1
|
Sirohi VK, Medrano TI, Kannan A, Bagchi IC, Cooke PS. Uterine-specific Ezh2 deletion enhances stromal cell senescence and impairs placentation, resulting in pregnancy loss. iScience 2023; 26:107028. [PMID: 37360688 PMCID: PMC10285549 DOI: 10.1016/j.isci.2023.107028] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/10/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
Maternal uterine remodeling facilitates embryo implantation, stromal cell decidualization and placentation, and perturbation of these processes may cause pregnancy loss. Enhancer of zeste homolog 2 (EZH2) is a histone methyltransferase that epigenetically represses gene transcription; loss of uterine EZH2 affects endometrial physiology and induces infertility. We utilized a uterine Ezh2 conditional knockout (cKO) mouse to determine EZH2's role in pregnancy progression. Despite normal fertilization and implantation, embryo resorption occurred mid-gestation in Ezh2cKO mice, accompanied by compromised decidualization and placentation. Western blot analysis revealed Ezh2-deficient stromal cells have reduced amounts of the histone methylation mark H3K27me3, causing upregulation of senescence markers p21 and p16 and indicating that enhanced stromal cell senescence likely impairs decidualization. Placentas from Ezh2cKO dams on gestation day (GD) 12 show architectural defects, including mislocalization of spongiotrophoblasts and reduced vascularization. In summary, uterine Ezh2 loss impairs decidualization, increases decidual senescence, and alters trophoblast differentiation, leading to pregnancy loss.
Collapse
Affiliation(s)
- Vijay K. Sirohi
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Theresa I. Medrano
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Athilakshmi Kannan
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Indrani C. Bagchi
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Paul S. Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| |
Collapse
|
2
|
Puscheck EE, Ruden X, Singh A, Abdulhasan M, Ruden DM, Awonuga AO, Rappolee DA. Using high throughput screens to predict miscarriages with placental stem cells and long-term stress effects with embryonic stem cells. Birth Defects Res 2022; 114:1014-1036. [PMID: 35979652 PMCID: PMC10108263 DOI: 10.1002/bdr2.2079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 11/10/2022]
Abstract
A problem in developmental toxicology is the massive loss of life from fertilization through gastrulation, and the surprising lack of knowledge of causes of miscarriage. Half to two-thirds of embryos are lost, and environmental and genetic causes are nearly equal. Simply put, it can be inferred that this is a difficult period for normal embryos, but that environmental stresses may cause homeostatic responses that move from adaptive to maladaptive with increasing exposures. At the lower 50% estimate, miscarriage causes greater loss-of-life than all cancers combined or of all cardio- and cerebral-vascular accidents combined. Surprisingly, we do not know if miscarriage rates are increasing or decreasing. Overshadowed by the magnitude of miscarriages, are insufficient data on teratogenic or epigenetic imbalances in surviving embryos and their stem cells. Superimposed on the difficult normal trajectory for peri-gastrulation embryos are added malnutrition, hormonal, and environmental stresses. An overarching hypothesis is that high throughput screens (HTS) using cultured viable reporter embryonic and placental stem cells (e.g., embryonic stem cells [ESC] and trophoblast stem cells [TSC] that report status using fluorescent reporters in living cells) from the pre-gastrulation embryo will most rapidly test a range of hormonal, environmental, nutritional, drug, and diet supplement stresses that decrease stem cell proliferation and imbalance stemness/differentiation. A second hypothesis is that TSC respond with greater sensitivity in magnitude to stress that would cause miscarriage, but ESC are stress-resistant to irreversible stemness loss and are best used to predict long-term health defects. DevTox testing needs more ESC and TSC HTS to model environmental stresses leading to miscarriage or teratogenesis and more research on epidemiology of stress and miscarriage. This endeavor also requires a shift in emphasis on pre- and early gastrulation events during the difficult period of maximum loss by miscarriage.
Collapse
Affiliation(s)
- Elizabeth E Puscheck
- CS Mott Center for Human Growth and Development, Department of Ob/Gyn, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan, USA
- Reproductive Stress 3M Inc, Grosse Pointe Farms, Michigan, USA
- Invia Fertility Clinics, Hoffman Estates, Illinois, USA
| | - Ximena Ruden
- CS Mott Center for Human Growth and Development, Department of Ob/Gyn, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Aditi Singh
- CS Mott Center for Human Growth and Development, Department of Ob/Gyn, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Mohammed Abdulhasan
- CS Mott Center for Human Growth and Development, Department of Ob/Gyn, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan, USA
- Reproductive Stress 3M Inc, Grosse Pointe Farms, Michigan, USA
| | - Douglas M Ruden
- CS Mott Center for Human Growth and Development, Department of Ob/Gyn, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan, USA
- Invia Fertility Clinics, Hoffman Estates, Illinois, USA
- Institute for Environmental Health Science, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Awoniyi O Awonuga
- CS Mott Center for Human Growth and Development, Department of Ob/Gyn, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Daniel A Rappolee
- CS Mott Center for Human Growth and Development, Department of Ob/Gyn, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan, USA
- Reproductive Stress 3M Inc, Grosse Pointe Farms, Michigan, USA
- Invia Fertility Clinics, Hoffman Estates, Illinois, USA
- Institute for Environmental Health Science, Wayne State University School of Medicine, Detroit, Michigan, USA
- Program for Reproductive Sciences and Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Biology, University of Windsor, Windsor, Ontario, Canada
| |
Collapse
|
3
|
Mesenchymal Stem Cells in Embryo-Maternal Communication under Healthy Conditions or Viral Infections: Lessons from a Bovine Model. Cells 2022; 11:cells11121858. [PMID: 35740987 PMCID: PMC9221285 DOI: 10.3390/cells11121858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 11/17/2022] Open
Abstract
Bovine mesenchymal stem cells are a relevant cell population found in the maternal reproductive tract that exhibits the immunomodulation capacity required to prevent embryo rejection. The phenotypic plasticity showed by both endometrial mesenchymal stem cells (eMSC) and embryonic trophoblast through mesenchymal to epithelial transition and epithelial to mesenchymal transition, respectively, is essential for embryo implantation. Embryonic trophoblast maintains active crosstalk via EVs and soluble proteins with eMSC and peripheral blood MSC (pbMSC) to ensure the retention of eMSC in case of pregnancy and induce the chemotaxis of pbMSC, critical for successful implantation. Early pregnancy-related proteins and angiogenic markers are detected as cargo in EVs and the soluble fraction of the embryonic trophectoderm secretome. The pattern of protein secretion in trophectoderm-EVs changes depending on their epithelial or mesenchymal phenotype and due to the uptake of MSC EVs. However, the changes in this EV-mediated communication between maternal and embryonic MSC populations infected by viruses that cause abortions in cattle are poorly understood. They are critical in the investigation of reproductive viral pathologies.
Collapse
|
4
|
Dickson MJ, Oh Y, Gruzdev A, Li R, Balaguer N, Kelleher AM, Spencer TE, Wu SP, DeMayo FJ. Inserting Cre recombinase into the Prolactin 8a2 gene for decidua-specific recombination in mice. Genesis 2022; 60:e23473. [PMID: 35475540 PMCID: PMC9422316 DOI: 10.1002/dvg.23473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/11/2022] [Accepted: 03/18/2022] [Indexed: 12/14/2022]
Abstract
An estimated 75% of unsuccessful pregnancies are due to implantation failure. Investigating the causes of implantation failure is difficult as decidualization and embryo implantation is a dynamic process. Here, we describe a new decidua-specific iCre recombinase mouse strain. Utilizing CRISPR/Cas9-based genome editing, a mouse strain was developed that expresses iCre recombinase under the control of the endogenous prolactin family 8, subfamily a, member 2 (Prl8a2) promoter. iCre recombinase activity was examined by crossing with mTmG/+ or Sun1-GFP reporter alleles. iCre activity initiated reporter expression at gestational day 5.5 in the primary decidual zone and continued into mid-gestation (gestational day 9.5), with expression highly concentrated in the anti-mesometrial region. No reporter expression was observed in the ovary, oviduct, pituitary, or skeletal muscle, supporting the tissue specificity of the Prl8a2iCre in the primary decidual zone. This novel iCre line will be a valuable tool for in vivo genetic manipulation and lineage tracing to investigate functions of genetic networks and cellular dynamics associated with decidualization and infertility.
Collapse
Affiliation(s)
- Mackenzie J Dickson
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, North Carolina, USA
| | - Yeongseok Oh
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, North Carolina, USA
| | - Artiom Gruzdev
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, North Carolina, USA
| | - Rong Li
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, North Carolina, USA
| | - Nuria Balaguer
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, North Carolina, USA
| | - Andrew M Kelleher
- Division of Animal Sciences and Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, Missouri, USA
| | - Thomas E Spencer
- Division of Animal Sciences and Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, Missouri, USA
| | - San-Pin Wu
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, North Carolina, USA
| | - Francesco J DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, North Carolina, USA
| |
Collapse
|
5
|
Ahn SH, Nguyen SL, Kim TH, Jeong JW, Arora R, Lydon JP, Petroff MG. Nuclear Progesterone Receptor Expressed by the Cortical Thymic Epithelial Cells Dictates Thymus Involution in Murine Pregnancy. Front Endocrinol (Lausanne) 2022; 13:846226. [PMID: 35498436 PMCID: PMC9046655 DOI: 10.3389/fendo.2022.846226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/14/2022] [Indexed: 12/14/2022] Open
Abstract
Progesterone is a gonadal pro-gestational hormone that is absolutely necessary for the success of pregnancy. Most notable actions of progesterone are observed in the female reproductive organs, the uterus and the ovary. Acting through the nuclear progesterone receptor (PGR), progesterone prepares the endometrium for implantation of the embryo. Interestingly, the maternal thymus also is a known expressor of Pgr; its absence is associated with murine pregnancy complications. However, the localization of its expression and its functional importance were not known. Here, we used a transgenic dual fluorescent reporter mouse model and genetic deletion of Pgr in Foxn1+ thymic epithelial cells (TEC) to demonstrate TEC-specific Pgr expression in pregnancy, especially in the cortex where thymocyte maturation occurs. Using our TEC-specific Pgr deletion mouse model, we demonstrate that TEC-specific Pgr is necessary for pregnancy-induced thymic involution in pregnancy. Our investigation reveals that PGR expression is upregulated in the cortical thymic epithelial cells during pregnancy, and that PGR expression is important for thymic involution during murine pregnancy.
Collapse
Affiliation(s)
- Soo Hyun Ahn
- Department of Pathobiology Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
| | - Sean L. Nguyen
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, United States
| | - Tae Hoon Kim
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI, United States
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI, United States
| | - Ripla Arora
- Department of Obstetrics, Gynecology, and Reproductive Biology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - John P. Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Margaret G. Petroff
- Department of Pathobiology Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
6
|
Oluwayiose OA, Marcho C, Wu H, Houle E, Krawetz SA, Suvorov A, Mager J, Pilsner JR. Paternal preconception phthalate exposure alters sperm methylome and embryonic programming. ENVIRONMENT INTERNATIONAL 2021; 155:106693. [PMID: 34120004 PMCID: PMC8292217 DOI: 10.1016/j.envint.2021.106693] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 05/21/2023]
Abstract
Preconception environmental conditions have been demonstrated to shape sperm epigenetics and subsequently offspring health and development. Our previous findings in humans showed that urinary anti-androgenic phthalate metabolites in males were associated with altered sperm methylation and blastocyst-stage embryo development. To corroborate this, we examined the effect of preconception exposure to di(2-ethylhexyl) phthalate (DEHP) on genome-wide DNA methylation and gene expression profiles in mice. Eight-week old C57BL/6J male mice were exposed to either a vehicle control, low, or high dose of DEHP (2.5 and 25 mg/kg/weight, respectively) for 67 days (~2 spermatogenic cycles) and were subsequently mated with unexposed females. Reduced representation bisulfite sequencing (RRBS) of epididymal sperm was performed and gastrulation stage embryos were collected for RRBS and transcriptome analyses in both embryonic and extra-embryonic lineages. Male preconception DEHP exposure resulted in 704 differentially methylated regions (DMRs; q-value < 0.05; ≥10% methylation change) in sperm, 1,716 DMRs in embryonic, and 3,181 DMRs in extra-embryonic tissue. Of these, 29 DMRs overlapped between sperm and F1 tissues, half of which showed concordant methylation changes between F0 and F1 generations. F1 transcriptomes at E7.5 were also altered by male preconception DEHP exposure including developmental gene families such as Hox, Gata, and Sox. Additionally, gene ontology analyses of DMRs and differentially expressed genes showed enrichment of multiple developmental processes including embryonic development, pattern specification and morphogenesis. These data indicate that spermatogenesis in adult may represent a sensitive window in which exposure to DEHP alters the sperm methylome as well as DNA methylation and gene expression in the developing embryo.
Collapse
Affiliation(s)
- Oladele A Oluwayiose
- Department of Environmental Health Sciences, University of Massachusetts Amherst, Amherst, MA, USA
| | - Chelsea Marcho
- Department of Environmental Health Sciences, University of Massachusetts Amherst, Amherst, MA, USA; Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, USA
| | - Haotian Wu
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, NY, USA
| | - Emily Houle
- Department of Environmental Health Sciences, University of Massachusetts Amherst, Amherst, MA, USA
| | - Stephen A Krawetz
- Department of Obstetrics and Gynecology & Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Alexander Suvorov
- Department of Environmental Health Sciences, University of Massachusetts Amherst, Amherst, MA, USA
| | - Jesse Mager
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, USA
| | - J Richard Pilsner
- Department of Environmental Health Sciences, University of Massachusetts Amherst, Amherst, MA, USA; Department of Obstetrics and Gynecology & Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
7
|
Uterine Notch2 facilitates pregnancy recognition and corpus luteum maintenance via upregulating decidual Prl8a2. PLoS Genet 2021; 17:e1009786. [PMID: 34460816 PMCID: PMC8432799 DOI: 10.1371/journal.pgen.1009786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 09/10/2021] [Accepted: 08/18/2021] [Indexed: 02/07/2023] Open
Abstract
The maternal recognition of pregnancy is a necessary prerequisite for gestation maintenance through prolonging the corpus luteum lifespan and ensuring progesterone production. In addition to pituitary prolactin and placental lactogens, decidual derived prolactin family members have been presumed to possess luteotropic effect. However, there was a lack of convincing evidence to support this hypothesis. Here, we unveiled an essential role of uterine Notch2 in pregnancy recognition and corpus luteum maintenance. Uterine-specific deletion of Notch2 did not affect female fertility. Nevertheless, the expression of decidual Prl8a2, a member of the prolactin family, was downregulated due to Notch2 ablation. Subsequently, we interrupted pituitary prolactin function to determine the luteotropic role of the decidua by employing the lipopolysaccharide-induced prolactin resistance model, or blocking the prolactin signaling by prolactin receptor-Fc fusion protein, or repressing pituitary prolactin release by dopamine receptor agonist bromocriptine, and found that Notch2-deficient females were more sensitive to these stresses and ended up in pregnancy loss resulting from abnormal corpus luteum function and insufficient serum progesterone level. Overexpression of Prl8a2 in Notch2 knockout mice rescued lipopolysaccharide-induced abortion, highlighting its luteotropic function. Further investigation adopting Rbpj knockout and DNMAML overexpression mouse models along with chromatin immunoprecipitation assay and luciferase analysis confirmed that Prl8a2 was regulated by the canonical Notch signaling. Collectively, our findings demonstrated that decidual prolactin members, under the control of uterine Notch signaling, assisted pituitary prolactin to sustain corpus luteum function and serum progesterone level during post-implantation phase, which was conducive to pregnancy recognition and maintenance. Progesterone secreted from the corpus luteum in the ovary is indispensable to pregnancy maintenance in both rodents and humans. Therefore, prolonged corpus luteum lifespan and sustainable progesterone production is a prerequisite for a successful pregnancy. In rodents, in addition to pituitary prolactin and placental lactogens, decidual derived factors have been presumed to possess luteotropic effects during the post-implantation stage. In this study, utilizing a mouse model with uterine specific deletion of Notch2, which displayed decreased level of decidual prolactin member Prl8a2, combined with multiple approaches to interrupt the pituitary prolactin signal, we demonstrated that decidual derived Prl8a2 assisted pituitary prolactin to sustain corpus luteum function and serum progesterone level during post-implantation phase, which was conducive to pregnancy recognition and maintenance. In addition, the expression of decidual Prl8a2 was under the direct control of the canonical Notch pathway. Together, we herein provide convincing evidence that decidual produced Prl8a2, modulated by uterine canonical Notch signaling, exhibits luteotropic functions and contributes to pregnancy maintenance.
Collapse
|
8
|
Sexually dimorphic effects of forkhead box a2 (FOXA2) and uterine glands on decidualization and fetoplacental development. Proc Natl Acad Sci U S A 2020; 117:23952-23959. [PMID: 32900950 DOI: 10.1073/pnas.2014272117] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Glands of the uterus are essential for pregnancy establishment. Forkhead box A2 (FOXA2) is expressed specifically in the glands of the uterus and a critical regulator of glandular epithelium (GE) differentiation, development, and function. Mice with a conditional deletion of FOXA2 in the adult uterus, created using the lactotransferrin iCre (Ltf-iCre) model, have a morphologically normal uterus with glands, but lack FOXA2-dependent GE-expressed genes, such as leukemia inhibitory factor (LIF). Adult FOXA2 conditional knockout (cKO; Ltf iCre/+ Foxa2 f/f ) mice are infertile due to defective embryo implantation arising from a lack of LIF, a critical implantation factor of uterine gland origin. However, intraperitoneal injections of LIF can initiate embryo implantation in the uterus of adult FOXA2 cKO mice with pregnancies maintained to term. Here, we tested the hypothesis that FOXA2-regulated genes in the uterine glands impact development of the decidua, placenta, and fetus. On gestational day 8.5, the antimesometrial and mesometrial decidua transcriptome was noticeably altered in LIF-replaced FOXA2 cKO mice. Viable fetuses were reduced in FOXA2 cKO mice on gestational days 12.5 and 17.5. Sex-dependent differences in fetal weight, placenta histoarchitecture, and the placenta and metrial gland transcriptome were observed between control and FOXA2 cKO mice. The transcriptome of the placenta with a female fetus was considerably more altered than the placenta with a male fetus in FOXA2 cKO dams. These studies reveal previously unrecognized sexually dimorphic effects of FOXA2 and uterine glands on fetoplacental development with potential impacts on offspring health into adulthood.
Collapse
|
9
|
Zhang Y, Yan L, Liu J, Cui S, Qiu J. cGMP-dependent protein kinase II determines β-catenin accumulation that is essential for uterine decidualization in mice. Am J Physiol Cell Physiol 2019; 317:C1115-C1127. [PMID: 31509448 DOI: 10.1152/ajpcell.00208.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In the early phase of pregnancy, decidualization is an indispensable event after mammal embryo implantation, accompanied by proliferation and differentiation of uterine stromal cells. Type II cGMP-dependent protein kinase (Prkg2) belongs to the family of serine/threonine kinase, which plays multiple roles in cellular signaling pathways to control proliferation and differentiation. However, the regulatory function and molecular mechanism of Prkg2 in decidualization are still unknown. In this study, we show that Prkg2 has a gradually increased expression pattern during peri-implantation and artificial decidualization, and the expression of Prkg2 is induced by estrogen and progesterone in the ovariectomized mouse uteri and primary cultured uterine stromal cells, the process of which is blocked by treating with estrogen receptor (ER) antagonist (ICI-182,780) and progesterone receptor (PR) antagonist (RU-486). Inhibition of Prkg2 activity by HA-100 promotes uterine stromal cell proliferation but compromises decidualization with decreased expression of prolactin family 8, subfamily a, member 2. In addition, the functional regulation of decidualization by Prkg2 is accomplished by its induced phosphorylation of glycogen synthase kinase-3β (GSK-3β) at serine-9, which results in accumulation of β-catenin in the decidual cells. Taken together, our findings demonstrate that estrogen and progesterone upregulate the expression of Prkg2 in uterine stromal cells depending on ER and PR; Prkg2 promotes phosphorylation of GSK-3β at serine-9 and inactivates it, leading to the accumulation of β-catenin and promoting the process of decidualization. In addition to revealing the regulatory mechanism of Prkg2 that ensures the success of uterine decidualization, our findings will contribute to the understanding in the maintenance of early pregnancy.
Collapse
Affiliation(s)
- Yang Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, People's Republic of China.,State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Lu Yan
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Jiali Liu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Sheng Cui
- College of Veterinary Medicine, Yangzhou University, Yangzhou, People's Republic of China.,State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Jingtao Qiu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, People's Republic of China
| |
Collapse
|
10
|
Tal R, Shaikh S, Pallavi P, Tal A, López-Giráldez F, Lyu F, Fang YY, Chinchanikar S, Liu Y, Kliman HJ, Alderman M, Pluchino N, Kayani J, Mamillapalli R, Krause DS, Taylor HS. Adult bone marrow progenitors become decidual cells and contribute to embryo implantation and pregnancy. PLoS Biol 2019; 17:e3000421. [PMID: 31513564 PMCID: PMC6742226 DOI: 10.1371/journal.pbio.3000421] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 08/09/2019] [Indexed: 12/22/2022] Open
Abstract
Decidua is a transient uterine tissue shared by mammals with hemochorial placenta and is essential for pregnancy. The decidua is infiltrated by many immune cells promoting pregnancy. Adult bone marrow (BM)-derived cells (BMDCs) differentiate into rare populations of nonhematopoietic endometrial cells in the uterus. However, whether adult BMDCs become nonhematopoietic decidual cells and contribute functionally to pregnancy is unknown. Here, we show that pregnancy mobilizes mesenchymal stem cells (MSCs) to the circulation and that pregnancy induces considerable adult BMDCs recruitment to decidua, where some differentiate into nonhematopoietic prolactin-expressing decidual cells. To explore the functional importance of nonhematopoietic BMDCs to pregnancy, we used Homeobox a11 (Hoxa11)-deficient mice, having endometrial stromal-specific defects precluding decidualization and successful pregnancy. Hoxa11 expression in BM is restricted to nonhematopoietic cells. BM transplant (BMT) from wild-type (WT) to Hoxa11-/- mice results in stromal expansion, gland formation, and marked decidualization otherwise absent in Hoxa11-/- mice. Moreover, in Hoxa11+/- mice, which have increased pregnancy losses, BMT from WT donors leads to normalized uterine expression of numerous decidualization-related genes and rescue of pregnancy loss. Collectively, these findings reveal that adult BMDCs have a previously unrecognized nonhematopoietic physiologic contribution to decidual stroma, thereby playing important roles in decidualization and pregnancy.
Collapse
Affiliation(s)
- Reshef Tal
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Shafiq Shaikh
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Pallavi Pallavi
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Aya Tal
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Francesc López-Giráldez
- Yale Center for Genome Analysis (YCGA), Yale University, New Haven, Connecticut, United States of America
| | - Fang Lyu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Yuan-Yuan Fang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Shruti Chinchanikar
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Ying Liu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Harvey J. Kliman
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Myles Alderman
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Nicola Pluchino
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Jehanzeb Kayani
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Ramanaiah Mamillapalli
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Diane S. Krause
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Hugh S. Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
11
|
Chen W, Zhang Q, Wang H, Tan D, Tan Y. Unique and independent role of the GABA B1 subunit in embryo implantation and uterine decidualization in mice. Genes Dis 2019; 8:79-86. [PMID: 33569516 PMCID: PMC7859463 DOI: 10.1016/j.gendis.2019.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/18/2019] [Accepted: 06/19/2019] [Indexed: 01/26/2023] Open
Abstract
Embryo implantation and decidualization are crucial for successful pregnancy, which include multiple genes and signaling pathways, while the precise mechanism regarding embryo implantation and decidualization has yet to be explored. The GABA which activates GABAA or GABAB receptors has been found playing an important role in early pregnancy. Here we seek to investigate whether GABAB receptors participate in embryo implantation in mice. This study first characterized the spatiotemporal expression pattern of GABAB receptors in the uterus during the peri-implantation period and found that GABAB1 expression was drastically upregulated in stromal cells on days 4–6, a period of embryo implantation and early stages of decidualization. Embryo delayed implantation and oil-induced decidualization models were further used to confirm that the GABAB1 was associated with embryo implantation and decidualization. We also found estrogen or progesterone had no directly effect on expression of GABAB1 in ovariectomized model. Because we were unable to detect significant GABAB2 which couples with GABAB1 to form whole GABAB receptors, and the agonist and antagonist of whole GABAB receptors had weak effect on the proliferation and differentiation of stromal cells as well, we excluded the possibility whole GABAB receptors function, and concluded it should be non-classical signals of GABAB1 involving in embryo implantation and decidualization. Future studies should focus on investigating the roles and mechanisms of GABAB1 during embryo implantation and decidualization.
Collapse
Affiliation(s)
- Wenhao Chen
- Laboratory Animal Center, Chongqing Medical University, Chongqing, 400016, PR China
| | - Qian Zhang
- Laboratory Animal Center, Chongqing Medical University, Chongqing, 400016, PR China
| | - Haibin Wang
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, Fujian, PR China.,Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen, 361102, Fujian, PR China
| | - Dongmei Tan
- Laboratory Animal Center, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yi Tan
- Laboratory Animal Center, Chongqing Medical University, Chongqing, 400016, PR China
| |
Collapse
|
12
|
Zhang Q, Hao J, Li G. Deletion of Prl7d1 causes placental defects at mid-pregnancy in mice. Mol Reprod Dev 2019; 86:696-713. [PMID: 31012985 DOI: 10.1002/mrd.23148] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 03/16/2019] [Accepted: 03/29/2019] [Indexed: 12/29/2022]
Abstract
Prolactin family 7, subfamily d, member 1 (Prl7d1), a member of the expanding prolactin family, is mainly expressed in the placental junctional zone (including trophoblast giant cells and spongiotrophoblast cells) with peak expression observed at 12 days postcoitum (dpc) in mice. Previous studies have shown that PRL7D1 is a key mediator of angiogenesis in vitro; however, its physiological roles in placental development in vivo have not been characterized. To address this issue, we deleted Prl7d1 in mice and demonstrated that its absence results in reduced litter size and fertility. Histologically, Prl7d1 mutants exhibited striking placental abnormalities at 12.5 dpc, including a reduction in the proportion of labyrinth layers and a significant increase in decidual natural killer cells, glycogen trophoblasts, and trophoblast giant cells in the junctional zone. Moreover, placentas from Prl7d1-null mice displayed a thickened decidual spiral artery. Notably, these negative effects were more pronounced in male fetuses. Further RNA-sequencing analysis showed that Prl7d1 deletion results in significant differences in the placental transcriptome profile between the two sexes of fetuses. Together, this study demonstrates that Prl7d1 possesses antiangiogenic properties in deciduas and inhibits the development of junctional zone, which potentially alters the functional capacity of the placenta to support optimal fetal growth. Moreover, of note, the role of Prl7d1 in the placenta is regulated in a fetal sex-specific manner.
Collapse
Affiliation(s)
- Qiong Zhang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Jie Hao
- Experimental Research Center, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Gang Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| |
Collapse
|
13
|
Gao Y, Wang Y, Zhou C, Kong S, Lu J, Wang H, Yang J. Ubiquitin-specific protease 7 (USP7) is essential for endometrial stromal cell decidualization in mice. Dev Growth Differ 2019; 61:176-185. [PMID: 30628051 DOI: 10.1111/dgd.12594] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 12/27/2022]
Abstract
Ubiquitin-specific protease 7 (USP7), a member of the deubiquitinating (DUB) enzyme family, regulates protein stability and has a well-characterized function in tumorigenesis. Given its critical role in growth and development, it was speculated to be involved in modulating processes in the female reproductive system but its exact role has not been elucidated. Decidualization is one of the key processes in pregnancy and aberrant decidualization is a cause of pregnancy failure. The uterine endometrium layer undergoes significant structural and functional changes during decidualization in preparation for and after embryo implantation. Here, we hypothesized that USP7 could be involved in mediating endometrial stromal cell (ESC) decidualization and set out to determine its function with a primary stromal cell culture. Using in situ hybridization and immunohistochemical techniques, we observed increased USP7 expression during uterine decidualization and found that it was predominantly localized to the decidual zone in the post-implantation uterus. Since the ovarian hormones, progesterone (P4) and estrogen (E2), function in promoting stroma decidualization, we investigated their relationship with USP7 expression and found that they exert minimal influence. Moreover, increased USP7 expression observed during deciduoma development was found to be independent of blastocyst attachment. Using a specific USP7 inhibitor, HBX19818, we demonstrated an additional novel role for USP7 in endometrial stroma decidualization in mice during early pregnancy. Our findings could potentially be applied towards future research and development in female infertility.
Collapse
Affiliation(s)
- Yue Gao
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, China
| | - Yaqin Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, China
| | - Chan Zhou
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
- Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen, Fujian, China
| | - Shuangbo Kong
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
- Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen, Fujian, China
| | - Jinhua Lu
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
- Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen, Fujian, China
| | - Haibin Wang
- Reproductive Medical Center, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
- Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen, Fujian, China
| | - Jing Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, China
| |
Collapse
|
14
|
Nteeba J, Kubota K, Wang W, Zhu H, Vivian JL, Dai G, Soares MJ. Pancreatic prolactin receptor signaling regulates maternal glucose homeostasis. J Endocrinol 2019; 241:JOE-18-0518.R2. [PMID: 30798322 PMCID: PMC7189340 DOI: 10.1530/joe-18-0518] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 02/22/2019] [Indexed: 12/17/2022]
Abstract
Prolactin (PRL) signaling has been implicated in the regulation of glucose homeostatic adaptations to pregnancy. In this report, the PRL receptor (Prlr) gene was conditionally disrupted in the pancreas, creating an animal model which proved useful for investigating the biology and pathology of gestational diabetes including its impacts on fetal and placental development. In mice, pancreatic PRLR signaling was demonstrated to be required for pregnancy-associated changes in maternal β cell mass and function. Disruption of the Prlr gene in the pancreas resulted in fewer insulin producing cells, which failed to expand appropriately during pregnancy resulting in reduced blood insulin levels and maternal glucose intolerance. This inability to sustain normal blood glucose balance during pregnancy worsened with age and a successive pregnancy. The etiology of the insulin insufficiency was attributed to deficits in regulatory pathways controlling β cell development. Additionally, the disturbance in maternal blood glucose homeostasis, was associated with fetal overgrowth and dysregulation of inflammation and prolactin-associated transcripts in the placenta. Overall, these results indicate that the PRLR, acting within the pancreas, mediates maternal pancreatic adaptations to pregnancy and therefore its dysfunction may increase a woman's chances of becoming glucose intolerant during pregnancy.
Collapse
Affiliation(s)
- Jackson Nteeba
- Department of Pathology and Laboratory Medicine, Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Kaiyu Kubota
- Department of Pathology and Laboratory Medicine, Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Wenfang Wang
- Department of Clinical Laboratory Sciences, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Hao Zhu
- Department of Clinical Laboratory Sciences, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jay L Vivian
- Department of Pathology and Laboratory Medicine, Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Guoli Dai
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Michael J Soares
- Department of Pathology and Laboratory Medicine, Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, Kansas, USA
- Center for Perinatal Research, Children’s Research Institute, Children’s Mercy, Kansas City, Missouri, USA
| |
Collapse
|
15
|
Calle A, López-Martín S, Monguió-Tortajada M, Borràs FE, Yáñez-Mó M, Ramírez MÁ. Bovine endometrial MSC: mesenchymal to epithelial transition during luteolysis and tropism to implantation niche for immunomodulation. Stem Cell Res Ther 2019; 10:23. [PMID: 30635057 PMCID: PMC6330450 DOI: 10.1186/s13287-018-1129-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 12/10/2018] [Accepted: 12/27/2018] [Indexed: 12/20/2022] Open
Abstract
Background The uterus is a histologically dynamic organ, and the mechanisms coordinating its regeneration during the oestrous cycle and implantation are poorly understood. The aim of this study was to isolate, immortalize and characterize bovine endometrial mesenchymal stem cell (eMSC) lines from different oestrous cycle stages (embryo in the oviduct, embryo in the uterus or absence of embryo) and examine their migratory and immunomodulatory properties in an inflammatory or implantation-like environment, as well as possible changes in cell transdifferentiation. Methods eMSCs were isolated and analysed in terms of morphological features, expression of cell surface and intracellular markers of pluripotency, inmunocytochemical analyses, alkaline phosphatase activity, proliferation and osteogenic or chondrogenic differentiation capacities, as well as their ability to migrate in response to inflammatory (TNF-α or IL-1β) or implantation (IFN-τ) cytokines and their immunomodulatory effect in the proliferation of T cells. Results All eMSCs showed MSC properties such as adherence to plastic, high proliferative capacity, expression of CD44 and vimentin, undetectable expression of CD34 or MHCII, positivity for Pou5F1 and alkaline phosphatase activity. In the absence of an embryo, eMSC showed an apparent mesenchymal to epithelial transition state. eMSC during the entire oestrous cycle differentiated to osteogenic or chondrogenic lineages, showed the ability to suppress T cell proliferation and showed migratory capacity towards pro-inflammatory signal, while responded with a block in their migration to the embryo-derived pregnancy signal. Conclusion This study describes for the first time the isolation, immortalization and characterization of bovine mesenchymal stem cell lines from different oestrous cycle stages, with a clear mesenchymal pattern and immunomodulatory properties. Our study also reports that the migratory capacity of the eMSC was increased towards an inflammatory niche but was reduced in response to the expression of implantation cytokine by the embryo. The combination of both signals (pro-inflammatory and implantation) would ensure the retention of eMSC in case of pregnancy, to ensure the immunomodulation necessary in the mother for embryo survival. In addition, in the absence of an embryo, eMSC showed an apparent mesenchymal to epithelial transition state.
Collapse
Affiliation(s)
- Alexandra Calle
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Avenida Puerta de Hierro 12, local 10, 28040, Madrid, Spain
| | | | - Marta Monguió-Tortajada
- REMAR Group and Nephrology Service, Germans Trias i Pujol Health Science Institute & University Hospital, UAB, Badalona, Spain
| | - Francesc Enric Borràs
- REMAR Group and Nephrology Service, Germans Trias i Pujol Health Science Institute & University Hospital, UAB, Badalona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - María Yáñez-Mó
- Departamento de Biología Molecular, UAM, Madrid, Spain.,CBM-SO, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Miguel Ángel Ramírez
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Avenida Puerta de Hierro 12, local 10, 28040, Madrid, Spain.
| |
Collapse
|
16
|
Yang Y, Abdulhasan M, Awonuga A, Bolnick A, Puscheck EE, Rappolee DA. Hypoxic Stress Forces Adaptive and Maladaptive Placental Stress Responses in Early Pregnancy. Birth Defects Res 2018; 109:1330-1344. [PMID: 29105384 DOI: 10.1002/bdr2.1149] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 10/07/2017] [Indexed: 12/19/2022]
Abstract
This review focuses on hypoxic stress and its effects on the placental lineage and the earliest differentiation events in mouse and human placental trophoblast stem cells (TSCs). Although the placenta is a decidual organ at the end of pregnancy, its earliest rapid growth and function at the start of pregnancy precedes and supports growth and function of the embryo. Earliest function requires that TSCs differentiate, however, "hypoxia" supports rapid growth, but not differentiation of TSCs. Most of the literature on earliest placental "hypoxia" studies used 2% oxygen which is normoxic for TSCs. Hypoxic stress happens when oxygen level drops below 2%. It decreases anabolism, proliferation, potency/stemness and increases differentiation, despite culture conditions that would sustain proliferation and potency. Thus, to study the pathogenesis due to TSC dysfunction, it is important to study hypoxic stress below 2%. Many studies have been performed using 0.5 to 1% oxygen in cultured mouse TSCs. From all these studies, a small number has examined human trophoblast lines and primary first trimester placental hypoxic stress responses in culture. Some other stress stimuli, aside from hypoxic stress, are used to elucidate common and unique aspects of hypoxic stress. The key outcomes produced by hypoxic stress are mitochondrial, anabolic, and proliferation arrest, and this is coupled with stemness loss and differentiation. Hypoxic stress can lead to depletion of stem cells and miscarriage, or can lead to later dysfunctions in placentation and fetal development. Birth Defects Research 109:1330-1344, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yu Yang
- CS Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Mohammed Abdulhasan
- CS Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan
| | - Awoniyi Awonuga
- CS Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan
| | - Alan Bolnick
- CS Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan
| | - Elizabeth E Puscheck
- CS Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan
| | - Daniel A Rappolee
- CS Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Reproductive Endocrinology and Infertility, Wayne State University School of Medicine, Detroit, Michigan.,Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan.,Institutes for Environmental Health Science, Wayne state University School of Medicine, Detroit, Michigan.,Department of Biology, University of Windsor, Windsor, ON, Canada
| |
Collapse
|
17
|
Soares MJ, Iqbal K, Kozai K. Hypoxia and Placental Development. Birth Defects Res 2018; 109:1309-1329. [PMID: 29105383 DOI: 10.1002/bdr2.1135] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/04/2017] [Indexed: 12/17/2022]
Abstract
Hemochorial placentation is orchestrated through highly regulated temporal and spatial decisions governing the fate of trophoblast stem/progenitor cells. Trophoblast cell acquisition of specializations facilitating invasion and uterine spiral artery remodeling is a labile process, sensitive to the environment, and represents a process that is vulnerable to dysmorphogenesis in pathologic states. Hypoxia is a signal guiding placental development, and molecular mechanisms directing cellular adaptations to low oxygen tension are integral to trophoblast cell differentiation and placentation. Hypoxia can also be used as an experimental tool to investigate regulatory processes controlling hemochorial placentation. These developmental processes are conserved in mouse, rat, and human placentation. Consequently, elements of these developmental events can be modeled and hypotheses tested in trophoblast stem cells and in genetically manipulated rodents. Hypoxia is also a consequence of a failed placenta, yielding pathologies that can adversely affect maternal adjustments to pregnancy, fetal health, and susceptibility to adult disease. The capacity of the placenta for adaptation to environmental challenges highlights the importance of its plasticity in safeguarding a healthy pregnancy. Birth Defects Research 109:1309-1329, 2017.© 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas.,Fetal Health Research, Children's Research Institute, Children's Mercy, Kansas City, Missouri
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Keisuke Kozai
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
18
|
Bu P, Yagi S, Shiota K, Alam SMK, Vivian JL, Wolfe MW, Rumi MAK, Chakraborty D, Kubota K, Dhakal P, Soares MJ. Origin of a rapidly evolving homeostatic control system programming testis function. J Endocrinol 2017; 234:217-232. [PMID: 28576872 PMCID: PMC5529123 DOI: 10.1530/joe-17-0250] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 06/02/2017] [Indexed: 12/24/2022]
Abstract
Mammals share common strategies for regulating reproduction, including a conserved hypothalamic-pituitary-gonadal axis; yet, individual species exhibit differences in reproductive performance. In this report, we describe the discovery of a species-restricted homeostatic control system programming testis growth and function. Prl3c1 is a member of the prolactin gene family and its protein product (PLP-J) was discovered as a uterine cytokine contributing to the establishment of pregnancy. We utilized mouse mutagenesis of Prl3c1 and revealed its involvement in the regulation of the male reproductive axis. The Prl3c1-null male reproductive phenotype was characterized by testiculomegaly and hyperandrogenism. The larger testes in the Prl3c1-null mice were associated with an expansion of the Leydig cell compartment. Prl3c1 locus is a template for two transcripts (Prl3c1-v1 and Prl3c1-v2) expressed in a tissue-specific pattern. Prl3c1-v1 is expressed in uterine decidua, while Prl3c1-v2 is expressed in Leydig cells of the testis. 5'RACE, chromatin immunoprecipitation and DNA methylation analyses were used to define cell-specific promoter usage and alternative transcript expression. We examined the Prl3c1 locus in five murid rodents and showed that the testicular transcript and encoded protein are the result of a recent retrotransposition event at the Mus musculus Prl3c1 locus. Prl3c1-v1 encodes PLP-J V1 and Prl3c1-v2 encodes PLP-J V2. Each protein exhibits distinct intracellular targeting and actions. PLP-J V2 possesses Leydig cell-static actions consistent with the Prl3c1-null testicular phenotype. Analysis of the biology of the Prl3c1 gene has provided insight into a previously unappreciated homeostatic setpoint control system programming testicular growth and function.
Collapse
Affiliation(s)
- Pengli Bu
- Institute for Reproductive Health and Regenerative MedicineUniversity of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Pathology and Laboratory MedicineUniversity of Kansas Medical Center, Kansas City, Kansas, USA
| | - Shintaro Yagi
- Laboratory of Cellular BiochemistryVeterinary Medical Sciences/Animal Resource Sciences, The University of Tokyo, Tokyo, Japan
| | - Kunio Shiota
- Laboratory of Cellular BiochemistryVeterinary Medical Sciences/Animal Resource Sciences, The University of Tokyo, Tokyo, Japan
- Waseda Research Institute for Science and EngineeringWaseda University, Tokyo, Japan
| | - S M Khorshed Alam
- Institute for Reproductive Health and Regenerative MedicineUniversity of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Pathology and Laboratory MedicineUniversity of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jay L Vivian
- Institute for Reproductive Health and Regenerative MedicineUniversity of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Pathology and Laboratory MedicineUniversity of Kansas Medical Center, Kansas City, Kansas, USA
| | - Michael W Wolfe
- Institute for Reproductive Health and Regenerative MedicineUniversity of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Molecular and Integrative PhysiologyUniversity of Kansas Medical Center, Kansas City, Kansas, USA
| | - M A Karim Rumi
- Institute for Reproductive Health and Regenerative MedicineUniversity of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Pathology and Laboratory MedicineUniversity of Kansas Medical Center, Kansas City, Kansas, USA
| | - Damayanti Chakraborty
- Institute for Reproductive Health and Regenerative MedicineUniversity of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Pathology and Laboratory MedicineUniversity of Kansas Medical Center, Kansas City, Kansas, USA
| | - Kaiyu Kubota
- Institute for Reproductive Health and Regenerative MedicineUniversity of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Pathology and Laboratory MedicineUniversity of Kansas Medical Center, Kansas City, Kansas, USA
| | - Pramod Dhakal
- Institute for Reproductive Health and Regenerative MedicineUniversity of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Pathology and Laboratory MedicineUniversity of Kansas Medical Center, Kansas City, Kansas, USA
| | - Michael J Soares
- Institute for Reproductive Health and Regenerative MedicineUniversity of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Pathology and Laboratory MedicineUniversity of Kansas Medical Center, Kansas City, Kansas, USA
- Department of PediatricsUniversity of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
19
|
Deng W, Cha J, Yuan J, Haraguchi H, Bartos A, Leishman E, Viollet B, Bradshaw HB, Hirota Y, Dey SK. p53 coordinates decidual sestrin 2/AMPK/mTORC1 signaling to govern parturition timing. J Clin Invest 2016; 126:2941-54. [PMID: 27454290 DOI: 10.1172/jci87715] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/24/2016] [Indexed: 12/15/2022] Open
Abstract
Inflammation and oxidative stress are known risk factors for preterm birth (PTB); however, the mechanisms and pathways that influence this condition are not fully described. Previously, we showed that mTORC1 signaling is increased in mice harboring a uterine-specific deletion of transformation-related protein 53 (p53d/d mice), which exhibit premature decidual senescence that triggers spontaneous and inflammation-induced PTB. Treatment with the mTORC1 inhibitor rapamycin reduced the incidence of PTB in the p53d/d mice. Decidual senescence with heightened mTORC1 signaling is also a signature of human PTB. Here, we have identified an underlying mechanism for PTB and a potential therapeutic strategy for treating the condition. Treatment of pregnant p53d/d mice with either the antidiabetic drug metformin or the antioxidant resveratrol activated AMPK signaling and inhibited mTORC1 signaling in decidual cells. Both metformin and resveratrol protected against spontaneous and inflammation-induced PTB in p53d/d females. Using multiple approaches, we determined that p53 interacts with sestrins to coordinate an inverse relationship between AMPK and mTORC1 signaling that determines parturition timing. This signature was also observed in human decidual cells. Together, these results reveal that p53-dependent coordination of AMPK and mTORC1 signaling controls parturition timing and suggest that metformin and resveratrol have therapeutic potential to prevent PTB.
Collapse
|
20
|
Bu P, Alam SMK, Dhakal P, Vivian JL, Soares MJ. A Prolactin Family Paralog Regulates Placental Adaptations to a Physiological Stressor. Biol Reprod 2016; 94:107. [PMID: 26985002 PMCID: PMC4939737 DOI: 10.1095/biolreprod.115.138032] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/10/2016] [Indexed: 11/21/2022] Open
Abstract
The prolactin (PRL) family of hormones and cytokines participates in the regulation of optimal reproductive performance in the mouse and rat. Members of the PRL family are expressed in the anterior pituitary, uterus, and/or placenta. In the present study, we investigated the ontogeny of PRL family 7, subfamily b, member 1 (PRL7B1; also called PRL-like protein-N, PLP-N) expression in the developing mouse placenta and established a mouse model for investigating the biological function of PRL7B1. Transcripts for Prl7b1 were first detected on Gestation Day (d) 8.5. From gestation d8.5 through d14.5, Prl7b1 was expressed in trophoblast cells residing at the interface between maternal mesometrial decidua and the developing placenta. On gestation d17.5, the predominant cellular source of Prl7b1 mRNA was migratory trophoblast cells invading into the uterine mesometrial decidua. The Prl7b1 null mutant allele was generated via replacement of the endogenous Prl7b1 coding sequence with beta-galactosidase (LacZ) reporter and neomycin cassettes. The mutant Prl7b1 allele was successfully passed through the germline. Homozygous Prl7b1 mutant mice were viable and fertile. Under standard animal housing conditions, Prl7b1 had undetectable effects on placentation and pregnancy. Hypoxia exposure during pregnancy evoked adaptations in the organization of the wild-type placenta that were not observed in Prl7b1 null placentation sites. In summary, PRL7B1 is viewed as a part of a pathway regulating placental adaptations to physiological stressors.
Collapse
Affiliation(s)
- Pengli Bu
- Institute for Reproductive Health and Regenerative Medicine, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Sheikh M Khorshed Alam
- Institute for Reproductive Health and Regenerative Medicine, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Pramod Dhakal
- Institute for Reproductive Health and Regenerative Medicine, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Jay L Vivian
- Institute for Reproductive Health and Regenerative Medicine, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Michael J Soares
- Institute for Reproductive Health and Regenerative Medicine, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
21
|
Sferruzzi-Perri AN, Camm EJ. The Programming Power of the Placenta. Front Physiol 2016; 7:33. [PMID: 27014074 PMCID: PMC4789467 DOI: 10.3389/fphys.2016.00033] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 01/25/2016] [Indexed: 12/23/2022] Open
Abstract
Size at birth is a critical determinant of life expectancy, and is dependent primarily on the placental supply of nutrients. However, the placenta is not just a passive organ for the materno-fetal transfer of nutrients and oxygen. Studies show that the placenta can adapt morphologically and functionally to optimize substrate supply, and thus fetal growth, under adverse intrauterine conditions. These adaptations help meet the fetal drive for growth, and their effectiveness will determine the amount and relative proportions of specific metabolic substrates supplied to the fetus at different stages of development. This flow of nutrients will ultimately program physiological systems at the gene, cell, tissue, organ, and system levels, and inadequacies can cause permanent structural and functional changes that lead to overt disease, particularly with increasing age. This review examines the environmental regulation of the placental phenotype with particular emphasis on the impact of maternal nutritional challenges and oxygen scarcity in mice, rats and guinea pigs. It also focuses on the effects of such conditions on fetal growth and the developmental programming of disease postnatally. A challenge for future research is to link placental structure and function with clinical phenotypes in the offspring.
Collapse
Affiliation(s)
| | - Emily J Camm
- Department of Physiology, Development and Neuroscience, University of Cambridge Cambridge, UK
| |
Collapse
|
22
|
Qin A, Qin J, Jin Y, Xie W, Fan L, Jiang L, Mo F. DHEA improves the antioxidant capacity of endometrial stromal cells and improves endometrium receptivity via androgen receptor. Eur J Obstet Gynecol Reprod Biol 2016; 198:120-126. [DOI: 10.1016/j.ejogrb.2016.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 12/17/2015] [Accepted: 01/02/2016] [Indexed: 11/15/2022]
|
23
|
Alam SMK, Konno T, Soares MJ. Identification of target genes for a prolactin family paralog in mouse decidua. Reproduction 2016; 149:625-32. [PMID: 25926690 DOI: 10.1530/rep-15-0107] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Prolactin family 8, subfamily a, member 2 (PRL8A2; also called decidual prolactin-related protein; dPRP) is a member of the expanded prolactin family. PRL8A2 is expressed in the uterine decidua and contributes to pregnancy-dependent adaptations to hypoxia. The purpose of this study was to identify gene targets for PRL8A2 action within the uteroplacental compartment. Affymetrix DNA microarray analysis was performed for RNA samples from WT and Prl8a2 null tissues. Validation of the DNA microarray was performed using quantitative RT-PCR. Nine genes were confirmed with decreased expression in Prl8a2 null tissues (e.g., Klk7, Rimklb, Arhgef6, Calm4, Sprr2h, Prl4a1, Ccl27, Lipg, and Htra3). These include potential decidual, endothelial and trophoblast cell targets positively regulated by PRL8A2. A significant upregulation of Derl3, Herpud1, Creld2, Hsp90b1, Ddit3 and Hspa5 was identified in Prl8a2 null tissues, reflecting an increased endoplasmic reticulum (ER) stress response. ER stress genes were prominently expressed in the uterine decidua. We propose that PRL8A2 is a mediator of progesterone-dependent modulation of intrauterine responses to physiological stressors.
Collapse
Affiliation(s)
- S M Khorshed Alam
- Department of Pathology and Laboratory MedicineInstitute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Toshihiro Konno
- Department of Pathology and Laboratory MedicineInstitute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Michael J Soares
- Department of Pathology and Laboratory MedicineInstitute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| |
Collapse
|
24
|
Bloise E, Ortiga-Carvalho TM, Reis FM, Lye SJ, Gibb W, Matthews SG. ATP-binding cassette transporters in reproduction: a new frontier. Hum Reprod Update 2015; 22:164-81. [PMID: 26545808 DOI: 10.1093/humupd/dmv049] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 10/19/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The transmembrane ATP-binding cassette (ABC) transporters actively efflux an array of clinically relevant compounds across biological barriers, and modulate biodistribution of many physiological and pharmacological factors. To date, over 48 ABC transporters have been identified and shown to be directly and indirectly involved in peri-implantation events and fetal/placental development. They efflux cholesterol, steroid hormones, vitamins, cytokines, chemokines, prostaglandins, diverse xenobiotics and environmental toxins, playing a critical role in regulating drug disposition, immunological responses and lipid trafficking, as well as preventing fetal accumulation of drugs and environmental toxins. METHODS This review examines ABC transporters as important mediators of placental barrier functions and key reproductive processes. Expression, localization and function of all identified ABC transporters were systematically reviewed using PubMed and Google Scholar websites to identify relevant studies examining ABC transporters in reproductive tissues in physiological and pathophysiological states. Only reports written in English were incorporated with no restriction on year of publication. While a major focus has been placed on the human, extensive evidence from animal studies is utilized to describe current understanding of the regulation and function of ABC transporters relevant to human reproduction. RESULTS ABC transporters are modulators of steroidogenesis, fertilization, implantation, nutrient transport and immunological responses, and function as 'gatekeepers' at various barrier sites (i.e. blood-testes barrier and placenta) against potentially harmful xenobiotic factors, including drugs and environmental toxins. These roles appear to be species dependent and change as a function of gestation and development. The best-described ABC transporters in reproductive tissues (primarily in the placenta) are the multidrug transporters p-glycoprotein and breast cancer-related protein, the multidrug resistance proteins 1 through 5 and the cholesterol transporters ABCA1 and ABCG1. CONCLUSIONS The ABC transporters have various roles across multiple reproductive tissues. Knowledge of efflux direction, tissue distribution, substrate specificity and regulation of the ABC transporters in the placenta and other reproductive tissues is rapidly expanding. This will allow better understanding of the disposition of specific substrates within reproductive tissues, and facilitate development of novel treatments for reproductive disorders as well as improved approaches to protecting the developing fetus.
Collapse
Affiliation(s)
- E Bloise
- Laboratory of Translational Endocrinology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - T M Ortiga-Carvalho
- Laboratory of Translational Endocrinology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - F M Reis
- Division of Human Reproduction, Department of Obstetrics and Gynecology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - S J Lye
- Department of Physiology, Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, Canada M5S 1A8 Department Obstetrics & Gynecology, University of Toronto, Toronto, ON, Canada Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - W Gibb
- Department of Obstetrics & Gynecology, University of Ottawa, Ottawa, ON, Canada Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - S G Matthews
- Department of Physiology, Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, Canada M5S 1A8 Department Obstetrics & Gynecology, University of Toronto, Toronto, ON, Canada Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| |
Collapse
|
25
|
Kautz E, de Carvalho Papa P, Reichler IM, Gram A, Boos A, Kowalewski MP. In vitro decidualisation of canine uterine stromal cells. Reprod Biol Endocrinol 2015; 13:85. [PMID: 26242174 PMCID: PMC4526293 DOI: 10.1186/s12958-015-0066-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/16/2015] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND The uterine response to the presence of embryos is poorly understood in the domestic dog (Canis familiaris). The intimate embryo-maternal cross-talk, which begins following the hatching of blastocysts and embryo attachment leads to strong structural and functional remodelling of the uterus. A part of this process is decidualisation, comprising morphological and biochemical changes that result in formation of maternal stroma-derived decidual cells. These are an integral part of the canine placenta materna, which together with the maternal vascular endothelium are the only cells of the canine endotheliochorial placenta able to resist trophoblast invasion. These cells are also the only ones within the canine placenta expressing the progesterone receptor (PGR). Understanding the decidualisation process thus appears essential for understanding canine reproductive physiology. METHODS Here, we investigated the capability of canine uterine stromal cells to decidualise in vitro, thereby serving as a canine model of decidualisation. A dbcAMP-mediated approach was chosen during a time course of 24 - 72 h. Tissue material from six (n = 6) healthy, dioestric bitches was used (approximately 2 weeks after ovulation). Cells were characterized by differential staining, nearly 100 % of which were vimentin-positive. Scanning and transmission electron microscope analyses were applied, and morphological changes were recorded with a live cell imaging microscope. Expression of several decidualisation markers was investigated. RESULTS The in vitro cultured stromal cells acquired characteristics of decidual cells when incubated with 0.5 mM dbcAMP for 72 h. Their shape changed from elongated to rounded, while ultrastructural analysis revealed higher numbers of mitochondria and secretory follicles, and an increased proliferation rate. Elevated expression levels of IGF1, IGF2, PRLR and ERα were observed in decidualised cells; PRL and ERβ remained mostly below the detection limit, while PGR remained unaffected. The expression of smooth muscle α actin (αSMA), another decidualisation marker, was strongly induced. Among prostaglandin system members, levels of COX2 (PTGS2) and of PGE2-synthase (PTGES) were upregulated. Expression of the PGE2 receptors, PTGER2 and PTGER4, was clearly detectable. CONCLUSION An in vitro decidualisation model with canine uterine stromal cells was successfully established, allowing future, more detailed studies to be undertaken on the underlying molecular and endocrine mechanisms of canine decidualisation.
Collapse
Affiliation(s)
- Ewa Kautz
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, CH-8057, Zurich, Switzerland.
| | - Paula de Carvalho Papa
- Department of Surgery, Faculty of Veterinary Medicine and Animal Sciences, Sector of Anatomy, University of Sao Paulo, Sao Paulo, Brazil.
| | - Iris M Reichler
- Section of Small Animal Reproduction, Clinic of Reproductive Medicine, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.
| | - Aykut Gram
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, CH-8057, Zurich, Switzerland.
| | - Alois Boos
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, CH-8057, Zurich, Switzerland.
| | - Mariusz P Kowalewski
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, CH-8057, Zurich, Switzerland.
| |
Collapse
|
26
|
He H, Kong S, Liu F, Zhang S, Jiang Y, Liao Y, Jiang Y, Li Q, Wang B, Zhou Z, Wang H, Huo R. Rbbp7 Is Required for Uterine Stromal Decidualization in Mice1. Biol Reprod 2015; 93:13. [DOI: 10.1095/biolreprod.115.129015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 05/27/2015] [Indexed: 01/29/2023] Open
|
27
|
Mestre-Citrinovitz AC, Kleff V, Vallejo G, Winterhager E, Saragüeta P. A Suppressive Antagonism Evidences Progesterone and Estrogen Receptor Pathway Interaction with Concomitant Regulation of Hand2, Bmp2 and ERK during Early Decidualization. PLoS One 2015; 10:e0124756. [PMID: 25897495 PMCID: PMC4405574 DOI: 10.1371/journal.pone.0124756] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 03/06/2015] [Indexed: 01/01/2023] Open
Abstract
Progesterone receptor and estrogen receptor participate in growth and differentiation of the different rat decidual regions. Steroid hormone receptor antagonists were used to study steroid regulation of decidualization. Here we describe a suppressive interaction between progesterone receptor (onapristone) and estrogen receptor (ICI182780) antagonists and their relation to a rescue phenomenon with concomitant regulation of Hand2, Bmp2 and p-ERK1/2 during the early decidualization steps. Phenotypes of decidua development produced by antagonist treatments were characterized by morphology, proliferation, differentiation, angiogenesis and expression of signaling molecules. We found that suppression of progesterone receptor activity by onapristone treatment resulted in resorption of the implantation sites with concomitant decrease in progesterone and estrogen receptors, PCNA, KI67 antigen, DESMIN, CCND3, CX43, Prl8a2, and signaling players such as transcription factor Hand2, Bmp2 mRNAs and p-ERK1/2. Moreover, FGF-2 and Vegfa increased as a consequence of onapristone treatment. Implantation sites from antagonist of estrogen receptor treated rats developed all decidual regions, but showed an anomalous blood vessel formation at the mesometrial part of the decidua. The deleterious effect of onapristone was partially counteracted by the impairment of estrogen receptor activity with rescue of expression levels of hormone steroid receptors, proliferation and differentiation markers, and the induction of a probably compensatory increase in signaling molecules Hand2, Bmp2 and ERK1/2 activation compared to oil treated controls. This novel drug interaction during decidualization could be applied to pathological endometrial cell proliferation processes to improve therapies using steroid hormone receptor targets.
Collapse
Affiliation(s)
| | - Veronika Kleff
- Institut für Anatomie, Universaetsklinikum Duisburg-Essen, Essen, Germany
| | - Griselda Vallejo
- Instituto de Biología y Medicina Experimental, IByME-Conicet, Buenos Aires, Argentina
| | - Elke Winterhager
- Institut für Molekulare Biologie, Universaetsklinikum Duisburg-Essen, Essen, Germany
| | - Patricia Saragüeta
- Instituto de Biología y Medicina Experimental, IByME-Conicet, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
28
|
Prolactin (PRL) in Adipose Tissue: Regulation and Functions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 846:1-35. [DOI: 10.1007/978-3-319-12114-7_1] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
29
|
Henkes LE, Pru JK, Ashley RL, Anthony RV, Veeramachaneni DNR, Gates KC, Hansen TR. Embryo mortality in Isg15-/- mice is exacerbated by environmental stress. Biol Reprod 2014; 92:36. [PMID: 25505199 DOI: 10.1095/biolreprod.114.122002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The interferon-stimulated gene 15 (Isg15) encodes a ubiquitin-like protein that is induced in the endometrium by pregnancy in mice, humans, and ruminants. Because ISG15 is a component of the innate immune system, we hypothesized that development of the embryo, fetus, and postnatal pup may be impaired in mice lacking Isg15 (Isg15(-/-)) and that this development would be further impaired in response to environmental insults such as hypoxia. The number of implantation sites, resorption sites, dead embryos, and the changes in overall gross morphology of the uterus were evaluated in Isg15(-/-) mice on Days 7.5 and 12.5 postcoitum (dpc). Postnatal development also was monitored from birth to 12 wk of age. On 7.5 dpc, the number of implantation sites and serum progesterone concentrations were similar. However, embryo mortality increased (P < 0.05) in Isg15(-/-) dams by 12.5 dpc, resulting in smaller litter sizes (4.26 ± 0.21 embryos; n = 83 litters) compared to Isg15(+/+) females (7.78 ± 0.29 pups; n = 47 litters). Embryo mortality in Isg15(-/-) mice was further exacerbated to 70% when dams were stressed through housing under hypoxic conditions (PB = 445 mmHg; 6.5-12.5 dpc). Transmission electron microscopy revealed lesions in antimesometrial decidua as well as trophoblast cells adjacent to decidual cells on 7.5 dpc. ISG15 was localized to mesometrial decidua on 7.5 dpc. By 12.5 dpc, ISG15 was intensely localized to the labyrinth of the placenta. By 7.5 dpc, uterine natural killer cell migration into the mesometrial pole was diminished by 65% and was less prevalent in Isg15(-/-) compared to Isg15(+/+) deciduum. Postnatal growth rate of offspring that survived to birth from Isg15(-/-) and Isg15(+/+) dams was not different. Embryo mortality occurs in pregnant Isg15(-/-) mice, is exacerbated by environmental insults like maternal hypoxia, and might result from impaired early decidualization, vascular development, and formation of the labyrinth.
Collapse
Affiliation(s)
- Luiz E Henkes
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - James K Pru
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington
| | - Ryan L Ashley
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Russell V Anthony
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - D N Rao Veeramachaneni
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Katherine C Gates
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Thomas R Hansen
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
30
|
Perimenis P, Bouckenooghe T, Delplanque J, Moitrot E, Eury E, Lobbens S, Gosset P, Devisme L, Duvillie B, Abderrahmani A, Storme L, Fontaine P, Froguel P, Vambergue A. Placental antiangiogenic prolactin fragments are increased in human and rat maternal diabetes. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1783-93. [DOI: 10.1016/j.bbadis.2014.06.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 06/20/2014] [Accepted: 06/23/2014] [Indexed: 12/17/2022]
|
31
|
Hypoxic stress induces, but cannot sustain trophoblast stem cell differentiation to labyrinthine placenta due to mitochondrial insufficiency. Stem Cell Res 2014; 13:478-91. [PMID: 25239494 DOI: 10.1016/j.scr.2014.07.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/24/2014] [Accepted: 07/22/2014] [Indexed: 12/15/2022] Open
Abstract
Dysfunctional stem cell differentiation into placental lineages is associated with gestational diseases. Of the differentiated lineages available to trophoblast stem cells (TSC), elevated O2 and mitochondrial function are necessary to placental lineages at the maternal-placental surface and important in the etiology of preeclampsia. TSC lineage imbalance leads to embryonic failure during uterine implantation. Stress at implantation exacerbates stem cell depletion by decreasing proliferation and increasing differentiation. In an implantation site O2 is normally ~2%. In culture, exposure to 2% O2 and fibroblast growth factor 4 (FGF4) enabled the highest mouse TSC multipotency and proliferation. In contrast, hypoxic stress (0.5% O2) initiated the most TSC differentiation after 24h despite exposure to FGF4. However, hypoxic stress supported differentiation poorly after 4-7 days, despite FGF4 removal. At all tested O2 levels, FGF4 maintained Warburg metabolism; mitochondrial inactivity and aerobic glycolysis. However, hypoxic stress suppressed mitochondrial membrane potential and maintained low mitochondrial cytochrome c oxidase (oxidative phosphorylation/OxPhos), and high pyruvate kinase M2 (glycolysis) despite FGF4 removal. Inhibiting OxPhos inhibited optimum differentiation at 20% O2. Moreover, adding differentiation-inducing hyperosmolar stress failed to induce differentiation during hypoxia. Thus, differentiation depended on OxPhos at 20% O2; hypoxic and hyperosmolar stresses did not induce differentiation at 0.5% O2. Hypoxia-limited differentiation and mitochondrial inhibition and activation suggest that differentiation into two lineages of the labyrinthine placenta requires O2>0.5-2% and mitochondrial function. Stress-activated protein kinase increases an early lineage and suppresses later lineages in proportion to the deviation from optimal O2 for multipotency, thus it is the first enzyme reported to prioritize differentiation.
Collapse
|
32
|
Zuo RJ, Zhao YC, Lei W, Wang TS, Wang BC, Yang ZM. Crystallin αB acts as a molecular guard in mouse decidualization: regulation and function during early pregnancy. FEBS Lett 2014; 588:2944-51. [PMID: 24951838 DOI: 10.1016/j.febslet.2014.05.045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Revised: 05/08/2014] [Accepted: 05/28/2014] [Indexed: 02/06/2023]
Abstract
Although decidualization is crucial for the establishment of successful pregnancy, the molecular mechanism underlying decidualization remains poorly understood. Crystallin αB (CryAB), a small heat shock protein (sHSP), is up-regulated and phosphorylated in mouse decidua. In mouse primary endometrial stromal cells, CryAB is induced upon progesterone treatment via HIF1α. In addition, CryAB is strongly phosphorylated through the p38-MAPK pathway under stress or during in vitro decidualization. Knockdown of CryAB results in the increase of apoptosis of stromal cells and inhibits decidualization under oxidative or inflammatory stress. Our data indicate that CryAB protects decidualization against stress conditions.
Collapse
Affiliation(s)
- Ru-Juan Zuo
- School of Life Science, Xiamen University, Xiamen 361005, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yue-Chao Zhao
- School of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Wei Lei
- School of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Tong-Song Wang
- School of Science, Shantou University, Shantou 515063, China
| | - Bao-Cheng Wang
- School of Science, Shantou University, Shantou 515063, China
| | - Zeng-Ming Yang
- School of Life Science, Xiamen University, Xiamen 361005, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
33
|
Xu X, Leng JY, Gao F, Zhao ZA, Deng WB, Liang XH, Zhang YJ, Zhang ZR, Li M, Sha AG, Yang ZM. Differential expression and anti-oxidant function of glutathione peroxidase 3 in mouse uterus during decidualization. FEBS Lett 2014; 588:1580-9. [PMID: 24631040 DOI: 10.1016/j.febslet.2014.02.043] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 02/17/2014] [Accepted: 02/20/2014] [Indexed: 01/13/2023]
Abstract
Glutathione peroxidase 3 (GPX3) is an important member of antioxidant enzymes for reducing reactive oxygen species and maintaining the oxygen balance. Gpx3 mRNA is strongly expressed in decidual cells from days 5 to 8 of pregnancy. After pregnant mice are treated with GPX inhibitor for 3 days, pregnancy rate is significantly reduced. Progesterone stimulates Gpx3 expression through PR/HIF1α in mouse endometrial stromal cells. In the decidua, the high level of GPX3 expression is closely associated with the reduction of hydrogen peroxide (H2O2). Based on our data, GPX3 may play a major role in reducing H2O2 during decidualization.
Collapse
Affiliation(s)
- Xiu Xu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; School of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Jing-Yu Leng
- School of Life Science, Xiamen University, Xiamen 361005, China
| | - Fei Gao
- School of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Zhen-Ao Zhao
- School of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Wen-Bo Deng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Xiao-Huan Liang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yi-Juan Zhang
- School of Life Science, Xiamen University, Xiamen 361005, China
| | - Zhi-Rong Zhang
- School of Life Science, Xiamen University, Xiamen 361005, China
| | - Ming Li
- School of Life Science, Xiamen University, Xiamen 361005, China
| | - Ai-Guo Sha
- Reproductive Medicine Center, Bailu Hospital, Xiamen 361000, China
| | - Zeng-Ming Yang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
34
|
Rappolee DA, Zhou S, Puscheck EE, Xie Y. Stress responses at the endometrial-placental interface regulate labyrinthine placental differentiation from trophoblast stem cells. Reproduction 2013; 145:R139-55. [PMID: 23463790 DOI: 10.1530/rep-12-0240] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Development can happen in one of two ways. Cells performing a necessary function can differentiate from stem cells before the need for it arises and stress does not develop. Or need arises before function, stress develops and stress signals are part of the normal stimuli that regulate developmental mechanisms. These mechanisms adjust stem cell differentiation to produce function in a timely and proportional manner. In this review, we will interpret data from studies of null lethal mutants for placental stress genes that suggest the latter possibility. Acknowledged stress pathways participate in stress-induced and -regulated differentiation in two ways. These pathways manage the homeostatic response to maintain stem cells during the stress. Stress pathways also direct stem cell differentiation to increase the first essential lineage and suppress later lineages when stem cell accumulation is diminished. This stress-induced differentiation maintains the conceptus during stress. Pathogenic outcomes arise because population sizes of normal stem cells are first depleted by decreased accumulation. The fraction of stem cells is further decreased by differentiation that is induced to compensate for smaller stem cell populations. Analysis of placental lethal null mutant genes known to mediate stress responses suggests that the labyrinthine placenta develops during, and is regulated by, hypoxic stress.
Collapse
Affiliation(s)
- D A Rappolee
- CS Mott Center for Human Growth and Development, Wayne State University School of Medicine.
| | | | | | | |
Collapse
|
35
|
Zhang XH, Liang X, Liang XH, Wang TS, Qi QR, Deng WB, Sha AG, Yang ZM. The mesenchymal-epithelial transition during in vitro decidualization. Reprod Sci 2013; 20:354-60. [PMID: 23302397 DOI: 10.1177/1933719112472738] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The epithelial-mesenchymal transition plays a critical role in embryonic development, cancer progression, and metastasis. Decidualization is the process by which the fibroblast-like endometrial stromal cells differentiate into polygonal epithelial-like cells. However, it is still unclear whether mesenchymal-epithelial transition (MET) occurs during decidualization. The aim of this study was to examine whether decidualization causes the downregulation of some mesenchymal markers and upregulation of some epithelial markers in cultured uterine stromal cells. We showed that decidualization causes the downregulation of snail and vimentin expression, and upregulation of E-cadherin and cytokeratin expression. During in vitro decidualization, cultured stromal cells lose elongated shape and show epithelium-like characteristics. Our data suggest that the process of MET may exist during decidualization.
Collapse
Affiliation(s)
- Xiu-Hong Zhang
- School of Life Science, Xiamen University, Xiamen, PR China
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Kent LN, Ohboshi S, Soares MJ. Akt1 and insulin-like growth factor 2 (Igf2) regulate placentation and fetal/postnatal development. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2012; 56:255-61. [PMID: 22562201 DOI: 10.1387/ijdb.113407lk] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Phenotypic characterization of Akt1 and Igf2 null mice has revealed roles for each in the regulation of placentation, and fetal and postnatal growth. Insulin-like growth factor 2 (IGF2) is encoded by the Igf2 gene and influences cellular function, at least in part, through activation of an intracellular serine/threonine kinase called AKT1. Akt1 and Igf2 null mice were originally characterized on inbred and mixed genetic backgrounds, prohibiting direct comparisons of their phenotypes. The impact of loss of AKT1 or IGF2 on placental, fetal, and postnatal function were examined following transfer of Akt1 and Igf2 null mutations to an outbred CD1 genetic background. Disruption of IGF2 did not affect AKT expression or activation. Both Akt1-/- and Igf2-/- mice exhibited decreased placental weight, fetal weight and viability. Deregulation of placental growth was similar in Akt1 and Igf2 nulls; however, disruption of Igf2 had a more severe impact on prenatal survival and postnatal growth. Placental structure, including organization of junctional and labyrinth zones and development of the interstitial, invasive, trophoblast lineage, were similar in mutant and wild-type mice. Akt1 and Igf2 null mutations affected postnatal growth. The relative impact of each gene differed during pre-weaning versus post-weaning growth phases. AKT1 had a more significant role during pre-weaning growth, whereas IGF2 was a bigger contributor to post-weaning growth. Akt1 and Igf2 null mutations impact placental, fetal and postnatal growth. Placental phenotypes are similar; however, fetal and postnatal growth patterns are unique to each mutation.
Collapse
Affiliation(s)
- Lindsey N Kent
- Institute for Reproductive Health and Regenerative Medicine, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | |
Collapse
|
37
|
Nuño-Ayala M, Guillén N, Arnal C, Lou-Bonafonte JM, de Martino A, García-de-Jalón JA, Gascón S, Osaba L, Osada J, Navarro MA. Cystathionine β-synthase deficiency causes infertility by impairing decidualization and gene expression networks in uterus implantation sites. Physiol Genomics 2012; 44:702-16. [DOI: 10.1152/physiolgenomics.00189.2010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hyperhomocysteinemia has been reported in human reproduction as a risk factor for early pregnancy loss, preeclampsia, and congenital birth defects like spina bifida. Female infertility was also observed in cystathionine beta synthase-deficient mice ( Cbs-KO) as an animal model for severe hyperhomocysteinemia. The aim for the present research was to elucidate the time-point of pregnancy loss and to pinpoint gene and cellular changes involved in the underlying pathological mechanism. By mating 90-day-old wild-type and Cbs-KO female mice with their homologous male partners, we found that pregnancy loss in Cbs-KO occurred between the 8th and 12th gestation day during placenta formation. DNA microarrays were carried out on uterus from implantation and interimplantation samples obtained on day 8. The results allowed us to select genes potentially involved in embryo death; these were individually confirmed by RT-qPCR, and their expressions were also followed throughout pregnancy. We found that changes in expression of Calb1, Ttr, Expi, Inmt, Spink3, Rpgrip1, Krt15, Mt-4, Gzmc, Gzmb, Tdo2, and Afp were important for pregnancy success, since a different regulation in Cbs-KO mice was found. Also, differences in relationships among selected genes were observed, indicating a dysregulation of these genes in Cbs-KO females. In conclusion, our data provide more information on the gene expression cascade and its timely regulated process required for a successful pregnancy. In addition, we unveil new potential avenues to explore further investigations in pregnancy loss.
Collapse
Affiliation(s)
- Mario Nuño-Ayala
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Natalia Guillén
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Arnal
- Departamento de Patología Animal, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - José M. Lou-Bonafonte
- Departamento de Fisiología y Farmacología, Facultad de Ciencias de la Salud y del Deporte, Universidad de Zaragoza, Huesca, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Alba de Martino
- Departamento de Patología Animal, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
- Unidad de Anatomía Patológica, Instituto Aragonés de Ciencias de la Salud, IIS Aragón, Zaragoza, Spain
| | | | - Sonia Gascón
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Jesús Osada
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - María-Angeles Navarro
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
38
|
Salker MS, Christian M, Steel JH, Nautiyal J, Lavery S, Trew G, Webster Z, Al-Sabbagh M, Puchchakayala G, Föller M, Landles C, Sharkey AM, Quenby S, Aplin JD, Regan L, Lang F, Brosens JJ. Deregulation of the serum- and glucocorticoid-inducible kinase SGK1 in the endometrium causes reproductive failure. Nat Med 2011; 17:1509-13. [PMID: 22001908 DOI: 10.1038/nm.2498] [Citation(s) in RCA: 146] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 08/30/2011] [Indexed: 11/09/2022]
Abstract
Infertility and recurrent pregnancy loss (RPL) are prevalent but distinct causes of reproductive failure that often remain unexplained despite extensive investigations. Analysis of midsecretory endometrial samples revealed that SGK1, a kinase involved in epithelial ion transport and cell survival, is upregulated in unexplained infertility, most prominently in the luminal epithelium, but downregulated in the endometrium of women suffering from RPL. To determine the functional importance of these observations, we first expressed a constitutively active SGK1 mutant in the luminal epithelium of the mouse uterus. This prevented expression of certain endometrial receptivity genes, perturbed uterine fluid handling and abolished embryo implantation. By contrast, implantation was unhindered in Sgk1-/- mice, but pregnancy was often complicated by bleeding at the decidual-placental interface and fetal growth retardation and subsequent demise. Compared to wild-type mice, Sgk1-/- mice had gross impairment of pregnancy-dependent induction of genes involved in oxidative stress defenses. Relative SGK1 deficiency was also a hallmark of decidualizing stromal cells from human subjects with RPL and sensitized these cells to oxidative cell death. Thus, depending on the cellular compartment, deregulated SGK1 activity in cycling endometrium interferes with embryo implantation, leading to infertility, or predisposes to pregnancy complications by rendering the feto-maternal interface vulnerable to oxidative damage.
Collapse
Affiliation(s)
- Madhuri S Salker
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Campus, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Yu J, Wu J, Bagchi IC, Bagchi MK, Sidell N, Taylor RN. Disruption of gap junctions reduces biomarkers of decidualization and angiogenesis and increases inflammatory mediators in human endometrial stromal cell cultures. Mol Cell Endocrinol 2011; 344:25-34. [PMID: 21767601 PMCID: PMC3162129 DOI: 10.1016/j.mce.2011.04.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Revised: 04/14/2011] [Accepted: 04/17/2011] [Indexed: 11/22/2022]
Abstract
CONTEXT Uterine decidualization is critical to embryonic implantation and sustained pregnancy. OBJECTIVE To evaluate the role of gap junction intercellular communications and connexin (Cx) proteins in the morphological and biochemical differentiation of decidualized human endometrial stromal cell (ESC) cultures. DESIGN Translational cell biological study. SETTING Academic medical center. PATIENTS Endometrial tissue was provided by five healthy reproductive age women on no hormonal medication, undergoing laparoscopy in the early proliferative phase of the menstrual cycle. INTERVENTIONS Endometrial biopsy under general anesthesia, establishment and decidualization of ESC with 10 nM 17β-estradiol, 100 nM progesterone and 0.5 mM dibutyryl-cAMP (E/P/c), and manipulation of gap junctions in vitro via a combination of pharmacological or transgenic approaches. MAIN OUTCOME MEASURES Decidualized ESC evaluated morphologically for epithelioid transformation, gap junctions by dye diffusion and Cx43, prolactin, VEGF and IL-6 expression by RT-PCR, Western and ELISA methods. RESULTS Cx43 accumulation and functional gap junctions between decidualized ESC increase concomitantly with morphological differentiation following E/P/c treatment. Disruption of gap junctions using pharmacological inhibitors or Cx43 shRNA prevents morphological differentiation and inhibits prolactin and VEGF secretion. By contrast, IL-6 secretion from decidualized ESC is augmented by both approaches. CONCLUSIONS The findings suggest that decidualized ESC function as a coordinated secretory organ to regulate embryonic implantation via intercellular cooperation mediated by gap junctions. When adjacent cells can communicate through these junctions, decidual prolactin and VEGF secretion appears to be optimized for vascular development of the placental bed. Conversely, when intercellular communications are disrupted, angiogenesis is impaired and an inflammatory state is induced.
Collapse
Affiliation(s)
- Jie Yu
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA 30322, United States
| | | | | | | | | | | |
Collapse
|
40
|
Tian Z, Zhao ZA, Liang XH, Zhang XH, Sha AG, Zhang ZR, Yu YS, Yang ZM. Expression and function of fatty acid-binding protein 4 during mouse decidualization. Fertil Steril 2011; 95:2749-52.e1-5. [PMID: 21704217 DOI: 10.1016/j.fertnstert.2011.05.052] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Revised: 05/10/2011] [Accepted: 05/15/2011] [Indexed: 11/27/2022]
Abstract
Fatty acid-binding protein 4 (Fabp4) is highly expressed in the secondary decidual zone of mouse decidua and deciduoma and stromal cells under in vitro decidualization. Dtprp, a well-known marker of in vitro decidualization, is diminished by small interfering RNA against Fabp4 and FABP4 inhibitor and stimulated through Fabp4 overexpression.
Collapse
Affiliation(s)
- Zhen Tian
- Department of Biology, Santou University, Shantou, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Zhou S, Xie Y, Puscheck EE, Rappolee DA. Oxygen levels that optimize TSC culture are identified by maximizing growth rates and minimizing stress. Placenta 2011; 32:475-81. [PMID: 21511332 DOI: 10.1016/j.placenta.2011.03.013] [Citation(s) in RCA: 32] [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: 09/18/2010] [Revised: 03/24/2011] [Accepted: 03/25/2011] [Indexed: 11/17/2022]
Abstract
Accumulating data suggest that 20% O(2) causes human and mouse placental trophoblast stem cell (TSC) differentiation and suppresses proliferation. We tested the hypotheses that phosphorylated stress-activated protein kinase (pSAPK) levels report the optimal O(2) level for TSC culture, and that pSAPK responds to contradictory signals. We tested the dose range of 0-20% O(2) (0, 0.5, 2, and 20%) on five effects in cultured TSC. The results showed 1) TSC accumulation rates were highest at 2% O(2), lower at 20% and lowest at 0-0.5%; 2) pSAPK protein levels were lowest at 2% O(2), higher at 20%, and highest at 0-0.5%; 3) Cleaved caspase 3, an apoptosis marker, increased at 0.5% O(2), and was highest at 0% O(2); 4) Three markers for multipotency were highest at 2 and 20% and significantly decreased at 0.5%-0%; 5) In contrast three differentiation markers were lowest at 2% and highest at 0.5%-0%. Thus, 2% O(2) is the optimum as defined by lowest pSAPK and differentiation markers and highest growth rate and multipotency markers, without appreciable apoptosis. In addition, two lines of evidence suggest that fibroblast growth factor (FGF)4 does not directly activate SAPK. SAPK activity increases transiently with FGF4 removal at 2% O(2), but SAPK activity decreases when O(2) is switched from 20% to 2% with FGF4 present. Thus, SAPK is activated by contradictory signals, but activity decreases when either signal is removed. Taken together, the findings suggest that pSAPK senses suboptimal signals during TSC culture and probably in vivo.
Collapse
Affiliation(s)
- S Zhou
- CS Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | |
Collapse
|
42
|
Expression and localization pattern of ABCA1 in diverse human placental primary cells and tissues. Placenta 2011; 32:420-30. [PMID: 21501868 DOI: 10.1016/j.placenta.2011.03.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 03/06/2011] [Accepted: 03/07/2011] [Indexed: 11/20/2022]
Abstract
The ATP-binding cassette transporter A1 (ABCA1) mediates the transport of cholesterol, phospholipids, and other lipophilic molecules across cellular membranes. Recent data provide evidence that ABCA1 plays an important role in placental function but the exact cellular sites of ABCA1 action in the placenta remain controversial. To clarify this issue, we analyzed the cellular and subcellular localization of ABCA1 with immunocytochemistry, immunofluorescence and subsequent confocal or immunofluorescence microscopy in different types of isolated primary placenta cells: cytotrophoblast cells, amnion epithelial cells, villous macrophages (Hofbauer cells), and mesenchymal cells isolated from chorionic membrane and placental villi. After 12 h of cultivation, primary cytotrophoblast cells showed intensive membrane and cytoplasmic staining for ABCA1. After 24 h, with progressive syncytium formation, ABCA1 staining intensity was markedly reduced and ABCA1 was dispersed in the cytoplasm of the forming syncytial layer. In amnion epithelial cells, placental macrophages and mesenchymal cells, ABCA1 was predominantly localized at the cell membrane and cytoplasmic compartments partially corresponding to the endoplasmic reticulum. In these cell types, the ABCA1 staining intensity was not dependent on the cultivation time. In conclusion, ABCA1 shows marked expression levels in diverse placental cell types. The multitopic localization of ABCA1 in diverse human placental cells not all directly involved in materno-fetal exchange suggests that this protein may not only participate in transplacental lipid transport but could have additional regulatory functions.
Collapse
|
43
|
Xie Y, Awonuga AO, Zhou S, Puscheck EE, Rappolee DA. Interpreting the stress response of early mammalian embryos and their stem cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2011; 287:43-95. [PMID: 21414586 DOI: 10.1016/b978-0-12-386043-9.00002-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
This review analyzes and interprets the normal, pathogenic, and pathophysiological roles of stress and stress enzymes in mammalian development. Emerging data suggest that stem cells from early embryos are induced by stress to perform stress-enzyme-mediated responses that use the strategies of compensatory, prioritized, and reversible differentiation. These strategies have been optimized during evolution and in turn have aspects of energy conservation during stress that optimize and maximize the efficacy of the stress response. It is likely that different types of stem cells have varying degrees of flexibility in mediating compensatory and prioritized differentiation. The significance of this analysis and interpretation is that it will serve as a foundation for yielding tools for diagnosing, understanding normal and pathophysiological mechanisms, and providing methods for managing stress enzymes to improve short- and long-term reproductive outcomes.
Collapse
Affiliation(s)
- Y Xie
- CS Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | | | | | | |
Collapse
|
44
|
Alam SMK, Konno T, Rumi MAK, Dong Y, Weiner CP, Soares MJ. Prolactin family of the guinea pig, Cavia porcellus. Endocrinology 2010; 151:3918-28. [PMID: 20534723 PMCID: PMC2940522 DOI: 10.1210/en.2010-0239] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Prolactin (PRL) is a multifunctional hormone with prominent roles in regulating growth and reproduction. The guinea pig (Cavia porcellus) has been extensively used in endocrine and reproduction research. Thus far, the PRL cDNA and protein have not been isolated from the guinea pig. In the present study, we used information derived from the public guinea pig genome database as a tool for identifying guinea pig PRL and PRL-related proteins. Guinea pig PRL exhibits prominent nucleotide and amino acid sequence differences when compared with PRLs of other eutherian mammals. In contrast, guinea pig GH is highly conserved. Expression of PRL and GH in the guinea pig is prominent in the anterior pituitary, similar to known expression patterns of PRL and GH for other species. Two additional guinea pig cDNAs were identified and termed PRL-related proteins (PRLRP1, PRLRP2). They exhibited a more distant relationship to PRL and their expression was restricted to the placenta. Recombinant guinea pig PRL protein was generated and shown to be biologically active in the PRL-responsive Nb2 lymphoma cell bioassay. In contrast, recombinant guinea pig PRLRP1 protein did not exhibit PRL-like bioactivity. In summary, we have developed a new set of research tools for investigating the biology of the PRL family in an important animal model, the guinea pig.
Collapse
Affiliation(s)
- S M Khorshed Alam
- Department of Pathology and Laboratory Medicine, Institute of Maternal-Fetal Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | | | | | |
Collapse
|
45
|
Simmons DG, Rawn S, Davies A, Hughes M, Cross JC. Spatial and temporal expression of the 23 murine Prolactin/Placental Lactogen-related genes is not associated with their position in the locus. BMC Genomics 2008; 9:352. [PMID: 18662396 PMCID: PMC2527339 DOI: 10.1186/1471-2164-9-352] [Citation(s) in RCA: 187] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Accepted: 07/28/2008] [Indexed: 02/06/2023] Open
Abstract
Background The Prolactin (PRL) hormone gene family shows considerable variation among placental mammals. Whereas there is a single PRL gene in humans that is expressed by the pituitary, there are an additional 22 genes in mice including the placental lactogens (PL) and Prolactin-related proteins (PLPs) whose expression is limited to the placenta. To understand the regulation and potential functions of these genes, we conducted a detailed temporal and spatial expression study in the placenta between embryonic days 7.5 and E18.5 in three genetic strains. Results Of the 22 PRL/PL genes examined, only minor differences were observed among strains of mice. We found that not one family member has the same expression pattern as another when both temporal and spatial data were examined. There was also no correlation in expression between genes that were most closely related or between adjacent genes in the PRL/PL locus. Bioinformatic analysis of upstream regulatory regions identified conserved combinations (modules) of putative transcription factor binding sites shared by genes expressed in the same trophoblast subtype, supporting the notion that local regulatory elements, rather than locus control regions, specify subtype-specific expression. Further diversification in expression was also detected as splice variants for several genes. Conclusion In the present study, a detailed temporal and spatial placental expression map was generated for all murine PRL/PL family members from E7.5 to E18.5 of gestation in three genetic strains. This detailed analysis uncovered several new markers for some trophoblast cell types that will be useful for future analysis of placental structure in mutant mice with placental phenotypes. More importantly, several main conclusions about regulation of the locus are apparent. First, no two family members have the same expression pattern when both temporal and spatial data are examined. Second, most genes are expressed in multiple trophoblast cell subtypes though none were detected in the chorion, where trophoblast stem cells reside, or in syncytiotrophoblast of the labyrinth layer. Third, bioinformatic comparisons of upstream regulatory regions identified predicted transcription factor binding site modules that are shared by genes expressed in the same trophoblast subtype. Fourth, further diversification of gene products from the PRL/PL locus occurs through alternative splice isoforms for several genes.
Collapse
Affiliation(s)
- David G Simmons
- Department of Comparative Biology & Experimental Medicine, The University of Calgary, Calgary, Canada.
| | | | | | | | | |
Collapse
|
46
|
Alam SMK, Konno T, Sahgal N, Lu L, Soares MJ. Decidual cells produce a heparin-binding prolactin family cytokine with putative intrauterine regulatory actions. J Biol Chem 2008; 283:18957-68. [PMID: 18467328 DOI: 10.1074/jbc.m801826200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Pregnancy in mice and rats is associated with the production of a large family of hormones/cytokines related to prolactin (PRL). The hormones/cytokines are hypothesized to coordinate maternal and fetal adaptations to pregnancy. In this study, PRL-like protein-J (PLP-J, also known as PRL family 3, subfamily c, member 1 (Prl3c1)) is shown to be a product of the uterine decidua and a regulator of postimplantation intrauterine events. PLP-J-specific antibodies and a series of recombinant PLP-J proteins were generated and used to investigate PLP-J expression and as ligands for investigating biological targets. Decidual PLP-J migrates as a 29-kDa protein and localizes to a band of decidual cells surrounding the trophoblast cell layer on gestation day 8.5. PLP-J ligands specifically bound in situ to the surrounding uterine stromal cells and vasculature within the decidua of gestation day 8.5 implantation sites. We then investigated the in vitro actions of PLP-J on uterine stromal cells and endothelial cells. PLP-J specifically interacted with both cell populations. PLP-J promoted uterine stromal cell proliferation and inhibited endothelial cell proliferation. We determined that PLP-J does not interact with PRL receptors. Instead, PLP-J interacts with heparin-containing molecules, including syndecan-1, which is expressed in gestation day 8.5 pregnant uteri, as well as in uterine stromal cells and endothelial cells. The restricted expression of PLP-J and its specific interactions with uterine stromal cells and endothelial cells suggests that it acts locally and regulates decidual cell development and the endometrial vasculature.
Collapse
Affiliation(s)
- S M Khorshed Alam
- Institute of Maternal-Fetal Biology, Department of Pathology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | | | |
Collapse
|
47
|
Soares MJ, Konno T, Alam SMK. The prolactin family: effectors of pregnancy-dependent adaptations. Trends Endocrinol Metab 2007; 18:114-21. [PMID: 17324580 DOI: 10.1016/j.tem.2007.02.005] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2006] [Revised: 01/17/2007] [Accepted: 02/14/2007] [Indexed: 12/01/2022]
Abstract
Prolactin (PRL) is a hormone involved in many biological functions. In some species, there is a family of PRL-related genes; such is the case in the mouse and rat. The actions of members of the PRL family can be distinguished based on the involvement of the PRL receptor signaling pathway (classical versus nonclassical). Recent insights into the biology of the PRL family have been derived from mouse mutagenesis studies. There is compelling evidence suggesting that the PRL family contributes to the regulation of pregnancy-dependent adaptations to physiological stressors.
Collapse
Affiliation(s)
- Michael J Soares
- Institute of Maternal-Fetal Biology, Division of Cancer and Developmental Biology, Department of Pathology & Laboratory of Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | | | | |
Collapse
|