1
|
Kobayashi R, Tajika Y, Kohmaru J, Morita S, Horii T, Mizukami Y, Aikawa S, Hirota Y, Hatada I. The histone methyltransferase KMT2D is essential for embryo implantation via regulating precise differentiation of endometrial cells. Cell Death Discov 2024; 10:357. [PMID: 39117610 PMCID: PMC11310208 DOI: 10.1038/s41420-024-02134-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/01/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024] Open
Abstract
Embryo implantation failures are a major challenge in reproductive medicine, but the underlying mechanism remains poorly understood. Successful implantation requires dynamic remodeling of the endometrium through integrated proliferation and differentiation of endometrial cells including luminal epithelial, glandular epithelial, and stromal cells. Conversely, their disruption causes infertility. Spatiotemporal control of transcription is required for these processes; however, the underlying epigenetic regulation is largely unknown. In this study, we examined expression data from the human endometrium during implantation and discovered that expression of the histone lysine methyltransferase KMT2D was significantly suppressed in patients with recurrent implantation failure. Further study revealed that uterine deletion of Kmt2d in mice caused infertility due to implantation failure. Morphological analysis discovered a reduction in the number of uterine glands and aberrant differentiation of the luminal and glandular epithelium into stratified phenotypes in Kmt2d knockout uteri. Administration of leukemia inhibitory factor protein, which is expressed in uterine glands and is essential for implantation, did not rescue implantation failure in Kmt2d knockout mice, suggesting that infertility was not solely due to uterine gland dysfunction. RNA sequencing analysis revealed that Kmt2d knockout uteri displayed suppressed expression of genes involved in ion homeostasis, which may affect the uterine luminal morphology. Our study suggests that KMT2D plays an essential role in facilitating successful embryo implantation by regulating the coordinated differentiation of endometrial cells, providing valuable insights into unexplained implantation failures in women.
Collapse
Affiliation(s)
- Ryosuke Kobayashi
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Yuki Tajika
- Department of Anatomy, Gunma University Graduate School of Medicine, Maebashi, Japan
- Gunma Prefectural College of Health Sciences, Maebashi, Japan
| | - Junki Kohmaru
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Sumiyo Morita
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Takuro Horii
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Yoichi Mizukami
- Institute of Gene Research, Science Research Center, Yamaguchi University, Yamaguchi, Japan
| | - Shizu Aikawa
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Izuho Hatada
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan.
- Viral Vector Core, Gunma University Initiative for Advanced Research (GIAR), Gunma, Japan.
| |
Collapse
|
2
|
Schalich KM, Koganti PP, Castillo JM, Reiff OM, Cheong SH, Selvaraj V. The uterine secretory cycle: recurring physiology of endometrial outputs that setup the uterine luminal microenvironment. Physiol Genomics 2024; 56:74-97. [PMID: 37694291 DOI: 10.1152/physiolgenomics.00035.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 09/12/2023] Open
Abstract
Conserved in female reproduction across all mammalian species is the estrous cycle and its regulation by the hypothalamic-pituitary-gonadal (HPG) axis, a collective of intersected hormonal events that are crucial for ensuring uterine fertility. Nonetheless, knowledge of the direct mediators that synchronously shape the uterine microenvironment for successive yet distinct events, such as the transit of sperm and support for progressive stages of preimplantation embryo development, remain principally deficient. Toward understanding the timed endometrial outputs that permit luminal events as directed by the estrous cycle, we used Bovidae as a model system to uniquely surface sample and study temporal shifts to in vivo endometrial transcripts that encode for proteins destined to be secreted. The results revealed the full quantitative profile of endometrial components that shape the uterine luminal microenvironment at distinct phases of the estrous cycle (estrus, metestrus, diestrus, and proestrus). In interpreting this comprehensive log of stage-specific endometrial secretions, we define the "uterine secretory cycle" and extract a predictive understanding of recurring physiological actions regulated within the uterine lumen in anticipation of sperm and preimplantation embryonic stages. This repetitive microenvironmental preparedness to sequentially provide operative support was a stable intrinsic framework, with only limited responses to sperm or embryos if encountered in the lumen within the cyclic time period. In uncovering the secretory cycle and unraveling realistic biological processes, we present novel foundational knowledge of terminal effectors controlled by the HPG axis to direct a recurring sequence of vital functions within the uterine lumen.NEW & NOTEWORTHY This study unravels the recurring sequence of changes within the uterus that supports vital functions (sperm transit and development of preimplantation embryonic stages) during the reproductive cycle in female Ruminantia. These data present new systems knowledge in uterine reproductive physiology crucial for setting up in vitro biomimicry and artificial environments for assisted reproduction technologies for a range of mammalian species.
Collapse
Affiliation(s)
- Kasey M Schalich
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, United States
| | - Prasanthi P Koganti
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, United States
| | - Juan M Castillo
- Department of Clinical Sciences, Veterinary College, Cornell University, Ithaca, New York, United States
| | - Olivia M Reiff
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, United States
| | - Soon Hon Cheong
- Department of Clinical Sciences, Veterinary College, Cornell University, Ithaca, New York, United States
| | - Vimal Selvaraj
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, United States
| |
Collapse
|
3
|
Gao X, Yao X, Li X, Liang Y, Liu Z, Wang Z, Li K, Li Y, Zhang G, Wang F. Roles of WNT6 in Sheep Endometrial Epithelial Cell Cycle Progression and Uterine Glands Organogenesis. Vet Sci 2021; 8:vetsci8120316. [PMID: 34941843 PMCID: PMC8708052 DOI: 10.3390/vetsci8120316] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/22/2021] [Accepted: 12/04/2021] [Indexed: 11/22/2022] Open
Abstract
The uterus, as part of the female reproductive tract, is essential for embryo survival and in the maintenance of multiple pregnancies in domestic animals. This study was conducted to investigate the effects of WNT6 on Hu sheep endometrial epithelial cells (EECs) and uterine glands (UGs) in Hu sheep, with high prolificacy rates. In the present study, Hu sheep with different fecundity, over three consecutive pregnancies, were divided into two groups: high prolificacy rate group (HP, litter size = 3) and low prolificacy rate group (LP, litter size = 1). A comparative analysis of the endometrial morphology was performed by immunofluorescence. RNA-seq was used to analyze the gene’s expression in endometrium of HP and LP Hu sheep, providing a candidate gene, which was investigated in EECs and organoid culture. Firstly, higher density of UGs was found in the HP Hu sheep groups (p < 0.05). The RNA-seq data revealed the importance of the WNT signaling pathway and WNT6 gene in Hu sheep endometrium. Functionally, WNT6 could promote the cell cycle progression of EECs via WNT/β-catenin signal and enhance UGs organogenesis. Taken together, WNT6 is a crucial regulator for sheep endometrial development; this finding may offer a new insight into understanding the regulatory mechanism of sheep prolificacy.
Collapse
Affiliation(s)
- Xiaoxiao Gao
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (X.G.); (X.Y.); (X.L.); (Y.L.); (Z.L.); (Z.W.); (K.L.); (Y.L.); (G.Z.)
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaolei Yao
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (X.G.); (X.Y.); (X.L.); (Y.L.); (Z.L.); (Z.W.); (K.L.); (Y.L.); (G.Z.)
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaodan Li
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (X.G.); (X.Y.); (X.L.); (Y.L.); (Z.L.); (Z.W.); (K.L.); (Y.L.); (G.Z.)
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing 210095, China
| | - Yaxu Liang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (X.G.); (X.Y.); (X.L.); (Y.L.); (Z.L.); (Z.W.); (K.L.); (Y.L.); (G.Z.)
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing 210095, China
| | - Zifei Liu
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (X.G.); (X.Y.); (X.L.); (Y.L.); (Z.L.); (Z.W.); (K.L.); (Y.L.); (G.Z.)
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhibo Wang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (X.G.); (X.Y.); (X.L.); (Y.L.); (Z.L.); (Z.W.); (K.L.); (Y.L.); (G.Z.)
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing 210095, China
| | - Kang Li
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (X.G.); (X.Y.); (X.L.); (Y.L.); (Z.L.); (Z.W.); (K.L.); (Y.L.); (G.Z.)
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing 210095, China
| | - Yingqi Li
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (X.G.); (X.Y.); (X.L.); (Y.L.); (Z.L.); (Z.W.); (K.L.); (Y.L.); (G.Z.)
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing 210095, China
| | - Guomin Zhang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (X.G.); (X.Y.); (X.L.); (Y.L.); (Z.L.); (Z.W.); (K.L.); (Y.L.); (G.Z.)
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing 210095, China
| | - Feng Wang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (X.G.); (X.Y.); (X.L.); (Y.L.); (Z.L.); (Z.W.); (K.L.); (Y.L.); (G.Z.)
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing 210095, China
- Correspondence: ; Tel.: +86-025-84395381
| |
Collapse
|
4
|
Owens CE, Daniels KM, Ealy AD, Knowlton KF, Cockrum RR. Graduate Student Literature Review: Potential mechanisms of interaction between bacteria and the reproductive tract of dairy cattle. J Dairy Sci 2020; 103:10951-10960. [PMID: 32896395 DOI: 10.3168/jds.2019-18050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 06/29/2020] [Indexed: 11/19/2022]
Abstract
Although the presence of bacteria has been characterized throughout the reproductive tracts of multiple species, how these bacteria may interact with the host has yet to be described. Previous reviews have described how pathogenic bacteria interact with the reproductive tract to cause infections such as metritis. This review aimed to summarize the knowledge related to pathogenic and nonpathogenic bacteria in various locations of the bovine reproductive tract and the possible mechanisms underlying host-microbe interactions during gametogenesis and early pregnancy. Lactic acid bacteria such as Lactobacillus seem to be beneficial in multiple areas of the reproductive tract: they have been associated with increased oocyte quality when in follicular fluid and secrete reactive oxygen species that are beneficial during placental angiogenesis. However, other bacteria, including Enterococcus, Staphylococcus, and Streptococcus, may modulate T helper cells that inhibit maternal recognition of pregnancy. Available data on the reproductive microbiome focus on variations in microbial communities and their associations with reproductive performance. However, research on these host-microbiome interactions may provide more insight on how bacteria affect fertility.
Collapse
Affiliation(s)
- C E Owens
- Department of Dairy Science, Virginia Polytechnic Institute and State University, Blacksburg 24061.
| | - K M Daniels
- Department of Dairy Science, Virginia Polytechnic Institute and State University, Blacksburg 24061
| | - A D Ealy
- Department of Animal and Poultry Science, Virginia Polytechnic Institute and State University, Blacksburg 24061
| | - K F Knowlton
- Department of Dairy Science, Virginia Polytechnic Institute and State University, Blacksburg 24061
| | - R R Cockrum
- Department of Dairy Science, Virginia Polytechnic Institute and State University, Blacksburg 24061
| |
Collapse
|
5
|
Ban Z, Knöspel F, Schneider MR. Shedding light into the black box: Advances in in vitro systems for studying implantation. Dev Biol 2020; 463:1-10. [DOI: 10.1016/j.ydbio.2020.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/01/2020] [Accepted: 04/13/2020] [Indexed: 12/17/2022]
|
6
|
Kelleher AM, DeMayo FJ, Spencer TE. Uterine Glands: Developmental Biology and Functional Roles in Pregnancy. Endocr Rev 2019; 40:1424-1445. [PMID: 31074826 PMCID: PMC6749889 DOI: 10.1210/er.2018-00281] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/15/2019] [Indexed: 12/18/2022]
Abstract
All mammalian uteri contain glands in the endometrium that develop only or primarily after birth. Gland development or adenogenesis in the postnatal uterus is intrinsically regulated by proliferation, cell-cell interactions, growth factors and their inhibitors, as well as transcription factors, including forkhead box A2 (FOXA2) and estrogen receptor α (ESR1). Extrinsic factors regulating adenogenesis originate from other organs, including the ovary, pituitary, and mammary gland. The infertility and recurrent pregnancy loss observed in uterine gland knockout sheep and mouse models support a primary role for secretions and products of the glands in pregnancy success. Recent studies in mice revealed that uterine glandular epithelia govern postimplantation pregnancy establishment through effects on stromal cell decidualization and placental development. In humans, uterine glands and, by inference, their secretions and products are hypothesized to be critical for blastocyst survival and implantation as well as embryo and placental development during the first trimester before the onset of fetal-maternal circulation. A variety of hormones and other factors from the ovary, placenta, and stromal cells impact secretory function of the uterine glands during pregnancy. This review summarizes new information related to the developmental biology of uterine glands and discusses novel perspectives on their functional roles in pregnancy establishment and success.
Collapse
Affiliation(s)
- Andrew M Kelleher
- Division of Animal Sciences, University of Missouri, Columbia, Missouri
| | - Francesco J DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute on Environmental Health Sciences, Research Triangle Park, Durham, North Carolina
| | - Thomas E Spencer
- Division of Animal Sciences, University of Missouri, Columbia, Missouri.,Department of Obstetrics, Gynecology, and Women's Health, University of Missouri, Columbia, Missouri
| |
Collapse
|
7
|
Harlow K, Ferreira CR, Sobreira TJP, Casey T, Stewart K. Lipidome profiles of postnatal day 2 vaginal swabs reflect fat composition of gilt's postnatal diet. PLoS One 2019; 14:e0215186. [PMID: 31557164 PMCID: PMC6762109 DOI: 10.1371/journal.pone.0215186] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 09/11/2019] [Indexed: 11/18/2022] Open
Abstract
We hypothesized that postnatal development of the vagina is impacted by early nutritional environment. Our objective was to determine if lipid profiles of vaginal swabs were different between postnatal gilts suckled by sow or fed milk replacer the first 48 h after birth, with or without a lard-based fat supplement. Gilts (>1.3 kg) were selected at birth across 8 litters and assigned to one of four treatments: 1) suckled by sow (S, n = 8); 2) suckled by sow plus administration of a fat supplement (SF, n = 5); 3) bottle-fed solely milk replacer (B, n = 8); or 4) bottle-fed solely milk replacer plus administration of a fat supplement (BF, n = 7). At 48 h postnatal, vaginal swabs of gilts were taken with a cytology brush, and lipids were extracted for analysis using multiple reaction monitoring (MRM)-profiling. Lipids extracted from serum collected at 48 h from gilts, milk collected at 24 h from sows, and milk replacer were also analyzed with MRM-profiling. Receiver operating characteristic curve analysis found 18 lipids recovered from vaginal swabs that highly distinguished between S and B gilts [area-under-the-curve (AUC) > 0.9], including phosphatidylethanolamine with 34 carbons and four unsaturations in the fatty acyl residues [PE (34:4)]. Twelve lipids from vaginal swabs highly correlated (r > 0.6; p < 0.01) with nutrition source. Lipids with greater abundance in milk replacer drove association. For example, mean intensity of PE (34:4) was 149-fold higher in milk replacer than colostrum. Consequently, PE (34:4) was found to have 1.6- and 2.12-fold higher levels in serum and vaginal swab samples (p < 0.001), respectively, of B gilts as compared to S gilts. Findings support that vaginal swabs can be used to noninvasively study effects of perinatal nutrition on tissue composition.
Collapse
Affiliation(s)
- KaLynn Harlow
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Christina R. Ferreira
- Metabolomics Core, Bindley Science Center, Purdue University, West Lafayette, Indiana, United States of America
| | - Tiago J. P. Sobreira
- Metabolomics Core, Bindley Science Center, Purdue University, West Lafayette, Indiana, United States of America
| | - Theresa Casey
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, United States of America
- * E-mail:
| | - Kara Stewart
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, United States of America
| |
Collapse
|
8
|
Spencer TE, Kelleher AM, Bartol FF. Development and Function of Uterine Glands in Domestic Animals. Annu Rev Anim Biosci 2019; 7:125-147. [DOI: 10.1146/annurev-animal-020518-115321] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
All mammalian uteri contain glands that synthesize or transport and secrete substances into the uterine lumen. Uterine gland development, or adenogenesis, is uniquely a postnatal event in sheep and pigs and involves differentiation of glandular epithelium from luminal epithelium, followed by invagination and coiling morphogenesis throughout the stroma. Intrinsic transcription factors and extrinsic factors from the ovary and pituitary as well as the mammary gland (lactocrine) regulate uterine adenogenesis. Recurrent pregnancy loss is observed in the ovine uterine gland knockout sheep, providing unequivocal evidence that glands and their products are essential for fertility. Uterine gland hyperplasia and hypertrophy during pregnancy are controlled by sequential actions of hormones from the ovary and/or pituitary as well as the placenta. Gland-derived histotroph is transported by placental areolae for fetal growth. Increased knowledge of uterine gland biology is expected to improve pregnancy outcomes, as well as the health and productivity of mothers and their offspring.
Collapse
Affiliation(s)
- Thomas E. Spencer
- Division of Animal Sciences and Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, Missouri 65211, USA;,
| | - Andrew M. Kelleher
- Division of Animal Sciences and Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, Missouri 65211, USA;,
| | - Frank F. Bartol
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama 36849-5517, USA
| |
Collapse
|
9
|
Alarcón R, Ingaramo PI, Rivera OE, Dioguardi GH, Repetti MR, Demonte LD, Milesi MM, Varayoud J, Muñoz-de-Toro M, Luque EH. Neonatal exposure to a glyphosate-based herbicide alters the histofunctional differentiation of the ovaries and uterus in lambs. Mol Cell Endocrinol 2019; 482:45-56. [PMID: 30550814 DOI: 10.1016/j.mce.2018.12.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 12/10/2018] [Accepted: 12/10/2018] [Indexed: 12/12/2022]
Abstract
The aim of the present study was to compare the effect of oral and subcutaneous exposure to a glyphosate-based herbicide (GBH) on the female reproductive system, specifically in the ovaries and uterus of prepubertal lambs. To this end, ewe lambs were exposed to a s.c. (n: 5) or an oral (n: 5) environmentally relevant dose of GBH (2 mg/kg/day) or to vehicle (controls, n: 12), from postnatal day (PND) 1 to PND14. Serum glyphosate and aminomethylphosphonic acid (AMPA) concentrations were measured on PND15 and PND45. The ovaries and uterus were obtained and weighed on PND45. Ovarian follicular dynamics and uterine morphological features were determined by picrosirius-hematoxylin staining. The proliferation marker Ki67 was evaluated by immunohistochemistry in ovarian and uterine samples. Glyphosate but not AMPA was detected in serum of exposed lambs on PND15, whereas neither glyphosate nor AMPA were detected on PND45. Controls were negative for glyphosate and AMPA on PND15 and PND45. GBH exposure did not affect ovarian or uterine weight. However, on PND45, the ovary of GBH-exposed lambs showed altered follicular dynamics, increased proliferation of granulosa and theca cells, and decreased mRNA expression of FSHR and GDF9, whereas their uterus showed decreased cell proliferation but no alterations in the histomorphology or gene expression. In conclusion, GBH exposure altered the ovarian follicular dynamics and gene expression, and the proliferative activity of the ovaries and uterus of lambs. It is noteworthy that all the adverse effects found in the ovaries and uterus of both GBH-exposed groups were similar, independently of the administration route.
Collapse
Affiliation(s)
- Ramiro Alarcón
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Paola I Ingaramo
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Oscar E Rivera
- Instituto de Investigación sobre Producción Agropecuaria, Ambiente y Salud (IIPAAs), Facultad de Ciencias Agrarias, Universidad Nacional de Lomas de Zamora, Buenos Aires, Argentina
| | - Gisela H Dioguardi
- Instituto de Investigación sobre Producción Agropecuaria, Ambiente y Salud (IIPAAs), Facultad de Ciencias Agrarias, Universidad Nacional de Lomas de Zamora, Buenos Aires, Argentina
| | - María R Repetti
- Programa de Investigación y Análisis de Residuos y Contaminantes Químicos (PRINARC), Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Luisina D Demonte
- Programa de Investigación y Análisis de Residuos y Contaminantes Químicos (PRINARC), Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Mercedes M Milesi
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Jorgelina Varayoud
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Mónica Muñoz-de-Toro
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Enrique H Luque
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Litoral, Santa Fe, Argentina.
| |
Collapse
|
10
|
Glandular defects in the mouse uterus with sustained activation of TGF-beta signaling is associated with altered differentiation of endometrial stromal cells and formation of stromal compartment. PLoS One 2018; 13:e0209417. [PMID: 30550590 PMCID: PMC6294433 DOI: 10.1371/journal.pone.0209417] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 12/05/2018] [Indexed: 11/24/2022] Open
Abstract
Uterine gland development, also known as adenogenesis, is a key uterine morphogenic process indispensable for normal uterine function and fertility. Our earlier studies have reported that overactivation of TGFB receptor 1 (TGFBR1) in the mouse uterus using progesterone receptor (Pgr)-Cre recombinase causes female infertility, defective decidualization, and reduced uterine gland formation, a developmental milestone of postnatal uterus. To understand mechanisms that underpin the disrupted uterine gland formation in mice with sustained activation of TGFBR1, we raised the question of whether early postnatal adenogenesis was compromised in these mice. Experiments were designed using mice with constitutive activation of TGFBR1 driven by Pgr-Cre to determine the timing of adenogenic defects and potential mechanisms associated with dysregulation of adenogenic genes, luminal epithelial cell proliferation and endometrial fibrotic changes. Uterine tissues from mice with constitutive activation of TGFBR1 were collected during the critical time window of adenogenesis and analyzed together with age-matched controls. Multiple approaches including immunohistochemistry, immunofluorescence, Trichrome staining, quantitative real-time PCR, western blot, conditional knockout and human endometrial cell culture were utilized. TGFBR1 activation in the mouse uterus suppressed adenogenesis during postnatal uterine development, concomitant with the aberrant differentiation of uterine stromal cells. Analysis of transcript expression of WNT pathway components revealed dysregulation of adenogenesis-associated genes. Notably, the adenogenic defects occurred in spite of the increased proliferation of uterine luminal epithelial cells, accompanied by increased expression of genes associated with fibrotic changes. Moreover, the adenogenic defects were alleviated in mice where TGFBR1 was activated in presumably half of the complement of uterine cells. Our results suggest that altered differentiation of endometrial stromal cells and formation of stromal compartment promote adenogenic defects.
Collapse
|
11
|
Uterine Glands Agenesia in the Mare. J Equine Vet Sci 2017. [DOI: 10.1016/j.jevs.2017.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
12
|
Different enzymatic antioxidative pathways operate within the sheep caruncular and intercaruncular endometrium throughout the estrous cycle and early pregnancy. Theriogenology 2017; 99:111-118. [DOI: 10.1016/j.theriogenology.2017.05.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 05/03/2017] [Accepted: 05/22/2017] [Indexed: 12/12/2022]
|
13
|
Lopez Merlo M, Faya M, Priotto M, Barbeito C, Gobello C. Development and proliferation of feline endometrial glands from fetal life to ovarian cyclicity. Theriogenology 2017; 99:119-123. [DOI: 10.1016/j.theriogenology.2017.05.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/23/2017] [Accepted: 05/30/2017] [Indexed: 10/19/2022]
|
14
|
Hayashi KG, Hosoe M, Kizaki K, Fujii S, Kanahara H, Takahashi T, Sakumoto R. Differential gene expression profiling of endometrium during the mid-luteal phase of the estrous cycle between a repeat breeder (RB) and non-RB cows. Reprod Biol Endocrinol 2017; 15:20. [PMID: 28335821 PMCID: PMC5364712 DOI: 10.1186/s12958-017-0237-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 03/03/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Repeat breeding directly affects reproductive efficiency in cattle due to an increase in services per conception and calving interval. This study aimed to investigate whether changes in endometrial gene expression profile are involved in repeat breeding in cows. Differential gene expression profiles of the endometrium were investigated during the mid-luteal phase of the estrous cycle between repeat breeder (RB) and non-RB cows using microarray analysis. METHODS The caruncular (CAR) and intercaruncular (ICAR) endometrium of both ipsilateral and contralateral uterine horns to the corpus luteum were collected from RB (inseminated at least three times but not pregnant) and non-RB cows on Day 15 of the estrous cycle (4 cows/group). Global gene expression profiles of these endometrial samples were analyzed with a 15 K custom-made oligo-microarray for cattle. Immunohistochemistry was performed to investigate the cellular localization of proteins of three identified transcripts in the endometrium. RESULTS Microarray analysis revealed that 405 and 397 genes were differentially expressed in the CAR and ICAR of the ipsilateral uterine horn of RB, respectively when compared with non-RB cows. In the contralateral uterine horn, 443 and 257 differentially expressed genes were identified in the CAR and ICAR of RB, respectively when compared with non-RB cows. Gene ontology analysis revealed that genes involved in development and morphogenesis were mainly up-regulated in the CAR of RB cows. In the ICAR of both the ipsilateral and contralateral uterine horns, genes related to the metabolic process were predominantly enriched in the RB cows when compared with non-RB cows. In the analysis of the whole uterus (combining the data above four endometrial compartments), RB cows showed up-regulation of 37 genes including PRSS2, GSTA3 and PIPOX and down-regulation of 39 genes including CHGA, KRT35 and THBS4 when compared with non-RB cows. Immunohistochemistry revealed that CHGA, GSTA3 and PRSS2 proteins were localized in luminal and glandular epithelial cells and stroma of the endometrium. CONCLUSION The present study showed that endometrial gene expression profiles are different between RB and non-RB cows. The identified candidate endometrial genes and functions in each endometrial compartment may contribute to bovine reproductive performance.
Collapse
Affiliation(s)
- Ken-Go Hayashi
- 0000 0000 9191 6962grid.419600.aDivision of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, 305-0901 Japan
| | - Misa Hosoe
- 0000 0001 2222 0432grid.416835.dDivision of Animal Sciences, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, 305-8602 Japan
| | - Keiichiro Kizaki
- 0000 0001 0018 0409grid.411792.8Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, 020-8550 Japan
| | - Shiori Fujii
- 0000 0000 9191 6962grid.419600.aDivision of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, 305-0901 Japan
| | - Hiroko Kanahara
- 0000 0000 9191 6962grid.419600.aDivision of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, 305-0901 Japan
| | - Toru Takahashi
- 0000 0001 0018 0409grid.411792.8Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, 020-8550 Japan
| | - Ryosuke Sakumoto
- 0000 0000 9191 6962grid.419600.aDivision of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, 305-0901 Japan
| |
Collapse
|
15
|
Young CH, Rothfuss HM, Gard PF, Muth A, Thompson PR, Ashley RL, Cherrington BD. Citrullination regulates the expression of insulin-like growth factor-binding protein 1 (IGFBP1) in ovine uterine luminal epithelial cells. Reproduction 2016; 153:1-10. [PMID: 29565015 DOI: 10.1530/rep-16-0494] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 09/28/2016] [Accepted: 10/10/2016] [Indexed: 01/19/2023]
Abstract
There are five peptidylarginine deiminase (PAD) isozymes designated as PADs 1, 2, 3, 4 and 6, and many are expressed in female reproductive tissues. These enzymes post-translationally convert positively charged arginine amino acids into neutral citrulline residues. Targets for PAD-catalyzed citrullination include arginine residues on histone tails, which results in chromatin decondensation and changes in gene expression. Some of the first studies examining PADs found that they are localized to rodent uterine epithelial cells. Despite these findings, the function of PAD-catalyzed citrullination in uterine epithelial cells is still unknown. To address this, we first examined PAD expression in uterine cross-sections from pregnant ewes on gestation day 25 (d25). Immunohistochemistry revealed that the levels of PADs 2 and 4 are robust in luminal and glandular epithelia compared with those of PADs 1 and 3. As PADs 2 and 4 have well-characterized roles in histone citrullination, we next hypothesized that PADs citrullinate histones in these uterine cells. Examination of caruncle lysates from pregnant ewes on gestation d25 and an ovine luminal epithelial (OLE) cell line shows that histone H3 arginine residues 2, 8, 17 and 26 are citrullinated, but histone H4 arginine 3 is not. Using a pan-PAD inhibitor, we next attenuated histone citrullination in OLE cells, which resulted in a significant decrease in the expression of insulin-like growth factor-binding protein 1 (IGFBP1) mRNA. As IGFBP1 is important for the migration and attachment of the trophectoderm to uterine endometrium, our results suggest that PAD-catalyzed citrullination may be an important post-translational mechanism for the establishment of pregnancy in ewes.
Collapse
Affiliation(s)
- Coleman H Young
- Department of Zoology and PhysiologyUniversity of Wyoming, Laramie, Wyoming, USA
| | - Heather M Rothfuss
- Department of Zoology and PhysiologyUniversity of Wyoming, Laramie, Wyoming, USA
| | - Philip F Gard
- Department of Zoology and PhysiologyUniversity of Wyoming, Laramie, Wyoming, USA
| | - Aaron Muth
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Paul R Thompson
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Ryan L Ashley
- Department of Animal and Range SciencesNew Mexico State University, Las Cruces, New Mexico, USA
| | - Brian D Cherrington
- Department of Zoology and PhysiologyUniversity of Wyoming, Laramie, Wyoming, USA
| |
Collapse
|
16
|
Jeong W, Seo H, Sung Y, Ka H, Song G, Kim J. Lysophosphatidic Acid (LPA) Receptor 3-Mediated LPA Signal Transduction Pathways: A Possible Relationship with Early Development of Peri-Implantation Porcine Conceptus. Biol Reprod 2016; 94:104. [PMID: 27030044 DOI: 10.1095/biolreprod.115.137174] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 03/16/2016] [Indexed: 11/01/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a phospholipid with a variety of fatty acyl groups that mediates diverse biological effects on various types of cells through specific G protein-coupled receptors. LPA appears to play a significant role in many reproductive processes, including luteolysis, implantation, and placentation. Our previous study in pigs demonstrated that LPA and the LPA receptor system are present at the maternal-conceptus interface and that LPA increases uterine endometrial expression of prostaglandin-endoperoxide synthase 2 (PTGS2) through LPA receptor 3 (LPAR3). However, the role of LPA in conceptuses during early pregnancy has not been determined. Therefore, this study examined the effects of LPA in cell proliferation, migration, and activation of the intracellular signaling pathway in porcine conceptuses by using an established porcine trophectoderm (pTr) cell line isolated from Day 12 conceptuses. All examined LPA species with various fatty acid lengths increased proliferation and migration of pTr cells as the dosage increased. Immunoblot analyses found that LPA activated intracellular signaling molecules, extracellular signal-regulated kinase 1/2 (ERK1/2), ribosomal protein S6 kinase 90 kDa (P90RSK), ribosomal protein S6 (RPS6), and P38 in pTr cells. Furthermore, LPA increased expression of PTGS2 and urokinase-type plasminogen activator (PLAU), and the LPA-induced increases in PTGS2 and PLAU expression were inhibited by LPAR3 siRNA. Collectively, these results showed that LPA promotes proliferation, migration, and differentiation of pTr cells by activating the ERK1/2-P90RSK-RPS6 and P38 pathways, indicating that the LPA-LPAR3 system may be involved in the development of trophoblast during early pregnancy in pigs.
Collapse
Affiliation(s)
- Wooyoung Jeong
- Department of Animal Resources Science, Dankook University, Cheonan, Republic of Korea
| | - Heewon Seo
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Yujin Sung
- Department of Animal Resources Science, Dankook University, Cheonan, Republic of Korea
| | - Hakhyun Ka
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Jinyoung Kim
- Department of Animal Resources Science, Dankook University, Cheonan, Republic of Korea
| |
Collapse
|
17
|
Lopez Merlo M, Faya M, Blanco P, Carransa A, Barbeito C, Gobello C. Failure of a single dose of medroxyprogesterone acetate to induce uterine infertility in postnatally treated domestic cats. Theriogenology 2016; 85:718-23. [DOI: 10.1016/j.theriogenology.2015.10.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 10/04/2015] [Accepted: 10/07/2015] [Indexed: 11/17/2022]
|
18
|
Säfholm M, Jansson E, Fick J, Berg C. Molecular and histological endpoints for developmental reproductive toxicity in Xenopus tropicalis: Levonorgestrel perturbs anti-Müllerian hormone and progesterone receptor expression. Comp Biochem Physiol C Toxicol Pharmacol 2016; 181-182:9-18. [PMID: 26689642 DOI: 10.1016/j.cbpc.2015.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 12/02/2015] [Accepted: 12/07/2015] [Indexed: 02/02/2023]
Abstract
There is an increasing concern regarding the risks associated with developmental exposure to endocrine disrupting chemicals and the consequences for reproductive capability. The present study aimed to refine the Xenopus (Silurana) tropicalis test system for developmental reproductive toxicity by characterising molecular and histological features of sexual development, and to explore effects of exposure to the progestagen levonorgestrel (LNG). Larvae were exposed to LNG (0, 3, 30, 300 ng/L) over the first three weeks of development, encompassing the beginning of gonadal differentiation. mRNA levels of amh (anti-Müllerian hormone), amhr2 (amh receptor 2), ipgr (intracellular progesterone receptor), mpgr beta (membrane progesterone receptor beta), and cyp19a1 (cytochrome p450 19a1) were quantified in larvae and juveniles (4 weeks post-metamorphosis). Relative cyp19a1 and amh expression was used as a molecular marker for phenotypic sex of larvae. Gonadal and Müllerian duct development were characterised histologically in juveniles. Compared to controls, LNG exposure increased the expression of amh and ipgr in male larvae. In juveniles, mpgr beta expression was increased in both sexes and amhr2 expression was decreased in males, implying persistent effects of developmental progestagen exposure on amh and pgr expression signalling. No effects of LNG on the gonadal or Müllerian duct development were found, implying that the exposure window was not critical with regard to these endpoints. In juveniles, folliculogenesis had initiated and the Müllerian ducts were larger in females than in males. This new knowledge on sexual development in X. tropicalis is useful in the development of early life-stage endpoints for developmental reproductive toxicity.
Collapse
Affiliation(s)
- Moa Säfholm
- Uppsala University, Department of Organismal Biology, Norbyvägen 18A, 752 36 Uppsala, Sweden.
| | - Erika Jansson
- Uppsala University, Department of Organismal Biology, Norbyvägen 18A, 752 36 Uppsala, Sweden.
| | - Jerker Fick
- Umeå University, Department of Chemistry, KBC 6A, Linnaeus väg 6, 901 87 Umeå, Sweden.
| | - Cecilia Berg
- Uppsala University, Department of Organismal Biology, Norbyvägen 18A, 752 36 Uppsala, Sweden.
| |
Collapse
|
19
|
Bhusane K, Bhutada S, Chaudhari U, Savardekar L, Katkam R, Sachdeva G. Secrets of Endometrial Receptivity: Some Are Hidden in Uterine Secretome. Am J Reprod Immunol 2016; 75:226-36. [DOI: 10.1111/aji.12472] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/02/2015] [Indexed: 01/03/2023] Open
Affiliation(s)
- Kashmira Bhusane
- Primate Biology Laboratory; National Institute for Research in Reproductive Health; Indian Council of Medical Research; Mumbai India
| | - Sumit Bhutada
- Primate Biology Laboratory; National Institute for Research in Reproductive Health; Indian Council of Medical Research; Mumbai India
| | - Uddhav Chaudhari
- Primate Biology Laboratory; National Institute for Research in Reproductive Health; Indian Council of Medical Research; Mumbai India
| | - Lalita Savardekar
- Primate Biology Laboratory; National Institute for Research in Reproductive Health; Indian Council of Medical Research; Mumbai India
| | - Rajendra Katkam
- Primate Biology Laboratory; National Institute for Research in Reproductive Health; Indian Council of Medical Research; Mumbai India
| | - Geetanjali Sachdeva
- Primate Biology Laboratory; National Institute for Research in Reproductive Health; Indian Council of Medical Research; Mumbai India
| |
Collapse
|
20
|
Jeong W, Song G, Kim J. Mitogen activated protein kinase pathway-dependent effects of platelet-derived growth factor on migration of trophectoderm cells. Biochem Biophys Res Commun 2015; 463:575-81. [PMID: 26043684 DOI: 10.1016/j.bbrc.2015.05.098] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 05/29/2015] [Indexed: 11/29/2022]
Abstract
Successful development of the conceptus and implantation requires an intimate trophic connection between maternal uterus and conceptus mediated by local regulators including growth factors. Platelet-derived growth factor (PDGF) acts as a chemotactic factor for a variety of cell types. Current studies have determined that PDGF participates in rapid growth and development of cleavage stage embryos, but PDGF-induced effects on the growth and development of peri-implantation conceptus remains unknown. In the present study, PDGF induced phosphorylation of ERK1/2, AKT and RPS6 proteins in porcine trophectoderm (pTr) cells in a dose- and time-dependent manner. Addition of U0126 (an inhibitor of ERK1/2) or LY294002 (a PI3K inhibitor) blocked PDGF-induced effects on phosphorylation of signaling proteins. Combinations of PDGF and U0126 decreased PDGF-induced p-ERK1/2 and p-AKT1, but combinations of PDGF and LY294002 blocked only PDGF-induced AKT phosphorylation. Furthermore, PDGF significantly induced pTr cell migration and these stimulatory effects were blocked by U0126 and LY294002. Immunoreactive p-ERK1/2 and p-RPS6 proteins were abundant in pTr cells treated with PDGF, but U0126 reduced PDGF-induced p-ERK1/2 and p-RPS6 levels to basal amounts. Present study suggests that PDGF secreted into the maternal-conceptus microenvironment stimulates pTr cell migration through signal transduction cascades mediated by the ERK1/2 MAPK and AKT1 pathways.
Collapse
Affiliation(s)
- Wooyoung Jeong
- Department of Animal Resources Science, Dankook University, Cheonan 330-714, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 136-713, Republic of Korea
| | - Jinyoung Kim
- Department of Animal Resources Science, Dankook University, Cheonan 330-714, Republic of Korea.
| |
Collapse
|
21
|
Säfholm M, Jansson E, Fick J, Berg C. Mixture effects of levonorgestrel and ethinylestradiol: estrogenic biomarkers and hormone receptor mRNA expression during sexual programming. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 161:146-153. [PMID: 25703176 DOI: 10.1016/j.aquatox.2015.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 12/22/2014] [Accepted: 02/08/2015] [Indexed: 06/04/2023]
Abstract
Synthetic progesterone (progestins) and estrogens are widely used pharmaceuticals. Given that their simultaneous unintentional exposure occurs in wildlife and also in human infants, data on mixture effects of combined exposures to these hormones during development is needed. Using the Xenopus (Silurana) tropicalis test system we investigated mixture effects of levonorgestrel (LNG) and ethinylestradiol (EE2) on hormone sensitive endpoints. After larval exposure to LNG (0.1nM), or EE2 (0.1nM) singly, or in combination with LNG (0.01, 0.1, 1.0nM), the gonadal sex ratio was determined histologically and hepatic mRNA levels of genes encoding vitellogenin (vtg beta1) and the estrogen (esr1, esr2), progesterone (ipgr) and androgen (ar) receptors were quantified using quantitative PCR. All EE2-exposed groups showed female-biased sex ratios and increased vtg beta1 mRNA levels compared with the controls. Compared with the EE2-alone group (positive control) there were no significant alterations in vtg beta1 levels or in sex ratios in the co-exposure groups. Exposure to LNG-alone caused an increase in ar mRNA levels in females, but not in males, compared to the controls and the co-exposed groups, indicating that co-exposure to EE2 counteracted the LNG-induced ar levels. No treatment related impacts on the mRNA expression of esr1, esr2, and ipgr in female tadpoles were found, suggesting that these endpoints are insensitive to long-term exposure to estrogen or progestin. Due to the EE2-induced female-biased sex ratios, the mRNA expression data for the low number of males in the EE2-exposed groups were not statistically analyzed. In conclusion, our results suggest that induced vtg expression is a robust biomarker for estrogenic activity in exposure scenarios involving both estrogens and progestins. Developmental exposure to LNG caused an induction of hepatic ar mRNA expression that was antagonized by combined exposure to EE2 and LNG. To our knowledge this is the first study to report effects of combined exposures to EE2 and LNG during the period of sexual programming.
Collapse
Affiliation(s)
- Moa Säfholm
- Uppsala University, Department of Environmental Toxicology, Centre for Reproductive Biology in Uppsala, Norbyvägen 18A, 752 36 Uppsala, Sweden.
| | - Erika Jansson
- Uppsala University, Department of Environmental Toxicology, Centre for Reproductive Biology in Uppsala, Norbyvägen 18A, 752 36 Uppsala, Sweden.
| | - Jerker Fick
- Umeå University, Department of Chemistry, KBC 6A, Linnaeus väg 6, 901 87 Umeå, Sweden.
| | - Cecilia Berg
- Uppsala University, Department of Environmental Toxicology, Centre for Reproductive Biology in Uppsala, Norbyvägen 18A, 752 36 Uppsala, Sweden.
| |
Collapse
|
22
|
Filant J, Spencer TE. Uterine glands: biological roles in conceptus implantation, uterine receptivity and decidualization. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2015; 58:107-16. [PMID: 25023676 DOI: 10.1387/ijdb.130344ts] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
All mammalian uteri contain glands in the endometrium that synthesize or transport and secrete substances essential for survival and development of the conceptus (embryo/fetus and associated extraembryonic membranes). This review summarizes information related to the biological roles of uterine glands and their secretions in uterine receptivity, blastocyst/conceptus survival and implantation, and stromal cell decidualization. Studies with the ovine uterine gland knockout (UGKO) model support a primary role for uterine glands and, by inference, their secretions present in uterine luminal fluid histrotroph for conceptus survival and development. In rodents, studies with mutant and progesterone-induced UGKO mice found that uterine glands and their secretions are unequivocally required for establishment of uterine receptivity and blastocyst implantation and also may influence blastocyst trophectoderm activation and stromal cell decidualization in the uterus. Similarly in humans, histotroph from uterine glands appears critical for blastocyst implantation, uterine receptivity, and conceptus nutrition during the first trimester and uterine glands likely have a role in stromal cell decidualization. An increased understanding of uterine gland biology is important for diagnosis, prevention and treatment of fertility problems, particularly infertility and recurrent pregnancy loss, in domestic animals and humans.
Collapse
Affiliation(s)
- Justyna Filant
- Department of Animal Sciences and Center for Reproductive Biology, Washington State University, Pullman, WA, USA.
| | | |
Collapse
|
23
|
Spencer TE, Hansen TR. Implantation and Establishment of Pregnancy in Ruminants. REGULATION OF IMPLANTATION AND ESTABLISHMENT OF PREGNANCY IN MAMMALS 2015; 216:105-35. [DOI: 10.1007/978-3-319-15856-3_7] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
24
|
Abstract
All mammalian uteri contain glands in the endometrium that synthesize or transport and secrete substances essential for survival and development of the conceptus (embryo/fetus and associated extraembryonic membranes). This review summarizes information related to the biological roles of uterine glands and their secretions in blastocyst/conceptus survival and implantation, uterine receptivity, and stromal cell decidualization in humans and animal models. The infertility and recurrent pregnancy loss observed in the ovine uterine gland knockout (UGKO) model unequivocally supports a primary role for uterine glands and, by inference, their secretions present in uterine luminal fluid in survival and development of the conceptus. Further, studies with mutant and progesterone-induced UGKO mice found that uterine glands and their secretions are required for establishment of uterine receptivity and blastocyst implantation as well as stromal cell decidualization. Similarly in humans, uterine glands and their secretory products are likely critical regulators of blastocyst implantation, uterine receptivity, and conceptus growth and development during the first trimester. Circumstantial evidence suggests that deficient glandular activity may be a causative factor in pregnancy failure and complications in humans. Thus, an increased understanding of uterine gland biology is important for diagnosis, prevention, and treatment of fertility and pregnancy problems in mammals.
Collapse
Affiliation(s)
- Thomas E Spencer
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington
| |
Collapse
|
25
|
Jeong W, Song G, Bazer FW, Kim J. Insulin-like growth factor I induces proliferation and migration of porcine trophectoderm cells through multiple cell signaling pathways, including protooncogenic protein kinase 1 and mitogen-activated protein kinase. Mol Cell Endocrinol 2014; 384:175-84. [PMID: 24508636 DOI: 10.1016/j.mce.2014.01.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 01/08/2014] [Accepted: 01/31/2014] [Indexed: 11/27/2022]
Abstract
During early pregnancy, the developing conceptus is dependent upon a wide range of growth factors and nutrients that are secreted by or transported by uterine epithelia into the uterus at the maternal-conceptus interface for successful implantation and placentation. Among these factors, insulin-like growth factor-I (IGF-I) is known to play an important role in development of the early embryo and uterine endometrium. However, few studies have been conducted with pigs to determine IGF-I-induced functional effects on peri-implantation embryos such as activation of cell signaling cascades responsible for growth, proliferation and differentiation of cells of the conceptus. Therefore, the aim of this study was to analyze mRNA expression of endometrial IGF-I and its receptor, to examine the functional role of IGF-I on primary porcine trophectoderm (pTr) cells and to assess potential signaling pathways responsible for biological activities of IGF-1. In the present study, expression of endometrial type I IGF receptor (IGF-IR) mRNA increased significantly from Day 10 to Day 12 of pregnancy and the increase was greater for pregnant than cyclic gilts. Both IGF-I and IGF-IR mRNAs were abundant in endometrial luminal-, glandular epithelia, and stratum compactum stroma on Day 12 of pregnancy. In addition, IGF-I significantly induced phosphorylation of AKT1, ERK1/2 and RPS6 in a time- and concentration-dependent manner in pTr cells. Immunofluorescence microscopy revealed that IGF-I treated pTr cells exhibited increased abundance of phosphorylated (p)-AKT1 and p-ERK1/2 MAPK proteins in the nucleus and cytoplasm, and p-RPS6 proteins in the cytosol as compared to non-treated pTr cells. In the presence of the ERK1/2 MAPK inhibitor (U0126), IGF-I-induced AKT1 phosphorylation was not affected, whereas the PI3K inhibitor (LY294002) decreased IGF-I-induced phosphorylation of ERK1/2 and AKT1 proteins, and both the PI3K-AKT1 and ERK1/2 MAPK pathways were blocked by LY294002. Furthermore, IGF-I significantly stimulated both proliferation and migration of pTr cells, but these effects were blocked by P38 inhibitor (SB203580), U0126, MTOR inhibitor (rapamycin) and LY294002. Taken together, these results indicate that IGF-I coordinately regulates multiple cell signaling pathways including PI3K-AKT1-RPS6 and ERK1/2 MAPK signaling pathways that are critical to proliferation, migration and survival of trophectoderm cells during early pregnancy in pigs.
Collapse
Affiliation(s)
- Wooyoung Jeong
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Gwonhwa Song
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Fuller W Bazer
- Center for Animal Biotechnology and Genomics and Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Jinyoung Kim
- Department of Animal Resources Science, Dankook University, Cheonan, Republic of Korea.
| |
Collapse
|
26
|
Jeong W, Kim J, Bazer FW, Song G. Stimulatory Effect of Vascular Endothelial Growth Factor on Proliferation and Migration of Porcine Trophectoderm Cells and Their Regulation by the Phosphatidylinositol-3-Kinase-AKT and Mitogen-Activated Protein Kinase Cell Signaling Pathways. Biol Reprod 2014; 90:50. [PMID: 24451985 DOI: 10.1095/biolreprod.113.115873] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Vascular endothelial growth factor (VEGF), a potent stimulator for angiogenesis, is likely to regulate implantation by stimulating endometrial angiogenesis and vascular permeability. In addition to known angiogenetic effects, VEGF has been suggested to participate in development of the early embryo as a mediator of fetal-maternal dialogue. Current studies have determined VEGF in terms of its role in endometrial vascular events, but VEGF-induced effects on the peri-implantation conceptus (embryo and extraembryonic membranes) remains unknown. In the present study, endometrial VEGF, VEGF receptor-1 (VEGFR-1), and VEGF receptor-2 (VEGFR-2) mRNAs increased significantly during the peri-implantation period of pregnancy as compared to the estrous cycle. Expression of VEGF, VEGFR-1, and VEGFR-2 mRNAs was abundant in endometrial luminal and glandular epithelia, endothelial blood vessels, and scattered cells in the stroma and conceptus trophectoderm. In addition, porcine trophectoderm (pTr) cells treated with VEGF exhibited increased abundance of phosphorylated (p)-AKT1, p-ERK1/2, p-p70RSK, p-RPS6, and p-4EBP1 in a time-dependent manner. The addition of U0126, an inhibitor of ERK1/2, inhibited VEGF-induced ERK1/2 phosphorylation, but AKT1 phosphorylation was not affected. The addition of LY294002, a PI3K inhibitor, decreased VEGF-induced phosphorylation of ERK1/2 and AKT1. Furthermore, VEGF significantly stimulated proliferation and migration of pTr cells, but these effects were blocked by SB203580, U0126, rapamycin, and LY294002, which inhibit p38 MAPK, ERK1/2, mTOR, and PI3K, respectively. These results suggest that VEGF is critical to successful growth and development of pTr during early pregnancy and that VEGF-induced stimulatory effect is coordinately regulated by multiple cell signaling pathways, including PI3K-AKT1 and MAPK signaling pathways.
Collapse
Affiliation(s)
- Wooyoung Jeong
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
27
|
Stewart CA, Wang Y, Bonilla-Claudio M, Martin JF, Gonzalez G, Taketo MM, Behringer RR. CTNNB1 in mesenchyme regulates epithelial cell differentiation during Müllerian duct and postnatal uterine development. Mol Endocrinol 2013; 27:1442-54. [PMID: 23904126 DOI: 10.1210/me.2012-1126] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Müllerian duct differentiation and development into the female reproductive tract is essential for fertility, but mechanisms regulating these processes are poorly understood. WNT signaling is critical for proper development of the female reproductive tract as evident by the phenotypes of Wnt4, Wnt5a, Wnt7a, and β-catenin (Ctnnb1) mutant mice. Here we extend these findings by determining the effects of constitutive CTNNB1 activation within the mesenchyme of the developing Müllerian duct and its differentiated derivatives. This was accomplished by crossing Amhr2-Cre knock-in mice with Ctnnb1 exon (ex) 3(f/f) mice. Amhr2-Cre(Δ/+); Ctnnb1 ex3(f/+) females did not form an oviduct, had smaller uteri, endometrial gland defects, and were infertile. At the cellular level, stabilization of CTNNB1 in the mesenchyme caused alterations within the epithelium, including less proliferation, delayed uterine gland formation, and induction of an epithelial-mesenchymal transition (EMT) event. This EMT event is observed before birth and is complete within 5 days after birth. Misexpression of estrogen receptor α in the epithelia correlated with the EMT before birth, but not after. These studies indicate that regulated CTNNB1 in mesenchyme is important for epithelial cell differentiation during female reproductive tract development.
Collapse
Affiliation(s)
- C Allison Stewart
- Department of Genetics, University of Texas MD Anderson Cancer Center, University of Texas Graduate School of Biomedical Sciences, Houston, Texas 77030, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Cooke PS, Spencer TE, Bartol FF, Hayashi K. Uterine glands: development, function and experimental model systems. Mol Hum Reprod 2013; 19:547-58. [PMID: 23619340 DOI: 10.1093/molehr/gat031] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Development of uterine glands (adenogenesis) in mammals typically begins during the early post-natal period and involves budding of nascent glands from the luminal epithelium and extensive cell proliferation in these structures as they grow into the surrounding stroma, elongate and mature. Uterine glands are essential for pregnancy, as demonstrated by the infertility that results from inhibiting the development of these glands through gene mutation or epigenetic strategies. Several genes, including forkhead box A2, beta-catenin and members of the Wnt and Hox gene families, are implicated in uterine gland development. Progestins inhibit uterine epithelial proliferation, and this has been employed as a strategy to develop a model in which progestin treatment of ewes for 8 weeks from birth produces infertile adults lacking uterine glands. More recently, mouse models have been developed in which neonatal progestin treatment was used to permanently inhibit adenogenesis and adult fertility. These studies revealed a narrow and well-defined window in which progestin treatments induced permanent infertility by impairing neonatal gland development and establishing endometrial changes that result in implantation defects. These model systems are being utilized to better understand the molecular mechanisms underlying uterine adenogenesis and endometrial function. The ability of neonatal progestin treatment in sheep and mice to produce infertility suggests that an approach of this kind may provide a contraceptive strategy with application in other species. Recent studies have defined the temporal patterns of adenogenesis in uteri of neonatal and juvenile dogs and work is underway to determine whether neonatal progestin or other steroid hormone treatments might be a viable contraceptive approach in this species.
Collapse
Affiliation(s)
- Paul S Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA.
| | | | | | | |
Collapse
|
29
|
Aralla M, Groppetti D, Caldarini L, Cremonesi F, Arrighi S. Morphological evaluation of the placenta and fetal membranes during canine pregnancy from early implantation to term. Res Vet Sci 2013; 95:15-22. [PMID: 23485173 DOI: 10.1016/j.rvsc.2013.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 12/12/2012] [Accepted: 02/01/2013] [Indexed: 11/26/2022]
Abstract
To describe the histological changes of fetal adnexa throughout the physiological pregnancy, canine samples were obtained during natural delivery and caesarean section, as well as during ovariohysterectomy performed at any stage of undesired pregnancies (N=12). The first period of pregnancy (multiple samples collected at 10, 12, 14 days) was consistent with pre- and peri-implantation events, i.e. apposition and initial invasion steps into the endometrium. The second period (multiple samples collected at 18, 38, 40, 45 days) was related to the development of extra-embryonic structures, placenta establishment and labyrinth formation. At the end of this period the maximum morphological complexity of the endotheliochorial placenta was achieved, characterized by complete erosion of the endometrial epithelium and underlying interstitium with exposure of maternal capillaries to the chorial cells. The third period of gestation (multiple samples collected at 50, 53, 57, 60, 63 days) was characterized by enhancement either of placental and extra-embryonic tissues.
Collapse
Affiliation(s)
- Marina Aralla
- Department of Health, Animal Sciences and Food Safety, Università degli Studi di Milano, 10 Via Celoria, I-20133 Milan, Italy.
| | | | | | | | | |
Collapse
|
30
|
Zhang S, Lin H, Kong S, Wang S, Wang H, Wang H, Armant DR. Physiological and molecular determinants of embryo implantation. Mol Aspects Med 2013; 34:939-80. [PMID: 23290997 DOI: 10.1016/j.mam.2012.12.011] [Citation(s) in RCA: 381] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 12/25/2012] [Accepted: 12/26/2012] [Indexed: 01/19/2023]
Abstract
Embryo implantation involves the intimate interaction between an implantation-competent blastocyst and a receptive uterus, which occurs in a limited time period known as the window of implantation. Emerging evidence shows that defects originating during embryo implantation induce ripple effects with adverse consequences on later gestation events, highlighting the significance of this event for pregnancy success. Although a multitude of cellular events and molecular pathways involved in embryo-uterine crosstalk during implantation have been identified through gene expression studies and genetically engineered mouse models, a comprehensive understanding of the nature of embryo implantation is still missing. This review focuses on recent progress with particular attention to physiological and molecular determinants of blastocyst activation, uterine receptivity, blastocyst attachment and uterine decidualization. A better understanding of underlying mechanisms governing embryo implantation should generate new strategies to rectify implantation failure and improve pregnancy rates in women.
Collapse
Affiliation(s)
- Shuang Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China; Graduate School of the Chinese Academy of Sciences, Beijing 100039, PR China
| | | | | | | | | | | | | |
Collapse
|
31
|
Bazer FW. Contributions of an animal scientist to understanding the biology of the uterus and pregnancy. Reprod Fertil Dev 2013; 25:129-47. [DOI: 10.1071/rd12266] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
I developed a passion for reproductive biology when taking a course in Physiology of Reproduction at Louisiana State University while preparing to apply for Veterinary School at Texas A&M University. My career path changed. I entered graduate school, obtained a Ph.D. and have enjoyed an academic career conducting research in uterine biology and pregnancy in animal science departments at the University of Florida and at Texas A&M University. My contributions to science include: (1) identification of molecules secreted by or transported by uterine epithelia into the uterine lumen that are critical to successful establishment and maintenance of pregnancy, (2) discovery of steroids and proteins required for pregnancy-recognition signalling and their mechanisms of action in pigs and ruminants, (3) patterns of fetal–placental development and placental transport of nutrients, (4) identification of links between nutrients and components of histotroph that affect fetal–placental development, (5) characterising aspects of the endocrinology of pregnancy and (6) contributing to efforts to exploit the therapeutic value of interferon tau, particularly for treatment of autoimmune and inflammatory diseases. Current research focuses on select nutrients in the uterine lumen, specifically amino acids, glucose and fructose, that affect conceptus development, the therapeutic potential for interferon tau, stromal–epithelial cell signalling whereby progesterone and oestrogen act via steroid receptors in uterine stromal cells to stimulate secretion of growth factors (e.g. fibroblast growth factors and hepatocyte growth factor) that regulate uterine epithelial cells and conceptus trophectoderm, and roles of toll-like receptors expressed by uterine epithelia and conceptus trophectoderm in pregnancy.
Collapse
|
32
|
Spencer TE, Dunlap KA, Filant J. Comparative developmental biology of the uterus: insights into mechanisms and developmental disruption. Mol Cell Endocrinol 2012; 354:34-53. [PMID: 22008458 DOI: 10.1016/j.mce.2011.09.035] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 09/19/2011] [Accepted: 09/22/2011] [Indexed: 01/30/2023]
Abstract
The uterus is an essential organ for reproduction in mammals that derives from the Müllerian duct. Despite the importance of the uterus for the fertility and health of women and their offspring, relatively little is known about the hormonal, cellular and molecular mechanisms that regulate development of the Müllerian duct and uterus. This review aims to summarize the hormonal, cellular and molecular mechanisms and pathways governing development of the Müllerian duct and uterus as well as highlight developmental programming effects of endocrine disruptor compounds. Organogenesis, morphogenesis, and functional differentiation of the uterus are complex, multifactorial processes. Disruption of uterine development in the fetus and neonate by genetic defects and exposure to endocrine disruptor compounds can cause infertility and cancer in the adult and their offspring via developmental programming. Clear conservation of some factors and pathways are observed between species; therefore, comparative biology is useful to identify candidate genes and pathways underlying congenital abnormalities in humans.
Collapse
Affiliation(s)
- Thomas E Spencer
- Center for Reproductive Biology, Department of Animal Sciences, Washington State University, Pullman, WA 99164-6310, USA.
| | | | | |
Collapse
|
33
|
Bazer FW, Song G, Kim J, Erikson DW, Johnson GA, Burghardt RC, Gao H, Carey Satterfield M, Spencer TE, Wu G. Mechanistic mammalian target of rapamycin (MTOR) cell signaling: effects of select nutrients and secreted phosphoprotein 1 on development of mammalian conceptuses. Mol Cell Endocrinol 2012; 354:22-33. [PMID: 21907263 DOI: 10.1016/j.mce.2011.08.026] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Revised: 08/16/2011] [Accepted: 08/17/2011] [Indexed: 01/30/2023]
Abstract
Morphological differentiation of uterine glands in mammals is a postnatal event vulnerable to adverse effects of endocrine disruptors. Exposure of ewe lambs to a progestin from birth to postnatal day 56 prevents development of uterine glands and, as adults, the ewes are unable to exhibit estrous cycles or maintain pregnancy. Uterine epithelia secrete proteins and transport nutrients into the uterine lumen necessary for conceptus development, pregnancy recognition signaling and implantation, including arginine and secreted phosphoprotein 1 (SPP1). Arginine can be metabolized to nitric oxide and to polyamines or act directly to activate MTOR cell signaling to stimulate proliferation, migration, and mRNA translation in trophectoderm cells. SPP1 binds αvβ3 and α5β1 integrins and induces focal adhesion assembly, adhesion and migration of conceptus trophectoderm cells during implantation. Thus, arginine and SPP1 mediate growth, migration, cytoskeletal remodeling and adhesion of trophectoderm essential for pregnancy recognition signaling and implantation.
Collapse
Affiliation(s)
- Fuller W Bazer
- Center for Animal Biotechnology and Genomics and Department of Animal Science, Texas A&M University, College Station, TX, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Franco HL, Rubel CA, Large MJ, Wetendorf M, Fernandez-Valdivia R, Jeong JW, Spencer TE, Behringer RR, Lydon JP, Demayo FJ. Epithelial progesterone receptor exhibits pleiotropic roles in uterine development and function. FASEB J 2011; 26:1218-27. [PMID: 22155565 DOI: 10.1096/fj.11-193334] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The ovarian steroid progesterone, acting through the progesterone receptor (PR), coordinates endometrial epithelial-stromal cell communication, which is critical for its development and function. PR expression in these cellular compartments is under tight temporal and endocrine control. Although ex vivo studies demonstrated the importance of stromal PR expression, they failed to show a role for epithelial PR in uterine function. Here, the in vivo role of PR in the uterine epithelium is defined using floxed PR (PR(f/f)) mice crossed to Wnt7a-Cre mice. Progesterone was unable to stimulate the expression of its epithelial target genes, including Ihh, in the Wnt7a-Cre(+)PR(f/-) mice. Analysis was conducted on Ihh to determine whether PR directly regulates epithelial gene transcription. ChIP-on-chip analysis identified PR binding sites in the 5'-flanking region of Ihh. Cotransfection of the proximal Ihh promoter with PR demonstrated that PR directly regulates Ihh transcription. Female Wnt7a-Cre(+)PR(f/-) mice are infertile due to defects in embryo attachment, stromal cell decidualization, and the inability to cease estrogen-induced epithelial cell proliferation. Finally, progesterone was unable to inhibit neonatal endometrial glandular development in Wnt7a-Cre(+)PR(f/-) mice. Thus, epithelial PR is necessary for the regulation of progesterone epithelial target gene expression, as well as uterine function and development.
Collapse
Affiliation(s)
- Heather L Franco
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Stewart CA, Fisher SJ, Wang Y, Stewart MD, Hewitt SC, Rodriguez KF, Korach KS, Behringer RR. Uterine gland formation in mice is a continuous process, requiring the ovary after puberty, but not after parturition. Biol Reprod 2011; 85:954-64. [PMID: 21734259 DOI: 10.1095/biolreprod.111.091470] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Uterine gland formation occurs postnatally in an ovary- and steroid-independent manner in many species, including humans. Uterine glands secrete substances that are essential for embryo survival. Disruption of gland development during the postnatal period prevents gland formation, resulting in infertility. Interestingly, stabilization of beta-catenin (CTNNB1) in the uterine stroma causes a delay in gland formation rather than a complete absence of uterine glands. Thus, to determine if a critical postnatal window for gland development exists in mice, we tested the effects of extending the endocrine environment of pregnancy on uterine gland formation by treating neonatal mice with estradiol, progesterone, or oil for 5 days. One uterine horn was removed before puberty, and the other was collected at maturity. Some mice were also ovariectomized before puberty. The hormone-treated mice exhibited a delay in uterine gland formation. Hormone-treatment increased the abundance of uterine CTNNB1 and estrogen receptor alpha (ESR1) before puberty, indicating possible mechanisms for delayed gland formation. Despite having fewer glands, progesterone-treated mice were fertile, suggesting that a threshold number of glands is required for pregnancy. Mice that were ovariectomized before puberty did not undergo further uterine growth or gland development. Finally, to establish the role of the ovary in postpartum uterine gland regeneration, mice were either ovariectomized or given a sham surgery after parturition, and uteri were evaluated 1 wk later. We found that the ovary is not required for uterine growth or gland development following parturition. Thus, uterine gland development occurs continuously in mice and requires the ovary after puberty, but not after parturition.
Collapse
Affiliation(s)
- C Allison Stewart
- Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Kvarnryd M, Grabic R, Brandt I, Berg C. Early life progestin exposure causes arrested oocyte development, oviductal agenesis and sterility in adult Xenopus tropicalis frogs. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2011; 103:18-24. [PMID: 21392492 DOI: 10.1016/j.aquatox.2011.02.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 02/04/2011] [Accepted: 02/08/2011] [Indexed: 05/30/2023]
Abstract
Levonorgestrel (LNG) is a commonly used pharmaceutical progestin found in the environment. Information on the long-term toxicity of progestins following early life exposure is scant. We investigated the effects of developmental LNG exposure on sex differentiation, reproductive organ development and fertility in the model frog Xenopus tropicalis. Tadpoles were exposed to 0, 0.06 or 0.5nM LNG via the water from hatching until metamorphosis. At metamorphosis effects on gonadal differentiation were evaluated using a subsample of frogs. Remaining animals were held unexposed for nine months, at which time reproductive organ structure, function and fertility were determined. LNG exposure severely impaired oviduct and ovary development and fertility. All adult females in the 0.5nM group (n=10) completely lacked oviducts. They also displayed a significantly larger fraction of immature oocytes, arrested in meiotic prophase, than control females. Upon mating with unexposed males, only one of 11 LNG-exposed females laid eggs, whereas all control females did. No effects on testicular development, sperm count or male fertility were observed. At metamorphosis, no effects on sex ratio or gonadal histology were evident. The effects on ovarian and oviductal development were detected at adult age but not at metamorphosis, emphasising the importance of investigating the long-term consequences of developmental exposure. This is the first developmental reproductive toxicity study of a progestin in an aquatic vertebrate. Considering that several progestins are present in contaminated surface waters, further investigation into the sensitivity of frogs to progestins is warranted to understand the risk such compounds may pose to wild frog populations.
Collapse
Affiliation(s)
- Moa Kvarnryd
- Department of Environmental Toxicology, Uppsala University, Center for Reproductive Biology in Uppsala (CRU), Uppsala University, Norbyvägen 18A, 752 36 Uppsala, Sweden.
| | | | | | | |
Collapse
|
38
|
Yang QE, Giassetti MI, Ealy AD. Fibroblast growth factors activate mitogen-activated protein kinase pathways to promote migration in ovine trophoblast cells. Reproduction 2011; 141:707-14. [PMID: 21310815 DOI: 10.1530/rep-10-0541] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Fibroblast growth factors (FGFs) 2 and FGF10 are uterine- and conceptus-derived factors that mediate trophoblast activities in cattle and sheep. To extend our understanding of how FGFs may control peri-implantation development in ruminants, we determined whether FGF2 and FGF10 impact trophoblast cell migration. Transwell inserts containing 8 μm pores were used to examine whether FGF2 or FGF10 supplementation increased oTr1 cell migration. Supplementation with 0.5 ng/ml FGF2 or FGF10 did not affect oTr1 cell migration number, but exposure to 5 or 50 ng/ml FGF2 or FGF10 increased (P<0.05) oTr1 cell migration when compared with controls. The involvement of specific MAP kinase (MAPK) cascades in mediating this FGF response was examined by using pharmacological inhibitors of specific MAPKs. Western blot analysis indicated that FGF2 and FGF10 increased phosphorylation status of MAPKs 1, 3, 8, 9, and 14. Exposure to specific inhibitors blocked FGF induction of each MAPK. Exposure to inhibitors before supplementation with FGF2 or FGF10 prevented FGF induction of cell migration, indicating that each of these signaling molecules was required for FGF effects. A final series of studies examined whether FGF2 and FGF10 also mediated the migration of a bovine trophoblast line (CT1 cell). Increases in migration were detected in each cell line by supplementing 5 or 50 ng/ml FGF2 or FGF10 (P<0.05). In summary, FGF2 and FGF10 regulate migratory activity of ovine trophoblast cells through MAPK-dependent pathways. These outcomes provide further evidence that FGFs function as mediators of peri-implantation conceptus development in cattle and sheep.
Collapse
Affiliation(s)
- Qi En Yang
- Department of Animal Sciences, DH Barron Reproductive and Perinatal Biology Research Program, University of Florida, PO Box 110910, Gainesville, Florida 32611-0910, USA
| | | | | |
Collapse
|
39
|
Jones C, Aplin J, Burton G. First Trimester Histiotrophe Shows Altered Sialylation Compared with Secretory Phase Glycoconjugates in Human Endometrium. Placenta 2010; 31:576-80. [DOI: 10.1016/j.placenta.2010.04.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Revised: 04/22/2010] [Accepted: 04/26/2010] [Indexed: 11/28/2022]
|
40
|
Hugentobler SA, Sreenan JM, Humpherson PG, Leese HJ, Diskin MG, Morris DG. Effects of changes in the concentration of systemic progesterone on ions, amino acids and energy substrates in cattle oviduct and uterine fluid and blood. Reprod Fertil Dev 2010; 22:684-94. [PMID: 20353728 DOI: 10.1071/rd09129] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Accepted: 09/30/2009] [Indexed: 12/20/2022] Open
Abstract
Early embryo loss is a major factor affecting the conception rate in cattle. Up to 40% of cattle embryos die within 3 weeks of fertilisation while they are nutritionally dependent on oviduct and uterine fluids for their survival. Inadequate systemic progesterone is one of the factors contributing to this loss. We have characterised the effects of changes in systemic progesterone on amino acid, ion and energy substrate composition of oviduct and uterine fluids on Days 3 and 6, respectively, of the oestrus cycle in cattle. Oviduct and uterine fluids were collected in situ following infusion of progesterone. There was no effect of progesterone on oviduct fluid secretion rate; however, uterine fluid secretion rate was lowered. Progesterone increased uterine glucose, decreased oviduct sulfate and, to a lesser degree, oviduct sodium, but had no effect on any of the ions in the uterus. The most marked effect of progesterone was on oviducal amino acid concentrations, with a twofold increase in glycine, whereas in the uterus only valine was increased. These results provide novel information on the maternal environment of the early cattle embryo and provide further evidence of progesterone regulation of oviduct amino acid concentrations in cattle.
Collapse
Affiliation(s)
- S A Hugentobler
- Animal Bioscience Department, Teagasc, Mellows Campus, Athenry, Galway, Ireland
| | | | | | | | | | | |
Collapse
|
41
|
Post natal oestrogen administration stimulates precocious endometrial gland development in the horse. Equine Vet J 2010; 41:678-84. [DOI: 10.2746/042516409x429428] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
42
|
Song GH, Han JY, Spencer TE, Bazer FW. Interferon Tau in the Ovine Uterus. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2009. [DOI: 10.5187/jast.2009.51.6.471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
43
|
Bagnell CA, Steinetz BG, Bartol FF. Milk-Borne Relaxin and the Lactocrine Hypothesis for Maternal Programming of Neonatal Tissues. Ann N Y Acad Sci 2009; 1160:152-7. [DOI: 10.1111/j.1749-6632.2009.03834.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
44
|
Kim J, Song G, Gao H, Farmer JL, Satterfield MC, Burghardt RC, Wu G, Johnson GA, Spencer TE, Bazer FW. Insulin-like growth factor II activates phosphatidylinositol 3-kinase-protooncogenic protein kinase 1 and mitogen-activated protein kinase cell Signaling pathways, and stimulates migration of ovine trophectoderm cells. Endocrinology 2008; 149:3085-94. [PMID: 18339715 DOI: 10.1210/en.2007-1367] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
IGF-II, a potent stimulator of cellular proliferation, differentiation, and development, regulates uterine function and conceptus growth in several species. In situ hybridization analyses found that IGF-II mRNA was most abundant in the caruncular endometrial stroma of both cyclical and pregnant ewes. In the intercaruncular endometrium, IGF-II mRNA transitioned from stroma to luminal epithelium between d 14 and 20 of pregnancy. IGF-II mRNA was present in all cells of the conceptus but was particularly abundant in the yolk sac. Immunohistochemical analyses revealed that phosphorylated (p)-protooncogenic protein kinase 1, p-ribosomal protein S6 kinase, p-ERK1/2, and p-P38 MAPK proteins were present at low levels in a majority of endometrial cells but were most abundant in the nuclei of endometrial luminal epithelium and conceptus trophectoderm of pregnant ewes. In mononuclear trophectoderm cells isolated from d-15 conceptuses, IGF-II increased the abundance of p-pyruvate dehydrogenase kinase 1, p-protooncogenic protein kinase 1, p-glycogen synthase kinase 3B, p-FK506 binding protein 12-rapamycin associated protein 1, and p-ribosomal protein S6 kinase protein within 15 min, and the increase was maintained for 90 min. IGF-II also elicited a rapid increase in p-ERK1/2 and p-P38 MAPK proteins that was maximal at 15 or 30 min posttreatment. Moreover, IGF-II increased migration of trophectoderm cells. Collectively, these results support the hypothesis that IGF-II coordinately activates multiple cell signaling pathways critical to survival, growth, and differentiation of the ovine conceptus during early pregnancy.
Collapse
Affiliation(s)
- Jinyoung Kim
- Center for Animal Biotechnology and Genomics, Department of Animal Science, Texas A&M University, College Station, Texas 77843-2471, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Hayashi K, O'Connell AR, Juengel JL, McNatty KP, Davis GH, Bazer FW, Spencer TE. Postnatal uterine development in Inverdale ewe lambs. Reproduction 2008; 135:357-65. [PMID: 18299429 DOI: 10.1530/rep-07-0323] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Postnatal development of the uterus involves, particularly, development of uterine glands. Studies with ovariectomized ewe lambs demonstrated a role for ovaries in uterine growth and endometrial gland development between postnatal days (PNDs) 14 and 56. The uterotrophic ovarian factor(s) is presumably derived from the large numbers of growing follicles in the neonatal ovary present after PND 14. The Inverdale gene mutation (FecXI) results in an increased ovulation rate in heterozygous ewes; however, homozygous ewes (II) are infertile and have 'streak' ovaries that lack normal developing of preantral and antral follicles. Uteri were obtained on PND 56 to determine whether postnatal uterine development differs between wild-type (++) and II Inverdale ewes. When compared with wild-type ewes, uterine weight of II ewes was 52% lower, and uterine horn length tended to be shorter, resulting in a 68% reduction in uterine weight:length ratio in II ewes. Histomorphometrical analyses determined that endometria and myometria of II ewes were thinner and intercaruncular endometrium contained 38% fewer endometrial glands. Concentrations of estradiol in the neonatal ewes were low and not different between ++ and II ewes, but II ewes had lower concentrations of testosterone and inhibin-alpha between PNDs 14 and 56. Receptors for androgen and activin were detected in the neonatal uteri of both ++ and II ewes. These results support the concept that developing preantral and/or antral follicles of the ovary secrete uterotrophic factors, perhaps testosterone or inhibin-alpha, that acts in an endocrine manner to stimulate uterine growth and endometrial gland development in the neonatal ewes.
Collapse
Affiliation(s)
- Kanako Hayashi
- Department of Animal Science, Center for Animal Biotechnology and Genomics, Texas A and M University, 442 Kleberg Center, 2471 TAMU, College Station, Texas 77843-2471, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Spencer TE, Sandra O, Wolf E. Genes involved in conceptus–endometrial interactions in ruminants: insights from reductionism and thoughts on holistic approaches. Reproduction 2008; 135:165-79. [DOI: 10.1530/rep-07-0327] [Citation(s) in RCA: 204] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
This review summarizes new knowledge on expression of genes and provides insights into approaches for study of conceptus–endometrial interactions in ruminants with emphasis on the peri-implantation stage of pregnancy. Conceptus–endometrial interactions in ruminants are complex and involve carefully orchestrated temporal and spatial alterations in gene expression regulated by hormones from the ovary and conceptus. Progesterone is the hormone of pregnancy and acts on the uterus to stimulate blastocyst survival, growth, and development. Inadequate progesterone levels or a delayed rise in progesterone is associated with pregnancy loss. The mononuclear trophectoderm cells of the elongating blastocyst synthesize and secrete interferon-τ (IFNT), the pregnancy recognition signal. Trophoblast giant binucleate cells begin to differentiate and produce hormones including chorionic somatomammotropin 1 (CSH1 or placental lactogen). A number of genes, induced or stimulated by progesterone, IFNT, and/or CSH1 in a cell-specific manner, are implicated in trophectoderm adhesion to the endometrial luminal epithelium and regulation of conceptus growth and differentiation. Transcriptional profiling experiments are beginning to unravel the complex dynamics of conceptus–endometrial interactions in cattle and sheep. Future experiments should incorporate physiological models of pregnancy loss and be complemented by metabolomic studies of uterine lumen contents to more completely define factors required for blastocyst survival, growth, and implantation. Both reduction and holistic approaches will be important to understand the multifactorial phenomenon of recurrent pregnancy loss and provide a basis for new strategies to improve pregnancy outcome and reproductive efficiency in cattle and other domestic animals.
Collapse
|
47
|
Tranguch S, Wang H, Daikoku T, Xie H, Smith DF, Dey SK. FKBP52 deficiency-conferred uterine progesterone resistance is genetic background and pregnancy stage specific. J Clin Invest 2007; 117:1824-34. [PMID: 17571166 PMCID: PMC1888571 DOI: 10.1172/jci31622] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Accepted: 04/17/2007] [Indexed: 11/17/2022] Open
Abstract
Immunophilin FKBP52 serves as a cochaperone to govern normal progesterone (P(4)) receptor (PR) function. Using Fkbp52(-/-) mice, we show intriguing aspects of uterine P(4)/PR signaling during pregnancy. Implantation failure is the major phenotype found in these null females, which is conserved on both C57BL6/129 and CD1 backgrounds. However, P(4) supplementation rescued implantation and subsequent decidualization in CD1, but not C57BL6/129, null females. Surprisingly, experimentally induced decidualization in the absence of blastocysts failed in Fkbp52(-/-) mice on either background even with P(4) supplementation, suggesting that embryonic signals complement uterine signaling for this event. Another interesting finding was that while P(4) at higher than normal pregnancy levels conferred PR signaling sufficient for implantation in CD1 null females, these levels were inefficient in maintaining pregnancy to full term. However, elevating P(4) levels further restored PR signaling to a level optimal for successful term pregnancy with normal litter size. Collectively, the results show that the indispensability of FKBP52 in uterine P(4)/PR signaling is a function of genetic disparity and is pregnancy stage specific. Since there is evidence for a correlation between P(4) supplementation and reduced risks of P(4)-resistant recurrent miscarriages and remission of endometriosis, these findings have clinical implications for genetically diverse populations of women.
Collapse
Affiliation(s)
- Susanne Tranguch
- Department of Pediatrics, Department of Cell and Developmental Biology, Department of Pharmacology, and Division of Reproductive and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, Arizona, USA
| | - Haibin Wang
- Department of Pediatrics, Department of Cell and Developmental Biology, Department of Pharmacology, and Division of Reproductive and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, Arizona, USA
| | - Takiko Daikoku
- Department of Pediatrics, Department of Cell and Developmental Biology, Department of Pharmacology, and Division of Reproductive and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, Arizona, USA
| | - Huirong Xie
- Department of Pediatrics, Department of Cell and Developmental Biology, Department of Pharmacology, and Division of Reproductive and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, Arizona, USA
| | - David F. Smith
- Department of Pediatrics, Department of Cell and Developmental Biology, Department of Pharmacology, and Division of Reproductive and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, Arizona, USA
| | - Sudhansu K. Dey
- Department of Pediatrics, Department of Cell and Developmental Biology, Department of Pharmacology, and Division of Reproductive and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, Arizona, USA
| |
Collapse
|
48
|
Wang CK, Robinson RS, Flint APF, Mann GE. Quantitative analysis of changes in endometrial gland morphology during the bovine oestrous cycle and their association with progesterone levels. Reproduction 2007; 134:365-71. [PMID: 17660245 DOI: 10.1530/rep-06-0133] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
This study describes a digital technique for uterine morphometry and its application to endometrial structure during the bovine oestrous cycle. Neither the number nor the size of uterine gland ducts changed during the cycle but a reduction in total endometrial area from days 0 to 8 after oestrus led to an increase in the proportion of the endometrium occupied by gland ducts (gland duct density). This effect on day 8 was maintained to day 16. When endometrial morphology was related to circulating progesterone concentrations on days 5 and 8 of the luteal phase, no relationships were found on day 5, but on day 8, a high progesterone concentration was associated with an increased number of gland ducts. Furthermore, in animals slaughtered on day 8, a high progesterone concentration on day 5 was associated with decreased gland duct size, though a simultaneous decrease in endometrial area led to an increase in gland duct density. The results suggest that contrary to expectation, endometrial glands do not grow and regress during the oestrous cycle, although cyclic changes in endometrial area controlled by progesterone lead to changes in gland duct density.
Collapse
Affiliation(s)
- C K Wang
- Division of Animal Physiology, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK
| | | | | | | |
Collapse
|
49
|
Gray CA, Abbey CA, Beremand PD, Choi Y, Farmer JL, Adelson DL, Thomas TL, Bazer FW, Spencer TE. Identification of endometrial genes regulated by early pregnancy, progesterone, and interferon tau in the ovine uterus. Biol Reprod 2005; 74:383-94. [PMID: 16251498 DOI: 10.1095/biolreprod.105.046656] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
During early pregnancy in ruminants, progesterone (P4) from the corpus luteum and interferon tau (IFNT) from the conceptus act on the endometrium to regulate genes important for uterine receptivity and conceptus growth. The use of the uterine gland knockout (UGKO) ewe has demonstrated the critical role of epithelial secretions in regulation of conceptus survival and growth. A custom ovine cDNA array was used to identify alterations in gene expression of endometria from Day 14 cyclic, pregnant, and UGKO ewes (study 1) and from cyclic ewes treated with P4 or P4 with ZK 136,317 antiprogestin and control proteins or IFNT (study 2). In study 1, expression of 47 genes was more than 2-fold different between Day 14 pregnant and cyclic endometria, whereas 23 genes was different between Day 14 cyclic and UGKO endometria. In study 2, 70 genes were different due to P4 alone, 74 genes were affected by IFNT in a P4-dependent manner, and 180 genes were regulated by IFNT in a P4-independent manner. In each study, an approximately equal number of genes were found to be activated or repressed in each group. Endometrial genes increased by pregnancy and P4 and/or IFNT include B2M, CTSL, CXCL10, G1P3, GRP, IFI27, IFIT1, IFITM3, LGALS15, MX1, POSTN, RSAD2, and STAT5A. Transcripts decreased by pregnancy and P4 and/or IFNT include COL3A1, LUM, PTMA, PUM1, RPL9, SPARC, and VIM. Identification and analysis of these hormonally responsive genes will help define endometrial pathways critical for uterine support of peri-implantation conceptus survival, growth, and implantation.
Collapse
Affiliation(s)
- C Allison Gray
- Center for Animal Biotechnology and Genomics, Texas A&M University, College Station, Texas 77843, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hayashi K, Spencer TE. Estrogen disruption of neonatal ovine uterine development: effects on gene expression assessed by suppression subtraction hybridization. Biol Reprod 2005; 73:752-60. [PMID: 15972882 DOI: 10.1095/biolreprod.105.042812] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Inappropriate exposure of neonatal sheep to estrogen during critical developmental periods inhibits or retards endometrial gland morphogenesis and reduces uterine growth. Studies were conducted to identify mechanisms mediating estrogen disruption of neonatal ovine uterine development by analysis of candidate growth factor systems and using suppression subtraction hybridization (SSH). In study 1, sheep were exposed either to corn oil as a control or to estradiol valerate (EV) from birth to Postnatal Day (PND) 14, which ablated endometrial gland development. Estradiol valerate decreased uterine FGF7 (fibroblast growth factor 7) and MET (hepatocyte growth factor receptor) expression and increased INHBA (inhibin betaA). The SSH identified a number of genes responsive to EV, which included GSTM3 (glutathione S-transferase), IDH1 (cytosolic NADP-isocitrate dehydrogenase), PECI (peroxisomal D(3),D(2)-enoyl-coenzyme A isomerase), OAS1 (2',5'-oligoadenylate 40/46-kDa synthetase), IGFBP3 (insulin-like growth factor-binding protein-3), TEGT (testis-enhanced gene transcript), CXCL10 (interferon-gamma-inducible protein 10), and IGLV (immunoglobulin V). These mRNAs were expressed predominantly in the endometrial epithelia (GSTM3, IDH1, PEC1, OAS1, and TEGT), stroma (IGFBP3), or immune cells (CXCL10 and IGLV). In study 2, effects of estrogen exposure on uterine gene expression were determined during three different critical developmental periods (PNDs 0-14, 14- 28, and 42-56). Estrogen exposure decreased expression of the SSH-identified genes, particularly those from PNDs 0-14. These studies suggest that estrogen disruption of postnatal uterine development involves period-specific effects on expression of genes predominantly in the endometrial epithelium. The SSH-identified, estrogen-disrupted genes represent new candidate regulators of postnatal endometrial adenogenesis.
Collapse
Affiliation(s)
- Kanako Hayashi
- Center for Animal Biotechnology and Genomics and Department of Animal Science, Texas A&M University, College Station, 77843-2471, USA
| | | |
Collapse
|