1
|
Fiorentino G, Merico V, Zanoni M, Comincini S, Sproviero D, Garofalo M, Gagliardi S, Cereda C, Lin CJ, Innocenti F, Taggi M, Vaiarelli A, Ubaldi FM, Rienzi L, Cimadomo D, Garagna S, Zuccotti M. Extracellular vesicles secreted by cumulus cells contain microRNAs that are potential regulatory factors of mouse oocyte developmental competence. Mol Hum Reprod 2024; 30:gaae019. [PMID: 38745364 PMCID: PMC12054941 DOI: 10.1093/molehr/gaae019] [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: 06/30/2023] [Revised: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
The role of cumulus cells (CCs) in the acquisition of oocyte developmental competence is not yet fully understood. In a previous study, we matured cumulus-denuded fully-grown mouse oocytes to metaphase II (MII) on a feeder layer of CCs (FL-CCs) isolated from developmentally competent (FL-SN-CCs) or incompetent (FL-NSN-CCs) SN (surrounded nucleolus) or NSN (not surrounding nucleolus) oocytes, respectively. We observed that oocytes cultured on the former could develop into blastocysts, while those matured on the latter arrested at the 2-cell stage. To investigate the CC factors contributing to oocyte developmental competence, here we focused on the CCs' release into the medium of extracellular vesicles (EVs) and on their miRNA content. We found that, during the 15-h transition to MII, both FL-SN-CCs and FL-NSN-CCs release EVs that can be detected, by confocal microscopy, inside the zona pellucida (ZP) or the ooplasm. The majority of EVs are <200 nm in size, which is compatible with their ability to cross the ZP. Next-generation sequencing of the miRNome of FL-SN-CC versus FL-NSN-CC EVs highlighted 74 differentially expressed miRNAs, with 43 up- and 31 down-regulated. Although most of these miRNAs do not have known roles in the ovary, in silico functional analysis showed that seven of these miRNAs regulate 71 target genes with specific roles in meiosis resumption (N = 24), follicle growth (N = 23), fertilization (N = 1), and the acquisition of oocyte developmental competence (N = 23). Overall, our results indicate CC EVs as emerging candidates of the CC-to-oocyte communication axis and uncover a group of miRNAs as potential regulatory factors.
Collapse
Affiliation(s)
- Giulia Fiorentino
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology ‘Lazzaro Spallanzani’, University of Pavia, Pavia, Italy
| | - Valeria Merico
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology ‘Lazzaro Spallanzani’, University of Pavia, Pavia, Italy
| | - Mario Zanoni
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology ‘Lazzaro Spallanzani’, University of Pavia, Pavia, Italy
| | - Sergio Comincini
- Functional Genomics Laboratory, Department of Biology and Biotechnology ‘Lazzaro Spallanzani’, University of Pavia, Pavia, Italy
| | - Daisy Sproviero
- IFOM, IFOM—The FIRC Institute of Molecular Oncology, Milan, Italy
| | - Maria Garofalo
- Molecular Biology and Transcriptomics Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Stella Gagliardi
- Molecular Biology and Transcriptomics Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Cristina Cereda
- Department of Pediatrics, Center of Functional Genomics and Rare Diseases, Buzzi Children’s Hospital, Milan, Italy
| | - Chih-Jen Lin
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Federica Innocenti
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | - Marilena Taggi
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | - Alberto Vaiarelli
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | | | - Laura Rienzi
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
- Department of Biomolecular Sciences, University of Urbino “Carlo Bo”, Urbino, Italy
| | - Danilo Cimadomo
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | - Silvia Garagna
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology ‘Lazzaro Spallanzani’, University of Pavia, Pavia, Italy
| | - Maurizio Zuccotti
- Laboratory of Biology and Biotechnology of Reproduction, Department of Biology and Biotechnology ‘Lazzaro Spallanzani’, University of Pavia, Pavia, Italy
| |
Collapse
|
2
|
González Altamiranda EA, Arias ME, Kaiser GG, Mucci NC, Odeón AC, Felmer RN. Upregulation of interferon-alpha gene in bovine embryos produced in vitro in response to experimental infection with noncytophatic bovine-viral-diarrhea virus. Mol Biol Rep 2020; 47:9959-9965. [PMID: 33226564 PMCID: PMC7681760 DOI: 10.1007/s11033-020-05958-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 10/29/2020] [Indexed: 12/20/2022]
Abstract
In-vitro fertilization is a routine livestock-breeding technique widely used around the world. Several studies have reported the interaction of bovine viral-diarrhea virus (BVDV) with gametes and in-vitro-produced (IVP) bovine embryos. Since, gene expression in BVDV-infected IVP bovine embryos is scarcely addressed. The aim of this work was to evaluate the differential expression of genes involved in immune and inflammatory response. Groups of 20-25 embryos on Day 6 (morula stage) were exposed (infected) or not (control) to an NCP-BVDV strain in SOF medium. After 24 h, embryos that reached expanded blastocyst stage were washed. Total RNA of each embryo group was extracted to determine the transcription levels of 9 specific transcripts related with antiviral and inflammatory response by SYBR Green real time quantitative (RT-qPCR). Culture media and an aliquot of the last embryos wash on Day 7 were analyzed by titration and virus isolation, respectively. A conventional PCR confirmed BVDV presence in IVP embryos. A significantly higher expression of interferon-α was observed in blastocysts exposed to NCP-BVDV compared to the controls (p < 0.05). In this study, the upregulation of INFα and TLR7 genes involved in inflammatory and immune response in BVDV-infected IVP bovine embryos is a new finding in this field. This differential expression suggest that embryonic cells could function in a manner like immune cells by recognizing and responding early to interaction with viral pathogens. These results provide new insights into the action of BVDV on the complex molecular pathways controlling bovine early embryonic development.
Collapse
Affiliation(s)
- Erika A González Altamiranda
- Consejo Nacional de Investigaciones Científicas y Técnicas CONICET, Buenos Aires, Argentina. .,Laboratorio de Virología Veterinaria, Instituto de Innovación para la Producción Agropecuaria y el Desarrollo Sostenible INTA CONICET, Balcarce, Argentina.
| | - María E Arias
- Laboratorio de Reproducción Centro de Biotecnologia Reproductiva CEBIOR-BIOREN Facultad de Ciencias Agrícolas y Forestales, Universidad de La Frontera, Temuco, Chile
| | - Germán G Kaiser
- Laboratorio de Biotecnología de la Reproducción, Instituto de Innovación para la Producción Agropecuaria y el Desarrollo Sostenible INTA CONICET, Balcarce, Argentina
| | - Nicolás C Mucci
- Laboratorio de Biotecnología de la Reproducción, Instituto de Innovación para la Producción Agropecuaria y el Desarrollo Sostenible INTA CONICET, Balcarce, Argentina
| | - Anselmo C Odeón
- Facultad de Ciencias Agrarias, Universidad Nacional de Mar del Plata, Balcarce, Argentina
| | - Ricardo N Felmer
- Laboratorio de Reproducción Centro de Biotecnologia Reproductiva CEBIOR-BIOREN Facultad de Ciencias Agrícolas y Forestales, Universidad de La Frontera, Temuco, Chile
| |
Collapse
|
3
|
Wydooghe E, Vandaele L, Heras S, De Sutter P, Deforce D, Peelman L, De Schauwer C, Van Soom A. Autocrine embryotropins revisited: how do embryos communicate with each other in vitro when cultured in groups? Biol Rev Camb Philos Soc 2015; 92:505-520. [PMID: 26608222 DOI: 10.1111/brv.12241] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 10/15/2015] [Accepted: 10/21/2015] [Indexed: 01/10/2023]
Abstract
In the absence of the maternal genital tract, preimplantation embryos can develop in vitro in culture medium where all communication with the oviduct or uterus is absent. In several mammalian species, it has been observed that embryos cultured in groups thrive better than those cultured singly. Here we argue that group-cultured embryos are able to promote their own development in vitro by the production of autocrine embryotropins that putatively serve as a communication tool. The concept of effective communication implies an origin, a signalling agent, and finally a recipient that is able to decode the message. We illustrate this concept by demonstrating that preimplantation embryos are able to secrete autocrine factors in several ways, including active secretion, passive outflow, or as messengers bound to a molecular vehicle or transported within extracellular vesicles. Likewise, we broaden the traditional view that inter-embryo communication is dictated mainly by growth factors, by discussing a wide range of other biochemical messengers including proteins, lipids, neurotransmitters, saccharides, and microRNAs, all of which can be exchanged among embryos cultured in a group. Finally, we describe how different classes of messenger molecules are decoded by the embryo and influence embryo development by triggering different pathways. When autocrine embryotropins such as insulin-like growth factor-I (IGF-I) or platelet activating factor (PAF) bind to their appropriate receptor, the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) pathway will be activated which is important for embryo survival. On the other hand, the mitogen-activated protein kinase (MAPK) pathway is activated when compounds such as hyaluronic acid and serotonin bind to their respective receptors, thereby acting as growth factors. By activating the peroxisome-proliferator-activated receptor family (PPAR) pathway, lipophilic autocrine factors such as prostaglandins or fatty acids have both survival and anti-apoptotic functions. In conclusion, considering different types of messenger molecules simultaneously will be crucial to understanding more comprehensively how embryos communicate with each other in group-culture systems. This approach will assist in the development of novel media for single-embryo culture.
Collapse
Affiliation(s)
- Eline Wydooghe
- Department of Reproduction, Obstetrics, and Herd Health, Faculty of Veterinary Medicine, Ghent University, B-9820, Merelbeke, Belgium
| | - Leen Vandaele
- Animal Sciences Unit, Institute for Agricultural and Fisheries Research (ILVO), B-9090, Melle, Belgium
| | - Sonia Heras
- Department of Reproduction, Obstetrics, and Herd Health, Faculty of Veterinary Medicine, Ghent University, B-9820, Merelbeke, Belgium
| | - Petra De Sutter
- Department of Reproductive Medicine, University Hospital, Ghent University, B-9000, Ghent, Belgium
| | - Dieter Deforce
- Laboratory for Pharmaceutical Biotechnology, Ghent University, B-9000, Ghent, Belgium
| | - Luc Peelman
- Laboratory of Animal Genetics, Faculty of Veterinary Medicine, Ghent University, B-9820, Merelbeke, Belgium
| | - Catharina De Schauwer
- Department of Reproduction, Obstetrics, and Herd Health, Faculty of Veterinary Medicine, Ghent University, B-9820, Merelbeke, Belgium
| | - Ann Van Soom
- Department of Reproduction, Obstetrics, and Herd Health, Faculty of Veterinary Medicine, Ghent University, B-9820, Merelbeke, Belgium
| |
Collapse
|
4
|
Wu FR, Ding B, Qi B, Shang MB, Yang XX, Liu Y, Li WY. Sequence analysis, expression patterns and transcriptional regulation of mouse Ifrg15 during preimplantation embryonic development. Gene 2012; 507:119-24. [PMID: 22871540 DOI: 10.1016/j.gene.2012.07.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 07/06/2012] [Accepted: 07/17/2012] [Indexed: 12/21/2022]
Abstract
Ifrg15 is a newly identified interferon alpha responsive gene and is implicated in a wide variety of physiological roles in mammals. In the present study, multiple alignments of the deduced amino acids of 10 eutherian mammalian IFRG15/Ifrg15s isolated from open genomic database revealed that they were highly conserved. Real-time PCR showed that mouse Ifrg15 mRNA was expressed in MII stage oocytes and preimplantation embryos, and its highest value peaked at the stage of mouse blastocysts. To understand the effect of three development-related genes on the promoter activity of mouse Ifrg15, promoter analysis using luciferase assays in COS-7 cells were performed. The results showed that the transcription of mouse Ifrg15 was suppressed by Oct4 and Nanog when transfected with the longest Ifrg15 promoter reporter gene. After the relatively shorter promoters were co-transfected with Oct4, c-Myc and Nanog, the relative luciferase activities of Ifrg15 were gradually increased. These in vitro results data and expression profiles of Ifrg15 as revealed by real-time PCR partly indicated that Ifrg15 transcription might be either potentially regulated or dependent on the post-transcriptional effects of IFN-α mediated by the three genes indirectly. Our data suggested that the mouse Ifrg15 might interact with these key development-related genes and play significant roles on the mouse preimplantation embryos development, especially for the development of mouse blastocysts.
Collapse
Affiliation(s)
- Feng-Rui Wu
- School of Life Science, Fuyang Teachers College, Anhui Province, Fuyang, China; Key Laboratory of Embryo Development and Reproductive Regulation, Anhui Province, Fuyang, China
| | | | | | | | | | | | | |
Collapse
|
5
|
In situ expression of 15 kDa interferon alpha responsive gene in the developing tooth germ of the mouse lower first molar. J Mol Histol 2010; 41:185-91. [DOI: 10.1007/s10735-010-9277-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2010] [Accepted: 07/02/2010] [Indexed: 12/18/2022]
|
6
|
Li L, Sherry B. IFN-alpha expression and antiviral effects are subtype and cell type specific in the cardiac response to viral infection. Virology 2009; 396:59-68. [PMID: 19896686 DOI: 10.1016/j.virol.2009.10.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Revised: 08/28/2009] [Accepted: 10/03/2009] [Indexed: 01/01/2023]
Abstract
The interferon-beta (IFN-beta) response is critical for protection against viral myocarditis in several mouse models, and IFN-alpha or -beta treatment is beneficial against human viral myocarditis. The IFN-beta response in cardiac myocytes and cardiac fibroblasts forms an integrated network for organ protection; however, the different IFN-alpha subtypes have not been studied in cardiac cells. We developed a quantitative RT-PCR assay that distinguishes between 13 highly conserved IFN-alpha subtypes and found that reovirus T3D induces five IFN-alpha subtypes in primary cardiac myocyte and fibroblast cultures: IFN-alpha1, -alpha2, -alpha4, -alpha5, and -alpha8/6. Murine IFN-alpha1, -alpha2, -alpha4, or -alpha5 treatment induced IRF7 and ISG56 and inhibited reovirus T3D replication in both cell types. This first investigation of IFN-alpha subtypes in cardiac cells for any virus demonstrates that IFN-alpha is induced in cardiac cells, that it is both subtype and cell type specific, and that it is likely important in the antiviral cardiac response.
Collapse
Affiliation(s)
- Lianna Li
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27606, USA
| | | |
Collapse
|
7
|
Xia C, Wang C, Zhang K, Qian C, Jing N. Induction of a high population of neural stem cells with anterior neuroectoderm characters from epiblast-like P19 embryonic carcinoma cells. Differentiation 2007; 75:912-27. [PMID: 17573917 DOI: 10.1111/j.1432-0436.2007.00188.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The epiblast, derived from the inner cell mass (ICM), represents the final embryonic founder cell population of mouse embryo and can give rise to all germ layer lineages including the neuroectoderm. The generation of neural stem cells from epiblast-like cells is of great value for studying the mechanism of neural determination during gastrulation stages of embryonic development. Mouse embryonic carcinoma (EC) P19 cells are equivalent to the epiblast of early post-implantation blastocysts. In this study, we establish a feasible induction system that allows rapid and efficient derivation of a high percentage ( approximately 95%) of neural stem cells from P19 EC cell in N2B27 serum-free medium. The induced neural stem cells bear anterior neuroectoderm characters, and can be efficiently caudalized by retinoic acid (RA). These neural stem cells have multilineage potential to differentiate into neurons, astrocytes, and oligodendrocytes. Mechanistic analysis indicates that inhibition of the bone morphogenetic protein (BMP) pathway may be the main reason for N2B27-neural induction, and that fibroblast growth factor (FGF) signaling is also involved in this process. This method will provide an in vitro system to dissect the molecular mechanisms involved in neural induction of early mouse embryos.
Collapse
Affiliation(s)
- Caihong Xia
- Laboratory of Molecular Cell Biology, Key Laboratory of Stem Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | |
Collapse
|
8
|
Tsang SL, Leung PC, Leung KK, Yau WL, Hardy MP, Mak NK, Leung KN, Fung MC. Characterization of murine interferon-alpha 12 (MuIFN-alpha12): biological activities and gene expression. Cytokine 2007; 37:138-49. [PMID: 17451966 DOI: 10.1016/j.cyto.2007.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2006] [Revised: 02/12/2007] [Accepted: 03/12/2007] [Indexed: 10/23/2022]
Abstract
Interferon alpha (IFN-alpha) belongs to the type I interferon family and consists of multiple subtypes in many species. In the mouse, there are at least 14 IFN-alpha genes and 3 IFN-alpha pseudogenes, the most recently identified of which are murine interferon-alpha 12 (MuIFN-alpha12), MuIFN-alpha13 and MuIFN-alpha14. To further study the biological activities of MuIFN-alpha12, we have produced a recombinant MuIFN-alpha12 (rMuIFN-alpha12) protein using COS-1 cells. rMuIFN-alpha12 was found to inhibit the growth of murine myeloid leukemia JCS cells. Flow cytofluorometric analysis with propidium iodide staining showed that the growth inhibitory activity of rMuIFN-alpha12 may be caused by the induction of apoptosis. Flow cytofluorometric analysis also revealed that rMuIFN-alpha12 was able to up-regulate the expression of MHC-I on both JCS cells and primary macrophages. Functional studies indicated that a MuIFN-alpha12 transgene could induce an anti-viral state in L929 cells against Influenza A virus. Moreover, expression of MuIFN-alpha12 was not detectable by RT-PCR in untreated, Influenza A virus infected, polyI:polyC induced L929 cells, or in a wide range of normal murine tissues. Taken together, this data shows that MuIFN-alpha12 is a protein with all the biological traits of a type I IFN.
Collapse
Affiliation(s)
- Sai Leong Tsang
- Department of Biology, The Chinese University of Hong Kong, Shatin, Hong Kong.
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Stein P, Zeng F, Pan H, Schultz RM. Absence of non-specific effects of RNA interference triggered by long double-stranded RNA in mouse oocytes. Dev Biol 2005; 286:464-71. [PMID: 16154556 DOI: 10.1016/j.ydbio.2005.08.015] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2005] [Revised: 08/11/2005] [Accepted: 08/12/2005] [Indexed: 11/27/2022]
Abstract
RNA interference (RNAi) is a conserved eukaryotic mechanism by which double-stranded RNA (dsRNA) triggers the sequence-specific degradation of homologous mRNAs. Recent concerns have arisen in mammalian systems about off-target effects of RNAi, as well as an interferon response. Most mammalian cells respond to long dsRNAs by inducing an antiviral response mediated by interferon that leads to general inhibition of protein synthesis and nonspecific degradation of mRNAs. Moreover, recent reports demonstrate that under certain conditions, short interfering RNAs (siRNAs, 21-25 bp) may activate the interferon system. Mouse oocytes and preimplantation embryos apparently lack this response, as potent and specific inhibition of gene expression triggered by long dsRNA is observed in these cells. In the present study, we analyzed the global pattern of gene expression by microarray analysis in transgenic mouse oocytes expressing long dsRNA and find no evidence of off-targeting. We also report that genes involved in the interferon response pathway are not expressed in mouse oocytes, even after exposure for an extended period of time to long dsRNA.
Collapse
Affiliation(s)
- Paula Stein
- Department of Biology, University of Pennsylvania, 415 South University Avenue, Philadelphia, PA 19104-6018, USA
| | | | | | | |
Collapse
|
10
|
van Pesch V, Lanaya H, Renauld JC, Michiels T. Characterization of the murine alpha interferon gene family. J Virol 2004; 78:8219-28. [PMID: 15254193 PMCID: PMC446145 DOI: 10.1128/jvi.78.15.8219-8228.2004] [Citation(s) in RCA: 160] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mouse and human genomes carry more than a dozen genes coding for closely related alpha interferon (IFN-alpha) subtypes. IFN-alpha, as well as IFN-beta, IFN-kappa, IFN-epsilon, and limitin, are thought to bind the same receptor, raising the question of whether different IFN subtypes possess specific functions. As some confusion existed in the identity and characteristics of mouse IFN-alpha subtypes, the availability of data from the mouse genome sequence prompted us to characterize the murine IFN-alpha family. A total of 14 IFN-alpha genes were detected in the mouse genome, in addition to three IFN-alpha pseudogenes. Four IFN-alpha genes (IFN-alpha1, IFN-alpha7/10, IFN-alpha8/6, and IFN-alpha11) exhibited surprising allelic divergence between 129/Sv and C57BL/6 mice. All IFN-alpha subtypes were found to be stable at pH 2 and to exhibit antiviral activity. Interestingly, some IFN subtypes (IFN-alpha4, IFN-alpha11, IFN-alpha12, IFN-beta, and limitin) showed higher biological activity levels than others, whereas IFN-alpha7/10 exhibited lower activity. Most murine IFN-alpha turned out to be N-glycosylated. However, no correlation was found between N-glycosylation and activity. The various IFN-alpha subtypes displayed a good correlation between their antiviral and antiproliferative potencies, suggesting that IFN-alpha subtypes did not diverge primarily to acquire specific biological activities but probably evolved to acquire specific expression patterns. In L929 cells, IFN genes activated in response to poly(I*C) transfection or to viral infection were, however, similar.
Collapse
Affiliation(s)
- Vincent van Pesch
- University of Louvain, Christian de Duve Institute of Cellular Pathology, MIPA-VIRO 74-49, B-1200 Brussels, Belgium
| | | | | | | |
Collapse
|
11
|
Truchet S, Chebrout M, Djediat C, Wietzerbin J, Debey P. Presence of permanently activated signal transducers and activators of transcription in nuclear interchromatin granules of unstimulated mouse oocytes and preimplantation embryos. Biol Reprod 2004; 71:1330-9. [PMID: 15189833 DOI: 10.1095/biolreprod.104.029405] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
We previously described that mouse oocytes and preimplantation embryos express the two subunits of interferon-gamma receptor. We now report that, despite the presence of STAT1 (signal transducer and activator of transcription 1) at both the mRNA and protein levels, interferon gamma (IFNgamma) as well as IFNalpha are unable to trigger massive nuclear translocation of STAT1 in these cells, even at high cytokine concentrations. Conversely, nuclear accumulation of STAT1 was readily observed in murine L929 somatic cells under the same conditions. However, in the absence of any stimulation, both tyrosine (Y701p) and serine (S727p) phosphorylated forms of STAT1 were already detected in the nuclei of oocytes and early embryos. Phosphorylated STAT1 appeared concentrated in large nuclear dots, which were identified by indirect immunofluorescence and electron microscopy as clusters of interchromatin granules (IGCs or speckles). A similar distribution was also observed for the serine (S727p) phosphorylated form of STAT3 as well as for tyrosine (Y689p) phosphorylated STAT2. Western blot analysis confirmed that STAT factors present in mouse oocytes are predominantly phosphorylated. In parallel, we showed that the transcription of two IFNgamma-target genes, namely interferon regulatory factor-1 (IRF-1) and suppressor of cytokine signaling-1 (SOCS-1) is indeed increased in two-cell embryos in response to IFNgamma. Altogether, our results suggest that, despite the lack of massive nuclear accumulation of STAT1 in response to exogenous IFNs and the permanent presence of phosphorylated STATs in the nucleus, JAK/ STAT pathways are functional during early development.
Collapse
Affiliation(s)
- Sandrine Truchet
- USM 503 MNHN, UMR 8646 CNRS-MNHN, U565 INSERM, Département Régulation, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, 75005 Paris, France.
| | | | | | | | | |
Collapse
|
12
|
Takahashi M, Takahashi H, Hamano S, Watanabe S, Inumaru S, Geshi M, Okuda K, Yokomizo Y, Okano A. Possible Role of Interferon-.TAU. on In Vitro Development of Bovine Embryos. J Reprod Dev 2003; 49:297-305. [PMID: 14967922 DOI: 10.1262/jrd.49.297] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The effect of interferon-tau on in vitro development of bovine embryos was investigated. After in vitro fertilization, embryos developed to the morula stage were cultured for 3 days in TCM-199 or CR1 medium containing BSA or FCS supplemented with or without recombinant IFN-tau produced by a baculovirus expression system. Addition of baculovirus-expressed IFN-tau (100 ng/ml) significantly promoted development to the blastocyst stage in both culture media. Addition of E. coli expressed IFN-tau (2 microg/ml) also significantly promoted the embryonic development. Supplementation of BSA or FCS did not affect the growth-promoting effect of IFN-tau. To determine whether the growth-promoting effect of IFN-tau is related to the interferon type I receptors that bind to type I interferon such as IFN-alpha, embryos were cultured with IFN-alpha. Although IFN-alpha significantly promoted the development, a much higher concentration (25 microg/ml) was required than IFN-tau. A reverse transcription polymerase chain reaction analysis revealed the expression of mRNA encoded type-I IFN receptor subunit from morula to blastocyst stage embryos. The overall results suggest a novel function for IFNs in promoting embryonic development and the effect may be related to type-I IFN receptor expressed in the early stages of preimplantation embryos.
Collapse
Affiliation(s)
- Masashi Takahashi
- National Agricultural Research Center for Kyushu Okinawa region, Kumamoto 861-1192, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Robert C, McGraw S, Massicotte L, Pravetoni M, Gandolfi F, Sirard MA. Quantification of housekeeping transcript levels during the development of bovine preimplantation embryos. Biol Reprod 2002; 67:1465-72. [PMID: 12390877 DOI: 10.1095/biolreprod.102.006320] [Citation(s) in RCA: 167] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
In mammals, the study of gene expression in the preimplantation embryo has been difficult because the standard procedures used to quantify mRNA generally require large amounts of starting material. The development of protocols using different quantitative strategies generally involving the polymerase chain reaction (PCR) has provided new tools for exploration of gene expression in preimplantation embryos. However, the use of an internal standard, often referred as a housekeeping gene, is essential to normalize the mRNA levels. RNA levels of eight housekeeping genes were quantified using real time PCR throughout the preimplantation period of the bovine embryo to find the most suitable gene to be used as standard. Histone H2a was the best internal standard because the transcript levels were constant across the preimplantation period. Linear amplification of antisense RNA using the T7 promotor for in vitro transcription of the entire RNA pool was evaluated as a suitable way to preamplify the starting material prior to quantification and was effective in providing accurate RNA abundance profiles throughout the preimplantation period. However, the amplification appears to be template dependent because the amplification factors were higher for some genes.
Collapse
Affiliation(s)
- Claude Robert
- Department of Animal Sciences, Centre de Recherche en Biologie de la Reproduction, Laval University, Québec, Canada G1K 7P4
| | | | | | | | | | | |
Collapse
|
14
|
Truchet S, Wietzerbin J, Debey P. Mouse oocytes and preimplantation embryos bear the two sub-units of interferon-gamma receptor. Mol Reprod Dev 2001; 60:319-30. [PMID: 11599043 DOI: 10.1002/mrd.1094] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Cytokines and growth factors play important roles in implantation and maintenance of pregnancy, but also during early development. Among them interferon-gamma (IFNgamma) is highly expressed by mammalian trophoblast cells during implantation and seems to be involved in some cases of pregnancy loss. In the present study we investigated the possible presence of IFNgamma receptors (IFNGR) on mouse oocytes and preimplantation embryos. The two receptor chains IFNgammaRalpha (IFNGR-1) and IFNgammaRbeta (IFNGR-2) have been detected by indirect immunofluorescence at the surface of mouse oocytes (in germinal vesicle and metaphase II stages), as well as at all stages of in vitro embryo development from the one-cell to blastocyst stage. IFNGR appeared to colocalize partly with ganglioside GM1 at the cell surface of oocytes and embryos, indicating a possible preferential localization of this receptor in "rafts" microdomains. This was analyzed in more detail using software developed in the laboratory. IFNgamma was found to bind to its receptor at all stages analyzed. RT-PCR and Southern blot experiments confirmed the presence of the transcriptionally regulated IFNGR-2 chain mRNA, in mouse oocytes and preimplantation embryos. These results show, for the first time, that mouse oocytes and preimplantation embryos bear a complete and theoretically functional IFNGR, suggesting that this cytokine could play a role during early development.
Collapse
MESH Headings
- Animals
- Blastocyst/immunology
- Blastocyst/metabolism
- Cleavage Stage, Ovum/immunology
- Cleavage Stage, Ovum/metabolism
- Female
- Fluorescent Antibody Technique, Indirect
- Interferon-gamma/metabolism
- Membrane Microdomains/immunology
- Membrane Microdomains/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Oocytes/immunology
- Oocytes/metabolism
- Pregnancy
- Protein Subunits
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Interferon/chemistry
- Receptors, Interferon/genetics
- Receptors, Interferon/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Interferon gamma Receptor
Collapse
Affiliation(s)
- S Truchet
- INRA 806/MNHN EA 2703, IFR 63 Muséum National d'Histoire Naturelle, Institut de Biologie Physico-Chimique, 13, rue P. et M. Curie, 75005 Paris, France
| | | | | |
Collapse
|
15
|
Li Q, Zhang M, Kumar S, Zhu LJ, Chen D, Bagchi MK, Bagchi IC. Identification and implantation stage-specific expression of an interferon-alpha-regulated gene in human and rat endometrium. Endocrinology 2001; 142:2390-400. [PMID: 11356686 DOI: 10.1210/endo.142.6.8101] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Implantation of the developing blastocyst is regulated by multiple effectors, such as steroid hormones, growth factors, and cytokines. To understand how these diverse signaling pathways interact to modulate uterine gene expression, we employed a gene expression screen technique to identify the molecules that are induced in the periimplantation rat uterus. Here we report the isolation of a complementary DNA representing a novel gene, interferon-regulated gene 1 (IRG1). This gene exhibits significant homology to interferon (IFN)-alpha/beta-inducible human genes p27 and 6-16, indicating that these genes may belong to the same family. Consistent with this finding, expression of IRG1 messenger RNA (mRNA) in rat uterus increased about 20-fold in response to IFNalpha. Uterine expression of IRG1 was also stimulated by estrogen and was partially inhibited by an antiestrogen, ICI 182,780. In pregnant rats, IRG1 expression was high on day 1, but declined on days 2 and 3. The level of IRG1 mRNA again rose transiently on day 4 immediately preceding implantation. In situ hybridization analysis localized the IRG1 mRNA expression in the endometrial epithelium and the surrounding stroma. Interestingly, the expression of p27, which shows high homology to IRG1, was strongly enhanced in human endometrium during the midsecretory phase of the menstrual cycle, overlapping the putative window of implantation. Both IRG1 and p27 mRNAs are therefore induced in the endometrium in an implantation stage-specific manner. We also observed a synergistic interaction between IFNalpha and estrogen receptor signaling pathways that led to maximal induction of p27 mRNA in Ishikawa cells. Although the functional roles of IRG1 and p27 remain unclear, we describe for the first time, identification of a gene family regulated by IFNalpha in both rodent and human uteri. More importantly, our studies reveal that a complex interplay between the steroid hormone and IFN pathways regulates the expression of these genes in the endometrium at the time of implantation.
Collapse
Affiliation(s)
- Q Li
- The Population Council and Rockefeller University, New York, NY 10021, USA
| | | | | | | | | | | | | |
Collapse
|
16
|
Aguirre A, Castro-Palomino N, De la Fuente J, Ovidio Castro FO. Expression of human erythropoietin transgenes and of the endogenous WAP gene in the mammary gland of transgenic rabbits during gestation and lactation. Transgenic Res 1998; 7:311-7. [PMID: 9859219 DOI: 10.1023/a:1008882332133] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
An understanding of the expression of transgenes in the mammary gland during gestation and lactation is crucial for the use of transgenic mammals as bioreactors. Here we describe the temporal pattern of expression of the endogenous rabbit WAP gene and human erythropoietin (hEPO) transgenes under the control of rabbit WAP promoter and 3' flanking sequences. The endogenous rabbit WAP gene was expressed throughout gestation including the day of mating, as well as during lactation in transgenic rabbits bearing a minigene construct. In non-pregnant cycling females, WAP expression was found independent of transgenic status; however, WAP expression was not detected in non-cycling females. The significance of this new finding is not clear at present. hEPO mRNA was detected in mammary gland biopsies from pregnant transgenic rabbits only on day 28 of gestation. During lactation, transcripts were present in mammary gland biopsy samples taken on days 0, 7, 14 and 21. A sharp decline in the levels of transcripts was found for an hEPOcDNA gene construct at the end of lactation (day 28). Although the levels of hEPO were too low to allow a conclusion to be drawn regarding temporal or position-dependent expression, this finding may reflect an integration position effect.
Collapse
Affiliation(s)
- A Aguirre
- Mammalian Cell Genetics Division, Centro de Ingeniería Genética y Biotechnología, La Habana, Cuba
| | | | | | | |
Collapse
|