1
|
Dominguez EM, Moreno-Irusta A, Scott RL, Iqbal K, Soares MJ. TFAP2C is a key regulator of intrauterine trophoblast cell invasion and deep hemochorial placentation. JCI Insight 2024; 10:e186471. [PMID: 39625795 PMCID: PMC11790029 DOI: 10.1172/jci.insight.186471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/26/2024] [Indexed: 12/11/2024] Open
Abstract
Transcription factor AP-2 gamma (TFAP2C) has been identified as a key regulator of the trophoblast cell lineage and hemochorial placentation. The rat possesses deep placentation characterized by extensive intrauterine trophoblast cell invasion, which resembles human placentation. Tfap2c is expressed in multiple trophoblast cell lineages, including invasive trophoblast cells situated within the uterine-placental interface of the rat placentation site. Global genome editing was used to explore the biology of Tfap2c in rat placenta development. Homozygous global disruption of Tfap2c resulted in prenatal lethality. Heterozygous global disruption of Tfap2c was associated with diminished invasive trophoblast cell infiltration into the uterus. The role of TFAP2C in the invasive trophoblast cell lineage was explored using Cre-lox conditional mutagenesis. Invasive trophoblast cell-specific disruption of Tfap2c resulted in inhibition of intrauterine trophoblast cell invasion and intrauterine and postnatal growth restriction. The invasive trophoblast cell lineage was not impaired following conditional monoallelic disruption of Tfap2c. In summary, TFAP2C contributes to the progression of distinct stages of placental development. TFAP2C is a driver of early events in trophoblast cell development and reappears later in gestation as an essential regulator of the invasive trophoblast cell lineage. A subset of TFAP2C actions on trophoblast cells are dependent on gene dosage.
Collapse
Affiliation(s)
- Esteban M. Dominguez
- Institute for Reproductive and Developmental Sciences, Department of Pathology & Laboratory Medicine, and
| | - Ayelen Moreno-Irusta
- Institute for Reproductive and Developmental Sciences, Department of Pathology & Laboratory Medicine, and
| | - Regan L. Scott
- Institute for Reproductive and Developmental Sciences, Department of Pathology & Laboratory Medicine, and
| | - Khursheed Iqbal
- Institute for Reproductive and Developmental Sciences, Department of Pathology & Laboratory Medicine, and
| | - Michael J. Soares
- Institute for Reproductive and Developmental Sciences, Department of Pathology & Laboratory Medicine, and
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, Kansas, USA
- Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy, Kansas City, Missouri, USA
| |
Collapse
|
2
|
Dominguez EM, Moreno-Irusta A, Scott RL, Iqbal K, Soares MJ. TFAP2C is a key regulator of intrauterine trophoblast cell invasion and deep hemochorial placentation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.31.621324. [PMID: 39554130 PMCID: PMC11565979 DOI: 10.1101/2024.10.31.621324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Transcription factor AP-2 gamma ( TFAP2C ) has been identified as a key regulator of the trophoblast cell lineage and hemochorial placentation. The rat possesses deep placentation characterized by extensive intrauterine trophoblast cell invasion, which resembles human placentation. Tfap2c is expressed in multiple trophoblast cell lineages, including invasive trophoblast cells situated within the uterine-placental interface of the rat placentation site. Global genome-editing was used to explore the biology of Tfap2c in rat placenta development. Homozygous global disruption of Tfap2c resulted in prenatal lethality. Heterozygous global disruption of Tfap2c was associated with diminished invasive trophoblast cell infiltration into the uterus. The role of TFAP2C in the invasive trophoblast cell lineage was explored using Cre-lox conditional mutagenesis. Invasive trophoblast cell-specific disruption of Tfap2c resulted in inhibition of intrauterine trophoblast cell invasion and intrauterine and postnatal growth restriction. The invasive trophoblast cell lineage was not impaired following conditional monoallelic disruption of Tfap2c . In summary, TFAP2C contributes to the progression of distinct stages of placental development. TFAP2C is a driver of early events in trophoblast cell development and reappears later in gestation as an essential regulator of the invasive trophoblast cell lineage. A subset of TFAP2C actions on trophoblast cells are dependent on gene dosage.
Collapse
|
3
|
Home P, Ghosh A, Kumar RP, Ray S, Gunewardena S, Kumar R, Dasgupta P, Roy N, Saha A, Ouseph MM, Leone GW, Paul S. A Single Trophoblast Layer Acts as the Gatekeeper at the Endothelial-Hematopoietic Crossroad in the Placenta. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.12.603303. [PMID: 39071312 PMCID: PMC11275844 DOI: 10.1101/2024.07.12.603303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
During embryonic development the placental vasculature acts as a major hematopoietic niche, where endothelial to hematopoietic transition ensures emergence of hematopoietic stem cells (HSCs). However, the molecular mechanisms that regulate the placental hematoendothelial niche are poorly understood. Using a parietal trophoblast giant cell (TGC)-specific knockout mouse model and single-cell RNA-sequencing, we show that the paracrine factors secreted by the TGCs are critical in the development of this niche. Disruptions in the TGC-specific paracrine signaling leads to the loss of HSC population and the concomitant expansion of a KDR+/DLL4+/PROM1+ hematoendothelial cell-population in the placenta. Combining single-cell transcriptomics and receptor-ligand pair analyses, we also define the parietal TGC-dependent paracrine signaling network and identify Integrin signaling as a fundamental regulator of this process. Our study elucidates novel mechanisms by which non-autonomous signaling from the primary parietal TGCs maintain the delicate placental hematopoietic-angiogenic balance and ensures embryonic and extraembryonic development.
Collapse
Affiliation(s)
- Pratik Home
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Present address: XenoTech, A BioIVT Company, 1101 W Cambridge Cir Dr, Kansas City, KS 66103
| | - Ananya Ghosh
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Present address: Department of Urology, University of California San Francisco, 35, Medical 12 Center Way, San Francisco, CA 94143
| | - Ram Parikshan Kumar
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Institute for Reproductive Health and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Soma Ray
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Rajnish Kumar
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Purbasa Dasgupta
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Namrata Roy
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Abhik Saha
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Madhu M. Ouseph
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065
| | - Gustavo W. Leone
- Department of Biochemistry, Medical College of Wisconsin, WI 53226, USA
| | - Soumen Paul
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Institute for Reproductive Health and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
4
|
Sairenji TJ, Masuda S, Higuchi Y, Miyazaki M, Yajima H, Kwan Ee O, Fujiwara Y, Araki T, Shimokawa N, Koibuchi N. Plasma prolactin axis shift from placental to pituitary origin in late prepartum mice. Endocr J 2024; 71:661-674. [PMID: 38749736 DOI: 10.1507/endocrj.ej23-0724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/17/2024] Open
Abstract
The placenta secretes a prolactin (PRL)-like hormone PRL3B1 (placental lactogen II), a luteotropic hormone essential for maintaining pregnancy until labor in mice. A report from 1984 examined the secretion pattern of PRL3B1 in prepartum mice. In the current study, we found contradictory findings in the secretion pattern that invalidate the previous report. By measuring maternal plasma PRL3B1 and PRL every 4 hrs from gestational day 17 (G17), we newly discovered that maternal plasma PRL3B1 levels decrease rapidly in prepartum C57BL/6 mice. Interestingly, the onset of this decline coincided with the PRL surge at G18, demonstrating a plasma prolactin axis shift from placental to pituitary origin. We also found that maternal plasma progesterone regression precedes the onset of the PRL shift. The level of Prl3b1 mRNA was determined by RT-qPCR in the placenta and remained stable until parturition, implying that PRL3B1 peptide production or secretion was suppressed. We hypothesized that production of the PRL family, the 25 paralogous PRL proteins exclusively expressed in mice placenta, would decrease alongside PRL3B1 during this period. To investigate this hypothesis and to seek proteomic changes, we performed a shotgun proteome analysis of the placental tissue using data-independent acquisition mass spectrometry (DIA-MS). Up to 5,891 proteins were identified, including 17 PRL family members. Relative quantitative analysis between embryonic day 17 (E17) and E18 placentas showed no significant difference in the expression of PRL3B1 and most PRL family members except PRL7C1. These results suggest that PRL3B1 secretion from the placenta is suppressed at G18 (E18).
Collapse
Affiliation(s)
- Taku James Sairenji
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Shinnosuke Masuda
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
- Laboratory of Epigenetics and Metabolism, Institute of Molecular and Cellular Regulations, Gunma 371-8512, Japan
| | - Yuya Higuchi
- Department of Clinical Pharmacology and Therapeutics, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Mitsue Miyazaki
- Department of Clinical Pharmacology and Therapeutics, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
- Department of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Sciences, Aomori 036-8564, Japan
- Department of Nutrition, Takasaki University of Health and Welfare, Gunma 370-0033, Japan
| | - Hiroyuki Yajima
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Oh Kwan Ee
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Yuki Fujiwara
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Takuya Araki
- Department of Clinical Pharmacology and Therapeutics, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Noriaki Shimokawa
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
- Department of Nutrition, Takasaki University of Health and Welfare, Gunma 370-0033, Japan
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| |
Collapse
|
5
|
Kuraku S, Kaiya H, Tanaka T, Hyodo S. Evolution of Vertebrate Hormones and Their Receptors: Insights from Non-Osteichthyan Genomes. Annu Rev Anim Biosci 2023; 11:163-182. [PMID: 36400012 DOI: 10.1146/annurev-animal-050922-071351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Homeostatic control and reproductive functions of humans are regulated at the molecular levels largely by peptide hormones secreted from endocrine and/or neuroendocrine cells in the central nervous system and peripheral organs. Homologs of those hormones and their receptors function similarly in many vertebrate species distantly related to humans, but the evolutionary history of the endocrine system involving those factors has been obscured by the scarcity of genome DNA sequence information of some taxa that potentially contain their orthologs. Focusing on non-osteichthyan vertebrates, namely jawless and cartilaginous fishes, this article illustrates how investigating genome sequence information assists our understanding of the diversification of vertebrate gene repertoires in four broad themes: (a) the presence or absence of genes, (b) multiplication and maintenance of paralogs, (c) differential fates of duplicated paralogs, and (d) the evolutionary timing of gene origins.
Collapse
Affiliation(s)
- Shigehiro Kuraku
- Molecular Life History Laboratory, Department of Genomics and Evolutionary Biology, National Institute of Genetics, Mishima, Japan; .,Department of Genetics, Sokendai (Graduate University for Advanced Studies), Mishima, Japan.,Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, Japan
| | - Hiroyuki Kaiya
- Grandsoul Research Institute of Immunology, Inc., Uda, Japan
| | - Tomohiro Tanaka
- Department of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Susumu Hyodo
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Japan
| |
Collapse
|
6
|
Carter AM. Evolution of Placental Hormones: Implications for Animal Models. Front Endocrinol (Lausanne) 2022; 13:891927. [PMID: 35692413 PMCID: PMC9176407 DOI: 10.3389/fendo.2022.891927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/11/2022] [Indexed: 11/15/2022] Open
Abstract
Human placenta secretes a variety of hormones, some of them in large amounts. Their effects on maternal physiology, including the immune system, are poorly understood. Not one of the protein hormones specific to human placenta occurs outside primates. Instead, laboratory and domesticated species have their own sets of placental hormones. There are nonetheless several examples of convergent evolution. Thus, horse and human have chorionic gonadotrophins with similar functions whilst pregnancy-specific glycoproteins have evolved in primates, rodents, horses, and some bats, perhaps to support invasive placentation. Placental lactogens occur in rodents and ruminants as well as primates though evolved through duplication of different genes and with functions that only partially overlap. There are also placental hormones, such as the pregnancy-associated glycoproteins of ruminants, that have no equivalent in human gestation. This review focusses on the evolution of placental hormones involved in recognition and maintenance of pregnancy, in maternal adaptations to pregnancy and lactation, and in facilitating immune tolerance of the fetal semiallograft. The contention is that knowledge gained from laboratory and domesticated mammals can translate to a better understanding of human placental endocrinology, but only if viewed in an evolutionary context.
Collapse
Affiliation(s)
- Anthony M. Carter
- Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
7
|
Kang J, Liu Y, Zhang Y, Yan W, Wu Y, Su R. The Influence of the Prolactins on the Development of the Uterus in Neonatal Mice. Front Vet Sci 2022; 9:818827. [PMID: 35252420 PMCID: PMC8891943 DOI: 10.3389/fvets.2022.818827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
The endometrial gland is one of the most important components of the mammalian uterus. However, few studies have been conducted on the regulatory mechanisms of adenogenesis during the development of endometrium. In the present study, we detected the genes expression of 35 different prolactin family members (PRLs) together with the prolactin receptor (PRL-R) in the endometrium of neonatal mice along with the adenogenesis process, to address which prolactin-like genes play a key role during gland development in mice. We found that: (1) The expression of Prl1a1, Prl3d1, Prl5a1, Prl7a1, Prl7a2, Prl7d1, Prl8a6, Prl8a8, and Prl8a9 genes were significantly increased along with the development of uterine glands. Prl7c1 and Prl8a1 were observably up-regulated on Postnatal day 5 (PND5) when the uterine glandular bud invagination begins. Prl3a1, Prl3b1, and Prl7b1 suddenly increased significantly on PND9. But, Prl3c1 and Prl8a2 were markedly down-regulated on PND5 and the expression of Prl6a1 and Prlr were stable extremely. (2) After continuous injection of Progesterone (P4), a well-known method to suppress the endometrial adenogenesis, the expression of Prl1a1, Prl3d1, Prl5a1, Prl7a1, Prl7a2, Prl7d1, Prl8a6, Prl8a8, Prl8a9, and Prlr were suppressed on PND7. And on PND9, Prl1a1, Prl3d1, Prl8a6, Prl8a8, and Prl8a9 were significantly inhibited. (3) Further analysis of the epithelial and stroma showed that these PRLs were mainly expressed in the endometrial stroma of neonatal mice. Our results indicate that multiple PRLs are involved in uterine development and endometrial adenogenesis. Continued progesterone therapy may alter the expression pattern of these PRLs in endometrial stromal cells, thereby altering the interaction and communication between stroma and epithelium, and ultimately leading to complete suppression of endometrial adenogenesis.
Collapse
|
8
|
Effects of Maternal Diabetes and Diet on Gene Expression in the Murine Placenta. Genes (Basel) 2022; 13:genes13010130. [PMID: 35052470 PMCID: PMC8775503 DOI: 10.3390/genes13010130] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/08/2022] [Accepted: 01/10/2022] [Indexed: 11/16/2022] Open
Abstract
Adverse exposures during pregnancy have been shown to contribute to susceptibility for chronic diseases in offspring. Maternal diabetes during pregnancy is associated with higher risk of pregnancy complications, structural birth defects, and cardiometabolic health impairments later in life. We showed previously in a mouse model that the placenta is smaller in diabetic pregnancies, with reduced size of the junctional zone and labyrinth. In addition, cell migration is impaired, resulting in ectopic accumulation of spongiotrophoblasts within the labyrinth. The present study had the goal to identify the mechanisms underlying the growth defects and trophoblast migration abnormalities. Based upon gene expression assays of 47 candidate genes, we were able to attribute the reduced growth of diabetic placenta to alterations in the Insulin growth factor and Serotonin signaling pathways, and provide evidence for Prostaglandin signaling deficiencies as the possible cause for abnormal trophoblast migration. Furthermore, our results reinforce the notion that the exposure to maternal diabetes has particularly pronounced effects on gene expression at midgestation time points. An implication of these findings is that mechanisms underlying developmental programming act early in pregnancy, during placenta morphogenesis, and before the conceptus switches from histiotrophic to hemotrophic nutrition.
Collapse
|
9
|
Martínez-Alarcón O, García-López G, Guerra-Mora JR, Molina-Hernández A, Diaz-Martínez NE, Portillo W, Díaz NF. Prolactin from Pluripotency to Central Nervous System Development. Neuroendocrinology 2022; 112:201-214. [PMID: 33934093 DOI: 10.1159/000516939] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/30/2021] [Indexed: 11/19/2022]
Abstract
Prolactin (PRL) is a versatile hormone that exerts more than 300 functions in vertebrates, mainly associated with physiological effects in adult animals. Although the process that regulates early development is poorly understood, evidence suggests a role of PRL in the early embryonic development regarding pluripotency and nervous system development. Thus, PRL could be a crucial regulator in oocyte preimplantation and maturation as well as during diapause, a reversible state of blastocyst development arrest that shares metabolic, transcriptomic, and proteomic similarities with pluripotent stem cells in the naïve state. Thus, we analyzed the role of the hormone during those processes, which involve the regulation of its receptor and several signaling cascades (Jak/Mapk, Jak/Stat, and PI3k/Akt), resulting in either a plethora of physiological actions or their dysregulation, a factor in developmental disorders. Finally, we propose models to improve the knowledge on PRL function during early development.
Collapse
Affiliation(s)
- Omar Martínez-Alarcón
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| | - Guadalupe García-López
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| | - José Raúl Guerra-Mora
- Departamento de Neurociencias, Instituto Nacional de Cancerología, Ciudad de México, Mexico
- Departamento de Cirugia Experimental, Instituto Nacional de Nutrición, Ciudad de México, Mexico
| | - Anayansi Molina-Hernández
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| | - Néstor Emmanuel Diaz-Martínez
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica CONACYT, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, Guadalajara, Mexico
| | - Wendy Portillo
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, UNAM, Quéretaro, Mexico
| | - Néstor Fabián Díaz
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| |
Collapse
|
10
|
Wallis M. Do some viruses use growth hormone, prolactin and their receptors to facilitate entry into cells?: Episodic evolution of hormones and receptors suggests host-virus arms races; related placental lactogens may provide protective viral decoys. Bioessays 2021; 43:e2000268. [PMID: 33521987 DOI: 10.1002/bies.202000268] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/30/2020] [Accepted: 12/30/2020] [Indexed: 12/14/2022]
Abstract
The molecular evolution of pituitary growth hormone and prolactin in mammals shows two unusual features: episodes of markedly accelerated evolution and, in some species, complex families of related proteins expressed in placenta and resulting from multiple gene duplications. Explanations of these phenomena in terms of physiological adaptations seem unconvincing. Here, I propose an alternative explanation, namely that these evolutionary features reflect the use of the hormones (and their receptors) as viral receptors. Episodes of rapid evolution can then be explained as due to "arms races" in which changes in the hormone lead to reduced interaction with the virus, and subsequent changes in the virus counteract this. Placental paralogues of the hormones could provide decoys that bind viruses, and protect the foetus against infection. The hypothesis implies that the extensive changes introduced into growth hormone, prolactin and their receptors during the course of mammalian evolution reflect viral interactions, not endocrine adaptations.
Collapse
Affiliation(s)
- Michael Wallis
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, UK
| |
Collapse
|
11
|
Short prolactin isoforms are expressed in photoreceptors of canine retinas undergoing retinal degeneration. Sci Rep 2021; 11:460. [PMID: 33432105 PMCID: PMC7801730 DOI: 10.1038/s41598-020-80691-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/21/2020] [Indexed: 12/18/2022] Open
Abstract
Prolactin (PRL) hormone functions as a pleiotropic cytokine with a protective role in the retina. We recently identified by transcriptome profiling that PRL is one of the most highly upregulated mRNAs in the retinas of mutant rcd1 (PDE6B) and xlpra2 (RPGR) dogs at advanced stages of photoreceptor disease. In the present study, we have identified the expression of a short PRL isoform that lacks exon 1 in canine retinas and analyzed the time-course of expression and localization of this isoform in the retinas of these two models. Using laser capture microdissection to isolate RNA from each of the retinal cellular layers, we found by qPCR that this short PRL isoform is expressed in photoreceptors of degenerating retinas. We confirmed by in situ hybridization that its expression is localized to the outer nuclear layer and begins shortly after the onset of disease at the time of peak photoreceptor cell death in both models. PRL protein was also detected only in mutant dog retinas. Our results call for further investigations into the role of this novel PRL isoform in retinal degeneration.
Collapse
|
12
|
Pérez-Roque L, Núñez-Gómez E, Rodríguez-Barbero A, Bernabéu C, López-Novoa JM, Pericacho M. Pregnancy-Induced High Plasma Levels of Soluble Endoglin in Mice Lead to Preeclampsia Symptoms and Placental Abnormalities. Int J Mol Sci 2020; 22:ijms22010165. [PMID: 33375253 PMCID: PMC7795873 DOI: 10.3390/ijms22010165] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/17/2022] Open
Abstract
Preeclampsia is a pregnancy-specific disease of high prevalence characterized by the onset of hypertension, among other maternal or fetal signs. Its etiopathogenesis remains elusive, but it is widely accepted that abnormal placentation results in the release of soluble factors that cause the clinical manifestations of the disease. An increased level of soluble endoglin (sEng) in plasma has been proposed to be an early diagnostic and prognostic biomarker of this disease. A pathogenic function of sEng involving hypertension has also been reported in several animal models with high levels of plasma sEng not directly dependent on pregnancy. The aim of this work was to study the functional effect of high plasma levels of sEng in the pathophysiology of preeclampsia in a model of pregnant mice, in which the levels of sEng in the maternal blood during pregnancy replicate the conditions of human preeclampsia. Our results show that wild type pregnant mice carrying human sEng-expressing transgenic fetuses (fWT(hsEng+)) present high plasma levels of sEng with a timing profile similar to that of human preeclampsia. High plasma levels of human sEng (hsEng) are associated with hypertension, proteinuria, fetal growth restriction, and the release of soluble factors to maternal plasma. In addition, fWT(hsEng+) mice also present placental alterations comparable to those caused by the poor remodeling of the spiral arteries characteristic of preeclampsia. In vitro and ex vivo experiments, performed in a human trophoblast cell line and human placental explants, show that sEng interferes with trophoblast invasion and the associated pseudovasculogenesis, a process by which cytotrophoblasts switch from an epithelial to an endothelial phenotype, both events being related to remodeling of the spiral arteries. Our findings provide a novel and useful animal model for future research in preeclampsia and reveal a much more relevant role of sEng in preeclampsia than initially proposed.
Collapse
Affiliation(s)
- Lucía Pérez-Roque
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (L.P.-R.); (E.N.-G.); (A.R.-B.); (J.M.L.-N.)
- Renal and Cardiovascular Physiopathology Unit, Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain
| | - Elena Núñez-Gómez
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (L.P.-R.); (E.N.-G.); (A.R.-B.); (J.M.L.-N.)
- Renal and Cardiovascular Physiopathology Unit, Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain
| | - Alicia Rodríguez-Barbero
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (L.P.-R.); (E.N.-G.); (A.R.-B.); (J.M.L.-N.)
- Renal and Cardiovascular Physiopathology Unit, Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain
| | - Carmelo Bernabéu
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040 Madrid, Spain;
| | - José M. López-Novoa
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (L.P.-R.); (E.N.-G.); (A.R.-B.); (J.M.L.-N.)
- Renal and Cardiovascular Physiopathology Unit, Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain
| | - Miguel Pericacho
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (L.P.-R.); (E.N.-G.); (A.R.-B.); (J.M.L.-N.)
- Renal and Cardiovascular Physiopathology Unit, Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain
- Correspondence:
| |
Collapse
|
13
|
Cattini PA, Jin Y, Jarmasz JS, Noorjahan N, Bock ME. Obesity and regulation of human placental lactogen production in pregnancy. J Neuroendocrinol 2020; 32:e12859. [PMID: 32500948 DOI: 10.1111/jne.12859] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/19/2020] [Accepted: 04/21/2020] [Indexed: 12/16/2022]
Abstract
The four genes coding for placental members of the human (h) growth hormone (GH) family include two that code independently for placental lactogen (PL), also known as chorionic somatomammotrophin hormone, one that codes for placental growth hormone (PGH) and a pseudogene for which RNA but no protein product is reported. These genes are expressed preferentially in the villus syncytiotrophoblast of the placenta in pregnancy. In higher primates, the placental members, including hPL and PGH, are the result of multiple duplication events of the GH gene. This contrasts with rodents and ruminants, where PLs result from duplication of the prolactin (PRL) gene. Thus, unlike their mouse counterparts, the hPL and PGH hormones bind both lactogenic and somatogenic receptors with varying affinity. Roles influenced by nutrient availability in both metabolic control in pregnancy and maternal behaviour are supported. However, the effect maternal obesity has on the activation of placental members of the hGH gene family, particularly the expression and function of those genes, is poorly understood. Evidence from partially humanised hGH/PL transgenic mice indicates that both the remote upstream hPL locus control region (LCR) and more gene-related regulatory regions are required for placental expression in vivo. Furthermore, a specific pattern of interactions between the LCR and hPL gene promoter regions is detected in term placenta chromatin from women with a normal body mass index (BMI) in the range 18.5-25 kg m-2 by chromosome conformation capture assay. This pattern is disrupted with maternal obesity (class II BMI > 35 kg m-2 ) and associated with a > 40% decrease in term hPL RNA levels, as well as serum hPL but not PRL levels, during pregnancy. The relative importance of the chromosomal architecture and predicted properties for transcription factor participation in terms of hPL production and response to obesity are considered, based on comparison with components required for efficient human pituitary GH gene expression.
Collapse
Affiliation(s)
- Peter A Cattini
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - Yan Jin
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - Jessica S Jarmasz
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - Noshin Noorjahan
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - Margaret E Bock
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
14
|
Mouillet JF, Goff J, Sadovsky E, Sun H, Parks T, Chu T, Sadovsky Y. Transgenic expression of human C19MC miRNAs impacts placental morphogenesis. Placenta 2020; 101:208-214. [PMID: 33017713 DOI: 10.1016/j.placenta.2020.09.069] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 08/28/2020] [Accepted: 09/28/2020] [Indexed: 12/26/2022]
Abstract
INTRODUCTION The chromosome 19 miRNA cluster (C19MC) encodes a large family of microRNAs (miRNAs) that are abundantly expressed in the placenta of higher primates and also in certain cancers. In the placenta, miRNAs from this cluster account for nearly 40% of all miRNAs present in trophoblasts. However, the function of these miRNAs in the placenta remains poorly understood. Recent observations reveal a role for these miRNAs in cell migration, and suggest that they are involved in the development and function of the human placenta. Here, we examine the placenta in transgenic mice expressing the human C19MC miRNAs. METHODS We produced transgenic mice using pronuclear microinjection of a bacterial artificial chromosome plasmid carrying the entire human C19MC locus and derived a homozygous line using crossbreeding. We performed morphological characterization and profiled gene expression changes in the placentas of the transgenic mice. RESULTS C19MC transgenic mice delivered on time with no gross malformations. The placentas of transgenic mice expressed C19MC miRNAs and were larger than wild type placentas. Histologically, we found that the transgenic placenta exhibited projections of spongiotrophoblasts that penetrated deep into the labyrinth. Gene expression analysis revealed alterations in the expression of several genes involved in cell migration, with evidence of enhanced cell proliferation. DISCUSSION Mice that were humanized for transgenically overexpressed C19MC miRNAs exhibit enlarged placentas with aberrant delineation of cell layers. The observed phenotype and the related gene expression changes suggest disrupted migration of placental cell subpopulations.
Collapse
Affiliation(s)
- Jean-Francois Mouillet
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Julie Goff
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Elena Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Huijie Sun
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tony Parks
- Department of Laboratory Medicine and Pathobiology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Tianjiao Chu
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
15
|
Kang ML, Goo JTT, Lee DJK. CHOP Protocol: streamlining access to definitive intervention for major trauma victims. Singapore Med J 2020; 62:620-622. [PMID: 32728086 DOI: 10.11622/smedj.2020113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Min Li Kang
- Department of Surgery, Khoo Teck Puat Hospital, Singapore
| | | | | |
Collapse
|
16
|
Gardner S, Grindstaff JL, Campbell P. Placental genotype affects early postpartum maternal behaviour. ROYAL SOCIETY OPEN SCIENCE 2019; 6:190732. [PMID: 31598302 PMCID: PMC6774950 DOI: 10.1098/rsos.190732] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 08/20/2019] [Indexed: 05/06/2023]
Abstract
The mammalian placenta is a source of endocrine signals that prime the onset of maternal care at parturition. While consequences of placental dysfunction for offspring growth are well defined, how altered placental signalling might affect maternal behaviour is unstudied in a natural system. In the cross between sympatric mouse species, Mus musculus domesticus and Mus spretus, hybrid placentas are undersized and show misexpression of genes critical to placental endocrine function. Using this cross, we quantified the effects of placental dysregulation on maternal and anxiety-like behaviours in mice that differed only in pregnancy type. Relative to mothers of conspecific litters, females exposed to hybrid placentas did not differ in anxiety-like behaviours but were slower to retrieve 1-day-old pups and spent less time in the nest on the night following parturition. Early deficits in maternal responsiveness were not explained by reduced ultrasonic vocalization production in hybrid pups and there was no effect of pup genotype on measures of maternal behaviour and physiology collected after the first 24 h postpartum. These results suggest that placental dysregulation leads to poor maternal priming, the effect of which is alleviated by continued exposure to pups. This study provides new insight into the placental mediation of mother-offspring interactions.
Collapse
Affiliation(s)
- Sarah Gardner
- Department of Integrative Biology, Oklahoma State University, Stillwater, OK, USA
- Department of Evolution, Ecology, and Organismal Biology, University of California Riverside, Riverside, CA, USA
- Author for correspondence: Sarah Gardner e-mail:
| | | | - Polly Campbell
- Department of Integrative Biology, Oklahoma State University, Stillwater, OK, USA
- Department of Evolution, Ecology, and Organismal Biology, University of California Riverside, Riverside, CA, USA
| |
Collapse
|
17
|
Soares MJ, Varberg KM, Iqbal K. Hemochorial placentation: development, function, and adaptations. Biol Reprod 2019; 99:196-211. [PMID: 29481584 DOI: 10.1093/biolre/ioy049] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/21/2018] [Indexed: 11/12/2022] Open
Abstract
Placentation is a reproductive adaptation that permits fetal growth and development within the protected confines of the female reproductive tract. Through this important role, the placenta also determines postnatal health and susceptibility to disease. The hemochorial placenta is a prominent feature in primate and rodent development. This manuscript provides an overview of the basics of hemochorial placental development and function, provides perspectives on major discoveries that have shaped placental research, and thoughts on strategies for future investigation.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas, USA and the Center for Perinatal Research, Children΄s Research Institute, Children΄s Mercy, Kansas City, Missouri, USA
| | - Kaela M Varberg
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
18
|
Napso T, Yong HEJ, Lopez-Tello J, Sferruzzi-Perri AN. The Role of Placental Hormones in Mediating Maternal Adaptations to Support Pregnancy and Lactation. Front Physiol 2018; 9:1091. [PMID: 30174608 PMCID: PMC6108594 DOI: 10.3389/fphys.2018.01091] [Citation(s) in RCA: 245] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 07/23/2018] [Indexed: 12/12/2022] Open
Abstract
During pregnancy, the mother must adapt her body systems to support nutrient and oxygen supply for growth of the baby in utero and during the subsequent lactation. These include changes in the cardiovascular, pulmonary, immune and metabolic systems of the mother. Failure to appropriately adjust maternal physiology to the pregnant state may result in pregnancy complications, including gestational diabetes and abnormal birth weight, which can further lead to a range of medically significant complications for the mother and baby. The placenta, which forms the functional interface separating the maternal and fetal circulations, is important for mediating adaptations in maternal physiology. It secretes a plethora of hormones into the maternal circulation which modulate her physiology and transfers the oxygen and nutrients available to the fetus for growth. Among these placental hormones, the prolactin-growth hormone family, steroids and neuropeptides play critical roles in driving maternal physiological adaptations during pregnancy. This review examines the changes that occur in maternal physiology in response to pregnancy and the significance of placental hormone production in mediating such changes.
Collapse
Affiliation(s)
- Tina Napso
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Hannah E J Yong
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Jorge Lopez-Tello
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
19
|
Ocampo Daza D, Larhammar D. Evolution of the growth hormone, prolactin, prolactin 2 and somatolactin family. Gen Comp Endocrinol 2018; 264:94-112. [PMID: 29339183 DOI: 10.1016/j.ygcen.2018.01.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/29/2017] [Accepted: 01/11/2018] [Indexed: 12/30/2022]
Abstract
Growth hormone (GH), prolactin (PRL), prolactin 2 (PRL2) and somatolactin (SL) belong to the same hormone family and have a wide repertoire of effects including development, osmoregulation, metabolism and stimulation of growth. Both the hormone and the receptor family have been proposed to have expanded by gene duplications in early vertebrate evolution. A key question is how hormone-receptor preferences have arisen among the duplicates. The first step to address this is to determine the time window for these duplications. Specifically, we aimed to see if duplications resulted from the two basal vertebrate tetraploidizations (1R and 2R). GH family genes from a broad range of vertebrate genomes were investigated using a combination of sequence-based phylogenetic analyses and comparisons of synteny. We conclude that the PRL and PRL2 genes arose from a common ancestor in 1R/2R, as shown by neighboring gene families. No other gene duplicates were preserved from these tetraploidization events. The ancestral genes that would give rise to GH and PRL/PRL2 arose from an earlier duplication; most likely a local gene duplication as they are syntenic in several species. Likewise, some evidence suggests that SL arose from a local duplication of an ancestral GH/SL gene in the same time window, explaining the lack of similarity in chromosomal neighbors to GH, PRL or PRL2. Thus, the basic triplet of ancestral GH, PRL/PRL2 and SL genes appear to be unexpectedly ancient. Following 1R/2R, only SL was duplicated in the teleost-specific tetraploidization 3R, resulting in SLa and SLb. These time windows contrast with our recent report that the corresponding receptor genes GHR and PRLR arose through a local duplication in jawed vertebrates and that both receptor genes duplicated further in 3R, which reveals a surprising asynchrony in hormone and receptor gene duplications.
Collapse
Affiliation(s)
- Daniel Ocampo Daza
- Department of Neuroscience, Science for Life Laboratory, Uppsala University, Box 593, SE-75124 Uppsala, Sweden.
| | - Dan Larhammar
- Department of Neuroscience, Science for Life Laboratory, Uppsala University, Box 593, SE-75124 Uppsala, Sweden
| |
Collapse
|
20
|
Soares MJ, Iqbal K, Kozai K. Hypoxia and Placental Development. Birth Defects Res 2018; 109:1309-1329. [PMID: 29105383 DOI: 10.1002/bdr2.1135] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/04/2017] [Indexed: 12/17/2022]
Abstract
Hemochorial placentation is orchestrated through highly regulated temporal and spatial decisions governing the fate of trophoblast stem/progenitor cells. Trophoblast cell acquisition of specializations facilitating invasion and uterine spiral artery remodeling is a labile process, sensitive to the environment, and represents a process that is vulnerable to dysmorphogenesis in pathologic states. Hypoxia is a signal guiding placental development, and molecular mechanisms directing cellular adaptations to low oxygen tension are integral to trophoblast cell differentiation and placentation. Hypoxia can also be used as an experimental tool to investigate regulatory processes controlling hemochorial placentation. These developmental processes are conserved in mouse, rat, and human placentation. Consequently, elements of these developmental events can be modeled and hypotheses tested in trophoblast stem cells and in genetically manipulated rodents. Hypoxia is also a consequence of a failed placenta, yielding pathologies that can adversely affect maternal adjustments to pregnancy, fetal health, and susceptibility to adult disease. The capacity of the placenta for adaptation to environmental challenges highlights the importance of its plasticity in safeguarding a healthy pregnancy. Birth Defects Research 109:1309-1329, 2017.© 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas.,Fetal Health Research, Children's Research Institute, Children's Mercy, Kansas City, Missouri
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Keisuke Kozai
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
21
|
|
22
|
Legg-St Pierre CB, Mackova M, Miskiewicz EI, Hemmings DG, Unniappan S, MacPhee DJ. Insulinotropic nucleobindin-2/nesfatin-1 is dynamically expressed in the haemochorial mouse and human placenta. Reprod Fertil Dev 2018; 30:519-532. [DOI: 10.1071/rd16486] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 07/29/2017] [Indexed: 12/12/2022] Open
Abstract
The placenta is the physiological bridge between mother and fetus and has life-sustaining functions during pregnancy, including metabolic regulation, fetal protection and hormone secretion. Nucleobindin-2 (NUCB2) is a calcium- and DNA-binding protein and precursor of nesfatin-1, a signalling peptide with multiple functions, including regulation of energy homeostasis and glucose transport. These are also key functions of the placenta, yet NUCB2/nesfatin-1 expression has never been comprehensively studied in this organ. In the present study, mouse placental samples from Embryonic Day (E) 7.5 to E17.5 and human chorionic villi from the first and second trimester, as well as term pregnancy, were analysed for NUCB2/nesfatin-1 expression by immunohistochemistry with an antiserum that recognised both NUCB2 and nesfatin-1. From E7.5 to E9.5, NUCB2/nesfatin-1 was expressed in the ectoplacental cone, then parietal trophoblast giant cells and early spongiotrophoblast. At E10.5–12.5, NUCB2/nesfatin-1 expression became detectable in the developing labyrinth. From E12.5 and onwards, NUCB2/nesfatin-1 was expressed in the glycogen trophoblast cells, as well as highly expressed in syncytiotrophoblast, sinusoidal trophoblast giant cells and fetal capillary endothelial cells of the labyrinth. In all trimesters of human pregnancy, NUCB2/nesfatin-1 was highly expressed in syncytiotrophoblast. In addition, there was a significant increase in NUCB2 expression in human primary trophoblast cells induced to syncytialise. Thus, the haemochorial mammalian placenta is a novel source of NUCB2/nesfatin-1 and likely a site of its action, with potential roles in glucose homeostasis and/or nutrient sensing.
Collapse
|
23
|
Akison LK, Nitert MD, Clifton VL, Moritz KM, Simmons DG. Review: Alterations in placental glycogen deposition in complicated pregnancies: Current preclinical and clinical evidence. Placenta 2017; 54:52-58. [PMID: 28117144 DOI: 10.1016/j.placenta.2017.01.114] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/04/2017] [Accepted: 01/11/2017] [Indexed: 11/16/2022]
Abstract
Normal placental function is essential for optimal fetal growth. Transport of glucose from mother to fetus is critical for fetal nutrient demands and can be stored in the placenta as glycogen. However, the function of this glycogen deposition remains a matter of debate: It could be a source of fuel for the placenta itself or a storage reservoir for later use by the fetus in times of need. While the significance of placental glycogen remains elusive, mounting evidence indicates that altered glycogen metabolism and/or deposition accompanies many pregnancy complications that adversely affect fetal development. This review will summarize histological, biochemical and molecular evidence that glycogen accumulates in a) placentas from a variety of experimental rodent models of perturbed pregnancy, including maternal alcohol exposure, glucocorticoid exposure, dietary deficiencies and hypoxia and b) placentas from human pregnancies with complications including preeclampsia, gestational diabetes mellitus and intrauterine growth restriction (IUGR). These pregnancies typically result in altered fetal growth, developmental abnormalities and/or disease outcomes in offspring. Collectively, this evidence suggests that changes in placental glycogen deposition is a common feature of pregnancy complications, particularly those associated with altered fetal growth.
Collapse
Affiliation(s)
- Lisa K Akison
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Marloes Dekker Nitert
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia; Centre for Clinical Research, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Vicki L Clifton
- Mater Medical Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD, 4101, Australia
| | - Karen M Moritz
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, 4072, Australia; Child Health Research Centre, The University of Queensland, Centre for Children's Health Research, South Brisbane, QLD, 4101, Australia
| | - David G Simmons
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, 4072, Australia; Mater Medical Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD, 4101, Australia
| |
Collapse
|
24
|
Cross JC. Adaptability and potential for treatment of placental functions to improve embryonic development and postnatal health. Reprod Fertil Dev 2017; 28:75-82. [PMID: 27062876 DOI: 10.1071/rd15342] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
For an organ that is so critical for life in eutherian mammals, the placenta hardly gets the attention that it deserves. The placenta does a series of remarkable things, including implanting the embryo in the uterus, negotiating with the mother for nutrients but also protecting her health during pregnancy, helping establish normal metabolic and cardiovascular function for life postnatally (developmental programming) and initiating changes that prepare the mother to care for and suckle her young after birth. Different lines of evidence in experimental animals suggest that the development and function of the placenta are adaptable. This means that some of the changes observed in pathological pregnancies may represent attempts to mitigate the impact of fetal growth and development. Key and emerging concepts are reviewed here concerning how we may view the placenta diagnostically and therapeutically in pregnancy complications, focusing on information from experimental studies in mice, sheep and cattle, as well as association studies from humans. Hundreds of different genes have been shown to underlie normal placental development and function, some of which have promise as tractable targets for intervention in pregnancies at risk for poor fetal growth.
Collapse
Affiliation(s)
- James C Cross
- Departments of Comparative Biology and Experimental Medicine, Biochemistry and Molecular Biology, Medical Genetics, and Obstetrics and Gynecology, University of Calgary, Calgary, Alberta T2N 4N1, Canada.
| |
Collapse
|
25
|
Abstract
Evolutionary gene duplication, developmental endoreduplication and selective gene amplification are alternative strategies for increasing gene copy number. When these processes occur together, things get really interesting, and new work shows that is the lifestyle of cells in the placenta.
Collapse
Affiliation(s)
- James C Cross
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, Alberta, T1S 1A2 Canada.
| |
Collapse
|
26
|
Ilekis JV, Tsilou E, Fisher S, Abrahams VM, Soares MJ, Cross JC, Zamudio S, Illsley NP, Myatt L, Colvis C, Costantine MM, Haas DM, Sadovsky Y, Weiner C, Rytting E, Bidwell G. Placental origins of adverse pregnancy outcomes: potential molecular targets: an Executive Workshop Summary of the Eunice Kennedy Shriver National Institute of Child Health and Human Development. Am J Obstet Gynecol 2016; 215:S1-S46. [PMID: 26972897 DOI: 10.1016/j.ajog.2016.03.001] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 02/11/2016] [Accepted: 03/01/2016] [Indexed: 12/26/2022]
Abstract
Although much progress is being made in understanding the molecular pathways in the placenta that are involved in the pathophysiology of pregnancy-related disorders, a significant gap exists in the utilization of this information for the development of new drug therapies to improve pregnancy outcome. On March 5-6, 2015, the Eunice Kennedy Shriver National Institute of Child Health and Human Development of the National Institutes of Health sponsored a 2-day workshop titled Placental Origins of Adverse Pregnancy Outcomes: Potential Molecular Targets to begin to address this gap. Particular emphasis was given to the identification of important molecular pathways that could serve as drug targets and the advantages and disadvantages of targeting these particular pathways. This article is a summary of the proceedings of that workshop. A broad number of topics were covered that ranged from basic placental biology to clinical trials. This included research in the basic biology of placentation, such as trophoblast migration and spiral artery remodeling, and trophoblast sensing and response to infectious and noninfectious agents. Research findings in these areas will be critical for the formulation of the development of future treatments and the development of therapies for the prevention of a number of pregnancy disorders of placental origin that include preeclampsia, fetal growth restriction, and uterine inflammation. Research was also presented that summarized ongoing clinical efforts in the United States and in Europe that has tested novel interventions for preeclampsia and fetal growth restriction, including agents such as oral arginine supplementation, sildenafil, pravastatin, gene therapy with virally delivered vascular endothelial growth factor, and oxygen supplementation therapy. Strategies were also proposed to improve fetal growth by the enhancement of nutrient transport to the fetus by modulation of their placental transporters and the targeting of placental mitochondrial dysfunction and oxidative stress to improve placental health. The roles of microRNAs and placental-derived exosomes, as well as messenger RNAs, were also discussed in the context of their use for diagnostics and as drug targets. The workshop discussed the aspect of safety and pharmacokinetic profiles of potential existing and new therapeutics that will need to be determined, especially in the context of the unique pharmacokinetic properties of pregnancy and the hurdles and pitfalls of the translation of research findings into practice. The workshop also discussed novel methods of drug delivery and targeting during pregnancy with the use of macromolecular carriers, such as nanoparticles and biopolymers, to minimize placental drug transfer and hence fetal drug exposure. In closing, a major theme that developed from the workshop was that the scientific community must change their thinking of the pregnant woman and her fetus as a vulnerable patient population for which drug development should be avoided, but rather be thought of as a deprived population in need of more effective therapeutic interventions.
Collapse
Affiliation(s)
- John V Ilekis
- Pregnancy and Perinatology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Department of Health and Human Services, Bethesda, MD.
| | - Ekaterini Tsilou
- Obstetric and Pediatric Pharmacology and Therapeutics Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Department of Health and Human Services, Bethesda, MD.
| | - Susan Fisher
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA
| | - Vikki M Abrahams
- Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine; New Haven, CT
| | - Michael J Soares
- Institute of Reproductive Health and Regenerative Medicine and Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - James C Cross
- Comparative Biology and Experimental Medicine, University of Calgary Health Sciences Centre, Calgary, Alberta, Canada
| | - Stacy Zamudio
- Department of Obstetrics and Gynecology, Hackensack University Medical Center, Hackensack, NJ
| | - Nicholas P Illsley
- Department of Obstetrics and Gynecology, Hackensack University Medical Center, Hackensack, NJ
| | - Leslie Myatt
- Center for Pregnancy and Newborn Research, University of Texas Health Science Center, San Antonio, TX
| | - Christine Colvis
- Therapeutics Discovery Program, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD
| | - Maged M Costantine
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX
| | - David M Haas
- Department of Obstetrics and Gynecology Indiana University, Indianapolis, IN
| | | | - Carl Weiner
- University of Kansas Medical Center, Kansas City, KS
| | - Erik Rytting
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX
| | - Gene Bidwell
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
27
|
Al-Soudy AS, Nakanishi T, Mizuno S, Hasegawa Y, Shawki HH, Katoh MC, Basha WA, Ibrahim AE, El-Shemy HA, Iseki H, Yoshiki A, Hiromori Y, Nagase H, Takahashi S, Oishi H, Sugiyama F. Germline recombination in a novel Cre transgenic line, Prl3b1-Cre mouse. Genesis 2016; 54:389-97. [PMID: 27124574 DOI: 10.1002/dvg.22944] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/12/2016] [Accepted: 04/24/2016] [Indexed: 11/09/2022]
Abstract
Spermatogenesis is a complex and highly regulated process by which spermatogonial stem cells differentiate into spermatozoa. To better understand the molecular mechanisms of the process, the Cre/loxP system has been widely utilized for conditional gene knockout in mice. In this study, we generated a transgenic mouse line that expresses Cre recombinase under the control of the 2.5 kbp of the Prolactin family 3, subfamily b, member 1 (Prl3b1) gene promoter (Prl3b1-cre). Prl3b1 was initially reported to code for placental lactogen 2 (PL-2) protein in placenta along with increased expression toward the end of pregnancy. PL-2 was found to be expressed in germ cells in the testis, especially in spermatocytes. To analyze the specificity and efficiency of Cre recombinase activity in Prl3b1-cre mice, the mice were mated with reporter R26GRR mice, which express GFP ubiquitously before and tdsRed exclusively after Cre recombination. The systemic examination of Prl3b1-cre;R26GRR mice revealed that tdsRed-positive cells were detected only in the testis and epididymis. Fluorescence imaging of Prl3b1-cre;R26GRR testes suggested that Cre-mediated recombination took place in the germ cells with approximately 74% efficiency determined by in vitro fertilization. In conclusion, our results suggest that the Prl3b1-cre mice line provides a unique resource to understand testicular germ-cell development. genesis 54:389-397, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Al-Sayed Al-Soudy
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Ibaraki, Japan.,Animal Genetic Resource Department, National Gene Bank, Giza, Egypt.,Department of Anatomy and Embryology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Tsuyoshi Nakanishi
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, Gifu, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoshikazu Hasegawa
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hossam H Shawki
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Ibaraki, Japan.,Animal Genetic Resource Department, National Gene Bank, Giza, Egypt.,Department of Anatomy and Embryology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Megumi C Katoh
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Ibaraki, Japan.,Department of Anatomy and Embryology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Walaa A Basha
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Ibaraki, Japan.,Department of Anatomy and Embryology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan.,Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Abdelaziz E Ibrahim
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Ibaraki, Japan.,Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Hany A El-Shemy
- Cairo University Research Park, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Hiroyoshi Iseki
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Ibaraki, Japan.,International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Atsushi Yoshiki
- Experimental Animal Division, Riken BioResource Center, Tsukuba, Japan
| | - Youhei Hiromori
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, Gifu, Japan.,Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Aichi, Japan
| | - Hisamitsu Nagase
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, Gifu, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Ibaraki, Japan.,Department of Anatomy and Embryology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan.,International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hisashi Oishi
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Ibaraki, Japan.,Department of Anatomy and Embryology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
28
|
Bu P, Alam SMK, Dhakal P, Vivian JL, Soares MJ. A Prolactin Family Paralog Regulates Placental Adaptations to a Physiological Stressor. Biol Reprod 2016; 94:107. [PMID: 26985002 PMCID: PMC4939737 DOI: 10.1095/biolreprod.115.138032] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/10/2016] [Indexed: 11/21/2022] Open
Abstract
The prolactin (PRL) family of hormones and cytokines participates in the regulation of optimal reproductive performance in the mouse and rat. Members of the PRL family are expressed in the anterior pituitary, uterus, and/or placenta. In the present study, we investigated the ontogeny of PRL family 7, subfamily b, member 1 (PRL7B1; also called PRL-like protein-N, PLP-N) expression in the developing mouse placenta and established a mouse model for investigating the biological function of PRL7B1. Transcripts for Prl7b1 were first detected on Gestation Day (d) 8.5. From gestation d8.5 through d14.5, Prl7b1 was expressed in trophoblast cells residing at the interface between maternal mesometrial decidua and the developing placenta. On gestation d17.5, the predominant cellular source of Prl7b1 mRNA was migratory trophoblast cells invading into the uterine mesometrial decidua. The Prl7b1 null mutant allele was generated via replacement of the endogenous Prl7b1 coding sequence with beta-galactosidase (LacZ) reporter and neomycin cassettes. The mutant Prl7b1 allele was successfully passed through the germline. Homozygous Prl7b1 mutant mice were viable and fertile. Under standard animal housing conditions, Prl7b1 had undetectable effects on placentation and pregnancy. Hypoxia exposure during pregnancy evoked adaptations in the organization of the wild-type placenta that were not observed in Prl7b1 null placentation sites. In summary, PRL7B1 is viewed as a part of a pathway regulating placental adaptations to physiological stressors.
Collapse
Affiliation(s)
- Pengli Bu
- Institute for Reproductive Health and Regenerative Medicine, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Sheikh M Khorshed Alam
- Institute for Reproductive Health and Regenerative Medicine, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Pramod Dhakal
- Institute for Reproductive Health and Regenerative Medicine, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Jay L Vivian
- Institute for Reproductive Health and Regenerative Medicine, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Michael J Soares
- Institute for Reproductive Health and Regenerative Medicine, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
29
|
Alam SMK, Konno T, Soares MJ. Identification of target genes for a prolactin family paralog in mouse decidua. Reproduction 2016; 149:625-32. [PMID: 25926690 DOI: 10.1530/rep-15-0107] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Prolactin family 8, subfamily a, member 2 (PRL8A2; also called decidual prolactin-related protein; dPRP) is a member of the expanded prolactin family. PRL8A2 is expressed in the uterine decidua and contributes to pregnancy-dependent adaptations to hypoxia. The purpose of this study was to identify gene targets for PRL8A2 action within the uteroplacental compartment. Affymetrix DNA microarray analysis was performed for RNA samples from WT and Prl8a2 null tissues. Validation of the DNA microarray was performed using quantitative RT-PCR. Nine genes were confirmed with decreased expression in Prl8a2 null tissues (e.g., Klk7, Rimklb, Arhgef6, Calm4, Sprr2h, Prl4a1, Ccl27, Lipg, and Htra3). These include potential decidual, endothelial and trophoblast cell targets positively regulated by PRL8A2. A significant upregulation of Derl3, Herpud1, Creld2, Hsp90b1, Ddit3 and Hspa5 was identified in Prl8a2 null tissues, reflecting an increased endoplasmic reticulum (ER) stress response. ER stress genes were prominently expressed in the uterine decidua. We propose that PRL8A2 is a mediator of progesterone-dependent modulation of intrauterine responses to physiological stressors.
Collapse
Affiliation(s)
- S M Khorshed Alam
- Department of Pathology and Laboratory MedicineInstitute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Toshihiro Konno
- Department of Pathology and Laboratory MedicineInstitute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Michael J Soares
- Department of Pathology and Laboratory MedicineInstitute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| |
Collapse
|
30
|
Triebel J, Bertsch T, Bollheimer C, Rios-Barrera D, Pearce CF, Hüfner M, Martínez de la Escalera G, Clapp C. Principles of the prolactin/vasoinhibin axis. Am J Physiol Regul Integr Comp Physiol 2015; 309:R1193-203. [PMID: 26310939 PMCID: PMC4666935 DOI: 10.1152/ajpregu.00256.2015] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/25/2015] [Indexed: 12/18/2022]
Abstract
The hormonal family of vasoinhibins, which derive from the anterior pituitary hormone prolactin, are known for their inhibiting effects on blood vessel growth, vasopermeability, and vasodilation. As pleiotropic hormones, vasoinhibins act in multiple target organs and tissues. The generation, secretion, and regulation of vasoinhibins are embedded into the organizational principle of an axis, which integrates the hypothalamus, the pituitary, and the target tissue microenvironment. This axis is designated as the prolactin/vasoinhibin axis. Disturbances of the prolactin/vasoinhibin axis are associated with the pathogenesis of retinal and cardiac diseases and with diseases occurring during pregnancy. New phylogenetical, physiological, and clinical implications are discussed.
Collapse
Affiliation(s)
- Jakob Triebel
- Institute for Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Paracelsus Medical University, Nuremberg, Germany;
| | - Thomas Bertsch
- Institute for Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Paracelsus Medical University, Nuremberg, Germany
| | - Cornelius Bollheimer
- Institute for Biomedicine of Aging, Friedrich-Alexander Universität Erlangen-Nürnberg, Nuremberg, Germany
| | - Daniel Rios-Barrera
- European Molecular Biology Laboratory, Developmental Biology Unit, Directors' Research, Heidelberg, Germany
| | - Christy F Pearce
- Southern Colorado Maternal Fetal Medicine, St. Francis Medical Campus, Centura Health, Colorado Springs, Colorado
| | | | | | - Carmen Clapp
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| |
Collapse
|
31
|
Rawn SM, Huang C, Hughes M, Shaykhutdinov R, Vogel HJ, Cross JC. Pregnancy Hyperglycemia in Prolactin Receptor Mutant, but Not Prolactin Mutant, Mice and Feeding-Responsive Regulation of Placental Lactogen Genes Implies Placental Control of Maternal Glucose Homeostasis. Biol Reprod 2015; 93:75. [PMID: 26269505 DOI: 10.1095/biolreprod.115.132431] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 07/27/2015] [Indexed: 12/23/2022] Open
Abstract
Pregnancy is often viewed as a conflict between the fetus and mother over metabolic resources. Insulin resistance occurs in mothers during pregnancy but does not normally lead to diabetes because of an increase in the number of the mother's pancreatic beta cells. In mice, this increase is dependent on prolactin (Prl) receptor signaling but the source of the ligand has been unclear. Pituitary-derived Prl is produced during the first half of pregnancy in mice but the placenta produces Prl-like hormones from implantation to term. Twenty-two separate mouse genes encode the placenta Prl-related hormones, making it challenging to assess their roles in knockout models. However, because at least four of them are thought to signal through the Prl receptor, we analyzed Prlr mutant mice and compared their phenotypes with those of Prl mutants. We found that whereas Prlr mutants develop hyperglycemia during gestation, Prl mutants do not. Serum metabolome analysis showed that Prlr mutants showed other changes consistent with diabetes. Despite the metabolic changes, fetal growth was normal in Prlr mutants. Of the four placenta-specific, Prl-related hormones that have been shown to interact with the Prlr, their gene expression localizes to different endocrine cell types. The Prl3d1 gene is expressed by trophoblast giant cells both in the labyrinth layer, sitting on the arterial side where maternal blood is highest in oxygen and nutrients, and in the junctional zone as maternal blood leaves the placenta. Expression increases during the night, though the increase in the labyrinth is circadian whereas it occurs only after feeding in the junctional zone. These data suggest that the placenta has a sophisticated endocrine system that regulates maternal glucose metabolism during pregnancy.
Collapse
Affiliation(s)
- Saara M Rawn
- Department of Comparative Biology & Experimental Medicine, University of Calgary, Calgary, Alberta, Canada Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Carol Huang
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
| | - Martha Hughes
- Department of Comparative Biology & Experimental Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rustem Shaykhutdinov
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Hans J Vogel
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - James C Cross
- Department of Comparative Biology & Experimental Medicine, University of Calgary, Calgary, Alberta, Canada Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
32
|
Aydin E, Kloos DP, Gay E, Jonker W, Hu L, Bullwinkel J, Brown JP, Manukyan M, Giera M, Singh PB, Fundele R. A hypomorphic Cbx3 allele causes prenatal growth restriction and perinatal energy homeostasis defects. J Biosci 2015; 40:325-38. [DOI: 10.1007/s12038-015-9520-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
33
|
Goyvaerts L, Lemaire K, Arijs I, Auffret J, Granvik M, Van Lommel L, Binart N, in’t Veld P, Schuit F, Schraenen A. Prolactin receptors and placental lactogen drive male mouse pancreatic islets to pregnancy-related mRNA changes. PLoS One 2015; 10:e0121868. [PMID: 25816302 PMCID: PMC4376745 DOI: 10.1371/journal.pone.0121868] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 02/19/2015] [Indexed: 12/03/2022] Open
Abstract
Pregnancy requires a higher functional beta cell mass and this is associated with profound changes in the gene expression profile of pancreatic islets. Taking Tph1 as a sensitive marker for pregnancy-related islet mRNA expression in female mice, we previously identified prolactin receptors and placental lactogen as key signalling molecules. Since beta cells from male mice also express prolactin receptors, the question arose whether male and female islets have the same phenotypic resilience at the mRNA level during pregnancy. We addressed this question in vitro, by stimulating cultured islets with placental lactogen and in vivo, by transplanting male or female islets into female acceptor mice. Additionally, the islet mRNA expression pattern of pregnant prolactin receptor deficient mice was compared with that of their pregnant wild-type littermates. When cultured with placental lactogen, or when transplanted in female recipients that became pregnant (day 12.5), male islets induced the 'islet pregnancy gene signature', which we defined as the 12 highest induced genes in non-transplanted female islets at day 12.5 of pregnancy. In addition, serotonin immunoreactivity and beta cell proliferation was also induced in these male transplanted islets at day 12.5 of pregnancy. In order to further investigate the importance of prolactin receptors in these mRNA changes we used a prolactin receptor deficient mouse model. For the 12 genes of the signature, which are highly induced in control pregnant mice, no significant induction of mRNA transcripts was found at day 9.5 of pregnancy. Together, our results support the key role of placental lactogen as a circulating factor that can trigger the pregnancy mRNA profile in both male and female beta cells.
Collapse
Affiliation(s)
- Lotte Goyvaerts
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Katleen Lemaire
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Ingrid Arijs
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Gasthuisberg, KU Leuven, Leuven, Belgium
| | - Julien Auffret
- Inserm U693, Faculté de Médecine Paris-Sud, Université Paris-Sud, Le Kremlin-Bicêtre, France
| | - Mikaela Granvik
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Leentje Van Lommel
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Nadine Binart
- Inserm U693, Faculté de Médecine Paris-Sud, Université Paris-Sud, Le Kremlin-Bicêtre, France
| | - Peter in’t Veld
- Department of Pathology, Vrije Universiteit Brussel, Jette, Belgium
| | - Frans Schuit
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Anica Schraenen
- Gene Expression Unit, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
34
|
Effect of duplicate genes on mouse genetic robustness: an update. BIOMED RESEARCH INTERNATIONAL 2014; 2014:758672. [PMID: 25110693 PMCID: PMC4119742 DOI: 10.1155/2014/758672] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 06/15/2014] [Accepted: 06/16/2014] [Indexed: 12/28/2022]
Abstract
In contrast to S. cerevisiae and C. elegans, analyses based on the current knockout (KO) mouse phenotypes led to the conclusion that duplicate genes had almost no role in mouse genetic robustness. It has been suggested that the bias of mouse KO database toward ancient duplicates may possibly cause this knockout duplicate puzzle, that is, a very similar proportion of essential genes (PE) between duplicate genes and singletons. In this paper, we conducted an extensive and careful analysis for the mouse KO phenotype data and corroborated a strong effect of duplicate genes on mouse genetics robustness. Moreover, the effect of duplicate genes on mouse genetic robustness is duplication-age dependent, which holds after ruling out the potential confounding effect from coding-sequence conservation, protein-protein connectivity, functional bias, or the bias of duplicates generated by whole genome duplication (WGD). Our findings suggest that two factors, the sampling bias toward ancient duplicates and very ancient duplicates with a proportion of essential genes higher than that of singletons, have caused the mouse knockout duplicate puzzle; meanwhile, the effect of genetic buffering may be correlated with sequence conservation as well as protein-protein interactivity.
Collapse
|
35
|
YANG QIAOZHEN, HAO JIE, HE MIAO, CHEN MAOXIN, LI GANG. Localization and expression patterns of prolactin-like protein J in mouse testis. Mol Med Rep 2014; 10:255-61. [DOI: 10.3892/mmr.2014.2218] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 03/07/2014] [Indexed: 11/06/2022] Open
|
36
|
Yang X, Meyer K, Friedl A. STAT5 and prolactin participate in a positive autocrine feedback loop that promotes angiogenesis. J Biol Chem 2013; 288:21184-21196. [PMID: 23729680 DOI: 10.1074/jbc.m113.481119] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have shown previously that the murine prolactin/growth hormone family member proliferin plays a pivotal role in angiogenesis induced by the FGF2/STAT5 signaling cascade. To delineate the signaling pathway downstream of STAT5 in the human system, where proliferin does not exist, we expressed constitutively active (CA) or dominant-negative (DN) mutant STAT5A in hCMEC/D3 human brain endothelial cells. We found that conditioned medium from CA-STAT5A- but not from DN-STAT5A-overexpressing endothelial cells (EC) is sufficient to induce EC migration and tube formation but not proliferation, indicating that STAT5A regulates the secretion of autocrine proangiogenic factors. We identified prolactin (PRL) as a candidate autocrine factor. CA-STAT5A expression stimulates PRL production at the RNA and protein level, and STAT5A binds to the PRL promoter region, suggesting direct transcriptional regulation. Medium conditioned by CA-STAT5A-overexpressing EC induces phosphorylation of the PRL receptor and activates MAPK. Knockdown of PRL expression by shRNA or blocking of PRL activity with neutralizing antibodies removed the CA-STAT5A-dependent proangiogenic activity from the conditioned medium of EC. The addition of recombinant PRL restores this activity. STAT5A-induced PRL in the conditioned medium can activate STAT5, STAT1, and to a lesser extent STAT3 in hCMEC/D3 cells, suggesting the existence of a positive feedback loop between STAT5 and PRL that promotes angiogenesis. Furthermore, we find that VEGF, a potent proangiogenic factor, is induced by activation of STAT5A, and VEGF induction depends on PRL expression. These observations demonstrate a STAT5/PRL/VEGF signaling cascade in human brain EC and implicate PRL and VEGF as autocrine regulators of EC migration, invasion, and tube formation.
Collapse
Affiliation(s)
- Xinhai Yang
- From the Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin 53705
| | - Kristy Meyer
- From the Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin 53705
| | - Andreas Friedl
- From the Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin 53705,; Pathology and Laboratory Medicine Service, William S. Middleton Memorial Veterans Hospital, Department of Veterans Affairs Medical Center, Madison, Wisconsin 53705, and; UW Carbone Cancer Center, Madison, Wisconsin 53792.
| |
Collapse
|
37
|
Rappolee DA, Zhou S, Puscheck EE, Xie Y. Stress responses at the endometrial-placental interface regulate labyrinthine placental differentiation from trophoblast stem cells. Reproduction 2013; 145:R139-55. [PMID: 23463790 DOI: 10.1530/rep-12-0240] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Development can happen in one of two ways. Cells performing a necessary function can differentiate from stem cells before the need for it arises and stress does not develop. Or need arises before function, stress develops and stress signals are part of the normal stimuli that regulate developmental mechanisms. These mechanisms adjust stem cell differentiation to produce function in a timely and proportional manner. In this review, we will interpret data from studies of null lethal mutants for placental stress genes that suggest the latter possibility. Acknowledged stress pathways participate in stress-induced and -regulated differentiation in two ways. These pathways manage the homeostatic response to maintain stem cells during the stress. Stress pathways also direct stem cell differentiation to increase the first essential lineage and suppress later lineages when stem cell accumulation is diminished. This stress-induced differentiation maintains the conceptus during stress. Pathogenic outcomes arise because population sizes of normal stem cells are first depleted by decreased accumulation. The fraction of stem cells is further decreased by differentiation that is induced to compensate for smaller stem cell populations. Analysis of placental lethal null mutant genes known to mediate stress responses suggests that the labyrinthine placenta develops during, and is regulated by, hypoxic stress.
Collapse
Affiliation(s)
- D A Rappolee
- CS Mott Center for Human Growth and Development, Wayne State University School of Medicine.
| | | | | | | |
Collapse
|
38
|
Carter AM. Evolution of Placental Function in Mammals: The Molecular Basis of Gas and Nutrient Transfer, Hormone Secretion, and Immune Responses. Physiol Rev 2012; 92:1543-76. [DOI: 10.1152/physrev.00040.2011] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Placenta has a wide range of functions. Some are supported by novel genes that have evolved following gene duplication events while others require acquisition of gene expression by the trophoblast. Although not expressed in the placenta, high-affinity fetal hemoglobins play a key role in placental gas exchange. They evolved following duplications within the beta-globin gene family with convergent evolution occurring in ruminants and primates. In primates there was also an interesting rearrangement of a cassette of genes in relation to an upstream locus control region. Substrate transfer from mother to fetus is maintained by expression of classic sugar and amino acid transporters at the trophoblast microvillous and basal membranes. In contrast, placental peptide hormones have arisen largely by gene duplication, yielding for example chorionic gonadotropins from the luteinizing hormone gene and placental lactogens from the growth hormone and prolactin genes. There has been a remarkable degree of convergent evolution with placental lactogens emerging separately in the ruminant, rodent, and primate lineages and chorionic gonadotropins evolving separately in equids and higher primates. Finally, coevolution in the primate lineage of killer immunoglobulin-like receptors and human leukocyte antigens can be linked to the deep invasion of the uterus by trophoblast that is a characteristic feature of human placentation.
Collapse
Affiliation(s)
- Anthony M. Carter
- Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
39
|
Featherstone K, White MRH, Davis JRE. The prolactin gene: a paradigm of tissue-specific gene regulation with complex temporal transcription dynamics. J Neuroendocrinol 2012; 24:977-90. [PMID: 22420298 PMCID: PMC3505372 DOI: 10.1111/j.1365-2826.2012.02310.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transcription of numerous mammalian genes is highly pulsatile, with bursts of expression occurring with variable duration and frequency. The presence of this stochastic or 'noisy' expression pattern has been relatively unexplored in tissue systems. The prolactin gene provides a model of tissue-specific gene regulation resulting in pulsatile transcription dynamics in both cell lines and endocrine tissues. In most cell culture models, prolactin transcription appears to be highly variable between cells, with differences in transcription pulse duration and frequency. This apparently stochastic transcription is constrained by a transcriptional refractory period, which may be related to cycles of chromatin remodelling. We propose that prolactin transcription dynamics result from the summation of oscillatory cellular inputs and by regulation through chromatin remodelling cycles. Observations of transcription dynamics in cells within pituitary tissue show reduced transcriptional heterogeneity and can be grouped into a small number of distinct patterns. Thus, it appears that the tissue environment is able to reduce transcriptional noise to enable coordinated tissue responses to environmental change. We review the current knowledge on the complex tissue-specific regulation of the prolactin gene in pituitary and extra-pituitary sites, highlighting differences between humans and rodent experimental animal models. Within this context, we describe the transcription dynamics of prolactin gene expression and how this may relate to specific processes occurring within the cell.
Collapse
Affiliation(s)
- K Featherstone
- Developmental Biomedicine Research Group, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK.
| | | | | |
Collapse
|
40
|
Yang X, Qiao D, Meyer K, Pier T, Keles S, Friedl A. Angiogenesis induced by signal transducer and activator of transcription 5A (STAT5A) is dependent on autocrine activity of proliferin. J Biol Chem 2011; 287:6490-502. [PMID: 22199350 DOI: 10.1074/jbc.m111.254631] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Multiple secreted factors induce the formation of new blood vessels (angiogenesis). The signal transduction events that orchestrate the numerous cellular activities required for angiogenesis remain incompletely understood. We have shown previously that STAT5 plays a pivotal role in angiogenesis induced by FGF2 and FGF8b. To delineate the signaling pathway downstream of STAT5, we expressed constitutively active (CA) or dominant-negative (DN) mutant STAT5A in mouse brain endothelial cells (EC). We found that the conditioned medium from CA-STAT5A but not from dominant-negative STAT5A overexpressing EC is sufficient to induce EC invasion and tube formation, indicating that STAT5A regulates the secretion of autocrine proangiogenic factors. Conversely, CA-STAT5A-induced conditioned medium had no effect on EC proliferation. Using a comparative genome-wide transcription array screen, we identified the prolactin family member proliferin (PLF1 and PLF4) as a candidate autocrine factor. The CA-STAT5A-dependent transcription and secretion of PLF by EC was confirmed by quantitative RT-PCR and Western blotting, respectively. CA-STAT5A binds to the PLF1 promoter region, suggesting a direct transcriptional regulation. Knockdown of PLF expression by shRNA or by blocking of PLF activity with neutralizing antibodies removed the CA-STAT5A-dependent proangiogenic activity from the conditioned medium of EC. Similarly, the ability of concentrated conditioned medium from CA-STAT5A transfected EC to induce angiogenesis in Matrigel plugs in vivo was abolished when PLF was depleted from the medium. These observations demonstrate a FGF/STAT5/PLF signaling cascade in EC and implicate PLF as autocrine regulator of EC invasion and tube formation.
Collapse
Affiliation(s)
- Xinhai Yang
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53792, USA
| | | | | | | | | | | |
Collapse
|
41
|
Bridging sequence diversity and tissue-specific expression by DNA methylation in genes of the mouse prolactin superfamily. Mamm Genome 2011; 23:336-45. [PMID: 22193412 DOI: 10.1007/s00335-011-9383-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 11/25/2011] [Indexed: 12/14/2022]
Abstract
Much of the DNA in genomes is organized within gene families and hierarchies of gene superfamilies. DNA methylation is the main epigenetic event involved in gene silencing and genome stability. In the present study, we analyzed the DNA methylation status of the prolactin (PRL) superfamily to obtain insight into its tissue-specific expression and the evolution of its sequence diversity. The PRL superfamily in mice consists of two dozen members, which are expressed in a tissue-specific manner. The genes in this family have CpG-less sequences, and they are located within a 1-Mb region as a gene cluster on chromosome 13. We tentatively grouped the family into several gene clusters, depending on location and gene orientation. We found that all the members had tissue-dependent differentially methylated regions (T-DMRs) around the transcription start site. The T-DMRs are hypermethylated in nonexpressing tissues and hypomethylated in expressing cells, supporting the idea that the expression of the PRL superfamily genes is subject to epigenetic regulation. Interestingly, the DNA methylation patterns of T-DMRs are shared within a cluster, while the patterns are different among the clusters. Finally, we reconstituted the nucleotide sequences of T-DMRs by converting TpG to CpG based on the consideration of a possible conversion of 5-methylcytosine to thymine by spontaneous deamination during the evolutionary process. On the phylogenic tree, the reconstituted sequences were well matched with the DNA methylation pattern of T-DMR and orientation. Our study suggests that DNA methylation is involved in tissue-specific expression and sequence diversity during evolution.
Collapse
|
42
|
Dai G, Bustamante JJ, Zou Y, Myronovych A, Bao Q, Kumar S, Soares MJ. Maternal hepatic growth response to pregnancy in the mouse. Exp Biol Med (Maywood) 2011; 236:1322-32. [PMID: 21969712 DOI: 10.1258/ebm.2011.011076] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Pregnancy is characterized by physiological adjustments in the maternal compartment. In this investigation, the influence of pregnancy on maternal liver was examined in CD-1 mice. Dramatic changes were observed in the size of the maternal liver during pregnancy. Livers doubled in weight from the non-pregnant state to day 18 of pregnancy. The pregnancy-induced hepatomegaly was a physiological event of liver growth confirmed by DNA content increase and detection of hepatocyte hyperplasia and hypertrophy. Growth of the liver was initiated following implantation and peaked at parturition. The expression and/or activities of key genes known to regulate liver regeneration, a phenomenon of liver growth compensatory to liver mass loss, were investigated. The results showed that pregnancy-dependent liver growth was associated with interleukin (IL)-6, tumor necrosis factor α, c-Jun and IL-1β, but independent of hepatocyte growth factor, fibroblast growth factor 1, tumor necrosis factor receptor 1, constitutive androstane receptor and pregnane X receptor. Furthermore, maternal liver growth was associated with the activation of hepatic signal transducer and activator of transcription 3, β-catenin and epidermal growth factor receptor, but pregnancy did not activate hepatic c-Met. The findings suggest that the molecular mechanisms regulating pregnancy-induced liver growth and injury-induced liver regeneration exhibit overlapping features but are not identical. In summary, the liver of the mouse adapts to the demands of pregnancy via a dramatic growth response driven by hepatocyte proliferation and size increase.
Collapse
Affiliation(s)
- Guoli Dai
- Department of Biology, Center for Regenerative Biology and Medicine, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Knox K, Leuenberger D, Penn AA, Baker JC. Global hormone profiling of murine placenta reveals Secretin expression. Placenta 2011; 32:811-6. [PMID: 21944867 DOI: 10.1016/j.placenta.2011.08.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 08/10/2011] [Accepted: 08/30/2011] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To elucidate and categorize the murine placental hormones expressed across gestation, including the expression of hormones with previously undescribed roles. STUDY DESIGN Expression levels of all genes with known or predicted hormone activity expressed in two separate tissues, the placenta and maternal decidua, were assessed across a timecourse spanning the full lifetime of the placenta. Novel expression patterns were confirmed by in situ hybridization and protein level measurements. RESULTS A combination of temporal and spatial information defines five groups that can accurately predict the patterns of uncharacterized hormones. Our analysis identified Secretin, a novel placental hormone that is expressed specifically by the trophoblast at levels many times greater than in any other tissue. CONCLUSIONS The characteristics of Secretin fit the paradigm of known placental hormones and suggest that it may play an important role during pregnancy.
Collapse
Affiliation(s)
- K Knox
- Department of Genetics, Stanford University, Stanford, CA 94062, USA
| | | | | | | |
Collapse
|
44
|
Zhao L, Hao J, Hu J, Wang Q, Lü Z, Wang L, Yu Q, Wang Y, Li G. Expression of proliferin-related protein in testis and the biological significance in testosterone production. Mol Cell Endocrinol 2011; 343:25-31. [PMID: 21679748 DOI: 10.1016/j.mce.2011.05.046] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Revised: 04/29/2011] [Accepted: 05/31/2011] [Indexed: 12/29/2022]
Abstract
Proliferin-related protein (PRP) was originally identified as an angiogenesis inhibitor in mouse placentas. Indeed, the tissue expression of PRP has mainly been documented in placentas. We report herein for the first time that PRP is expressed in male rat testes. Immunocytochemical and in situ hybridization results showed positive PRP immunostaining in Leydig cells. Immunofluorescent staining of PRP in the TM3 Leydig cell line indicates that PRP is located within the cytoplasm. The expression pattern of PRP in rat testis exhibited an age-related increase. HCG significantly up-regulated the level of expression of PRP in TM3 cells via the PKA pathway. To elucidate the function of PRP, experiments were conducted to examine the consequences of lentiviral-mediated RNA interference (RNAi) of PRP on testosterone production and expression of several genes involved in steroidogenesis. PRP silencing caused a decrease in HCG-stimulated testosterone production. In addition, PRP silencing attenuated the increase in PRLR mRNA following HCG stimulation. Moreover, the enhanced effect of PRL on HCG-induced testosterone production was also weakened following PRP silencing, indicating that PRP may be involved in PRL function through an effect on PRL receptor expression in response to stimuli. Taken together, these data suggest that PRP is regulated by HCG and plays roles in male reproduction, such as testosterone production.
Collapse
Affiliation(s)
- Lina Zhao
- School of Public Health, Chongqing Medical University, Chongqing, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Emera D, Casola C, Lynch VJ, Wildman DE, Agnew D, Wagner GP. Convergent evolution of endometrial prolactin expression in primates, mice, and elephants through the independent recruitment of transposable elements. Mol Biol Evol 2011; 29:239-47. [PMID: 21813467 DOI: 10.1093/molbev/msr189] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Prolactin (PRL) is a multifunctional signaling molecule best known for its role in regulating lactation in mammals. Systemic PRL is produced by the anterior pituitary, but extrapituitary PRL has also been detected in many tissues including the human endometrium. Prolactin is essential for pregnancy in rodents and one of the most dramatically induced genes in the endometrium during human pregnancy. The promoter for human endometrial Prl is located about 5.8 kb upstream of the pituitary promoter and is derived from a transposable element called MER39. Although it has been shown that prolactin is expressed in the pregnant endometrium of a few mammals other than humans, MER39 has been described as primate specific. Thus, in an effort to understand mechanisms of prolactin regulatory evolution, we sought to determine how uterine prolactin is transcribed in species that lack MER39. Using a variety of complementary strategies, including reverse transcriptase-polymerase chain reaction, 5' rapid amplification of cDNA ends, and whole-transcriptome sequencing, we show that endometrial Prl expression is not a shared character of all placental mammals, as it is not expressed in rabbits, pigs, dogs, or armadillos. We show that in primates, mice, and elephants, prolactin mRNA is transcribed in the pregnant endometrium from alternative promoters, different from the pituitary promoter and different from each other. Moreover, we demonstrate that the spider monkey promoter derives from the long terminal repeat (LTR) element MER39 as in humans, the mouse promoter derives from the LTR element MER77, and the elephant promoter derives from the lineage-specific LINE retrotransposon L1-2_LA. We also find surprising variation of transcriptional start sites within these transposable elements and of Prl splice variants, suggesting a high degree of flexibility in the promoter architecture even among closely related species. Finally, the three groups shown here to express endometrial prolactin-the higher primates, the rodents, and the elephant-represent three of the four lineages that showed adaptive evolution of the Prl gene in an earlier study (Wallis M. 2000. Episodic evolution of protein hormones: molecular evolution of pituitary prolactin. J Mol Evol. 50:465-473), which supports our findings and suggests that the selective forces responsible for accelerated Prl evolution were in the endometrium. This is the first reported case of convergent evolution of gene expression through the independent recruitment of different transposable elements, highlighting the importance of transposable elements in gene regulatory, and potentially adaptive, evolution.
Collapse
Affiliation(s)
- Deena Emera
- Department of Ecology and Evolutionary Biology and Yale Systems Biology Institute, Yale University, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Hsiao EY, Patterson PH. Activation of the maternal immune system induces endocrine changes in the placenta via IL-6. Brain Behav Immun 2011; 25:604-15. [PMID: 21195166 PMCID: PMC3081363 DOI: 10.1016/j.bbi.2010.12.017] [Citation(s) in RCA: 290] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Revised: 12/21/2010] [Accepted: 12/22/2010] [Indexed: 02/08/2023] Open
Abstract
Activation of the maternal immune system in rodent models sets in motion a cascade of molecular pathways that ultimately result in autism- and schizophrenia-related behaviors in offspring. The finding that interleukin-6 (IL-6) is a crucial mediator of these effects led us to examine the mechanism by which this cytokine influences fetal development in vivo. Here we focus on the placenta as the site of direct interaction between mother and fetus and as a principal modulator of fetal development. We find that maternal immune activation (MIA) with a viral mimic, synthetic double-stranded RNA (poly(I:C)), increases IL-6 mRNA as well as maternally-derived IL-6 protein in the placenta. Placentas from MIA mothers exhibit increases in CD69+ decidual macrophages, granulocytes and uterine NK cells, indicating elevated early immune activation. Maternally-derived IL-6 mediates activation of the JAK/STAT3 pathway specifically in the spongiotrophoblast layer of the placenta, which results in expression of acute phase genes. Importantly, this parallels an IL-6-dependent disruption of the growth hormone-insulin-like growth factor (GH-IGF) axis that is characterized by decreased GH, IGFI and IGFBP3 levels. In addition, we observe an IL-6-dependent induction in pro-lactin-like protein-K (PLP-K) expression as well as MIA-related alterations in other placental endocrine factors. Together, these IL-6-mediated effects of MIA on the placenta represent an indirect mechanism by which MIA can alter fetal development.
Collapse
|
47
|
Saito K, Ogawa A, Toyofuku K, Hosoi Y, Soma M, Iha M, Kasuga K, Kojima I, Kobayashi M. Relationships between homeoprotein EGAM1C and the expression of the placental prolactin gene family in mouse placentae and trophoblast stem cells. Reproduction 2011; 141:259-68. [DOI: 10.1530/rep-10-0355] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The mouse Crxos gene encodes three structurally related homeoproteins, EGAM1, EGAM1N, and EGAM1C, as transcription and splicing variants. Recently, we identified the functions of EGAM1 and EGAM1N in the regulation of differentiation in mouse embryonic stem cells. However, the function of EGAM1C remains unknown. To explore the additional roles of these proteins, the ontogenic expression of the respective mRNAs in post implantation mouse embryos and extraembryonic tissues, particularly from embryonic day (E) 10.5 to E18.5, was analyzed. The expression of Egam1n mRNA was specifically detected in embryos throughout this period, whereas that of Egam1 was undetectable in any of the tissues examined. However, in the placenta, Egam1c mRNA and its encoded protein were detected after E16.5, and these expression levels increased by E18.5 immediately before partum. Quantitative RT-PCR and in situ hybridization analyses in placentae revealed that the spatial and temporal expression patterns of the Egam1c mRNA were related to some extent with those of Prl3a1 and Prl5a1 and partially overlapped that of Prl3b1, which are members of the placental prolactin (PRL) gene family. When EGAM1C was overexpressed moderately in mouse trophoblast stem cells as a model for undifferentiated and differentiating placental cell types, the expression levels of endogenous Prl3b1 and Prl5a1 were enhanced under both undifferentiated and differentiating culture conditions. These results indicated that EGAM1C may play a role in the expression of members of the placental PRL gene family, such as Prl3b1 and Prl5a1.
Collapse
|
48
|
Asanoma K, Rumi MAK, Kent LN, Chakraborty D, Renaud SJ, Wake N, Lee DS, Kubota K, Soares MJ. FGF4-dependent stem cells derived from rat blastocysts differentiate along the trophoblast lineage. Dev Biol 2011; 351:110-9. [PMID: 21215265 DOI: 10.1016/j.ydbio.2010.12.038] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Revised: 12/17/2010] [Accepted: 12/20/2010] [Indexed: 01/31/2023]
Abstract
Differentiated trophoblast cell lineages arise from trophoblast stem (TS) cells. To date such a stem cell population has only been established in the mouse. The objective of this investigation was to establish TS cell populations from rat blastocysts. Blastocysts were cultured individually on a feeder layer of rat embryonic fibroblasts (REFs) in fibroblast growth factor-4 (FGF4) and heparin supplemented culture medium. Once cell colonies were established REF feeder layers could be replaced with REF conditioned medium. The blastocyst-derived cell lines, in either proliferative or differentiated states, did not express genes indicative of ICM-derived tissues. In the proliferative state the cells expressed established stem cell-associated markers of TS cells. Cells ceased proliferation and differentiated when FGF4, heparin, and REF conditioned medium were removed. Differentiation was characterized by a decline of stem cell-associated marker gene expression, the appearance of large polyploid cells (trophoblast giant cells), and the expression of trophoblast differentiation-associated genes. Collectively, the data indicate that the rat blastocyst-derived cell lines not only possess many features characteristic of mouse TS cells but also possess some distinct properties. These rat TS cell lines represent valuable new in vitro models for analyses of mechanisms controlling TS cell renewal and differentiation.
Collapse
Affiliation(s)
- Kazuo Asanoma
- Institute for Reproductive Health and Regenerative Medicine, Department of Pathology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Zhong W, Xie Y, Abdallah M, Awonuga AO, Slater JA, Sipahi L, Puscheck EE, Rappolee DA. Cellular stress causes reversible, PRKAA1/2-, and proteasome-dependent ID2 protein loss in trophoblast stem cells. Reproduction 2010; 140:921-30. [PMID: 20876741 DOI: 10.1530/rep-10-0268] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stress reduces fertility, but the mechanisms mediating this are not understood. For a successful pregnancy, placental trophoblast stem cells (TSCs) in the implanting embryo proliferate and then a subpopulation differentiates to produce hormones. Normally, differentiation occurs when inhibitor of differentiation 2 (ID2) protein is lost in human and mouse placental stem cells. We hypothesize that stress enzyme-dependent differentiation occurs in association with insufficient TSC accumulation. We studied a well-defined model where TSC differentiation requires ID2 loss. The loss of ID2 derepresses the promoter of chorionic somatomammotropin hormone 1 (CSH1), the first hormone after implantation. Csh1 mRNA is known to be induced in stressed TSCs. In this study, we demonstrate that AMP-activated protein kinase (PRKAA1/2, aka AMPK) mediates the stress-induced proteasome-dependent loss of ID2 at high stress levels. At very low stress levels, PRKAA1/2 mediates metabolic adaptation exemplified by the inactivation of acetyl coA carboxylase by phosphorylation without ID2 loss. At the highest stress levels, irreversible TSC differentiation as defined by ID2 loss and slower cell accumulation occurs. However, lower stress levels lead to reversible differentiation accompanied by metabolic adaptation. These data support the hypothesis that PRKAA1/2 mediates preparation for differentiation that is induced by stress at levels where a significant decrease in cell accumulation occurs. This supports the interpretation that enzyme-mediated increases in differentiation may compensate when insufficient numbers of stem cells accumulate.
Collapse
Affiliation(s)
- W Zhong
- CS Mott Center for Human Growth and Development, Wayne State University School of Medicine, 275 East Hancock, Detroit, Michigan 48201, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Ovalbumin sensitization and challenge increases the number of lung cells possessing a mesenchymal stromal cell phenotype. Respir Res 2010; 11:127. [PMID: 20858250 PMCID: PMC2949728 DOI: 10.1186/1465-9921-11-127] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Accepted: 09/21/2010] [Indexed: 01/19/2023] Open
Abstract
Background Recent studies have indicated the presence of multipotent mesenchymal stromal cells (MSCs) in human lung diseases. Excess airway smooth muscle, myofibroblasts and activated fibroblasts have each been noted in asthma, suggesting that mesenchymal progenitor cells play a role in asthma pathogenesis. We therefore sought to determine whether MSCs are present in the lungs of ovalbumin (OVA)-sensitized and challenged mice, a model of allergic airways disease. Methods Balb/c mice were sensitized and challenged with PBS or OVA over a 25 day period. Flow cytometry as well as colony forming and differentiation potential were used to analyze the emergence of MSCs along with gene expression studies using immunochemical analyses, quantitative polymerase chain reaction (qPCR), and gene expression beadchips. Results A CD45-negative subset of cells expressed Stro-1, Sca-1, CD73 and CD105. Selection for these markers and negative selection against CD45 yielded a population of cells capable of adipogenic, osteogenic and chondrogenic differentiation. Lungs from OVA-treated mice demonstrated a greater average colony forming unit-fibroblast (CFU-F) than control mice. Sorted cells differed from unsorted lung adherent cells, exhibiting a pattern of gene expression nearly identical to bone marrow-derived sorted cells. Finally, cells isolated from the bronchoalveolar lavage of a human asthma patient showed identical patterns of cell surface markers and differentiation potential. Conclusions In summary, allergen sensitization and challenge is accompanied by an increase of MSCs resident in the lungs that may regulate inflammatory and fibrotic responses.
Collapse
|