1
|
Rapp K, Wei S, Roberts M, Yao S, Fei SS, Gao L, Ray K, Wang A, Godiah R, Han L. Transcriptional profiling of mucus production in rhesus macaque endocervical cells under hormonal regulation†. Biol Reprod 2024; 111:1045-1055. [PMID: 39115371 DOI: 10.1093/biolre/ioae121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/25/2024] [Accepted: 08/07/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE Endocervical mucus production is a key regulator of fertility throughout the menstrual cycle. With cycle-dependent variability in mucus quality and quantity, cervical mucus can either facilitate or block sperm ascension into the upper female reproductive tract. This study seeks to identify genes involved in the hormonal regulation of mucus production, modification, and regulation through profiling the transcriptome of endocervical cells from the non-human primate, the rhesus macaque (Macaca mulatta). INTERVENTION We treated differentiated primary endocervical cultures with estradiol (E2) and progesterone (P4) to mimic peri-ovulatory and luteal-phase hormonal changes. Using RNA-sequencing, we identified differential expression of gene pathways and mucus-producing and mucus-modifying genes in cells treated with E2 compared to hormone-free conditions and E2 compared to E2-primed cells treated with P4. MAIN OUTCOME MEASURES We pursued differential gene expression analysis on RNA-sequenced cells. Sequence validation was done using quantitative PCR (qPCR). RESULTS Our study identified 158 genes that show significant differential expression in E2-only conditions compared to hormone-free control and 250 genes that show significant differential expression in P4-treated conditions compared to E2-only conditions. From this list, we found hormone-induced changes in transcriptional profiles for genes across several classes of mucus production, including ion channels and enzymes involved in post-translational mucin modification that have not previously been described as hormonally regulated. CONCLUSION Our study is the first to use an in vitro culture system to create an epithelial cell-specific transcriptome of the endocervix. As a result, our study identifies new genes and pathways altered by sex steroids in cervical mucus production. SUMMARY SENTENCE In vitro hormonal regulation of mucus production, modification, and secretion was profiled using primary epithelial endocervical cells.
Collapse
Affiliation(s)
- Katrina Rapp
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Portland, OR, USA
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
| | - Shuhao Wei
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Portland, OR, USA
| | - Mackenzie Roberts
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Portland, OR, USA
| | - Shan Yao
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Portland, OR, USA
| | - Suzanne S Fei
- Bioinformatics & Biostatistics Core, Oregon National Primate Research Center, Portland, OR, USA
| | - Lina Gao
- Bioinformatics & Biostatistics Core, Oregon National Primate Research Center, Portland, OR, USA
| | - Karina Ray
- Bioinformatics & Biostatistics Core, Oregon National Primate Research Center, Portland, OR, USA
| | - Alexander Wang
- Bioinformatics & Biostatistics Core, Oregon National Primate Research Center, Portland, OR, USA
| | - Rachelle Godiah
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Portland, OR, USA
| | - Leo Han
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Portland, OR, USA
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
2
|
Zaniker EJ, Babayev E, Duncan FE. Common mechanisms of physiological and pathological rupture events in biology: novel insights into mammalian ovulation and beyond. Biol Rev Camb Philos Soc 2023; 98:1648-1667. [PMID: 37157877 PMCID: PMC10524764 DOI: 10.1111/brv.12970] [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: 08/11/2022] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/10/2023]
Abstract
Ovulation is a cyclical biological rupture event fundamental to fertilisation and endocrine function. During this process, the somatic support cells that surround the germ cell undergo a remodelling process that culminates in breakdown of the follicle wall and release of a mature egg. Ovulation is driven by known proteolytic and inflammatory pathways as well as structural alterations to the follicle vasculature and the fluid-filled antral cavity. Ovulation is one of several types of systematic remodelling that occur in the human body that can be described as rupture. Although ovulation is a physiological form of rupture, other types of rupture occur in the human body which can be pathological, physiological, or both. In this review, we use intracranial aneurysms and chorioamniotic membrane rupture as examples of rupture events that are pathological or both pathological and physiological, respectively, and compare these to the rupture process central to ovulation. Specifically, we compared existing transcriptomic profiles, immune cell functions, vascular modifications, and biomechanical forces to identify common processes that are conserved between rupture events. In our transcriptomic analysis, we found 12 differentially expressed genes in common among two different ovulation data sets and one intracranial aneurysm data set. We also found three genes that were differentially expressed in common for both ovulation data sets and one chorioamniotic membrane rupture data set. Combining analysis of all three data sets identified two genes (Angptl4 and Pfkfb4) that were upregulated across rupture systems. Some of the identified genes, such as Rgs2, Adam8, and Lox, have been characterised in multiple rupture contexts, including ovulation. Others, such as Glul, Baz1a, and Ddx3x, have not yet been characterised in the context of ovulation and warrant further investigation as potential novel regulators. We also identified overlapping functions of mast cells, macrophages, and T cells in the process of rupture. Each of these rupture systems share local vasoconstriction around the rupture site, smooth muscle contractions away from the site of rupture, and fluid shear forces that initially increase and then decrease to predispose one specific region to rupture. Experimental techniques developed to study these structural and biomechanical changes that underlie rupture, such as patient-derived microfluidic models and spatiotemporal transcriptomic analyses, have not yet been comprehensively translated to the study of ovulation. Review of the existing knowledge, transcriptomic data, and experimental techniques from studies of rupture in other biological systems yields a better understanding of the physiology of ovulation and identifies avenues for novel studies of ovulation with techniques and targets from the study of vascular biology and parturition.
Collapse
Affiliation(s)
- Emily J. Zaniker
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 10-109, Chicago, IL 60611, USA
| | - Elnur Babayev
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 10-109, Chicago, IL 60611, USA
| | - Francesca E. Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 10-109, Chicago, IL 60611, USA
| |
Collapse
|
3
|
Xu Q, Yao M, Tang C. RGS2 and female common diseases: a guard of women's health. J Transl Med 2023; 21:583. [PMID: 37649067 PMCID: PMC10469436 DOI: 10.1186/s12967-023-04462-3] [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: 07/05/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023] Open
Abstract
Currently, women around the world are still suffering from various female common diseases with the high incidence, such as ovarian cancer, uterine fibroids and preeclampsia (PE), and some diseases are even with the high mortality rate. As a negative feedback regulator in G Protein-Coupled Receptor signaling (GPCR), the Regulator of G-protein Signaling (RGS) protein family participates in regulating kinds of cell biological functions by destabilizing the enzyme-substrate complex through the transformation of hydrolysis of G Guanosine Triphosphate (GTP). Recent work has indicated that, the Regulator of G-protein Signaling 2 (RGS2), a member belonging to the RGS protein family, is closely associated with the occurrence and development of certain female diseases, providing with the evidence that RGS2 functions in sustaining women's health. In this review paper, we summarize the current knowledge of RGS2 in female common diseases, and also tap and discuss its therapeutic potential by targeting multiple mechanisms.
Collapse
Affiliation(s)
- Qiang Xu
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd, Hangzhou, 310052, People's Republic of China
| | - Mukun Yao
- Department of Gynecology, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China
| | - Chao Tang
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd, Hangzhou, 310052, People's Republic of China.
| |
Collapse
|
4
|
Rapp K, Wei S, Roberts M, Yao S, Fei SS, Gao L, Ray K, Wang A, Godiah R, Han L. Transcriptional profiling of mucus production and modification in rhesus macaque endocervical cells under hormonal regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.18.541362. [PMID: 37292621 PMCID: PMC10245652 DOI: 10.1101/2023.05.18.541362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Objective Endocervical mucus production is a key regulator of fertility throughout the menstrual cycle. With cycle-dependent variability in mucus quality and quantity, cervical mucus can either facilitate or block sperm ascension into the upper female reproductive tract. This study seeks to identify genes involved in the hormonal regulation of mucus production, modification, and regulation through profiling the transcriptome of endocervical cells from the non-human primate, the Rhesus Macaque (Macaca mulatta). Design Experimental. Setting Translational science laboratory. Intervention We treated differentiated primary endocervical cultures with estradiol (E2) and progesterone (P4) to mimic peri-ovulatory and luteal-phase hormonal changes. Using RNA-sequencing, we identified differential expression of gene pathways and mucus producing and modifying genes in cells treated with E2 compared to hormone-free conditions and E2 compared to E2-primed cells treated with P4. Main Outcome Measures We pursued differential gene expression analysis on RNA-sequenced cells. Sequence validation was done using qPCR. Results Our study identified 158 genes that show significant differential expression in E2-only conditions compared to hormone-free control, and 250 genes that show significant differential expression in P4-treated conditions compared to E2-only conditions. From this list, we found hormone-induced changes in transcriptional profiles for genes across several classes of mucus production, including ion channels and enzymes involved in post-translational mucin modification that have not previously been described as hormonally regulated. Conclusion Our study is the first to use an in vitro culture system to create an epithelial-cell specific transcriptome of the endocervix. As a result, our study identifies new genes and pathways that are altered by sex-steroids in cervical mucus production.
Collapse
|
5
|
Brady K, Liu HC, Hicks J, Long JA, Porter TE. Global gene expression analysis of the turkey hen hypothalamo-pituitary-gonadal axis during the preovulatory hormonal surge. Poult Sci 2023; 102:102547. [PMID: 36878099 PMCID: PMC10006860 DOI: 10.1016/j.psj.2023.102547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/04/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
The preovulatory hormonal surge (PS) consists of elevated circulating luteinizing hormone (LH) and progesterone levels and serves as the primary trigger for ovarian follicle ovulation. Increased LH and progesterone, produced by the pituitary and the granulosa layer of the largest ovarian follicle (F1), respectively, result from hypothalamic stimulation and steroid hormone feedback on the hypothalamo-pituitary-gonadal (HPG) axis. The hypothalamus, pituitary, F1 granulosa, and granulosa layer of the fifth largest follicle (F5) were isolated from converter turkey hens outside and during the PS and subjected to RNA sequencing (n = 6 per tissue). Differentially expressed genes were subjected to functional annotation using DAVID and IPA. A total of 12, 250, 1235, and 1938 DEGs were identified in the hypothalamus, pituitary, F1 granulosa, and F5 granulosa respectively (q<0.05, |fold change|>1.5, FPKM>1). Gene Ontology (GO) analysis revealed key roles for metabolic processes, steroid hormone feedback, and hypoxia induced gene expression changes. Upstream analysis identified a total of 4, 42, 126, and 393 potential regulators of downstream gene expression in the hypothalamus, pituitary, F1G, and F5G respectively, with a total of 63 potential regulators exhibiting differential expression between samples collected outside and during the PS (|z-score|>2). The results from this study serve to increase the current knowledge base surrounding the regulation of the PS in turkey hens. Through GO analysis, downstream processes and functions associated with the PS were linked to identified DEGs, and through upstream analysis, potential regulators of DEGs were identified for further analysis. Linking upstream regulators to the downstream PS and ovulation events could allow for genetic selection or manipulation of ovulation frequencies in turkey hens.
Collapse
Affiliation(s)
- Kristen Brady
- Animal Biosciences and Biotechnology Laboratory, BARC, ARS, USDA, Beltsville, MD 20705, United States.
| | - Hsiao-Ching Liu
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, United States
| | - Julie Hicks
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, United States
| | - Julie A Long
- Animal Biosciences and Biotechnology Laboratory, BARC, ARS, USDA, Beltsville, MD 20705, United States
| | - Tom E Porter
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, United States
| |
Collapse
|
6
|
Gholizadeh M, Esmaeili-Fard SM. Meta-analysis of genome-wide association studies for litter size in sheep. Theriogenology 2021; 180:103-112. [PMID: 34968818 DOI: 10.1016/j.theriogenology.2021.12.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 12/19/2021] [Accepted: 12/19/2021] [Indexed: 01/01/2023]
Abstract
Litter size and ovulation rate are important reproduction traits in sheep and have important impacts on the profitability of farm animals. To investigate the genetic architecture of litter size, we report the first meta-analysis of genome-wide association studies (GWAS) using 522 ewes and 564,377 SNPs from six sheep breeds. We identified 29 significant associations for litter size which 27 of which have not been reported in individual GWAS for each population. However, we could confirm the role of BMPR1B in prolificacy. Our gene set analysis discovered biological pathways related to cell signaling, communication, and adhesion. Functional clustering and enrichment using protein databases identified epidermal growth factor-like domain affecting litter size. Through analyzing protein-protein interaction data, we could identify hub genes like CASK, PLCB4, RPTOR, GRIA2, and PLCB1 that were enriched in most of the significant pathways. These genes have a role in cell proliferation, cell adhesion, cell growth and survival, and autophagy. Notably, identified SNPs were scattered on several different chromosomes implying different genetic mechanisms underlying variation of prolificacy in each breed. Given the different layers that make up the follicles and the need for communication and transfer of hormones and nutrients through these layers to the oocyte, the significance of pathways related to cell signaling and communication seems logical. Our results provide genetic insights into the litter size variation in different sheep breeds.
Collapse
Affiliation(s)
- Mohsen Gholizadeh
- Department of Animal Science, Faculty of Animal Science and Fisheries, Sari Agricultural Sciences and Natural Resources University, Sari, Iran.
| | - Seyed Mehdi Esmaeili-Fard
- Department of Animal Science, Faculty of Animal Science and Fisheries, Sari Agricultural Sciences and Natural Resources University, Sari, Iran
| |
Collapse
|
7
|
Choi Y, Jeon H, Akin JW, Curry TE, Jo M. The FOS/AP-1 Regulates Metabolic Changes and Cholesterol Synthesis in Human Periovulatory Granulosa Cells. Endocrinology 2021; 162:6309635. [PMID: 34171102 PMCID: PMC8315293 DOI: 10.1210/endocr/bqab127] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Indexed: 11/19/2022]
Abstract
FOS, a subunit of the activator protein-1 (AP-1) transcription factor, has been implicated in various cellular changes. In the human ovary, the expression of FOS and its heterodimeric binding partners JUN, JUNB, and JUND increases in periovulatory follicles. However, the specific role of the FOS/AP-1 remains elusive. The present study determined the regulatory mechanisms driving the expression of FOS and its partners and functions of FOS using primary human granulosa/lutein cells (hGLCs). Human chorionic gonadotropin (hCG) induced a biphasic increase in the expression of FOS, peaking at 1 to 3 hours and 12 hours. The levels of JUN proteins were also increased by hCG, with varying expression patterns. Coimmunoprecipitation analyses revealed that FOS is present as heterodimers with all JUN proteins. hCG immediately activated protein kinase A and p42/44MAPK signaling pathways, and inhibitors for these pathways abolished hCG-induced increases in the levels of FOS, JUN, and JUNB. To identify the genes regulated by FOS, high-throughput RNA sequencing was performed using hGLC treated with hCG ± T-5224 (FOS inhibitor). Sequencing data analysis revealed that FOS inhibition affects the expression of numerous genes, including a cluster of genes involved in the periovulatory process such as matrix remodeling, prostaglandin synthesis, glycolysis, and cholesterol biosynthesis. Quantitative PCR analysis verified hCG-induced, T-5224-regulated expression of a selection of genes involved in these processes. Consistently, hCG-induced increases in metabolic activities and cholesterol levels were suppressed by T-5224. This study unveiled potential downstream target genes of and a role for the FOS/AP-1 complex in metabolic changes and cholesterol biosynthesis in granulosa/lutein cells of human periovulatory follicles.
Collapse
Affiliation(s)
- Yohan Choi
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Hayce Jeon
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | | | - Thomas E Curry
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Misung Jo
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
- Correspondence: Misung Jo, PhD, Department of Obstetrics and Gynecology, Chandler Medical Center, 800 Rose Street, University of Kentucky, Lexington, KY 40536-0298, USA.
| |
Collapse
|
8
|
Yao YC, Song XT, Zhai YF, Liu S, Lu J, Xu X, Qi MY, Zhang JN, Huang H, Liu YF, Liu GS, Yuan H. Transcriptome analysis of sheep follicular development during prerecruitment, dominant, and mature stages after FSH superstimulation. Domest Anim Endocrinol 2021; 74:106563. [PMID: 33129139 DOI: 10.1016/j.domaniend.2020.106563] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/08/2020] [Accepted: 09/08/2020] [Indexed: 12/29/2022]
Abstract
Sheep is usually a monovular animal; superovulation technology is used to increase the number of offspring per individual and shorten generation intervals. To date, mature FSH superstimulatory treatments have been successfully used in sheep breeding, but much remains unknown about genes, pathways, and biological functions involved in follicular development. Therefore, in this study, we performed transcriptome profiling of small follicles (SFs; 2-2.5 mm), medium follicles (MFs; 3.5-4.5 mm), and large follicles (LFs; > 6 mm) in Mongolian ewes after FSH superstimulation. Furthermore, we identified differentially expressed genes and performed Kyoto Encyclopedia of Genes and Genomes pathway and Gene Ontology enrichment analyses in 3 separate pairwise comparisons. We found that ovarian steroidogenesis was significantly enriched in the SFs versus MFs analysis; the associated genes, cytochrome P450 family 19 (CYP19) and Hydroxy-delta-5-steroid dehydrogenase 3 beta- and steroid delta-isomerase 1 (HSD3B1), were significantly upregulated. Moreover, proline metabolism, glutathione metabolism, and PPAR signaling pathways were significantly enriched in the LFs versus SFs analysis; the associated genes, glutamate-cysteine ligase modifier subunit (GCLM) and cystathionine gamma-lyase (CTH), were significantly upregulated, whereas peroxisome proliferator-activated receptor gamma (PPARγ) was significantly downregulated. In summary, our study provides basic data and possible biological direction to further explore the molecular mechanism of sheep follicular development after FSH superstimulation.
Collapse
Affiliation(s)
- Y C Yao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang province, Harbin 150030, Heilongjiang, China
| | - X T Song
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang province, Harbin 150030, Heilongjiang, China
| | - Y F Zhai
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang province, Harbin 150030, Heilongjiang, China
| | - S Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang province, Harbin 150030, Heilongjiang, China
| | - J Lu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang province, Harbin 150030, Heilongjiang, China
| | - X Xu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang province, Harbin 150030, Heilongjiang, China
| | - M Y Qi
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, Heilongjiang, China
| | - J N Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang province, Harbin 150030, Heilongjiang, China
| | - H Huang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang province, Harbin 150030, Heilongjiang, China
| | - Y F Liu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, Heilongjiang, China
| | - G S Liu
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - H Yuan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang province, Harbin 150030, Heilongjiang, China.
| |
Collapse
|
9
|
Selvaraj S, Mondragon-Gonzalez R, Xu B, Magli A, Kim H, Lainé J, Kiley J, Mckee H, Rinaldi F, Aho J, Tabti N, Shen W, Perlingeiro RCR. Screening identifies small molecules that enhance the maturation of human pluripotent stem cell-derived myotubes. eLife 2019; 8:e47970. [PMID: 31710288 PMCID: PMC6845233 DOI: 10.7554/elife.47970] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 10/29/2019] [Indexed: 01/14/2023] Open
Abstract
Targeted differentiation of pluripotent stem (PS) cells into myotubes enables in vitro disease modeling of skeletal muscle diseases. Although various protocols achieve myogenic differentiation in vitro, resulting myotubes typically display an embryonic identity. This is a major hurdle for accurately recapitulating disease phenotypes in vitro, as disease commonly manifests at later stages of development. To address this problem, we identified four factors from a small molecule screen whose combinatorial treatment resulted in myotubes with enhanced maturation, as shown by the expression profile of myosin heavy chain isoforms, as well as the upregulation of genes related with muscle contractile function. These molecular changes were confirmed by global chromatin accessibility and transcriptome studies. Importantly, we also observed this maturation in three-dimensional muscle constructs, which displayed improved in vitro contractile force generation in response to electrical stimulus. Thus, we established a model for in vitro muscle maturation from PS cells.
Collapse
Affiliation(s)
- Sridhar Selvaraj
- Lillehei Heart Institute, Department of MedicineUniversity of MinnesotaMinneapolisUnited States
| | - Ricardo Mondragon-Gonzalez
- Lillehei Heart Institute, Department of MedicineUniversity of MinnesotaMinneapolisUnited States
- Departamento de Genética y Biología MolecularCentro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN)Ciudad de MéxicoMexico
| | - Bin Xu
- Department of Biomedical EngineeringUniversity of MinnesotaMinneapolisUnited States
| | - Alessandro Magli
- Lillehei Heart Institute, Department of MedicineUniversity of MinnesotaMinneapolisUnited States
- Stem Cell InstituteUniversity of MinnesotaMinneapolisUnited States
| | - Hyunkee Kim
- Lillehei Heart Institute, Department of MedicineUniversity of MinnesotaMinneapolisUnited States
| | - Jeanne Lainé
- Département de PhysiologieSorbonne Universités, Faculté de Médecine site Pitié-SalpêtrièreParisFrance
| | - James Kiley
- Lillehei Heart Institute, Department of MedicineUniversity of MinnesotaMinneapolisUnited States
| | - Holly Mckee
- Lillehei Heart Institute, Department of MedicineUniversity of MinnesotaMinneapolisUnited States
| | | | - Joy Aho
- Stem Cell DepartmentBio-TechneMinneapolisUnited States
| | - Nacira Tabti
- Département de PhysiologieSorbonne Universités, Faculté de Médecine site Pitié-SalpêtrièreParisFrance
| | - Wei Shen
- Lillehei Heart Institute, Department of MedicineUniversity of MinnesotaMinneapolisUnited States
- Department of Biomedical EngineeringUniversity of MinnesotaMinneapolisUnited States
- Stem Cell InstituteUniversity of MinnesotaMinneapolisUnited States
| | - Rita CR Perlingeiro
- Lillehei Heart Institute, Department of MedicineUniversity of MinnesotaMinneapolisUnited States
- Stem Cell InstituteUniversity of MinnesotaMinneapolisUnited States
| |
Collapse
|
10
|
Baufeld A, Koczan D, Vanselow J. L-lactate induces specific genome wide alterations of gene expression in cultured bovine granulosa cells. BMC Genomics 2019; 20:273. [PMID: 30953450 PMCID: PMC6451254 DOI: 10.1186/s12864-019-5657-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 03/29/2019] [Indexed: 12/11/2022] Open
Abstract
Background Previously, we could show that L-lactate affects cultured bovine granulosa cells (GC) in a specific manner driving the cells into an early pre-ovulatory phenotype. Here we studied genome wide effects in L-lactate-treated GC to further elucidate the underlying mechanisms that are responsible for the L-lactate induced transformation. Cultured estrogen producing GC treated either with L-lactate or vehicle control were subjected to mRNA microarray analysis. Results The analysis revealed 487 differentially expressed clusters, representing 461 annotated genes. Of these, 333 (= 318 genes) were identified as up- and 154 (= 143 genes) as down-regulated. As the top up-regulated genes we detected TXNIP, H19 and AHSG as well as our previously established marker transcripts RGS2 and PTX3. The top down-regulated genes included VNN1, SLC27A2 and GFRA1, but also MYC and the GC marker transcript CYP19A1. Pathway analysis with differentially expressed genes indicated “cAMP-mediated signaling” and “Axon guidance signaling” among the most affected pathways. Furthermore, estradiol, progesterone and Vegf were identified as potential upstream regulators. An effector network analysis by IPA provided first hints that processes of “angiogenesis” and “vascularization”, but also “cell movement” appeared to be activated, whereas “organismal death” was predicted to be inhibited. Conclusions Our data clearly show that L-lactate alters gene expression in cultured bovine GC in a broad, but obviously specific manner. Pathway analysis revealed that the mode of L-lactate action in GC initiates angiogenic processes, but also migratory events like cell movement and axonal guidance signaling, thus supporting the transformation of GC into an early luteal phenotype. Electronic supplementary material The online version of this article (10.1186/s12864-019-5657-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anja Baufeld
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Dirk Koczan
- Institute for Immunology, University of Rostock, 18055, Rostock, Germany
| | - Jens Vanselow
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| |
Collapse
|
11
|
Gérard N, Robin E. Cellular and molecular mechanisms of the preovulatory follicle differenciation and ovulation: What do we know in the mare relative to other species. Theriogenology 2019; 130:163-176. [PMID: 30921545 DOI: 10.1016/j.theriogenology.2019.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 02/19/2019] [Accepted: 03/14/2019] [Indexed: 02/06/2023]
Abstract
Terminal follicular differentiation and ovulation are essential steps of reproduction. They are induced by the increase in circulating LH, and lead to the expulsion from the ovary of oocytes ready to be fertilized. This review summarizes our current understanding of cellular and molecular pathways that control ovulation using a broad mammalian literature, with a specific focus to the mare, which is unique in some aspects of ovarian function in some cases. Essential steps and key factors are approached. The first part of this review concerns LH, receptors and signaling, addressing the description of the equine gonadotropin and cloning, signaling pathways that are activated following the binding of LH to its receptors, and implication of transcription factors which better known are CCAAT-enhancer-binding proteins (CEBP) and cAMP response element-binding protein (CREB). The second and major part is devoted to the cellular and molecular actors within follicular cells during preovulatory maturation. We relate to 1) molecules involved in vascular permeability and vasoconstriction, 2) involvement of neuropeptides, such as kisspeptin, neurotrophins and neuronal growth factor, neuropeptide Y (NPY), 3) the modification of steroidogenesis, steroids intrafollicular levels and enzymes activity, 4) the local inflammation, with the increase in prostaglandins synthesis, and implication of leukotrienes, cytokines and glucocorticoids, 5) extracellular matrix remodelling with involvement of proteases, antiproteases and inhibitors, as well as relaxin, and finaly 6) the implication of oxytocine, osteopontin, growth factors and reactive oxygen species. The third part describes our current knowledge on molecular aspect of in vivo cumulus-oocyte-complexe maturation, with a specific focus on signaling pathways, paracrine factors, and intracellular regulations that occur in cumulus cells during expansion, and in the oocyte during nuclear and cytoplasmic meiosis resumption. Our aim was to give an overall and comprehensive map of the regulatory mechanisms that intervene within the preovulatory follicle during differentiation and ovulation.
Collapse
Affiliation(s)
- Nadine Gérard
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France.
| | - Elodie Robin
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| |
Collapse
|
12
|
Schuermann Y, Rovani MT, Gasperin B, Ferreira R, Ferst J, Madogwe E, Gonçalves PB, Bordignon V, Duggavathi R. ERK1/2-dependent gene expression in the bovine ovulating follicle. Sci Rep 2018; 8:16170. [PMID: 30385793 PMCID: PMC6212447 DOI: 10.1038/s41598-018-34015-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/30/2018] [Indexed: 02/06/2023] Open
Abstract
Ovulation is triggered by gonadotropin surge-induced signaling cascades. To study the role of extracellular signal-regulated kinase 1/2 (ERK1/2) in bovine ovulation, we administered the pharmacological inhibitor, PD0325901, into the preovulatory dominant follicle by intrafollicular injection. Four of five cows treated with 50 µM PD0325901 failed to ovulate. To uncover the molecular basis of anovulation in ERK1/2-inhibited cows, we collected granulosa and theca cells from Vehicle and PD0325901 treated follicles. Next-generation sequencing of granulosa cell RNA revealed 285 differentially expressed genes between Vehicle and PD0325901-treated granulosa cells at 6 h post-GnRH. Multiple inflammation-related pathways were enriched among the differentially expressed genes. The ERK1/2 dependent LH-induced genes in granulosa cells included EGR1, ADAMTS1, STAT3 and TNFAIP6. Surprisingly, PD0325901 treatment did not affect STAR expression in granulosa cells at 6 h post-GnRH. Granulosa cells had higher STAR protein and theca cells had higher levels of STAR mRNA in ERK1/2-inhibited follicles. Further, both granulosa and theca cells of ERK1/2-inhibited follicles had higher expression of SLC16A1, a monocarboxylate transporter, transporting substances including β-hydroxybutyrate across the plasma membrane. Taken together, ERK1/2 plays a significant role in mediating LH surge-induced gene expression in granulosa and theca cells of the ovulating follicle in cattle.
Collapse
Affiliation(s)
- Yasmin Schuermann
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC, H9X 3V9, Canada
| | - Monique T Rovani
- Laboratory of Biotechnology and Animal Reproduction, BioRep, Veterinary Hospital, Federal University of Santa Maria, Santa Maria, 97105-900, Brazil
| | - Bernardo Gasperin
- Laboratory of Animal Reproduction-ReproPEL, Federal University of Pelotas, 96010-610, Capão do Leão, Brazil
| | - Rogério Ferreira
- Department of Animal Science, Santa Catarina State University, Santa Catarina, 88040-900, Brazil
| | - Juliana Ferst
- Laboratory of Biotechnology and Animal Reproduction, BioRep, Veterinary Hospital, Federal University of Santa Maria, Santa Maria, 97105-900, Brazil
| | - Ejimedo Madogwe
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC, H9X 3V9, Canada
| | - Paulo B Gonçalves
- Laboratory of Biotechnology and Animal Reproduction, BioRep, Veterinary Hospital, Federal University of Santa Maria, Santa Maria, 97105-900, Brazil
| | - Vilceu Bordignon
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC, H9X 3V9, Canada
| | - Raj Duggavathi
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC, H9X 3V9, Canada.
| |
Collapse
|
13
|
Yeh HY, Sun D, Peng YC, Wu YL. Regulation of the regulator of G protein signaling 2 expression and cellular localization by PKA and PKC pathways in mouse granulosa cells. Biochem Biophys Res Commun 2018; 503:950-955. [PMID: 29932914 DOI: 10.1016/j.bbrc.2018.06.101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 06/19/2018] [Indexed: 12/18/2022]
Abstract
G protein-coupled receptor (GPCR) activation-mediated PKA and PKC pathways have been recognized to be important in ovarian physiology. Expression of regulator of G-protein signaling 2 (RGS2) has been reported in ovarian granulosa cells. The detailed mechanisms in PKA- and PKC-regulated RGS2 expression and cellular translocation in granulosa cells remain mostly unclear. PKA activator 8-bromo-cAMP and PKC activator phorbol-12, 13-didecanoate appeared to rapidly elevate both protein and mRNA levels and promoter activation of RGS2 gene. Two consensus Sp1 elements within the shortest 78 bp fragment of RGS2 promoter sequence were essential for the full responsiveness to PKA and PKC. PKC activation appeared to increase the RGS2 translocation from nucleus to cytosol. PKA- and PKC-mediated RGS2 transcription in a Sp-1-dependent manner and a PKC-mediated RGS2 intracellular translocation were noted in granulosa cells.
Collapse
Affiliation(s)
- Hsiao-Yu Yeh
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - David Sun
- Department of Obstetrics and Gynecology, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Yen-Chun Peng
- Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yuh-Lin Wu
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
14
|
Perschbacher KJ, Deng G, Fisher RA, Gibson-Corley KN, Santillan MK, Grobe JL. Regulators of G protein signaling in cardiovascular function during pregnancy. Physiol Genomics 2018; 50:590-604. [PMID: 29702036 PMCID: PMC6139632 DOI: 10.1152/physiolgenomics.00037.2018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
G protein-coupled receptor signaling mechanisms are implicated in many aspects of cardiovascular control, and dysfunction of such signaling mechanisms is commonly associated with disease states. Investigators have identified a large number of regulator of G protein signaling (RGS) proteins that variously contribute to the modulation of intracellular second-messenger signaling kinetics. These many RGS proteins each interact with a specific set of second-messenger cascades and receptor types and exhibit tissue-specific expression patterns. Increasing evidence supports the contribution of RGS proteins, or their loss, in the pathogenesis of cardiovascular dysfunctions. This review summarizes the current understanding of the functional contributions of RGS proteins, particularly within the B/R4 family, in cardiovascular disorders of pregnancy including gestational hypertension, uterine artery dysfunction, and preeclampsia.
Collapse
Affiliation(s)
| | - Guorui Deng
- Department of Pharmacology, University of Iowa , Iowa City, Iowa
| | - Rory A Fisher
- Department of Pharmacology, University of Iowa , Iowa City, Iowa
| | - Katherine N Gibson-Corley
- Department of Pathology, University of Iowa , Iowa City, Iowa
- UIHC Center for Hypertension Research, University of Iowa , Iowa City, Iowa
| | - Mark K Santillan
- Department of Obstetrics & Gynecology, University of Iowa , Iowa City, Iowa
- UIHC Center for Hypertension Research, University of Iowa , Iowa City, Iowa
- Abboud Cardiovascular Research Center, University of Iowa , Iowa City, Iowa
| | - Justin L Grobe
- Department of Pharmacology, University of Iowa , Iowa City, Iowa
- UIHC Center for Hypertension Research, University of Iowa , Iowa City, Iowa
- Abboud Cardiovascular Research Center, University of Iowa , Iowa City, Iowa
- Fraternal Order of Eagles' Diabetes Research Center, University of Iowa , Iowa City, Iowa
- Obesity Education & Research Initiative, University of Iowa , Iowa City, Iowa
- Iowa Neuroscience Institute, University of Iowa , Iowa City, Iowa
| |
Collapse
|
15
|
Xu SS, Gao L, Xie XL, Ren YL, Shen ZQ, Wang F, Shen M, Eyϸórsdóttir E, Hallsson JH, Kiseleva T, Kantanen J, Li MH. Genome-Wide Association Analyses Highlight the Potential for Different Genetic Mechanisms for Litter Size Among Sheep Breeds. Front Genet 2018; 9:118. [PMID: 29692799 PMCID: PMC5902979 DOI: 10.3389/fgene.2018.00118] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/23/2018] [Indexed: 12/11/2022] Open
Abstract
Reproduction is an important trait in sheep breeding as well as in other livestock. However, despite its importance the genetic mechanisms of litter size in domestic sheep (Ovis aries) are still poorly understood. To explore genetic mechanisms underlying the variation in litter size, we conducted multiple independent genome-wide association studies in five sheep breeds of high prolificacy (Wadi, Hu, Icelandic, Finnsheep, and Romanov) and one low prolificacy (Texel) using the Ovine Infinium HD BeadChip, respectively. We identified different sets of candidate genes associated with litter size in different breeds: BMPR1B, FBN1, and MMP2 in Wadi; GRIA2, SMAD1, and CTNNB1 in Hu; NCOA1 in Icelandic; INHBB, NF1, FLT1, PTGS2, and PLCB3 in Finnsheep; ESR2 in Romanov and ESR1, GHR, ETS1, MMP15, FLI1, and SPP1 in Texel. Further annotation of genes and bioinformatics analyses revealed that different biological pathways could be involved in the variation in litter size of females: hormone secretion (FSH and LH) in Wadi and Hu, placenta and embryonic lethality in Icelandic, folliculogenesis and LH signaling in Finnsheep, ovulation and preovulatory follicle maturation in Romanov, and estrogen and follicular growth in Texel. Taken together, our results provide new insights into the genetic mechanisms underlying the prolificacy trait in sheep and other mammals, suggesting targets for selection where the aim is to increase prolificacy in breeding projects.
Collapse
Affiliation(s)
- Song-Song Xu
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Lei Gao
- Institute of Animal Husbandry and Veterinary Medicine, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
- State Key Laboratory of Sheep Genetic Improvement and Healthy Breeding, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
| | - Xing-Long Xie
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yan-Ling Ren
- Shandong Binzhou Academy of Animal Science and Veterinary Medicine Academy, Binzhou, China
| | - Zhi-Qiang Shen
- Shandong Binzhou Academy of Animal Science and Veterinary Medicine Academy, Binzhou, China
| | - Feng Wang
- Institute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing, China
| | - Min Shen
- Institute of Animal Husbandry and Veterinary Medicine, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
- State Key Laboratory of Sheep Genetic Improvement and Healthy Breeding, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, China
| | - Emma Eyϸórsdóttir
- Faculty of Natural Resources and Environmental Sciences, Agricultural University of Iceland, Borgarnes, Iceland
| | - Jón H. Hallsson
- Faculty of Natural Resources and Environmental Sciences, Agricultural University of Iceland, Borgarnes, Iceland
| | - Tatyana Kiseleva
- All-Russian Research Institute of Genetics and Farm Animal Breeding, Russian Academy of Sciences, Moscow, Russia
| | - Juha Kantanen
- Production Systems, Natural Resources Institute Finland, Jokioinen, Finland
| | - Meng-Hua Li
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
16
|
Lussier JG, Diouf MN, Lévesque V, Sirois J, Ndiaye K. Gene expression profiling of upregulated mRNAs in granulosa cells of bovine ovulatory follicles following stimulation with hCG. Reprod Biol Endocrinol 2017; 15:88. [PMID: 29100496 PMCID: PMC5670713 DOI: 10.1186/s12958-017-0306-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 10/16/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ovulation and luteinization of follicles are complex biological processes initiated by the preovulatory luteinizing hormone surge. The objective of this study was to identify genes that are differentially expressed in bovine granulosa cells (GC) of ovulatory follicles. METHODS Granulosa cells were collected during the first follicular wave of the bovine estrous cycle from dominant follicles (DF) and from ovulatory follicles (OF) obtained 24 h following injection of human chorionic gonadotropin (hCG). A granulosa cell subtracted cDNA library (OF-DF) was generated using suppression subtractive hybridization and screened. RESULTS Detection of genes known to be upregulated in bovine GC during ovulation, such as ADAMTS1, CAV1, EGR1, MMP1, PLAT, PLA2G4A, PTGES, PTGS2, RGS2, TIMP1, TNFAIP6 and VNN2 validated the physiological model and analytical techniques used. For a subset of genes that were identified for the first time, gene expression profiles were further compared by semiquantitative RT-PCR in follicles obtained at different developmental stages. Results confirmed an induction or upregulation of the respective mRNAs in GC of OF 24 h after hCG-injection compared with those of DF for the following genes: ADAMTS9, ARAF, CAPN2, CRISPLD2, FKBP5, GFPT2, KIT, KITLG, L3MBLT3, MRO, NUDT10, NUDT11, P4HA3, POSTN, PSAP, RBP1, SAT1, SDC4, TIMP2, TNC and USP53. In bovine GC, CRISPLD2 and POSTN mRNA were found as full-length transcript whereas L3MBLT3 mRNA was alternatively spliced resulting in a truncated protein missing the carboxy-terminal end amino acids, 774KNSHNEL780. Conversely, L3MBLT3 is expressed as a full-length mRNA in a bovine endometrial cell line. The 774KNSHNEL780 sequence is well conserved in all mammalian species and follows a SAM domain known to confer protein/protein interactions, which suggest a key function for these amino acids in the epigenetic control of gene expression. CONCLUSIONS We conclude that we have identified novel genes that are upregulated by hCG in bovine GC of OF, thereby providing novel insight into peri-ovulatory regulation of genes that contribute to ovulation and/or luteinization processes.
Collapse
Affiliation(s)
- Jacques G Lussier
- Centre de recherche en reproduction et fertilité, Faculté de médecine vétérinaire, Université de Montréal, 3200 Sicotte, St-Hyacinthe, Québec, J2S 2M2, Canada.
| | - Mame N Diouf
- Centre de recherche en reproduction et fertilité, Faculté de médecine vétérinaire, Université de Montréal, 3200 Sicotte, St-Hyacinthe, Québec, J2S 2M2, Canada
- Institut Sénégalais de Recherches Agricoles (ISRA) Laboratoire National de l'Elevage et de Recherches Vétérinaires (LNERV), BP 2057, Dakar-Hann, Sénégal
| | - Valérie Lévesque
- Centre de recherche en reproduction et fertilité, Faculté de médecine vétérinaire, Université de Montréal, 3200 Sicotte, St-Hyacinthe, Québec, J2S 2M2, Canada
| | - Jean Sirois
- Centre de recherche en reproduction et fertilité, Faculté de médecine vétérinaire, Université de Montréal, 3200 Sicotte, St-Hyacinthe, Québec, J2S 2M2, Canada
| | - Kalidou Ndiaye
- Centre de recherche en reproduction et fertilité, Faculté de médecine vétérinaire, Université de Montréal, 3200 Sicotte, St-Hyacinthe, Québec, J2S 2M2, Canada
| |
Collapse
|
17
|
Liu DT, Brewer MS, Chen S, Hong W, Zhu Y. Transcriptomic signatures for ovulation in vertebrates. Gen Comp Endocrinol 2017; 247:74-86. [PMID: 28111234 PMCID: PMC5410184 DOI: 10.1016/j.ygcen.2017.01.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/14/2017] [Accepted: 01/17/2017] [Indexed: 01/01/2023]
Abstract
The central roles of luteinizing hormone (LH), progestin and their receptors for initiating ovulation have been well established. However, signaling pathways and downstream targets such as proteases that are essential for the rupture of follicular cells are still unclear. Recently, we found anovulation in nuclear progestin receptor (Pgr) knockout (Pgr-KO) zebrafish, which offers a new model for examining genes and pathways that are important for ovulation and fertility. In this study, we examined expression of all transcripts using RNA-Seq in preovulatory follicular cells collected following the final oocyte maturation, but prior to ovulation, from wild-type (WT) or Pgr-KO fish. Differential expression analysis revealed 3567 genes significantly differentially expressed between WT and Pgr-KO fish (fold change⩾2, p<0.05). Among those, 1543 gene transcripts were significantly more expressed, while 2024 genes were significantly less expressed, in WT than those in Pgr-KO. We then retrieved and compared transcriptional data from online databases and further identified 661 conserved genes in fish, mice, and humans that showed similar levels of high (283 genes) or low (387) expression in animals that were ovulating compared to those with no ovulation. For the first time, ovulatory genes and their involved biological processes and pathways were also visualized using Enrichment Map and Cytoscape. Intriguingly, enrichment analysis indicated that the genes with higher expression were involved in multiple ovulatory pathways and processes such as inflammatory response, angiogenesis, cytokine production, cell migration, chemotaxis, MAPK, focal adhesion, and cytoskeleton reorganization. In contrast, the genes with lower expression were mainly involved in DNA replication, DNA repair, DNA methylation, RNA processing, telomere maintenance, spindle assembling, nuclear acid transport, catabolic processes, and nuclear and cell division. Our results indicate that a large set of genes (>3000) is differentially regulated in the follicular cells in zebrafish prior to ovulation, terminating programs such as growth and proliferation, and beginning processes including the inflammatory response and apoptosis. Further studies are required to establish relationships among these genes and an ovulatory circuit in the zebrafish model.
Collapse
Affiliation(s)
- Dong Teng Liu
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian Province 361102, People's Republic of China; Department of Biology, East Carolina University, Greenville, NC 27858, United States
| | - Michael S Brewer
- Department of Biology, East Carolina University, Greenville, NC 27858, United States
| | - Shixi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian Province 361102, People's Republic of China
| | - Wanshu Hong
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian Province 361102, People's Republic of China
| | - Yong Zhu
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian Province 361102, People's Republic of China; Department of Biology, East Carolina University, Greenville, NC 27858, United States.
| |
Collapse
|
18
|
Donadeu FX, Mohammed BT, Ioannidis J. A miRNA target network putatively involved in follicular atresia. Domest Anim Endocrinol 2017; 58:76-83. [PMID: 27664382 PMCID: PMC5145806 DOI: 10.1016/j.domaniend.2016.08.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/03/2016] [Accepted: 08/04/2016] [Indexed: 01/14/2023]
Abstract
In a previous microarray study, we identified a subset of micro RNAS (miRNAs), which expression was distinctly higher in atretic than healthy follicles of cattle. In the present study, we investigated the involvement of those miRNAs in granulosa and theca cells during atresia. Reverse Transcription-quantitative Polymerase Chain Reaction (RT-qPCR) confirmed that miR-21-5p/-3p, miR-150, miR-409a, miR-142-5p, miR-378, miR-222, miR-155, and miR-199a-5p were expressed at higher levels in atretic than healthy follicles (9-17 mm, classified based on steroidogenic capacity). All miRNAs except miR-21-3p and miR-378 were expressed at higher levels in theca than granulosa cells. The expression of 13 predicted miRNA targets was determined in follicular cells by RT-qPCR, revealing downregulation of HIF1A, ETS1, JAG1, VEGFA, and MSH2 in either or both cell types during atresia. Based on increases in miRNA levels simultaneous with decreases in target levels in follicular cells, several predicted miRNA target interactions were confirmed that are putatively involved in follicular atresia, namely miR-199a-5p/miR-155-HIF1A in granulosa cells, miR-155/miR-222-ETS1 in theca cells, miR-199a-5p-JAG1 in theca cells, miR-199a-5p/miR-150/miR-378-VEGFA in granulosa and theca cells, and miR-155-MSH2 in theca cells. These results offer novel insight on the involvement of miRNAs in follicle development by identifying a miRNA target network that is putatively involved in follicle atresia.
Collapse
Affiliation(s)
- F X Donadeu
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian EH25 9RG, UK.
| | - B T Mohammed
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian EH25 9RG, UK
| | - J Ioannidis
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian EH25 9RG, UK
| |
Collapse
|
19
|
Sayasith K, Sirois J. Molecular characterization of a disintegrin and metalloprotease-17 (ADAM17) in granulosa cells of bovine preovulatory follicles. Mol Cell Endocrinol 2015; 411:49-57. [PMID: 25917455 DOI: 10.1016/j.mce.2015.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 04/09/2015] [Accepted: 04/13/2015] [Indexed: 11/29/2022]
Abstract
A disintegrin and metalloprotease-17 (ADAM17) is thought to play a key role in the release of soluble and active epiregulin (EREG) and amphiregulin (AREG) in ovarian follicles but its transcriptional regulation in follicular cells remains largely unknown. The objectives of this study were to characterize the regulation of ADAM17 transcripts in bovine follicles prior to ovulation and to investigate its transcriptional control in bovine granulosa cells. To study the regulation of ADAM17 transcripts, RT-PCR analyses were performed using total RNA extracted from bovine follicles collected between 0 h and 24 h post-hCG. Results showed that levels of ADAM17 mRNA were low prior to hCG (0 h), markedly and transiently increased 6-12 h post-hCG (P <0.05), and returned to low baseline levels at 24 h post-hCG in granulosa and theca interna cells of preovulatory follicles. To determine the transcriptional control of ADAM17 expression, primary cultures of bovine granulosa cells were used. Forskolin (FSK) stimulation induced a pattern of ADAM17 mRNA up-regulation in vitro similar to that observed by hCG in vivo. 5'-Deletion mutagenesis studies identified a minimal region of the bovine ADAM17 promoter containing basal and FSK-inducible activities, which were dependent on the presence of a consensus AP1 cis-element. Electrophoretic mobility shift assays revealed an interaction between AP1 and the trans-acting factor Fra2. Chromatin immunoprecipitation assays confirmed an endogenous interaction between Fra2 and the ADAM17 promoter in granulosa cell cultures. FSK-inducible ADAM17 promoter activity and mRNA expression were suppressed by PKA and ERK1/2 inhibitors but not by a p38MAPK inhibitor, pointing to the importance of PKA and ERK1/2 signaling pathways in the up-regulation of bovine ADAM17 mRNA. Collectively, these findings describe the gonadotropin/FSK-dependent up-regulation of ADAM17 transcripts in bovine preovulatory follicles and unravel for the first time some of the molecular mechanisms involved in ADAM17 gene expression in granulosa cells of a monoovulatory species.
Collapse
Affiliation(s)
- Khampoun Sayasith
- Centre de Recherche en Reproduction Animale, Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec J2S 7C6, Canada.
| | - Jean Sirois
- Centre de Recherche en Reproduction Animale, Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec J2S 7C6, Canada
| |
Collapse
|