi-ken, Japan
| | - Azusa Ichino-Yamashita
- Laboratory of Molecular Biology, Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University, Sendai-shi, Miyagi-ken, Japan
| | - Kei Sato
- Laboratory of Molecular Biology, Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University, Sendai-shi, Miyagi-ken, Japan
- Division of Systems Virology, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo-to, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi-shi, Saitama-ken, Japan
| | - Yukihiko Sugimoto
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-Ku, Kumamoto-ken, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita-shi, Osaka-hu, Japan
| | - Katsuhiko Nishimori
- Laboratory of Molecular Biology, Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University, Sendai-shi, Miyagi-ken, Japan
- Department of Obesity and Inflammation Research, Fukushima Medical University, Fukushima-shi, Fukushima-ken, Japan
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University, Fukushima-shi, Fukushima-ken, Japan
- Correspondence: Katsuhiko Nishimori, PhD, Department of Obesity and Inflammation Research, Fukushima Medical University, 1 Hikarigaoka, Fukushima-shi, Fukushima-ken 960-1295, Japan. E-mail:
| |
Collapse
|
35
|
Zeng C, Zhu D, You J, Dong X, Yang B, Zhu H, He Q. Liquiritin, as a Natural Inhibitor of AKR1C1, Could Interfere With the Progesterone Metabolism. Front Physiol 2019; 10:833. [PMID: 31333491 PMCID: PMC6616128 DOI: 10.3389/fphys.2019.00833] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 06/17/2019] [Indexed: 11/13/2022] Open
Abstract
Low progesterone level is always linked with pre-term birth. Therefore, maintaining of progesterone level is vital during pregnancy. Aldo-keto reductase family one member C1 (AKR1C1) catalyzes the reduction of progesterone to its inactive form of 20-alpha-hydroxy-progesterone and thus limits the biological effect of progesterone. In our effort to identify the natural compound that would specifically inhibit AKR1C1, liquiritin was found to be a selective and potent inhibitor of AKR1C1. Kinetic analyses in the S-(+)-1,2,3,4-tetrahydro-1-naphthol (s-tetralol) catalyzed by AKR1C1 in the presence of the inhibitors suggest that liquiritin is a competitive inhibitor by targeting the residues Ala-27, Val-29, Ala-25, and Asn-56 of AKR1C1. In HEC-1-B cells, treatment with liquiritin results in 85.00% of reduction in progesterone metabolism, which is mediated by AKR1C1 enzymatic activity. Overall, our study not only identify liquiritin as an inhibitor against AKR1C1, but also reveal that liquiritin may be served as a potential intervention strategy for preventing pre-term birth caused by low progesterone level.
Collapse
Affiliation(s)
- Chenming Zeng
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Difeng Zhu
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, Hangzhou, China
| | - Jun You
- Zhejiang Cancer Hospital, Hangzhou, China
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Bo Yang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hong Zhu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
36
|
Mendelson CR, Gao L, Montalbano AP. Multifactorial Regulation of Myometrial Contractility During Pregnancy and Parturition. Front Endocrinol (Lausanne) 2019; 10:714. [PMID: 31708868 PMCID: PMC6823183 DOI: 10.3389/fendo.2019.00714] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/03/2019] [Indexed: 12/30/2022] Open
Abstract
The steroid hormones progesterone (P4) and estradiol-17β (E2), produced by the placenta in humans and the ovaries in rodents, serve crucial roles in the maintenance of pregnancy, and the initiation of parturition. Because of their critical importance for species survival, the mechanisms whereby P4 and its nuclear receptor (PR) maintain myometrial quiescence during pregnancy, and for the decline in P4/PR and increase in E2/estrogen receptor (ER) function leading to parturition, are multifaceted, cooperative, and redundant. These actions of P4/PR include: (1) PR interaction with proinflammatory transcription factors, nuclear factor κB (NF-κB), and activating protein 1 (AP-1) bound to promoters of proinflammatory and contractile/contraction-associated protein (CAP) genes and recruitment of corepressors to inhibit NF-κB and AP-1 activation of gene expression; (2) upregulation of inhibitors of proinflammatory transcription factor activation (IκBα, MKP-1); (3) induction of transcriptional repressors of CAP genes (e.g., ZEB1). In rodents and most other mammals, circulating maternal P4 levels remain elevated throughout most of pregnancy and decline precipitously near term. By contrast, in humans, circulating P4 levels and myometrial PR levels remain elevated throughout pregnancy and into labor. However, even in rodents, wherein P4 levels decline near term, P4 levels remain higher than the Kd for PR binding. Thus, parturition is initiated in all species by a series of molecular events that antagonize the P4/PR maintenance of uterine quiescence. These events include: direct interaction of inflammatory transcription factors (e.g., NF-κB, AP-1) with PR; increased expression of P4 metabolizing enzymes; increased expression of truncated/inhibitory PR isoforms; altered expression of PR coactivators and corepressors. This article will review various mechanisms whereby P4 acting through PR isoforms maintains myometrial quiescence during pregnancy as well as those that underlie the decline in PR function leading to labor. The roles of P4- and E2-regulated miRNAs in the regulation and integration of these mechanisms will also be considered.
Collapse
|
37
|
Fernando F, Boussata S, Jongejan A, van der Post JA, Afink G, Ris-Stalpers C. In silico analysis of the Mus musculus uterine gene expression landscape during pregnancy identifies putative upstream regulators for labour. PLoS One 2018; 13:e0204236. [PMID: 30235305 PMCID: PMC6147639 DOI: 10.1371/journal.pone.0204236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 09/05/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The molecular pathways involved in the transition from uterine quiescence to overt labour are mapped and form the currently established pharmacological targets for both the induction and inhibition of human labour. However, both spontaneous premature labour and functional dystocia occur and are difficult to treat adequately. The identification of upstream regulators involved in the onset and orchestration of labour pathways is essential to develop additional therapies that will contribute to the regulation of the timing of birth. OBJECTIVES To define uterine biological processes and their upstream activators involved in the transition from uterine quiescence to overt labour. STUDY DESIGN The uterus of non-pregnant and pregnant FVB M. musculus is collected at embryonic days (E) 6.5, 8.5, 10.5, 12.5, 15.5 and 17.5 and the uterine transcriptome is determined using the Illumina mouse Ref8v2 micro-array platform. K-means clustering and Ingenuity Pathway Analysis are applied to further dissect the transcriptome data. RESULTS From E6.5 to E17.5, 5405 genes are significantly differentially expressed and they segregate into 7 unique clusters. Five of the 7 clusters are enriched for genes involved in specific biological processes that include regulation of gene-expression, T-cell receptor activation, Toll-like receptor signalling and steroid metabolism. The identification of upstream activators for differentially expressed genes between consecutive time points highlights the E10.5 to E12.5 window during which the role from progesterone switches from an activated state to the inhibited state reflecting the process of functional progesterone withdrawal essential for the transgression from myometrial quiescence to synchronized contractions. For this time window in which 189 genes are differentially expressed we define 22 putative upstream activators of which NUPR1 and TBX2 are the most significant with respectively an activated and an inhibited status. CONCLUSIONS Gene expression profiling of mice uterus from E6.5 to E17.5 results in 7 unique gene expression clusters from early to late pregnancy that define the landscape of molecular events in ongoing pregnancy. In the current dataset progesterone is predicted as an activated upstream regulator and maintainer of myometrial quiescence and is active till E10.5. Progesterone is predicted as an inhibited upstream regulator at E12.5. We identify 22 upstream regulators in the E10.5 to E12.5 time window where the switch to progesterone withdrawal occurs. They are putative relevant upstream activators of labour.
Collapse
Affiliation(s)
- Febilla Fernando
- Reproductive Biology Laboratory Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Souad Boussata
- Reproductive Biology Laboratory Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Aldo Jongejan
- Department of Bioinformatics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Joris A. van der Post
- Women’s and Children’s Clinic, Department of Obstetrics and Gynaecology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Gijs Afink
- Reproductive Biology Laboratory Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Carrie Ris-Stalpers
- Reproductive Biology Laboratory Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Women’s and Children’s Clinic, Department of Obstetrics and Gynaecology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
38
|
Konings G, Brentjens L, Delvoux B, Linnanen T, Cornel K, Koskimies P, Bongers M, Kruitwagen R, Xanthoulea S, Romano A. Intracrine Regulation of Estrogen and Other Sex Steroid Levels in Endometrium and Non-gynecological Tissues; Pathology, Physiology, and Drug Discovery. Front Pharmacol 2018; 9:940. [PMID: 30283331 PMCID: PMC6157328 DOI: 10.3389/fphar.2018.00940] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 08/02/2018] [Indexed: 12/20/2022] Open
Abstract
Our understanding of the intracrine (or local) regulation of estrogen and other steroid synthesis and degradation expanded in the last decades, also thanks to recent technological advances in chromatography mass-spectrometry. Estrogen responsive tissues and organs are not passive receivers of the pool of steroids present in the blood but they can actively modify the intra-tissue steroid concentrations. This allows fine-tuning the exposure of responsive tissues and organs to estrogens and other steroids in order to best respond to the physiological needs of each specific organ. Deviations in such intracrine control can lead to unbalanced steroid hormone exposure and disturbances. Through a systematic bibliographic search on the expression of the intracrine enzymes in various tissues, this review gives an up-to-date view of the intracrine estrogen metabolisms, and to a lesser extent that of progestogens and androgens, in the lower female genital tract, including the physiological control of endometrial functions, receptivity, menopausal status and related pathological conditions. An overview of the intracrine regulation in extra gynecological tissues such as the lungs, gastrointestinal tract, brain, colon and bone is given. Current therapeutic approaches aimed at interfering with these metabolisms and future perspectives are discussed.
Collapse
Affiliation(s)
- Gonda Konings
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Linda Brentjens
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Bert Delvoux
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | | | - Karlijn Cornel
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | | | - Marlies Bongers
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Roy Kruitwagen
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Sofia Xanthoulea
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Andrea Romano
- GROW–School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
39
|
Berkane N, Liere P, Lefevre G, Alfaidy N, Nahed RA, Vincent J, Oudinet JP, Pianos A, Cambourg A, Rozenberg P, Galichon P, Rousseau A, Simon T, Schumacher M, Chabbert-Buffet N, Hertig A. Abnormal steroidogenesis and aromatase activity in preeclampsia. Placenta 2018; 69:40-49. [PMID: 30213483 DOI: 10.1016/j.placenta.2018.07.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 07/03/2018] [Accepted: 07/06/2018] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Estrogens and progesterone play critical roles in angiogenesis and vasodilation. Moreover, placental aromatase deficiency is detected in women with preeclampsia (PE) at delivery. We hypothesized that abnormal steroidogenesis occurs much earlier than typical PE diagnosis. Thus, we investigated whether the circulating steroid profile was already disturbed at 24-29 weeks of gestation in women with subsequent PE, and compared the profile with that of women with "placental" small gestational age (SGA) without PE. METHODS We selected nulliparous women (n = 90) from the MOMA trial, including women with PE (n = 25), SGA (n = 25), and controls (NP; n = 40), for plasma steroid profiling by gas chromatography/mass spectrometry and to measure placental growth factor and soluble fms-like tyrosine kinase-1. Placental aromatase expression was evaluated in a new set of women. RESULTS Compared with that of controls, the women with PE had a significantly lower estrone/androstenedione ratio, and exhibited a decreasing trend for estradiol and estrone levels. Lower estriol levels were observed in the SGA group compared to the NP group. Compared with that of controls, the women with PE and SGA had significantly higher levels of 20α-dihydroprogesterone (20α-DHP) and 20α-DHP/progesterone ratios. Pregnenolone sulfate levels were lower in the PE group than in the NP and SGA groups. Decreased expression of aromatase was observed in the PE group compared to the control group. DISCUSSION Preeclampsia appears to be characterized by specific steroidogenesis dysregulation long before PE diagnosis, highlighting potential new biomarkers of PE.
Collapse
Affiliation(s)
- Nadia Berkane
- Department of Gynecology and Obstetrics and Reproductive Medicine, Assistance Publique des Hôpitaux de Paris (APHP), Sorbonne University, Paris, France; Department of Gynecology and Obstetrics, University of Geneva Hospitals (HUG), Geneva, Switzerland
| | - Philippe Liere
- U1195 INSERM and University Paris-Sud, Kremlin-Bicêtre, France
| | - Guillaume Lefevre
- Department of Biochemistry and Hormonology, Assistance Publique des Hôpitaux de Paris (APHP), Sorbonne University, Paris, France
| | - Nadia Alfaidy
- U1036 INSERM, Biosciences and Biotechnology Institute, Grenoble, France
| | - Roland Abi Nahed
- U1036 INSERM, Biosciences and Biotechnology Institute, Grenoble, France
| | - Jessica Vincent
- Department of Gynecology and Obstetrics, University of Geneva Hospitals (HUG), Geneva, Switzerland
| | | | - Antoine Pianos
- U1195 INSERM and University Paris-Sud, Kremlin-Bicêtre, France
| | - Annie Cambourg
- U1195 INSERM and University Paris-Sud, Kremlin-Bicêtre, France
| | - Patrick Rozenberg
- Department of Obstetrics and Gynecology, Poissy-Saint Germain Hospital, Poissy, France
| | - Pierre Galichon
- Department of Nephrology, Assistance Publique des Hôpitaux de Paris (APHP), Sorbonne University, Tenon Hospital, Paris, France
| | - Alexandra Rousseau
- Department of Clinical Research Center-Est (URCEST), St. Antoine Hospital, Paris, France
| | - Tabassome Simon
- Department of Clinical Research Center-Est (URCEST), St. Antoine Hospital, Paris, France
| | | | - Nathalie Chabbert-Buffet
- Department of Gynecology and Obstetrics and Reproductive Medicine, Assistance Publique des Hôpitaux de Paris (APHP), Sorbonne University, Paris, France
| | - Alexandre Hertig
- Department of Nephrology, Assistance Publique des Hôpitaux de Paris (APHP), Sorbonne University, Tenon Hospital, Paris, France.
| |
Collapse
|
40
|
Wu SP, Li R, DeMayo FJ. Progesterone Receptor Regulation of Uterine Adaptation for Pregnancy. Trends Endocrinol Metab 2018; 29:481-491. [PMID: 29705365 PMCID: PMC6004243 DOI: 10.1016/j.tem.2018.04.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/01/2018] [Accepted: 04/03/2018] [Indexed: 01/01/2023]
Abstract
Progesterone acts through the progesterone receptor to direct physiological adaption of the uterus in preparation and completion of pregnancy. Genome-wide transcriptome and cistrome analyses have uncovered new members and novel modifiers of the progesterone signaling pathway. Genetically engineered mice allow functional assessment of newly identified genes in vivo and provide insights on the impact of progesterone receptor-dependent molecular mechanisms on pregnancy at the organ system level. Progesterone receptor isoforms collectively mediate progesterone signaling via their distinct and common downstream target genes, which makes the stoichiometry of isoforms relevant in modifying the progesterone activity. This review discusses recent advances on the discovery of the progesterone receptor network, with special focus on the endometrium at early pregnancy and myometrium during parturition.
Collapse
Affiliation(s)
- San-Pin Wu
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, NC 27709, USA
| | - Rong Li
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, NC 27709, USA
| | - Francesco J DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
41
|
Proietto S, Cortasa SA, Corso MC, Inserra PIF, Charif SE, Schmidt AR, Di Giorgio NP, Lux-Lantos V, Vitullo AD, Dorfman VB, Halperin J. Prolactin Is a Strong Candidate for the Regulation of Luteal Steroidogenesis in Vizcachas ( Lagostomus maximus). Int J Endocrinol 2018; 2018:1910672. [PMID: 30013596 PMCID: PMC6022330 DOI: 10.1155/2018/1910672] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 04/20/2018] [Accepted: 05/07/2018] [Indexed: 12/11/2022] Open
Abstract
Prolactin (PRL) is essential for the maintenance of the corpora lutea and the production of progesterone (P4) during gestation of mice and rats, which makes it a key factor for their successful reproduction. Unlike these rodents and the vast majority of mammals, female vizcachas (Lagostomus maximus) have a peculiar reproductive biology characterized by an ovulatory event during pregnancy that generates secondary corpora lutea with a consequent increment of the circulating P4. We found that, although the expression of pituitary PRL increased steadily during pregnancy, its ovarian receptor (PRLR) reached its maximum in midpregnancy and drastically decreased at term pregnancy. The luteinizing hormone receptor (LHR) exhibited a similar profile than PRLR. Maximum P4 and LH blood levels were recorded at midpregnancy as well. Remarkably, the P4-sinthesizing enzyme 3β-HSD accompanied the expression pattern of PRLR/LHR throughout gestation. Instead, the luteolytic enzyme 20α-HSD showed low expression at early and midpregnancy, but reached its maximum at the end of gestation, when PRLR/LHR/3ß-HSD expressions and circulating P4 were minimal. In conclusion, both the PRLR and LHR expressions in the ovary would define the success of gestation in vizcachas by modulating the levels of 20α-HSD and 3ß-HSD, which ultimately determine the level of serum P4 throughout gestation.
Collapse
Affiliation(s)
- S. Proietto
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - S. A. Cortasa
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - M. C. Corso
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - P. I. F. Inserra
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - S. E. Charif
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - A. R. Schmidt
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - N. P. Di Giorgio
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IByME), Ciudad Autónoma de Buenos Aires, Argentina
| | - V. Lux-Lantos
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IByME), Ciudad Autónoma de Buenos Aires, Argentina
| | - A. D. Vitullo
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - V. B. Dorfman
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - J. Halperin
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
42
|
Kusama K, Bai R, Imakawa K. Regulation of human trophoblast cell syncytialization by transcription factors STAT5B and NR4A3. J Cell Biochem 2018; 119:4918-4927. [PMID: 29377304 DOI: 10.1002/jcb.26721] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 01/24/2018] [Indexed: 11/08/2022]
Abstract
In human trophoblast cells, cyclic AMP or its inducer forskolin (FSK) activates two downstream signaling molecules, protein kinase A (PKA) and exchange protein directly activated by cAMP (EPAC), both of which induce syncytialization, cell fusion, and the production of human chorionic gonadotropin (hCG) and progesterone. However, a transcription factor other than GCM1 and molecular mechanisms associated with these events have not been well characterized. To identify novel transcription factors involved in syncytialization of cAMP-stimulated human choriocarcinoma BeWo cells, the microarray analysis was performed with RNAs extracted from PKA- or EPAC-selective cAMP analog-stimulated BeWo cells, from which two up-regulated transcription factors, STAT5 and NR4A3, were found. The knockdown of STAT5B decreased FSK-induced cell fusion and the expression of syncytialization markers, CGB, syncytin1, syncytin2, GCM1, and OVOL1, but NR4A3 knockdown increased FSK-induced cell fusion and the expression of CGB and syncytin2. These findings indicated that cAMP-PKA up-regulated STAT5B, followed by increase in syncytin2 expression through GCM1 and OVOL1, resulting in cell fusion and hCG production, while cAMP-PKA-up-regulated NR4A3 could decrease syncytin2 expression, and suggested that both positive and negative effects of STAT5B and NR4A3, respectively, are required to control the degree of syncytialization in human trophoblast cells.
Collapse
Affiliation(s)
- Kazuya Kusama
- Graduate School of Agricultural and Life Science, The University of Tokyo, Animal Resource Science Center, Kasama, Ibaraki, Japan
| | - Rulan Bai
- Graduate School of Agricultural and Life Science, The University of Tokyo, Animal Resource Science Center, Kasama, Ibaraki, Japan
| | - Kazuhiko Imakawa
- Graduate School of Agricultural and Life Science, The University of Tokyo, Animal Resource Science Center, Kasama, Ibaraki, Japan
| |
Collapse
|
43
|
Herington JL, O’Brien C, Robuck MF, Lei W, Brown N, Slaughter JC, Paria BC, Mahadevan-Jansen A, Reese J. Prostaglandin-Endoperoxide Synthase 1 Mediates the Timing of Parturition in Mice Despite Unhindered Uterine Contractility. Endocrinology 2018; 159:490-505. [PMID: 29029054 PMCID: PMC5761592 DOI: 10.1210/en.2017-00647] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 09/20/2017] [Indexed: 11/19/2022]
Abstract
Cyclooxygenase (COX)-derived prostaglandins stimulate uterine contractions and prepare the cervix for parturition. Prior reports suggest Cox-1 knockout (KO) mice exhibit delayed parturition due to impaired luteolysis, yet the mechanism for late-onset delivery remains unclear. Here, we examined key factors for normal onset of parturition to determine whether any could account for the delayed parturition phenotype. Pregnant Cox-1KO mice did not display altered timing of embryo implantation or postimplantation growth. Although messenger RNAs of contraction-associated proteins (CAPs) were differentially expressed between Cox-1KO and wild-type (WT) myometrium, there were no differences in CAP agonist-induced intracellular calcium release, spontaneous or oxytocin (OT)-induced ex vivo uterine contractility, or in vivo uterine contractile pressure. Delayed parturition in Cox-1KO mice persisted despite exogenous OT treatment. Progesterone (P4) withdrawal, by ovariectomy or administration of the P4-antagonist RU486, diminished the delayed parturition phenotype of Cox-1KO mice. Because antepartum P4 levels do not decline in Cox-1KO females, P4-treated WT mice were examined for the effect of this hormone on in vivo uterine contractility and ex vivo cervical dilation. P4-treated WT mice had delayed parturition but normal uterine contractility. Cervical distensibility was decreased in Cox-1KO mice on the day of expected delivery and reduced in WT mice with long-term P4 treatment. Collectively, these findings show that delayed parturition in Cox-1KO mice is the result of impaired luteolysis and cervical dilation, despite the presence of strong uterine contractions.
Collapse
Affiliation(s)
- Jennifer L. Herington
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Christine O’Brien
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232
| | - Michael F. Robuck
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Wei Lei
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215007, China
| | - Naoko Brown
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - James C. Slaughter
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, Tennessee, 37232
| | - Bibhash C. Paria
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | | | - Jeff Reese
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232
| |
Collapse
|
44
|
Yao Y, Li H, Ding J, Xia Y, Wang L. Progesterone impairs antigen-non-specific immune protection by CD8 T memory cells via interferon-γ gene hypermethylation. PLoS Pathog 2017; 13:e1006736. [PMID: 29155896 PMCID: PMC5714395 DOI: 10.1371/journal.ppat.1006736] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 12/04/2017] [Accepted: 11/05/2017] [Indexed: 12/11/2022] Open
Abstract
Pregnant women and animals have increased susceptibility to a variety of intracellular pathogens including Listeria monocytogenes (LM), which has been associated with significantly increased level of sex hormones such as progesterone. CD8 T memory(Tm) cell-mediated antigen-non-specific IFN-γ responses are critically required in the host defense against LM. However, whether and how increased progesterone during pregnancy modulates CD8 Tm cell-mediated antigen-non-specific IFN-γ production and immune protection against LM remain poorly understood. Here we show in pregnant women that increased serum progesterone levels are associated with DNA hypermethylation of IFN-γ gene promoter region and decreased IFN-γ production in CD8 Tm cells upon antigen-non-specific stimulation ex vivo. Moreover, IFN-γ gene hypermethylation and significantly reduced IFN-γ production post LM infection in antigen-non-specific CD8 Tm cells are also observed in pregnant mice or progesterone treated non-pregnant female mice, which is a reversible phenotype following demethylation treatment. Importantly, antigen-non-specific CD8 Tm cells from progesterone treated mice have impaired anti-LM protection when adoptive transferred in either pregnant wild type mice or IFN-γ-deficient mice, and demethylation treatment rescues the adoptive protection of such CD8 Tm cells. These data demonstrate that increased progesterone impairs immune protective functions of antigen-non-specific CD8 Tm cells via inducing IFN-γ gene hypermethylation. Our findings thus provide insights into a new mechanism through which increased female sex hormone regulate CD8 Tm cell functions during pregnancy. Increased female sex hormones during pregnancy generate a temporary immune suppression status in the pregnant that protect the developing fetus from maternal rejection but renders the pregnant highly susceptible to various pathogens. However, molecular mechanisms underlying such an increased maternal susceptibility to pathogens during pregnancy remain to be further understood. Here we show in pregnant women that increased progesterone levels are associated with IFN-γ gene hypermethylation and reduced IFN-γ production in peripheral CD8 Tm cells. By using murine models of LM infection, for the first time we show a causal relationship between increased level of progesterone, a characteristic female sex hormone of pregnancy, and increased susceptibility to Listeria monocytogenes, an intracellular bacterium that endangers both the pregnant and the fetus. Such an impact on anti-listeria host defense is mediated through progesterone-induced IFN-γ gene hypermethylation in CD8 Tm cells, resulting in impaired IFN-γ production and reduced immune protection by antigen-non-specific CD8 Tm cells. This study provides new insights into molecular mechanisms underlying the increased susceptibility to intracellular pathogens during pregnancy.
Collapse
Affiliation(s)
- Yushi Yao
- McMaster Immunology Research Center, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Hematology, Chinese PLA General Hospital, Beijing, China
| | - Hui Li
- Department of Clinical Nutrition, General Hospital of Chinese People's Armed Police Forces, Beijing, China
| | - Jie Ding
- Department of Hematology, Chinese PLA General Hospital, Beijing, China
| | - Yixin Xia
- Department of Obstetrics and Gynecology, General Hospital of Chinese People's Armed Police Forces, Beijing, China
| | - Lei Wang
- Department of Clinical Nutrition, General Hospital of Chinese People's Armed Police Forces, Beijing, China
- * E-mail:
| |
Collapse
|
45
|
Aghaeepour N, Ganio EA, Mcilwain D, Tsai AS, Tingle M, Van Gassen S, Gaudilliere DK, Baca Q, McNeil L, Okada R, Ghaemi MS, Furman D, Wong RJ, Winn VD, Druzin ML, El-Sayed YY, Quaintance C, Gibbs R, Darmstadt GL, Shaw GM, Stevenson DK, Tibshirani R, Nolan GP, Lewis DB, Angst MS, Gaudilliere B. An immune clock of human pregnancy. Sci Immunol 2017; 2:2/15/eaan2946. [PMID: 28864494 PMCID: PMC5701281 DOI: 10.1126/sciimmunol.aan2946] [Citation(s) in RCA: 319] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/27/2017] [Indexed: 12/21/2022]
Abstract
Themaintenance of pregnancy relies on finely tuned immune adaptations.We demonstrate that these adaptations are precisely timed, reflecting an immune clock of pregnancy in women delivering at term. Using mass cytometry, the abundance and functional responses of allmajor immune cell subsets were quantified in serial blood samples collected throughout pregnancy. Cell signaling–based Elastic Net, a regularized regressionmethod adapted from the elastic net algorithm, was developed to infer and prospectively validate a predictive model of interrelated immune events that accurately captures the chronology of pregnancy. Model components highlighted existing knowledge and revealed previously unreported biology, including a critical role for the interleukin-2–dependent STAT5ab signaling pathway in modulating T cell function during pregnancy. These findings unravel the precise timing of immunological events occurring during a term pregnancy and provide the analytical framework to identify immunological deviations implicated in pregnancy-related pathologies.
Collapse
Affiliation(s)
- Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - Edward A Ganio
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - David Mcilwain
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94121, USA
| | - Amy S Tsai
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - Martha Tingle
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - Sofie Van Gassen
- Department of Information Technology, Ghent University, and the Center for Inflammation Research, Ghent, Belgium
| | - Dyani K Gaudilliere
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - Quentin Baca
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - Leslie McNeil
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - Robin Okada
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - Mohammad S Ghaemi
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - David Furman
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Immunogenetics, Jose de San Martin Clinical Hospital, National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Ronald J Wong
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - Maurice L Druzin
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - Yaser Y El-Sayed
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - Cecele Quaintance
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - Ronald Gibbs
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - Gary L Darmstadt
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - Gary M Shaw
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - David K Stevenson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - Robert Tibshirani
- Departments of Biomedical Data Sciences and Statistics, Stanford University, Stanford, CA 94121, USA
| | - Garry P Nolan
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94121, USA
| | - David B Lewis
- Division of Pediatric Immunology and Allergy, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - Martin S Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121, USA
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121, USA.
| |
Collapse
|
46
|
O'Brien CM, Herington JL, Brown N, Pence IJ, Paria BC, Slaughter JC, Reese J, Mahadevan-Jansen A. In vivo Raman spectral analysis of impaired cervical remodeling in a mouse model of delayed parturition. Sci Rep 2017; 7:6835. [PMID: 28754971 PMCID: PMC5533720 DOI: 10.1038/s41598-017-07047-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 06/21/2017] [Indexed: 12/01/2022] Open
Abstract
Monitoring cervical structure and composition during pregnancy has high potential for prediction of preterm birth (PTB), a problem affecting 15 million newborns annually. We use in vivo Raman spectroscopy, a label-free, light-based method that provides a molecular fingerprint to non-invasively investigate normal and impaired cervical remodeling. Prostaglandins stimulate uterine contractions and are clinically used for cervical ripening during pregnancy. Deletion of cyclooxygenase-1 (Cox-1), an enzyme involved in production of these prostaglandins, results in delayed parturition in mice. Contrary to expectation, Cox-1 null mice displayed normal uterine contractility; therefore, this study sought to determine whether cervical changes could explain the parturition differences in Cox-1 null mice and gestation-matched wild type (WT) controls. Raman spectral changes related to extracellular matrix proteins, lipids, and nucleic acids were tracked over pregnancy and found to be significantly delayed in Cox-1 null mice at term. A cervical basis for the parturition delay was confirmed by other ex vivo tests including decreased tissue distensibility, hydration, and elevated progesterone levels in the Cox-1 null mice at term. In conclusion, in vivo Raman spectroscopy non-invasively detected abnormal remodeling in the Cox-1 null mouse, and clearly demonstrated that the cervix plays a key role in their delayed parturition.
Collapse
Affiliation(s)
- Christine M O'Brien
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37232, USA.,Biophotonics Center, Vanderbilt University, Nashville, TN, 37232, USA
| | - Jennifer L Herington
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Naoko Brown
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Isaac J Pence
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37232, USA.,Biophotonics Center, Vanderbilt University, Nashville, TN, 37232, USA
| | - Bibhash C Paria
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - James C Slaughter
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Jeff Reese
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37232, USA.,Biophotonics Center, Vanderbilt University, Nashville, TN, 37232, USA.,Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Anita Mahadevan-Jansen
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37232, USA. .,Biophotonics Center, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
47
|
Wu SP, DeMayo FJ. Progesterone Receptor Signaling in Uterine Myometrial Physiology and Preterm Birth. Curr Top Dev Biol 2017; 125:171-190. [PMID: 28527571 DOI: 10.1016/bs.ctdb.2017.03.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Myometrium holds the structural integrity for the uterus and generates force for parturition with its primary component, the smooth muscle cells. The progesterone receptor mediates progesterone-dependent signaling and connects to a network of pathways for regulation of contractility and inflammatory responses in myometrium. Dysfunctional progesterone signaling has been linked to pregnancy complications including preterm birth. In the present review, we summarize recent findings on modifiers and effectors of the progesterone receptor signaling. Discussions include novel conceptual discoveries and new development in legacy pathways such as the signal transducers NF-κB, ZEB, microRNA, and the unfolded protein response pathways. We also discuss the impact of progesterone receptor isoform composition and ligand accessibility in modification of the progesterone receptor genomic actions.
Collapse
Affiliation(s)
- San-Pin Wu
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, NC, United States
| | - Francesco J DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, NC, United States.
| |
Collapse
|
48
|
Clark NC, Pru CA, Yee SP, Lydon JP, Peluso JJ, Pru JK. Conditional Ablation of Progesterone Receptor Membrane Component 2 Causes Female Premature Reproductive Senescence. Endocrinology 2017; 158:640-651. [PMID: 28005395 PMCID: PMC5460782 DOI: 10.1210/en.2016-1701] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/21/2016] [Indexed: 01/04/2023]
Abstract
The nonclassical progesterone receptors progesterone receptor membrane component (PGRMC) 1 and PGRMC2 have been implicated in regulating cell survival of endometrial and ovarian cells in vitro and are abundantly expressed in these cell types. The objective of this study was to determine if Pgrmc1 and Pgrmc2 are essential for normal female reproduction. To accomplish this objective, Pgrmc1 and/or Pgrmc2 floxed mice (Pgrmc2fl/fl and Pgrmc1/2fl/fl) were crossed with Pgr-cre mice, which resulted in the conditional ablation of Pgrmc1 and/or Pgrmc2 from female reproductive tissues (i.e.,Pgrmc2d/d and Pgrmc1/2d/d mice). A breeding trial revealed that conditional ablation of Pgrmc2 initially led to subfertility, with Pgrmc2d/d female mice producing 47% fewer pups/litter than Pgrmc2fl/fl mice (P = 0.001). Pgrmc2d/d mice subsequently underwent premature reproductive senescence by parities 2 to 5, producing 37.8% fewer litters overall during the trial compared with Pgrmc2fl/fl mice (P = 0.020). Similar results were observed with Pgrmc1/2d/d mice. Based on ovarian morphology and serum P4, the subfertility/infertility was not due to faulty ovulation or luteal insufficiency. Rather an analysis of midgestation implantation sites revealed that postimplantation embryonic death was the major cause of the subfertility/infertility. As with our previous report of Pgrmc1d/d mice, Pgrmc2d/d and Pgrmc1/2d/d mice developed endometrial cysts consistent with accelerated aging of this tissue. Given the timing of postimplantation embryonic demise, uterine decidualization may be disrupted in mice deficient in PGRMC2 or PGRMC1/2. Overall, this study revealed that Pgrmc1 and/or Pgrmc2 are required for the maintenance of uterine histoarchitecture and normal female reproductive lifespan.
Collapse
Affiliation(s)
- Nicole C. Clark
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington 99164;
| | - Cindy A. Pru
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington 99164;
| | - Siu-Pok Yee
- Departments of Cell Biology and Obstetrics and Gynecology, University of Connecticut Health Center, Farmington, Connecticut 06030; and
| | - John P. Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - John J. Peluso
- Departments of Cell Biology and Obstetrics and Gynecology, University of Connecticut Health Center, Farmington, Connecticut 06030; and
| | - James K. Pru
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington 99164;
| |
Collapse
|
49
|
Baddela VS, Onteru SK, Singh D. A syntenic locus on buffalo chromosome 20: novel genomic hotspot for miRNAs involved in follicular-luteal transition. Funct Integr Genomics 2016; 17:321-334. [PMID: 27866284 DOI: 10.1007/s10142-016-0535-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 11/02/2016] [Accepted: 11/04/2016] [Indexed: 12/17/2022]
Abstract
The developmental reorganization of ovarian follicular granulosa cells (GC) during follicular maturation, ovulation, and luteinization require a well-controlled regulation of dynamic gene expression profiles. Recently, microRNAs (miRNAs) were found to be key players of ovarian follicular dynamics. The current study aimed to understand the miRNA regulatory role in follicular-luteal transition by characterizing the miRNA profile through miRNA-seq at different follicular (small, medium, and large) and luteal (early, mid, and late) stages in Indian water buffaloes, mono-ovulatory animals like humans. A total of 517 miRNAs were identified in follicular granulosa cells (GC) and corpus luteum (CL) together. Among them, 2 unique and 40 novel miRNAs were in GC; 15 unique and 45 novel miRNAs were in CL. Among the remaining 415 annotated common miRNAs between GC and CL, 43 have showed significant (p < 0.05) differential expression between GC and CL. Particularly, 39 and 4 miRNAs showed higher expression in CL and GC, respectively, with respect to each other. Genome mapping analysis revealed that 71.7% of differential miRNAs having higher expression in CL compared to GC, and 93% of the unique miRNAs in CL were mapped to a short chromosomal region of 0.7 Mb (67.4 to 68.1 Mb) on chromosome 21 of cows which is syntenic to the buffalo chromosome 20. Clustering of all these miRNAs at this locus suggests it as a chromosomal hotspot for miRNAs involved in follicular-luteal transition, especially for CL physiological functions.
Collapse
Affiliation(s)
- Vijay Simha Baddela
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute (Deemed University), Karnal, 132001, Haryana, India
| | - Suneel Kumar Onteru
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute (Deemed University), Karnal, 132001, Haryana, India
| | - Dheer Singh
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute (Deemed University), Karnal, 132001, Haryana, India.
| |
Collapse
|
50
|
Mi Y, Coonce M, Fiete D, Steirer L, Dveksler G, Townsend RR, Baenziger JU. Functional Consequences of Mannose and Asialoglycoprotein Receptor Ablation. J Biol Chem 2016; 291:18700-17. [PMID: 27405760 DOI: 10.1074/jbc.m116.738948] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Indexed: 11/06/2022] Open
Abstract
The mannose receptor (ManR, Mrc1) and asialoglycoprotein receptor (ASGR, Asgr1 and Asgr2) are highly abundant endocytic receptors expressed by sinusoidal endothelial cells and parenchymal cells in the liver, respectively. We genetically manipulated either receptor individually or in combination, revealing phenotypic changes in female and male mice associated with changes in circulating levels of many glycoproteins. Both receptors rise and fall in response to progesterone during pregnancy. Thirty percent of Asgr2(-/-) and 65% of Mrc1(-/-)Asgr2(-/-) mice are unable to initiate parturition at the end of pregnancy, whereas Mrc1(-/-) mice initiate normally. Twenty five percent of Mrc1(-/-)Asgr2(-/-) male mice develop priapism when mating due to thrombosis of the penile vein, but neither Mrc1(-/-) nor Asgr2(-/-) mice do so. The half-life for luteinizing hormone (LH) clearance increases in Mrc1(-/-) and Mrc1(-/-)Asgr2(-/-) mice but not in Asgr2(-/-) mice; however, LH and testosterone are elevated in all three knockouts. The ManR clears LH thus regulating testosterone production, whereas the ASGR appears to mediate clearance of an unidentified glycoprotein that increases LH levels. More than 40 circulating glycoproteins are elevated >3.0-fold in pregnant Mrc1(-/-)Asgr2(-/-) mice. Pregnancy-specific glycoprotein 23, undetectable in WT mice (<50 ng/ml plasma), reaches levels of 1-10 mg/ml in the plasma of Mrc1(-/-)Asgr2(-/-) and Asgr2(-/-) mice, indicating it is cleared by the ASGR. Elevation of multiple coagulation factors in Mrc1(-/-)Asgr2(-/-) mice may account for priapism seen in males. These male and female phenotypic changes underscore the key roles of the ManR and ASGR in controlling circulating levels of numerous glycoproteins critical for regulating reproductive hormones and blood coagulation.
Collapse
Affiliation(s)
- Yiling Mi
- From the Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Marcy Coonce
- From the Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Dorothy Fiete
- From the Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Lindsay Steirer
- From the Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Gabriela Dveksler
- From the Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri 63110
| | - R Reid Townsend
- From the Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Jacques U Baenziger
- From the Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|