1
|
Ventayol-Guirado M, Hernandez-Rodriguez J, Florit J, Llull-Alberti MV, Barragan R, Ferragut JF, Martorell J, Heine-Suñer D, Martinez I, Picornell A, Torres-Juan L, Peralta L. First follicular fluid cf-mtDNA sequencing unveils ovarian stimulation-induced mutations impacting oocyte quality and IVF success. J Assist Reprod Genet 2025:10.1007/s10815-025-03511-8. [PMID: 40405035 DOI: 10.1007/s10815-025-03511-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 05/05/2025] [Indexed: 05/24/2025] Open
Abstract
PURPOSE Cell-free mitochondrial DNA (cf-mtDNA) has emerged as a promising non-invasive method to predict embryo implantation potential. This study presents the first sequencing of cf-mtDNA in follicular fluid, aiming to explore the impact of ovarian stimulation treatment on the accumulation of mtDNA mutations and assess their implications for IVF outcomes. METHODS We selected 24 women aged 27 to 35 from the Assisted Reproduction Unit based on inclusion criteria requiring them to be undergoing their first IVF cycle without known infertility issues. For each participant, blood samples were collected prior to ovarian stimulation, and follicular fluid samples were obtained at the time of oocyte retrieval. Long-PCR amplification and next-generation sequencing were used to identify mtDNA mutations. RESULTS Ovarian stimulation-induced mtDNA mutations in follicular fluid were identified primarily in the D-loop region of the mitochondrial genome. These mutations were correlated with low oocyte counts and poor embryo quality, ultimately reducing IVF success rates. The accumulation of these mutations followed a time-dependent pattern, with longer stimulation durations resulting in higher mutation rates and lower pregnancy outcomes. Prolonged stimulations may exacerbate oxidative stress, negatively impacting oocyte quality. CONCLUSIONS By introducing the first sequencing of follicular fluid cf-mtDNA, this study establishes a direct link between ovarian stimulation-induced mtDNA mutations and reduced IVF efficacy, highlighting them as non-invasive biomarkers for predicting IVF outcomes. Future research may focus on optimizing stimulation protocols or incorporating antioxidants to minimize mtDNA mutations, improving oocyte quality and overall IVF success rates for patients.
Collapse
Affiliation(s)
- Marc Ventayol-Guirado
- Genomics of Health Research Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Balearic Islands, Spain.
| | - Jessica Hernandez-Rodriguez
- Genomics of Health Research Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Balearic Islands, Spain
- Molecular Diagnostics and Clinical Genetics Unit, Hospital Universitari Son Espases, 07120, Palma, Balearic Islands, Spain
| | - Joana Florit
- Genomics of Health Research Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Balearic Islands, Spain
| | - Maria Victoria Llull-Alberti
- Genomics of Health Research Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Balearic Islands, Spain
| | - Raquel Barragan
- Assisted Reproduction Unit, Hospital Universitari Son Espases, 07120, Palma, Balearic Islands, Spain
| | - Joana Francesca Ferragut
- Biology Department, Universitat de Les Illes Balears, 07122, Palma, Balearic Islands, Spain
- Institut Universitari d'Investigació en Ciències de La Salut (IUNICS), 07122, Palma, Balearic Islands, Spain
| | - Jaume Martorell
- Obstetrics and Gynecology Service, Hospital Universitari Son Espases, 07120, Palma, Balearic Islands, Spain
| | - Damian Heine-Suñer
- Genomics of Health Research Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Balearic Islands, Spain
- Molecular Diagnostics and Clinical Genetics Unit, Hospital Universitari Son Espases, 07120, Palma, Balearic Islands, Spain
| | - Iciar Martinez
- Genomics of Health Research Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Balearic Islands, Spain
- Molecular Diagnostics and Clinical Genetics Unit, Hospital Universitari Son Espases, 07120, Palma, Balearic Islands, Spain
| | - Antonia Picornell
- Biology Department, Universitat de Les Illes Balears, 07122, Palma, Balearic Islands, Spain
- Institut Universitari d'Investigació en Ciències de La Salut (IUNICS), 07122, Palma, Balearic Islands, Spain
| | - Laura Torres-Juan
- Genomics of Health Research Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Balearic Islands, Spain
- Molecular Diagnostics and Clinical Genetics Unit, Hospital Universitari Son Espases, 07120, Palma, Balearic Islands, Spain
| | - Laura Peralta
- Genomics of Health Research Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Balearic Islands, Spain
- Assisted Reproduction Unit, Hospital Universitari Son Espases, 07120, Palma, Balearic Islands, Spain
| |
Collapse
|
2
|
Li J, Zhu X, Zhu W, Li L, Wei H, Zhang S. Research Progress on the Impact of Human Chorionic Gonadotropin on Reproductive Performance in Sows. Animals (Basel) 2024; 14:3266. [PMID: 39595318 PMCID: PMC11591456 DOI: 10.3390/ani14223266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/01/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Human chorionic gonadotropin is a glycoprotein hormone produced by human or humanoid syncytiotrophoblasts that differentiate during pregnancy. Due to its superior stability and long-lasting effects compared to luteinizing hormone, it is often used to replace luteinizing hormone to regulate reproductive performance in sows. Human chorionic gonadotropin promotes oocyte maturation, follicle development, and luteinization, thereby increasing conception rates and supporting early embryonic development. In sow reproductive management, the application of human chorionic gonadotropin not only enhances ovulation synchrony but also improves the success rate of embryo implantation by regulating endometrial receptivity and immune mechanisms, significantly enhancing overall reproductive performance. This article primarily reviews the application of human chorionic gonadotropin in sow follicle development, luteal maintenance, and embryo implantation, providing theoretical support for its use in improving reproductive performance in sows.
Collapse
Affiliation(s)
| | | | | | | | | | - Shouquan Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agroanimal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510640, China; (J.L.); (X.Z.); (W.Z.); (L.L.); (H.W.)
| |
Collapse
|
3
|
Guzmán A, Rosales-Torres AM, Medina-Moctezuma ZB, González-Aretia D, Hernández-Coronado CG. Effects and action mechanism of gonadotropins on ovarian follicular cells: A novel role of Sphingosine-1-Phosphate (S1P). A review. Gen Comp Endocrinol 2024; 357:114593. [PMID: 39047797 DOI: 10.1016/j.ygcen.2024.114593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 07/02/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
Follicle-stimulating hormone (FSH) and luteinizing hormone (LH) control antral follicular growth by regulating several processes, such as the synthesis of hormones and signaling molecules, proliferation, survival, apoptosis, luteinization, and ovulation. To exert these effects, gonadotropins bind to their respective Gs protein-coupled receptors, activating the protein kinase A (PKA) pathway or recruiting Gq proteins to activate protein kinase C (PKC) signaling. Although the action mechanism of FSH and LH is clear, recently, it has been shown that both gonadotropins promote the synthesis of sphingosine-1-phosphate (S1P) in granulosa and theca cells through the activation of sphingosine kinase 1. Moreover, the inhibition of SPHKs reduces S1P synthesis, cell viability, and the proliferation of follicular cells in response to gonadotropins, and the addition of S1P to the culture medium increases the proliferation of granulosa and theca cells without apparent effects on sexual steroid synthesis. Therefore, we consider that S1P is a crucial signaling molecule that complements the canonical gonadotropin pathway to promote the proliferation and viability of granulosa and theca cells.
Collapse
Affiliation(s)
- A Guzmán
- Universidad Autónoma Metropolitana Unidad Xochimilco, Departamento Producción Agrícola y Animal, Ciudad de México, Mexico
| | - A M Rosales-Torres
- Universidad Autónoma Metropolitana Unidad Xochimilco, Departamento Producción Agrícola y Animal, Ciudad de México, Mexico
| | - Z B Medina-Moctezuma
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México, Mexico
| | - D González-Aretia
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México, Mexico
| | - C G Hernández-Coronado
- Universidad Autónoma Metropolitana Unidad Xochimilco, Departamento Producción Agrícola y Animal, Ciudad de México, Mexico.
| |
Collapse
|
4
|
Kalinderi K, Kalinderis M, Papaliagkas V, Fidani L. The Reproductive Lifespan of Ovarian Follicle. Reprod Sci 2024; 31:2604-2614. [PMID: 38816594 DOI: 10.1007/s43032-024-01606-8] [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: 12/18/2023] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
The functional unit within mammalian ovaries is the ovarian follicle. The development of the ovarian follicle is a lengthy process beginning from the time of embryogenesis, passing through multiple different stages of maturation. The purpose of this review is to describe the most basic events in the journey of ovarian follicle development, discussing the importance of ovarian reserve and highlighting the role of several factors that affect oocyte quality and quantity during aging including hormonal, genetic and epigenetic factors. Novel, promising anti-aging strategies are also discussed.
Collapse
Affiliation(s)
- Kallirhoe Kalinderi
- Laboratory of Medical Biology-Genetics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, GR-54124, Greece.
| | - Michail Kalinderis
- Department of Obstetrics and Gynaecology, St George's University Hospital NHS Trust, Blackshaw Road, Tooting, London, SW17 0QT, UK
| | - Vasileios Papaliagkas
- Department of Biomedical Sciences, School of Health Sciences, International Hellenic University, Thessaloniki, 57400, Greece
| | - Liana Fidani
- Laboratory of Medical Biology-Genetics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, GR-54124, Greece
| |
Collapse
|
5
|
Liu B, Liu Y, Li S, Chen P, Zhang J, Feng L. Depletion of placental brain-derived neurotrophic factor (BDNF) is attributed to premature ovarian insufficiency (POI) in mice offspring. J Ovarian Res 2024; 17:141. [PMID: 38982490 PMCID: PMC11232340 DOI: 10.1186/s13048-024-01467-4] [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: 12/11/2023] [Accepted: 06/29/2024] [Indexed: 07/11/2024] Open
Abstract
INTRODUCTION Premature ovarian insufficiency (POI) is one of the causes of female infertility. Unexplained POI is increasingly affecting women in their reproductive years. However, the etiology of POI is diverse and remains elusive. We and others have shown that brain-derived neurotrophic factor (BDNF) plays an important role in adult ovarian function. Here, we report on a novel role of BDNF in the Developmental Origins of POI. METHODS Placental BDNF knockout mice were created using CRISPR/CAS9. Homozygous knockout (cKO(HO)) mice didn't survive, while heterozygous knockout (cKO(HE)) mice did. BDNF reduction in cKO(HE) mice was confirmed via immunohistochemistry and Western blots. Ovaries were collected from cKO(HE) mice at various ages, analyzing ovarian metrics, FSH expression, and litter sizes. In one-month-old mice, oocyte numbers were assessed using super-ovulation, and oocyte gene expression was analyzed with smart RNAseq. Ovaries of P7 mice were studied with SEM, and gene expression was confirmed with RT-qPCR. Alkaline phosphatase staining at E11.5 and immunofluorescence for cyclinD1 assessed germ cell number and cell proliferation. RESULTS cKO(HE) mice had decreased ovarian function and litter size in adulthood. They were insensitive to ovulation induction drugs manifested by lower oocyte release after superovulation in one-month-old cKO(HE) mice. The transcriptome and SEM results indicate that mitochondria-mediated cell death or aging might occur in cKO(HE) ovaries. Decreased placental BDNF led to diminished primordial germ cell proliferation at E11.5 and ovarian reserve which may underlie POI in adulthood. CONCLUSION The current results showed decreased placental BDNF diminished primordial germ cell proliferation in female fetuses during pregnancy and POI in adulthood. Our findings can provide insights into understanding the underlying mechanisms of POI.
Collapse
Affiliation(s)
- Bin Liu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- Department of Reproduction, School of Medicine, Xinhua Hospital, Shanghai Jiao-Tong University, Shanghai, China
| | - Yongjie Liu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shuman Li
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Pingping Chen
- Department of Reproduction, School of Medicine, Xinhua Hospital, Shanghai Jiao-Tong University, Shanghai, China
| | - Jun Zhang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Liping Feng
- Department of Obstetrics and Gynaecology, Duke University, Durham, NC, USA.
| |
Collapse
|
6
|
Sakaue S, Weinand K, Isaac S, Dey KK, Jagadeesh K, Kanai M, Watts GFM, Zhu Z, Brenner MB, McDavid A, Donlin LT, Wei K, Price AL, Raychaudhuri S. Tissue-specific enhancer-gene maps from multimodal single-cell data identify causal disease alleles. Nat Genet 2024; 56:615-626. [PMID: 38594305 PMCID: PMC11456345 DOI: 10.1038/s41588-024-01682-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 02/07/2024] [Indexed: 04/11/2024]
Abstract
Translating genome-wide association study (GWAS) loci into causal variants and genes requires accurate cell-type-specific enhancer-gene maps from disease-relevant tissues. Building enhancer-gene maps is essential but challenging with current experimental methods in primary human tissues. Here we developed a nonparametric statistical method, SCENT (single-cell enhancer target gene mapping), that models association between enhancer chromatin accessibility and gene expression in single-cell or nucleus multimodal RNA sequencing and ATAC sequencing data. We applied SCENT to 9 multimodal datasets including >120,000 single cells or nuclei and created 23 cell-type-specific enhancer-gene maps. These maps were highly enriched for causal variants in expression quantitative loci and GWAS for 1,143 diseases and traits. We identified likely causal genes for both common and rare diseases and linked somatic mutation hotspots to target genes. We demonstrate that application of SCENT to multimodal data from disease-relevant human tissue enables the scalable construction of accurate cell-type-specific enhancer-gene maps, essential for defining noncoding variant function.
Collapse
Affiliation(s)
- Saori Sakaue
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kathryn Weinand
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Shakson Isaac
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Kushal K Dey
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Karthik Jagadeesh
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Masahiro Kanai
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Gerald F M Watts
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Zhu Zhu
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael B Brenner
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew McDavid
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA
| | - Laura T Donlin
- Hospital for Special Surgery, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Kevin Wei
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alkes L Price
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Kapper C, Oppelt P, Ganhör C, Gyunesh AA, Arbeithuber B, Stelzl P, Rezk-Füreder M. Minerals and the Menstrual Cycle: Impacts on Ovulation and Endometrial Health. Nutrients 2024; 16:1008. [PMID: 38613041 PMCID: PMC11013220 DOI: 10.3390/nu16071008] [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: 02/26/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
The role of minerals in female fertility, particularly in relation to the menstrual cycle, presents a complex area of study that underscores the interplay between nutrition and reproductive health. This narrative review aims to elucidate the impacts of minerals on key aspects of the reproductive system: hormonal regulation, ovarian function and ovulation, endometrial health, and oxidative stress. Despite the attention given to specific micronutrients in relation to reproductive disorders, there is a noticeable absence of a comprehensive review focusing on the impact of minerals throughout the menstrual cycle on female fertility. This narrative review aims to address this gap by examining the influence of minerals on reproductive health. Each mineral's contribution is explored in detail to provide a clearer picture of its importance in supporting female fertility. This comprehensive analysis not only enhances our knowledge of reproductive health but also offers clinicians valuable insights into potential therapeutic strategies and the recommended intake of minerals to promote female reproductive well-being, considering the menstrual cycle. This review stands as the first to offer such a detailed examination of minerals in the context of the menstrual cycle, aiming to elevate the understanding of their critical role in female fertility and reproductive health.
Collapse
Affiliation(s)
- Celine Kapper
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria; (C.K.); (P.O.); (B.A.)
| | - Peter Oppelt
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria; (C.K.); (P.O.); (B.A.)
- Department for Gynaecology, Obstetrics and Gynaecological Endocrinology, Kepler University Hospital, Johannes Kepler University Linz, 4020 Linz, Austria
| | - Clara Ganhör
- Division of Pathophysiology, Institute of Physiology and Pathophysiology, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
- Clinical Research Institute for Cardiovascular and Metabolic Diseases, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria
| | - Ayberk Alp Gyunesh
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria; (C.K.); (P.O.); (B.A.)
| | - Barbara Arbeithuber
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria; (C.K.); (P.O.); (B.A.)
| | - Patrick Stelzl
- Department for Gynaecology, Obstetrics and Gynaecological Endocrinology, Kepler University Hospital, Johannes Kepler University Linz, 4020 Linz, Austria
| | - Marlene Rezk-Füreder
- Experimental Gynaecology, Obstetrics and Gynaecological Endocrinology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria; (C.K.); (P.O.); (B.A.)
| |
Collapse
|
8
|
Lounas A, Breton Y, Lebrun A, Laflamme I, Vernoux N, Savage J, Tremblay MÈ, Pelletier M, Germain M, Richard FJ. The follicle-stimulating hormone triggers rapid changes in mitochondrial structure and function in porcine cumulus cells. Sci Rep 2024; 14:436. [PMID: 38172520 PMCID: PMC10764925 DOI: 10.1038/s41598-023-50586-3] [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: 08/28/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
Oocyte maturation is a key process during which the female germ cell undergoes resumption of meiosis and completes its preparation for embryonic development including cytoplasmic and epigenetic maturation. The cumulus cells directly surrounding the oocyte are involved in this process by transferring essential metabolites, such as pyruvate, to the oocyte. This process is controlled by cyclic adenosine monophosphate (cAMP)-dependent mechanisms recruited downstream of follicle-stimulating hormone (FSH) signaling in cumulus cells. As mitochondria have a critical but poorly understood contribution to this process, we defined the effects of FSH and high cAMP concentrations on mitochondrial dynamics and function in porcine cumulus cells. During in vitro maturation (IVM) of cumulus-oocyte complexes (COCs), we observed an FSH-dependent mitochondrial elongation shortly after stimulation that led to mitochondrial fragmentation 24 h later. Importantly, mitochondrial elongation was accompanied by decreased mitochondrial activity and a switch to glycolysis. During a pre-IVM culture step increasing intracellular cAMP, mitochondrial fragmentation was prevented. Altogether, the results demonstrate that FSH triggers rapid changes in mitochondrial structure and function in COCs involving cAMP.
Collapse
Affiliation(s)
- Amel Lounas
- Centre de recherche en reproduction, développement et santé intergénérationnelle (CRDSI), Département des sciences animales, Faculté des Sciences de l'agriculture et de l'alimentation, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Yann Breton
- Centre de recherche du CHU de Québec-Université Laval, Axe Maladies infectieuses et immunitaires, Département de microbiologie-infectiologie et d'immunologie, Faculté de médecine, Université Laval, Québec, QC, G1V4G2, Canada
| | - Ariane Lebrun
- Centre de recherche en reproduction, développement et santé intergénérationnelle (CRDSI), Département des sciences animales, Faculté des Sciences de l'agriculture et de l'alimentation, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Isabelle Laflamme
- Centre de recherche en reproduction, développement et santé intergénérationnelle (CRDSI), Département des sciences animales, Faculté des Sciences de l'agriculture et de l'alimentation, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Nathalie Vernoux
- Centre de recherche du CHU de Québec-Université Laval, Axe Neurosciences, Département de médecine moléculaire, Université Laval, Québec, QC, G1V 4G2, Canada
| | - Julie Savage
- Centre de recherche du CHU de Québec-Université Laval, Axe Neurosciences, Département de médecine moléculaire, Université Laval, Québec, QC, G1V 4G2, Canada
| | - Marie-Ève Tremblay
- Centre de recherche du CHU de Québec-Université Laval, Axe Neurosciences, Département de médecine moléculaire, Université Laval, Québec, QC, G1V 4G2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, V8W 2Y2, Canada
| | - Martin Pelletier
- Centre de recherche du CHU de Québec-Université Laval, Axe Maladies infectieuses et immunitaires, Département de microbiologie-infectiologie et d'immunologie, Faculté de médecine, Université Laval, Québec, QC, G1V4G2, Canada
| | - Marc Germain
- Département de biologie médicale, Université du Québec à Trois-Rivières, Québec, G8Z 4M3, Canada
| | - François J Richard
- Centre de recherche en reproduction, développement et santé intergénérationnelle (CRDSI), Département des sciences animales, Faculté des Sciences de l'agriculture et de l'alimentation, Université Laval, Québec, QC, G1V 0A6, Canada.
| |
Collapse
|
9
|
Jiang Y, Gao X, Liu Y, Yan X, Shi H, Zhao R, Chen ZJ, Gao F, Zhao H, Zhao S. Cellular atlases of ovarian microenvironment alterations by diet and genetically-induced obesity. SCIENCE CHINA. LIFE SCIENCES 2024; 67:51-66. [PMID: 37721638 DOI: 10.1007/s11427-023-2360-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 05/09/2023] [Indexed: 09/19/2023]
Abstract
Obesity, which can arise from genetic or environmental factors, has been shown to cause serious damages to the reproductive system. The ovary, as one of the primary regulators of female fertility, is a complex organ comprised of heterogeneous cell types that work together to maintain a normal ovarian microenvironment (OME). Despite its importance, the effect of obesity on the entire ovary remains poorly documented. In this study, we performed ovary single-cell and nanoscale spatial RNA sequencing to investigate how the OME changed under different kinds of obesity, including high-fat diet (HFD) induced obesity and Leptin ablation induced obesity (OB). Our results demonstrate that OB, but not HFD, dramatically altered the proportion of ovarian granulosa cells, theca-interstitial cells, luteal cells, and endothelial cells. Furthermore, based on the spatial dynamics of follicular development, we defined four subpopulations of granulosa cell and found that obesity drastically disrupted the differentiation of mural granulosa cells from small to large antral follicles. Functionally, HFD enhanced follicle-stimulating hormone (FSH) sensitivity and hormone conversion, while OB caused decreased sensitivity, inadequate steroid hormone conversion, and impaired follicular development. These differences can be explained by the differential expression pattern of the transcription factor Foxo1. Overall, our study provides a powerful and high-resolution resource for profiling obesity-induced OME and offers insights into the diverse effects of obesity on female reproductive disorders.
Collapse
Affiliation(s)
- Yonghui Jiang
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center of Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215000, China
| | - Xueying Gao
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center of Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
- Center for reproductive medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200127, China
| | - Yue Liu
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center of Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
| | - Xueqi Yan
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center of Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
| | - Huangcong Shi
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center of Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
| | - Rusong Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China
- National Research Center of Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215000, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China.
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, 250012, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China.
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China.
- Center for reproductive medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200127, China.
| | - Fei Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Science, Beijing, 100101, China.
| | - Han Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China.
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China.
- National Research Center of Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China.
| | - Shigang Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, China.
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, China.
- National Research Center of Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, China.
| |
Collapse
|
10
|
Chen X, Jin R, Yang A, Li J, Song Y, Zhao B, Chen Y, Wu X. Behavioral and Physiological Differences in Female Rabbits at Different Stages of the Estrous Cycle. Animals (Basel) 2023; 13:3414. [PMID: 37958169 PMCID: PMC10648029 DOI: 10.3390/ani13213414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Estrus involves a series of complex physiological signs and changes in behavior before ovulation, which play a crucial role in animal reproduction. However, there have been few studies that evaluate behaviors during the different stages of estrus cycle in female rabbits. Therefore, more detailed information is needed on distinguishing the various stages of the estrous cycle. This study explored the behavioral and physiological differences at various estrous cycle stages in female New Zealand White rabbits. The continuous recording method was employed to record the daily behaviors of twenty postpartum female rabbits during the estrous cycle. Compared with the diestrus stage, the duration of foraging and drinking behavior in estrus decreased significantly, and the frequency of grooming and biting behaviors increased (p < 0.05). Differences in reproductive hormone levels (FSH, LH, P4, and E2) and follicle development were measured at each stage via ELISA and HE staining. The FSH and LH levels showed an increasing trend and then decreased, with the lowest being in late estrus (p < 0.05). The P4 level was the lowest in estrus (p < 0.05), and E2 showed a gradually increasing trend. There was no significant difference in the number of primordial follicles at each stage, but the number of primary follicles in estrus was significantly higher than at the other stages (p < 0.05). To further understand the molecular regulation mechanism of the estrous cycle in female rabbits, we analyzed the ovarian transcription patterns of female rabbits in diestrus (D group) and estrus (E group) employing RNA-seq. A total of 967 differentially expressed genes (DEGs) were screened from the ovaries of female rabbits between the diestrus and estrus groups. A KEGG analysis of DEGs enriched in the estrogen signaling pathway, aldosterone synthesis, and secretion pathway, such as CYP19A1 and IGF1R, was performed. The rabbits' behavior, related physiological hormones, and molecular regulation also differed at different estrous cycle stages. The results provide recommendations for the adequate management practices of postpartum re-estrus and breeding female rabbits.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yang Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.C.); (R.J.); (A.Y.); (J.L.); (Y.S.); (B.Z.)
| | - Xinsheng Wu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.C.); (R.J.); (A.Y.); (J.L.); (Y.S.); (B.Z.)
| |
Collapse
|
11
|
Jia C, Zhang M, Liu X, Xu W, Xiong Y, Huang R, Li M, Li M. Transcriptome-wide m6A methylation profiling of Wuhua yellow-feathered chicken ovary revealed regulatory pathways underlying sexual maturation and low egg-laying performance. Front Genet 2023; 14:1284554. [PMID: 37928247 PMCID: PMC10622773 DOI: 10.3389/fgene.2023.1284554] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/10/2023] [Indexed: 11/07/2023] Open
Abstract
RNA N6-melthyladenosine (m6A) can play an important role in regulation of various biological processes. Chicken ovary development is closely related to egg laying performance, which is a process primarily controlled by complex gene regulations. In this study, transcriptome-wide m6A methylation of the Wuhua yellow-feathered chicken ovaries before and after sexual maturation was profiled to identify the potential molecular mechanisms underlying chicken ovary development. The results indicated that m6A levels of mRNAs were altered dramatically during sexual maturity. A total of 1,476 differential m6A peaks were found between these two stages with 662 significantly upregulated methylation peaks and 814 downregulated methylation peaks after sexual maturation. A positive correlation was observed between the m6A peaks and gene expression levels, indicating that m6A may play an important role in regulation of chicken ovary development. Functional enrichment analysis indicated that apoptosis related pathways could be the key molecular regulatory pathway underlying the poor reproductive performance of Wuhua yellow-feathered chicken. Overall, the various pathways and corresponding candidate genes identified here could be useful to facilitate molecular design breeding for improving egg production performance in Chinese local chicken breed, and it might also contribute to the genetic resource protection of valuable avian species.
Collapse
Affiliation(s)
- Congjun Jia
- College of Agricultural Engineering, Guangdong Meizhou Vocational and Technical College, Meizhou, China
- Meizhou Engineering Research Center for Veterinary Medicine and Natural Medicine, Meizhou, China
| | - Mengling Zhang
- College of Agricultural Engineering, Guangdong Meizhou Vocational and Technical College, Meizhou, China
- Meizhou Engineering Research Center for Veterinary Medicine and Natural Medicine, Meizhou, China
| | - Xiaoyan Liu
- College of Agricultural Engineering, Guangdong Meizhou Vocational and Technical College, Meizhou, China
- Meizhou Engineering Research Center for Veterinary Medicine and Natural Medicine, Meizhou, China
| | - Weilin Xu
- College of Agricultural Engineering, Guangdong Meizhou Vocational and Technical College, Meizhou, China
- Meizhou Engineering Research Center for Veterinary Medicine and Natural Medicine, Meizhou, China
| | - Yanqing Xiong
- College of Agricultural Engineering, Guangdong Meizhou Vocational and Technical College, Meizhou, China
- Meizhou Engineering Research Center for Veterinary Medicine and Natural Medicine, Meizhou, China
| | - Rihao Huang
- College of Agricultural Engineering, Guangdong Meizhou Vocational and Technical College, Meizhou, China
- Meizhou Engineering Research Center for Veterinary Medicine and Natural Medicine, Meizhou, China
| | - Meidi Li
- College of Agricultural Engineering, Guangdong Meizhou Vocational and Technical College, Meizhou, China
- Meizhou Engineering Research Center for Veterinary Medicine and Natural Medicine, Meizhou, China
| | - Mingna Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
12
|
Liu Q, Wei F, Wang J, Liu H, Zhang H, Liu M, Liu K, Ye Z. Molecular mechanisms regulating natural menopause in the female ovary: a study based on transcriptomic data. Front Endocrinol (Lausanne) 2023; 14:1004245. [PMID: 37564980 PMCID: PMC10411606 DOI: 10.3389/fendo.2023.1004245] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 07/03/2023] [Indexed: 08/12/2023] Open
Abstract
Introduction Natural menopause is an inevitable biological process with significant implications for women's health. However, the molecular mechanisms underlying menopause are not well understood. This study aimed to investigate the molecular and cellular changes occurring in the ovary before and after perimenopause. Methods Single-cell sequencing data from the GTEx V8 cohort (30-39: 14 individuals; 40-49: 37 individuals; 50-59: 61 individuals) and transcriptome sequencing data from ovarian tissue were analyzed. Seurat was used for single-cell sequencing data analysis, while harmony was employed for data integration. Cell differentiation trajectories were inferred using CytoTrace. CIBERSORTX assessed cell infiltration scores in ovarian tissue. WGCNA evaluated co-expression network characteristics in pre- and post-perimenopausal ovarian tissue. Functional enrichment analysis of co-expression modules was conducted using ClusterprofileR and Metascape. DESeq2 performed differential expression analysis. Master regulator analysis and signaling pathway activity analysis were carried out using MsViper and Progeny, respectively. Machine learning models were constructed using Orange3. Results We identified the differentiation trajectory of follicular cells in the ovary as ARID5B+ Granulosa -> JUN+ Granulosa -> KRT18+ Granulosa -> MT-CO2+ Granulosa -> GSTA1+ Granulosa -> HMGB1+ Granulosa. Genes driving Granulosa differentiation, including RBP1, TMSB10, SERPINE2, and TMSB4X, were enriched in ATP-dependent activity regulation pathways. Genes involved in maintaining the Granulosa state, such as DCN, ARID5B, EIF1, and HSP90AB1, were enriched in the response to unfolded protein and chaperone-mediated protein complex assembly pathways. Increased contents of terminally differentiated HMGB1+ Granulosa and GSTA1+ Granulosa were observed in the ovaries of individuals aged 50-69. Signaling pathway activity analysis indicated a gradual decrease in TGFb and MAPK pathway activity with menopause progression, while p53 pathway activity increased. Master regulator analysis revealed significant activation of transcription factors FOXR1, OTX2, MYBL2, HNF1A, and FOXN4 in the 30-39 age group, and GLI1, SMAD1, SMAD7, APP, and EGR1 in the 40-49 age group. Additionally, a diagnostic model based on 16 transcription factors (Logistic Regression L2) achieved reliable performance in determining ovarian status before and after perimenopause. Conclusion This study provides insights into the molecular and cellular mechanisms underlying natural menopause in the ovary. The findings contribute to our understanding of perimenopausal changes and offer a foundation for health management strategies for women during this transition.
Collapse
Affiliation(s)
- Quan Liu
- Binhu Hospital, Hefei First People’s Hospital, Hefei, Anhui, China
| | - Fangqin Wei
- Binhu Hospital, Hefei First People’s Hospital, Hefei, Anhui, China
| | - Jiannan Wang
- Binhu Hospital, Hefei First People’s Hospital, Hefei, Anhui, China
| | - Haiyan Liu
- Binhu Hospital, Hefei First People’s Hospital, Hefei, Anhui, China
| | - Hua Zhang
- Binhu Hospital, Hefei First People’s Hospital, Hefei, Anhui, China
| | - Min Liu
- Binhu Hospital, Hefei First People’s Hospital, Hefei, Anhui, China
| | - Kaili Liu
- Binhu Hospital, Hefei First People’s Hospital, Hefei, Anhui, China
| | - Zheng Ye
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, China
| |
Collapse
|
13
|
Zhang Y, Xu Y, Yu J, Wang X, Xue Q, Shang J, Yang X, Shan X. A premature luteinizing hormone surge without elevated progesterone levels has no adverse effect on cumulative live birth rate in patient undergoing a flexible GnRH antagonist protocol: a retrospective study. J Ovarian Res 2023; 16:119. [PMID: 37370146 DOI: 10.1186/s13048-023-01219-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/20/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND A premature luteinizing hormone (LH) surge refers to an endogenous LH peak that occurs before follicle maturation or human chorionic gonadotropin injection in the process of controlled ovarian hyperstimulation. The effect of premature LH surge on pregnancy outcomes in fresh embryo transfer cycles is still controversial. The aim of this study was to explore the effect of a premature LH surge without elevated progesterone levels on the cumulative pregnancy rate (CPR) and cumulative live birth rate (CLBR) of patients during a flexible GnRH antagonist protocol. METHODS A total of 730 infertile women undergoing IVF/ICSI were recruited for this retrospective study. Only women who either delivered a live infant or had no remaining frozen embryos after a single stimulation cycle were included in the analysis. During the study period, each patient underwent a flexible GnRH antagonist protocol. Women were divided into two groups according to the presence or absence of a premature LH surge. The primary outcome measures were the CPR and CLBR per ovarian stimulation cycle. The secondary outcome measures were the number of oocytes retrieved, fertilization rate, good-quality embryo rate, and clinical pregnancy rate. RESULTS Ninety-one women (12.47%) experienced a premature LH surge without elevated progesterone levels, and the other 639 (87.53%) women were assigned to the control group. The numbers of oocytes retrieved and fertilization rate were significantly greater in the premature LH surge group than in the control group. There was no significant difference between groups in the good-quality embryo rate, clinical pregnancy rate or live birth rate in the fresh embryo transfer cycle. The primary outcome measures, the CPR and CLBR per ovarian stimulation cycle, were not significantly different between the premature LH surge group and the control group. According to the analysis stratified by ovarian response (normal or high), there were no significant differences in pregnancy outcomes between the groups with and without a premature LH surge. CONCLUSIONS The retrospective study demonstrated that the patients experiencing a transient premature LH surge without progesterone elevation had equivalent pregnancy outcomes with those without a premature LH surge on a flexible GnRH antagonist protocol. The present conclusions need to be further validated in a prospective well-designed large-scale study.
Collapse
Affiliation(s)
- Yangyang Zhang
- Department of Obstetrics and Gynecology, Peking University First Hospital, 100034, Beijing, China
| | - Yang Xu
- Department of Obstetrics and Gynecology, Peking University First Hospital, 100034, Beijing, China.
| | - Jiao Yu
- Department of Obstetrics and Gynecology, Peking University First Hospital, 100034, Beijing, China
| | - Xi Wang
- Department of Obstetrics and Gynecology, Peking University First Hospital, 100034, Beijing, China
| | - Qing Xue
- Department of Obstetrics and Gynecology, Peking University First Hospital, 100034, Beijing, China
| | - Jing Shang
- Department of Obstetrics and Gynecology, Peking University First Hospital, 100034, Beijing, China
| | - Xiuli Yang
- Department of Obstetrics and Gynecology, Peking University First Hospital, 100034, Beijing, China
| | - Xuemin Shan
- Department of Obstetrics and Gynecology, Peking University First Hospital, 100034, Beijing, China
| |
Collapse
|
14
|
Li N, Zhou Y, Cai J, Wang Y, Zhou X, Hu M, Li Y, Zhang H, Li J, Cai B, Yuan X. A novel trans-acting lncRNA of ACTG1 that induces the remodeling of ovarian follicles. Int J Biol Macromol 2023:125170. [PMID: 37276900 DOI: 10.1016/j.ijbiomac.2023.125170] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/07/2023]
Abstract
Previous studies have implicated the attractive role of long noncoding RNAs (lncRNAs) in the remodeling of mammalian tissues. The migration of granulosa cells (GCs), which are the main supporting cells in ovarian follicles, stimulates the follicular remodeling. Here, with the cultured GCs as the follicular model, the actin gamma 1 (ACTG1) was observed to significantly promote the migration and proliferation while inhibit the apoptosis of GCs, suggesting that ACTG1 was required for ovarian remodeling. Moreover, we identified the trans-regulatory lncRNA of ACTG1 (TRLA), which was epigenetically targeted by histone H3 lysine 4 acetylation (H3K4ac). Mechanistically, the 2-375 nt of TRLA bound to ACTG1's mRNA to increase the expression of ACTG1. Furthermore, TRLA facilitated the migration and proliferation while inhibited the apoptosis of GCs, thereby accelerating follicular remodeling. Besides, TRLA acted as a ceRNA for miR-26a to increase the expression of high-mobility group AT-hook 1 (HMGA1). Collectively, TRLA induces the remodeling of ovarian follicles via complementary to ACTG1's mRNA and regulating miR-26a/HMGA1 axis in GCs. These observations revealed a novel and promising trans-acting lncRNA mechanism mediated by H3K4ac, and TRLA might be a new target to restore follicular remodeling and development.
Collapse
Affiliation(s)
- Nian Li
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yinqi Zhou
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Jiali Cai
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yifei Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Xiaofeng Zhou
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Mengting Hu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yubin Li
- Reproductive Medical Center, the First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong 510080, China
| | - Hao Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Jiaqi Li
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Bing Cai
- Reproductive Medical Center, the First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong 510080, China.
| | - Xiaolong Yuan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.
| |
Collapse
|
15
|
DEHP Decreases Steroidogenesis through the cAMP and ERK1/2 Signaling Pathways in FSH-Stimulated Human Granulosa Cells. Cells 2023; 12:cells12030398. [PMID: 36766740 PMCID: PMC9913623 DOI: 10.3390/cells12030398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
DEHP is an endocrine disruptor that interferes with the function of the female reproductive system. Several studies suggested that DEHP affects steroidogenesis in human and rodent granulosa cells (GC). Some studies have shown that DEHP can also affect the FSH-stimulated steroidogenesis in GC; however, the mechanism by which DEHP affects hormone-challenged steroidogenesis in human GC is not understood. Here, we analyzed the mechanism by which DEHP affects steroidogenesis in the primary culture of human cumulus granulosa cells (hCGC) stimulated with FSH. Cells were exposed to DEHP and FSH for 48 h, and steroidogenesis and the activation of cAMP and ERK1/2 were analyzed. The results show that DEHP decreases FSH-stimulated STAR and CYP19A1 expression, which is accompanied by a decrease in progesterone and estradiol production. DEHP lowers cAMP production and CREB phosphorylation in FSH but not cholera toxin- and forskolin-challenged hCGC. DEHP was not able to decrease steroidogenesis in cholera toxin- and forskolin-stimulated hCGC. Furthermore, DEHP decreases FSH-induced ERK1/2 phosphorylation. The addition of EGF rescued ERK1/2 phosphorylation in FSH- and DEHP-treated hCGC and prevented a decrease in steroidogenesis in the FSH- and DEHP-treated hCGC. These results suggest that DEHP inhibits the cAMP and ERK1/2 signaling pathways, leading to the inhibition of steroidogenesis in the FSH-stimulated hCGC.
Collapse
|
16
|
Sun X, Zhu H, Zhang C, Ilboudo JPHW, Zhao J, Ma C, Yan C, Liswaniso S, Qin N, Xu R. Transcriptomic analysis of ovarian follicles uncovers the crucial genes relevant to follicle selection and preovulatory hierarchy in hens. J Anim Sci 2023; 101:skad241. [PMID: 37453139 PMCID: PMC10414141 DOI: 10.1093/jas/skad241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023] Open
Abstract
Follicle selection and preovulatory hierarchy of hen ovaries were important stages of follicle development and crucially determining egg-laying performance. The selected follicles with a higher expression level of follicle-stimulating hormone receptor (FSHR) mRNA that facilitates response to FSH, and rapidly develops into preovulatory follicles with distinctive characteristics of granulosa cells (GCs) proliferation and differentiation. Identification of the key genes involved in these developmental events is helpful for elucidation of the molecular mechanism underlying egg-laying traits in chicken and other domestic fowl. Herein, the comparative transcriptomic analysis of ovarian prehierarchical follicles before selection (BSF), follicles at selection stage (ASF), and hierarchical follicles (HF) were implemented in the Jilin Black chicken (JB) and Lohmann Brown layer (LB) with the divergences in their egg-laying performance by RNA-sequencing. The results showed that nine deferentially expressed genes (DEGs), including STMN4, FABP3, ROBO2, RSPO4, and DMRT1 were revealed between follicles BSF and ASF; and seventeen DEGs, such as SLC6A15, SLITRK3, PRKG2 and TMC3 were mined between ASF and HF. These two group DEGs being co-expressed between BSF and ASF, and between ASF and HF were compared and substantiated in the JB and LB layers, respectively. Furthermore, 10 signaling pathways, such as cAMP signaling, PPAR signaling pathway, AMPK(Adenosine 5'-monophosphate (AMP)-activated protein kinase) pathway, and estrogen signaling pathway were also identified. Moreover, the roles of two representative candidates ROBO2 and PRKG2 genes presented as downregulated mRNA expression pattern in the transcriptomic profiles were further verified in vitro. The results demonstrated that downregulation of ROBO2 or PRKG2 significantly increased the expression levels of FSHR mRNA and protein with the boosted expression of CCND1, STAR, and BCL-2, whereas remarkably inhibited the expression of Caspase-3, consequently, brought about the decrease of GC apoptosis in the ovarian follicles, but increase of GC proliferation and differentiation serving as the hallmarks for follicle selection. It indicated that ROBO2 and PRKG2 may play indispensable roles in follicle selection and preovulatory hierarchy of hen ovaries separately. Our findings provided a comparative transcriptomic evidence for clarifying the molecular mechanism of the follicle development underlying egg-laying traits in chicken.
Collapse
Affiliation(s)
- Xue Sun
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Hongyan Zhu
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
- Department of Cell Biology and Genetics, College of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Changyan Zhang
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Jean Philippe Harold Wensesso Ilboudo
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Jinghua Zhao
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Chang Ma
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Chunchi Yan
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Simushi Liswaniso
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Ning Qin
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| | - Rifu Xu
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
| |
Collapse
|
17
|
Xia Q, Xie L, Wu Q, Cong J, Ma H, Li J, Cai W, Wu X. Elevated baseline LH/FSH ratio is associated with poor ovulatory response but better clinical pregnancy and live birth in Chinese women with PCOS after ovulation induction. Heliyon 2023; 9:e13024. [PMID: 36711322 PMCID: PMC9876966 DOI: 10.1016/j.heliyon.2023.e13024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Background What is the association between elevated baseline LH/FSH ratio and reproductive outcomes, especially ovulatory response, among Chinese women with polycystic ovary syndrome (PCOS) after ovulation induction. Methods This was a secondary analysis of a multicenter randomized trial in 1000 women with PCOS from 21 sites (27 hospitals) in Mainland China. LH and FSH levels before ovulation induction and the main outcomes including ovulation, biochemical pregnancy, clinical pregnancy, miscarriage, and live birth were measured. A linear regression model, logistic regression models and Cox proportional hazard regression model were used to estimate the association between LH/FSH ratios and reproductive outcomes in PCOS. Results LH/FSH ratio was significantly associated with age, body mass index (BMI), total testosterone (TT), estradiol (E2), free testosterone (FT), and antimullerian hormone (AMH). Anovulatory women had significantly higher LH/FSH ratio than ovulatory women (P = 0.003), especially in women with young age (P = 0.023), high BMI (P = 0.002), low E2 (P = 0.002), FT (P = 0.010), TT (P < 0.001) and AMH(P = 0.032). Women with elevated LH/FSH ratio were associated with lower ovulation (LH/FSH≥1 OR = 0.42, 95% CI, 0.26-0.68; LH/FSH≥2 OR = 0.32, 95% CI, 0.20-0.54; LH/FSH≥3 OR = 0.40, 95% CI 0.21-0.74) when compared with LH/FSH<1. The association was held after adjustment for treatment with or without the confounding factors. Although no association between LH/FSH ratio and biochemical pregnancy, women with 1 ≤ LH/FSH<2 were associated with higher clinical pregnancy (OR = 1.71; 95% CI, 1.09-2.67) and live birth (OR = 1.73; 95% CI, 1.09-2.75) compared to women with LH/FSH<1. Women with 2 ≤ LH/FSH<3 were associated with lower miscarriage rate (OR = 0.38; 95% CI, 0.16-0.93). Conclusions Elevated baseline LH/FSH ratio in women with PCOS was associated with poor ovulatory response, but women were more likely to achieve clinical pregnancy and live birth than women with normal LH/FSH. It suggests LH and FSH in women with PCOS may play a role in successful pregnancy despite of negative impact in ovulation.
Collapse
Affiliation(s)
- Qing Xia
- Department of Obstetrics and Gynaecology, Heilongjiang University of Chinese Medicine, Harbin 150001, China
- Department of Obstetrics and Gynaecology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100010, China
| | - Liangzhen Xie
- Department of Obstetrics and Gynaecology, Heilongjiang University of Chinese Medicine, Harbin 150001, China
| | - Qi Wu
- Department of Obstetrics and Gynaecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200000, China
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong City 999077, Hong Kong
| | - Jing Cong
- Department of Obstetrics and Gynaecology, Heilongjiang University of Chinese Medicine, Harbin 150001, China
| | - Hongli Ma
- Department of Obstetrics and Gynaecology, Heilongjiang University of Chinese Medicine, Harbin 150001, China
| | - Jian Li
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong City 999077, Hong Kong
- Department of Obstetrics and Gynecology, The Affiliated Hospital, Gui Zhou Medical University, Gui Zhou 550000, China
| | - Wangyu Cai
- Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Xiaoke Wu
- Department of Obstetrics and Gynaecology, Heilongjiang University of Chinese Medicine, Harbin 150001, China
- Heilongjiang Provincial Hospital, Harbin Institute of Technology, Harbin 150001, China
| |
Collapse
|
18
|
Huang C, Shi Q, Yan Y, Shen X, Shan H, Zhu Y, Sun H, Xing J, Kong N. Effect of Exogenous Luteinizing Hormone (LH) Supplementation on Clinical Pregnancy of Patients Receiving Long-Acting Gonadotropin-Releasing Hormone Agonist (GnRHa) Cycles: A Retrospective Cohort Study. Int J Womens Health 2022; 14:1691-1700. [PMID: 36536609 PMCID: PMC9759025 DOI: 10.2147/ijwh.s388726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/06/2022] [Indexed: 01/01/2025] Open
Abstract
PURPOSE Timely and moderate luteinizing hormone (LH) secretion plays critical roles in follicle development and maturation. However, the role of LH supplementation in in vitro fertilization/intracytoplasmic sperm injection and embryo transfer (IVF/ICSI-ET) cycles remains unclear. Can LH supplementation improve the clinical outcomes of patients who receive long-acting gonadotropin-releasing hormone agonist (GnRHa) pituitary downregulation in IVF/ICSI-ET cycles? PATIENTS AND METHODS This is a retrospective cohort study of 2600 long-acting GnRHa down-regulated IVF/ICSI cycles from 2017 to 2020 in our reproductive medicine centre of Nanjing Drum Tower Hospital. Total cycles were divided into two groups according to LH supplementation or not. In addition, we conducted a secondary analysis that used propensity-score matching to reduce the effects of confounding. RESULTS Exogenous LH addition was not significantly correlated with the clinical pregnancy rate (OR=0.910, 95% CI: 0.623-1.311, p=0.61) in logistic regression analysis. After propensity-score matching, we also found no significant association between LH supplementation and the clinical pregnancy rate (OR=0.792, 95% CI: 0.527-1.191, p=0.26). CONCLUSION There is no obvious effect of exogenous LH supplementation on the clinical pregnancy rate in non-selective patients receiving long-acting GnRHa IVF/ICSI-ET cycles, which suggests that exogenous LH addition is not always needed, which can help us avoid drug overuse to a certain extent.
Collapse
Affiliation(s)
- Chenyang Huang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Drum Tower Clinic Medical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, People’s Republic of China
| | - Qingqing Shi
- Center for Reproductive Medicine and Obstetrics and Gynecology, Drum Tower Clinic Medical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, People’s Republic of China
| | - Yuan Yan
- Center for Reproductive Medicine and Obstetrics and Gynecology, Drum Tower Clinic Medical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, People’s Republic of China
| | - Xiaoyue Shen
- Center for Reproductive Medicine and Obstetrics and Gynecology, Drum Tower Clinic Medical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, People’s Republic of China
| | - Huizhi Shan
- Center for Reproductive Medicine and Obstetrics and Gynecology, Drum Tower Clinic Medical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, People’s Republic of China
| | - Yingchun Zhu
- Center for Reproductive Medicine and Obstetrics and Gynecology, Drum Tower Clinic Medical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, People’s Republic of China
| | - Haixiang Sun
- Center for Reproductive Medicine and Obstetrics and Gynecology, Drum Tower Clinic Medical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, People’s Republic of China
| | - Jun Xing
- Center for Reproductive Medicine and Obstetrics and Gynecology, Drum Tower Clinic Medical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, People’s Republic of China
| | - Na Kong
- Center for Reproductive Medicine and Obstetrics and Gynecology, Drum Tower Clinic Medical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, People’s Republic of China
| |
Collapse
|
19
|
Paloviita P, Vuoristo S. The non-coding genome in early human development - Recent advancements. Semin Cell Dev Biol 2022; 131:4-13. [PMID: 35177347 DOI: 10.1016/j.semcdb.2022.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/08/2022] [Accepted: 02/08/2022] [Indexed: 12/14/2022]
Abstract
Not that long ago, the human genome was discovered to be mainly non-coding, that is comprised of DNA sequences that do not code for proteins. The initial paradigm that non-coding is also non-functional was soon overturned and today the work to uncover the functions of non-coding DNA and RNA in human early embryogenesis has commenced. Early human development is characterized by large-scale changes in genomic activity and the transcriptome that are partly driven by the coordinated activation and repression of repetitive DNA elements scattered across the genome. Here we provide examples of recent novel discoveries of non-coding DNA and RNA interactions and mechanisms that ensure accurate non-coding activity during human maternal-to-zygotic transition and lineage segregation. These include studies on small and long non-coding RNAs, transposable element regulation, and RNA tailing in human oocytes and early embryos. High-throughput approaches to dissect the non-coding regulatory networks governing early human development are a foundation for functional studies of specific genomic elements and molecules that has only begun and will provide a wider understanding of early human embryogenesis and causes of infertility.
Collapse
Affiliation(s)
- Pauliina Paloviita
- Department of Obstetrics and Gynaecology, University of Helsinki, 00014 Helsinki, Finland
| | - Sanna Vuoristo
- Department of Obstetrics and Gynaecology, University of Helsinki, 00014 Helsinki, Finland.
| |
Collapse
|
20
|
Ding N, Harlow SD, Randolph JF, Mukherjee B, Batterman S, Gold EB, Park SK. Perfluoroalkyl Substances and Incident Natural Menopause in Midlife Women: The Mediating Role of Sex Hormones. Am J Epidemiol 2022; 191:1212-1223. [PMID: 35292812 PMCID: PMC9393069 DOI: 10.1093/aje/kwac052] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 02/24/2022] [Accepted: 03/11/2022] [Indexed: 01/28/2023] Open
Abstract
Perfluoroalkyl and polyfluoroalkyl substances (PFAS) have been associated with earlier natural menopause; however, the underlying mechanisms are not well understood, particularly the extent to which this relationship is mediated by sex hormones. We analyzed data (1999-2017) on 1,120 premenopausal women from the Study of Women's Health Across the Nation (SWAN). Causal mediation analysis was applied to quantify the degree to which follicle-stimulating hormone (FSH) and estradiol levels could mediate the associations between PFAS and incident natural menopause. Participants with higher PFAS concentrations had shorter times to natural menopause, with a relative survival of 0.82 (95% confidence interval (CI): 0.69, 0.96) for linear perfluorooctane sulfonate (n-PFOS), 0.84 (95% CI: 0.69, 1.00) for sum of branched-chain perfluorooctane sulfonate (Sm-PFOS), 0.79 (95% CI: 0.66, 0.93) for linear-chain perfluorooctanoate (n-PFOA), and 0.84 (95% CI: 0.71, 0.97) for perfluorononanoate (PFNA), comparing the highest tertile of PFAS concentrations with the lowest. The proportion of the effect mediated through FSH was 8.5% (95% CI: -11.7, 24.0) for n-PFOS, 13.2% (95% CI: 0.0, 24.5) for Sm-PFOS, 26.9% (95% CI: 15.6, 38.4) for n-PFOA, and 21.7% (6.8, 37.0) for PFNA. No significant mediation by estradiol was observed. The effect of PFAS on natural menopause may be partially explained by variations in FSH concentrations.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sung Kyun Park
- Correspondence to Dr. Sung Kyun Park, Department of Epidemiology, School of Public Health, University of Michigan, M5541 SPH II, 1415 Washington Heights, Ann Arbor, MI 48109-2029 (e-mail: )
| |
Collapse
|
21
|
Wu YY, Li SY, Zhu HQ, Zhuang ZM, Shao M, Chen FL, Liu CS, Tang QF. Network pharmacology integrated with experimental validation reveals the regulatory mechanism of action of Hehuan Yin decoction in polycystic ovary syndrome with insulin resistance. JOURNAL OF ETHNOPHARMACOLOGY 2022; 289:115057. [PMID: 35121050 DOI: 10.1016/j.jep.2022.115057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/15/2022] [Accepted: 01/27/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hehuan Yin decoction (HHY), first recorded in the Jingyue Quanshu (published in 1624 A.D.), is composed of Albizia julibrissin Durazz. and Ampelopsis japonica (Thunb.) Makino. AIM OF THE STUDY This study aimed to investigate the mechanism of action of HHY in treating polycystic ovary syndrome with insulin resistance (PCOS-IR). MATERIALS AND METHODS Network pharmacology and molecular docking were used to predict active compounds, potential targets, and pathways for PCOS-IR treatment using HHY. Female Sprague-Dawley rats were administered letrozole (1 mg/kg) with a high-fat diet to establish a PCOS-IR model. Thereafter, symptoms, ovarian pathology, serum insulin resistance, and sex hormone levels were determined. Western blotting was used to determine the levels of PI3Kp85α, AKT, phospho (p)-AKT, and GSK3β in the ovaries of rats. RESULTS Network pharmacology revealed 58 components in HHY and 182 potential targets that were shared between HHY and PCOS-IR. HHY could potentially treat PCOS-IR via the insulin resistance, PI3K/AKT, HIF-1, and steroid hormone biosynthesis pathways. Molecular docking revealed that PI3K, AKT1, GSK3β, IRS1, and EGFR had high affinities to HHY compounds. In the PCOS-IR rats, HHY significantly normalised the symptoms and ovarian pathology, increased follicle-stimulating hormone (FSH) and oestradiol levels in the serum, and decreased the levels of fasting plasma glucose and fasting insulin, as well as the insulin resistance index. HHY also decreased the luteinising hormone (LH) and testosterone levels and the LH/FSH ratio in the PCOS-IR rats and increased the levels of PI3K, p-AKT, and GSK3β in ovary tissue, which indicated the activation of the PI3K/AKT pathway. CONCLUSIONS HHY can improve PCOS-IR symptoms via multiple pharmacological pathways and may be a potential alternative therapy for the treatment of PCOS-IR.
Collapse
Affiliation(s)
- Yuan-Yuan Wu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China
| | - Shu-Yu Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China
| | - Hui-Qing Zhu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China
| | - Zi-Ming Zhuang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China
| | - Meng Shao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China
| | - Fei-Long Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China
| | - Chang-Shun Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China.
| | - Qing-Fa Tang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China.
| |
Collapse
|
22
|
Differential expression of FSHR and LHR genes and proteins during development of rabbit ovarian follicles. ZYGOTE 2022; 30:577-583. [DOI: 10.1017/s0967199421000861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Summary
The development of an ovarian follicle is a complex process at the cellular and molecular level that is mainly regulated by follicle-stimulating hormone receptor (FSHR) and luteinizing hormone receptor (LHR). To elucidate the contribution of these receptors to ovarian follicle development, it is necessary to determine their expression profiles during this biological process. Therefore, this study aimed to investigate the relationship between ovarian development pattern and the differential ovarian expression pattern of FSHR and LHR genes as well as proteins at different developmental stages. Ovaries were collected from 30 New Zealand rabbits at day 0 (birth), week 2 (neonate), week 4 (cub), week 16 (maturity), and day 18 pregnancy. Ovarian histology, and gene as well as protein expression were determined using light microscopy, real-time PCR and western blotting, respectively. The results showed that the expression levels of FSHR mRNA and protein increased coincidently with age and the growth of ovarian follicles. The levels of LHR mRNA and protein remained low from the day of birth until week 4 and became significantly higher by week 16 coinciding with appearance of growing and antral follicles, which have a defined thecal layer. FSHR gene and protein expression decreased with pregnancy, whereas LHR increased, reaching a peak level during pregnancy. It can be concluded that changes in FSHR and LHR gene and protein expression could be related to the growth and development of follicles, indicating the regulatory role for these receptors in rabbit folliculogenesis.
Collapse
|
23
|
The Novel Competing Endogenous Long Noncoding RNA SM2 Regulates Gonadotropin Secretion in the Hu Sheep Anterior Pituitary by Targeting the Oar-miR-16b/TGF-β/SMAD2 Signaling Pathway. Cells 2022; 11:cells11060985. [PMID: 35326436 PMCID: PMC8947352 DOI: 10.3390/cells11060985] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/27/2022] [Accepted: 03/08/2022] [Indexed: 02/01/2023] Open
Abstract
Pituitary gonadotropins play a pivotal role in reproduction. Long noncoding RNAs (lncRNAs) have been identified as important regulators in the hypothalamic−pituitary−ovarian (HPO) axis associated with reproduction. However, the contributions of lncRNAs to pituitary gonadotropin secretion remain largely unknown. Therefore, this work was performed to uncover the functional mechanisms of the novel lncRNA TCONS_00083279 (lncRNA SM2) and its potential targeting pathway oar-miR-16b/TGF-beta/SMAD2, which is associated with gonadotropin secretion in sheep pituitary cells. In the present study, the lncRNA SM2 showed high expression levels in the sheep pituitary gland, and it was located in both the nucleus and the cytoplasm of pituitary cells. lncRNA SM2 knockdown inhibited pituitary cell proliferation and FSH and LH secretion. The function of the lncRNA SM2 was sponged by oar-miR-16b, and this regulated the growth and gonadotropin secretion of pituitary cells by modulating SMAD2, as shown by the dual-luciferase reporter assay. FSH and LH levels were both upregulated by SMAD2 overexpression. Moreover, the levels of the lncRNA SM2, SMAD2 and TGFR1, as well as FSH and LH, in sheep pituitary cells increased significantly under gonadotropin-releasing hormone (GnRH) stimulation (p < 0.05). This work illustrates that the lncRNA SM2 regulates gonadotropin secretion in the Hu sheep anterior pituitary by targeting the oar-miR-16b/TGF-β/SMAD2 signaling pathway, providing a valuable resource for understanding the molecular mechanisms underlying sheep reproduction.
Collapse
|
24
|
A potential role of fibrillin-1 (FBN1) mRNA and asprosin in follicular development in water buffalo. Theriogenology 2022; 178:67-72. [PMID: 34781067 DOI: 10.1016/j.theriogenology.2021.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 10/17/2021] [Accepted: 11/04/2021] [Indexed: 11/23/2022]
Abstract
Fibrillin-1 (FBN1) functions as a structural protein in the ovary, while the role of its protein product asprosin remains unknown. Both proteins are encoded by the FBN1 gene and when it is cleaved at the C-terminal end, asprosin is produced. Asprosin is associated with various metabolic parameters and sex-related hormones in women. One goal of this research was to quantify FBN1 and the presumed asprosin receptor, olfactory receptor family 4 subfamily M member 1 (OR4M1) mRNA in water buffalo granulosa cells and correlate them to aromatase (CYP19A1) gene expression. A second goal was to determine the effect of asprosin on follicular growth in vivo. In Exp. 1, ovaries were collected from a local slaughterhouse, follicular fluid and granulosa cells from small (<6 mm) and large (6-13 mm) follicles were aspirated, cellular RNA extracted for gene expression analysis, data analyzed using ANOVA, and Pearson correlation coefficients were calculated among FBN1, OR4M1, and CYP19A1 gene expression. In Exp. 2, an intra-follicular injection of asprosin (600 ng of asprosin/194 μL of PBS) or vehicle (200 μL of PBS; Controls) was given via the theca layer of the dominant follicle of synchronized cows (n = 5/group) 1 day after injection of PGF2α, follicle sizes were measured daily via transrectal ultrasonography for 3 days, a two-way repeated measures ANOVA was used to determine the effect of asprosin on growth rate of follicles from day 0-2, and Chi-square analysis for the percentage of cows ovulated 2 days following asprosin injections. In Exp. 1, FBN1 mRNA abundance was 1.9-fold greater in cells of follicular aspirates from small than large follicles (P < 0.05), but abundance of OR4M1 and CYP19A1 mRNA did not differ (P > 0.10) between the two sizes of follicles. Abundance of FBN1 mRNA was positively correlated with CYP19A1 (r = 0.55, P < 0.05) and OR4M1 mRNA (r = 0.50, P < 0.06) across follicle sizes. In Exp. 2, cows treated with asprosin revealed a greater follicle growth rate from day 0-2 (63.4% increase in diameter) than placebo cows (36.8% increase in diameter) post-injection, and more follicles from asprosin treatment vs. control group (100% vs. 20%; P < 0.05) ovulated within 2 days. These findings suggest that FBN1 may be developmentally regulated in follicular cells, and that asprosin may induce follicular growth in buffaloes, but further studies will be required to determine if asprosin directly regulates estradiol production during follicle development.
Collapse
|
25
|
Zheng X, Zheng Y, Qin D, Yao Y, Zhang X, Zhao Y, Zheng C. Regulatory Role and Potential Importance of GDF-8 in Ovarian Reproductive Activity. Front Endocrinol (Lausanne) 2022; 13:878069. [PMID: 35692411 PMCID: PMC9178251 DOI: 10.3389/fendo.2022.878069] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/15/2022] [Indexed: 11/13/2022] Open
Abstract
Growth differentiation factor-8 (GDF-8) is a member of the transforming growth factor-beta superfamily. Studies in vitro and in vivo have shown GDF-8 to be involved in the physiology and pathology of ovarian reproductive functions. In vitro experiments using a granulosa-cell model have demonstrated steroidogenesis, gonadotrophin responsiveness, glucose metabolism, cell proliferation as well as expression of lysyl oxidase and pentraxin 3 to be regulated by GDF-8 via the mothers against decapentaplegic homolog signaling pathway. Clinical data have shown that GDF-8 is expressed widely in the human ovary and has high expression in serum of obese women with polycystic ovary syndrome. GDF-8 expression in serum changes dynamically in patients undergoing controlled ovarian hyperstimulation. GDF-8 expression in serum and follicular fluid is correlated with the ovarian response and pregnancy outcome during in vitro fertilization. Blocking the GDF-8 signaling pathway is a potential therapeutic for ovarian hyperstimulation syndrome and ovulation disorders in polycystic ovary syndrome. GDF-8 has a regulatory role and potential importance in ovarian reproductive activity and may be involved in folliculogenesis, ovulation, and early embryo implantation.
Collapse
Affiliation(s)
- Xiaoling Zheng
- Department of Pharmacy, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongquan Zheng
- Department of Pharmacy, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Dongxu Qin
- Department of Pharmacy, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yao Yao
- Department of Pharmacy, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiao Zhang
- Department of Pharmacy, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yunchun Zhao
- Department of Pharmacy, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Caihong Zheng, ; Yunchun Zhao,
| | - Caihong Zheng
- Department of Pharmacy, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Women’s Reproductive Health Laboratory of Zhejiang Province, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Caihong Zheng, ; Yunchun Zhao,
| |
Collapse
|
26
|
Dynamics of Known Long Non-Coding RNAs during the Maternal-to-Zygotic Transition in Rabbit. Animals (Basel) 2021; 11:ani11123592. [PMID: 34944367 PMCID: PMC8698111 DOI: 10.3390/ani11123592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/05/2021] [Accepted: 12/14/2021] [Indexed: 01/06/2023] Open
Abstract
The control of pre-implantation development in mammals undergoes a maternal-to-zygotic transition (MZT) after fertilization. The transition involves maternal clearance and zygotic genome activation remodeling the terminal differentiated gamete to confer totipotency. In the study, we first determined the profile of long non-coding RNAs (lncRNAs) of mature rabbit oocyte, 2-cell, 4-cell, 8-cell, and morula embryos using RNA-seq. A total of 2673 known rabbit lncRNAs were identified. The lncRNAs exhibited dynamic expression patterns during pre-implantation development. Moreover, 107 differentially expressed lncRNAs (DE lncRNAs) were detected between mature oocyte and 2-cell embryo, while 419 DE lncRNAs were detected between 8-cell embryo and morula, consistent with the occurrence of minor and major zygotic genome activation (ZGA) wave of rabbit pre-implanted embryo. This study then predicted the potential target genes of DE lncRNAs based on the trans-regulation mechanism of lncRNAs. The GO and KEGG analyses showed that lncRNAs with stage-specific expression patterns promoted embryo cleavage and synchronic development by regulating gene transcription and translation, intracellular metabolism and organelle organization, and intercellular signaling transduction. The correlation analysis between mRNAs and lncRNAs identified that lncRNAs ENSOCUG00000034943 and ENSOCUG00000036338 may play a vital role in the late-period pre-implantation development by regulating ILF2 gene. This study also found that the sequential degradation of maternal lncRNAs occurred through maternal and zygotic pathways. Furthermore, the function analysis of the late-degraded lncRNAs suggested that these lncRNAs may play a role in the mRNA degradation in embryos via mRNA surveillance pathway. Therefore, this work provides a global view of known lncRNAs in rabbit pre-implantation development and highlights the role of lncRNAs in embryogenesis regulation.
Collapse
|
27
|
Lin Y, Chen Q, Zhu J, Teng Y, Huang X, Chen X. Progestin-Primed Ovarian Stimulation with Clomiphene Citrate Supplementation May Be More Feasible for Young Women with Diminished Ovarian Reserve Compared with Standard Progestin-Primed Ovarian Stimulation: A Retrospective Study. Drug Des Devel Ther 2021; 15:5087-5097. [PMID: 34992345 PMCID: PMC8710074 DOI: 10.2147/dddt.s338748] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 12/07/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose Patients and Methods Results Conclusion
Collapse
Affiliation(s)
- Yue Lin
- Reproductive Medicine Center, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Qianqian Chen
- Reproductive Medicine Center, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Jing Zhu
- Reproductive Medicine Center, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Yili Teng
- Reproductive Medicine Center, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Xuefeng Huang
- Reproductive Medicine Center, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Xia Chen
- Reproductive Medicine Center, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
- Correspondence: Xia Chen; Xuefeng Huang Reproductive Medicine Center, First Affiliated Hospital of Wenzhou Medical University, 96 Fuxue Road, Wenzhou, Zhejiang, People’s Republic of ChinaTel +86-577-88069380 Email ;
| |
Collapse
|
28
|
Recombinant luteinizing hormone supplementation in assisted reproductive technology: a review of literature. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2021. [DOI: 10.1186/s43043-021-00083-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Luteinizing hormone (LH) has the main role in ovarian function in both natural and artificial cycles. A normal LH concentration during controlled ovarian hyperstimulation is positively correlated to the number and quality of retrieved oocytes and resulting embryos.
Main body of the abstract
In this study, we reviewed whether rLH administration, adjunct to the ovarian stimulation regimen, could improve clinical outcomes. The literature review showed that rLH supplementation improves assisted reproductive technology (ART) outcomes among women with hypogonadotropic hypogonadism, and hyporesponsive women to follicle-stimulating hormone monotherapy. Besides, rLH supplementation has advantages for poor responder women 36–39 years of age. Even though the data suggested no priority regarding the LH source for improving ART outcome, women with different LH polymorphisms who did not respond similarly to ovarian stimulation may benefit from adjuvant rLH therapy.
Conclusion
rLH usage for improving ART outcome should be scrutinized via well-designed studies considering the subgroups of infertile women who benefit the most from rLH adjuvant therapy, the type of ovarian stimulation protocol to which rLH would be added, and also the exact dosage, as well as the proper timing (during or prior to a cycle).
Collapse
|
29
|
Zhang M, Tian Y, Zhang S, Yan H, Ge W, Han B, Yan Z, Cheng S, Shen W. The proliferation role of LH on porcine primordial germ cell-like cells (pPGCLCs) through ceRNA network construction. Clin Transl Med 2021; 11:e560. [PMID: 34709759 PMCID: PMC8516341 DOI: 10.1002/ctm2.560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The transdifferentiation of skin-derived stem cells (SDSCs) into primordial germ cell-like cells (PGCLCs) is one of the major breakthroughs in the field of stem cells research in recent years. This technology provides a new theoretical basis for the treatment of human infertility. However, the transdifferentiation efficiency of SDSCs to PGCLCs is very low, and scientists are still exploring ways to improve this efficiency or promote the proliferation of PGCLCs. This study aims to investigate the molecular mechanism of luteinising hormone (LH) to enhance porcine PGCLCs (pPGCLCs) proliferation. RESULTS In this study, we dissected the proliferation regulatory network of pPGCLCs by whole transcriptome sequencing, and the results showed that the pituitary-secreted reproductive hormone LH significantly promoted the proliferation of pPGCLCs. We combined whole transcriptome sequencing and related validation experiments to explore the mechanism of LH on the proliferation of pPGCLCs, and found that LH could affect the expression of Hippo signalling pathway-related mRNAs, miRNAs and lncRNAs in pPGCLCs. CONCLUSIONS For the first time, we found that LH promotes pPGCLCs proliferation through the competing endogenous RNA (ceRNA) regulatory networks and Hippo signalling pathway. This finding may help to elucidate the molecular mechanism by which LH promotes pPGCLCs proliferation.
Collapse
Affiliation(s)
- Ming‐Yu Zhang
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of ShandongQingdao Agricultural UniversityQingdaoChina
| | - Yu Tian
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of ShandongQingdao Agricultural UniversityQingdaoChina
| | - Shu‐Er Zhang
- Animal Husbandry General Station of Shandong ProvinceJinanChina
| | - Hong‐Chen Yan
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of ShandongQingdao Agricultural UniversityQingdaoChina
| | - Wei Ge
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of ShandongQingdao Agricultural UniversityQingdaoChina
| | - Bao‐Quan Han
- Urology DepartmentPeking University Shenzhen HospitalShenzhenChina
| | - Zi‐Hui Yan
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of ShandongQingdao Agricultural UniversityQingdaoChina
| | - Shun‐Feng Cheng
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of ShandongQingdao Agricultural UniversityQingdaoChina
| | - Wei Shen
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of ShandongQingdao Agricultural UniversityQingdaoChina
| |
Collapse
|
30
|
Lee EB, Chakravarthi VP, Wolfe MW, Rumi MAK. ERβ Regulation of Gonadotropin Responses during Folliculogenesis. Int J Mol Sci 2021; 22:ijms221910348. [PMID: 34638689 PMCID: PMC8508937 DOI: 10.3390/ijms221910348] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/11/2022] Open
Abstract
Gonadotropins are essential for regulating ovarian development, steroidogenesis, and gametogenesis. While follicle stimulating hormone (FSH) promotes the development of ovarian follicles, luteinizing hormone (LH) regulates preovulatory maturation of oocytes, ovulation, and formation of corpus luteum. Cognate receptors of FSH and LH are G-protein coupled receptors that predominantly signal through cAMP-dependent and cAMP-independent mechanisms that activate protein kinases. Subsequent vital steps in response to gonadotropins are mediated through activation or inhibition of transcription factors required for follicular gene expression. Estrogen receptors, classical ligand-activated transcriptional regulators, play crucial roles in regulating gonadotropin secretion from the hypothalamic-pituitary axis as well as gonadotropin function in the target organs. In this review, we discuss the role of estrogen receptor β (ERβ) regulating gonadotropin response during folliculogenesis. Ovarian follicles in Erβ knockout (ErβKO) mutant female mice and rats cannot develop beyond the antral state, lack oocyte maturation, and fail to ovulate. Theca cells (TCs) in ovarian follicles express LH receptor, whereas granulosa cells (GCs) express both FSH receptor (FSHR) and LH receptor (LHCGR). As oocytes do not express the gonadotropin receptors, the somatic cells play a crucial role during gonadotropin induced oocyte maturation. Somatic cells also express high levels of estrogen receptors; while TCs express ERα and are involved in steroidogenesis, GCs express ERβ and are involved in both steroidogenesis and folliculogenesis. GCs are the primary site of ERβ-regulated gene expression. We observed that a subset of gonadotropin-induced genes in GCs, which are essential for ovarian follicle development, oocyte maturation and ovulation, are dependent on ERβ. Thus, ERβ plays a vital role in regulating the gonadotropin responses in ovary.
Collapse
Affiliation(s)
- Eun B. Lee
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (V.P.C.)
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - V. Praveen Chakravarthi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (V.P.C.)
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA;
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Michael W. Wolfe
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA;
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - M. A. Karim Rumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (V.P.C.)
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Correspondence: ; Tel.: +1-913-588-8059
| |
Collapse
|
31
|
Recchia K, Jorge AS, Pessôa LVDF, Botigelli RC, Zugaib VC, de Souza AF, Martins DDS, Ambrósio CE, Bressan FF, Pieri NCG. Actions and Roles of FSH in Germinative Cells. Int J Mol Sci 2021; 22:10110. [PMID: 34576272 PMCID: PMC8470522 DOI: 10.3390/ijms221810110] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 12/21/2022] Open
Abstract
Follicle stimulating hormone (FSH) is produced by the pituitary gland in a coordinated hypothalamic-pituitary-gonadal (HPG) axis event, plays important roles in reproduction and germ cell development during different phases of reproductive development (fetal, neonatal, puberty, and adult life), and is consequently essential for fertility. FSH is a heterodimeric glycoprotein hormone of two dissociable subunits, α and β. The FSH β-subunit (FSHβ) function starts upon coupling to its specific receptor: follicle-stimulating hormone receptor (FSHR). FSHRs are localized mainly on the surface of target cells on the testis and ovary (granulosa and Sertoli cells) and have recently been found in testicular stem cells and extra-gonadal tissue. Several reproduction disorders are associated with absent or low FSH secretion, with mutation of the FSH β-subunit or the FSH receptor, and/or its signaling pathways. However, the influence of FSH on germ cells is still poorly understood; some studies have suggested that this hormone also plays a determinant role in the self-renewal of germinative cells and acts to increase undifferentiated spermatogonia proliferation. In addition, in vitro, together with other factors, it assists the process of differentiation of primordial germ cells (PGCLCs) into gametes (oocyte-like and SSCLCs). In this review, we describe relevant research on the influence of FSH on spermatogenesis and folliculogenesis, mainly in the germ cell of humans and other species. The possible roles of FSH in germ cell generation in vitro are also presented.
Collapse
Affiliation(s)
- Kaiana Recchia
- Department of Surgery, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo 01001-010, Brazil; (K.R.); (F.F.B.)
| | - Amanda Soares Jorge
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, Brazil; (A.S.J.); (L.V.d.F.P.); (R.C.B.); (V.C.Z.); (D.d.S.M.); (C.E.A.)
| | - Laís Vicari de Figueiredo Pessôa
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, Brazil; (A.S.J.); (L.V.d.F.P.); (R.C.B.); (V.C.Z.); (D.d.S.M.); (C.E.A.)
| | - Ramon Cesar Botigelli
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, Brazil; (A.S.J.); (L.V.d.F.P.); (R.C.B.); (V.C.Z.); (D.d.S.M.); (C.E.A.)
- Department of Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-970, Brazil
| | - Vanessa Cristiane Zugaib
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, Brazil; (A.S.J.); (L.V.d.F.P.); (R.C.B.); (V.C.Z.); (D.d.S.M.); (C.E.A.)
| | - Aline Fernanda de Souza
- Department Biomedical Science, Ontary Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Daniele dos Santos Martins
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, Brazil; (A.S.J.); (L.V.d.F.P.); (R.C.B.); (V.C.Z.); (D.d.S.M.); (C.E.A.)
| | - Carlos Eduardo Ambrósio
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, Brazil; (A.S.J.); (L.V.d.F.P.); (R.C.B.); (V.C.Z.); (D.d.S.M.); (C.E.A.)
| | - Fabiana Fernandes Bressan
- Department of Surgery, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo 01001-010, Brazil; (K.R.); (F.F.B.)
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, Brazil; (A.S.J.); (L.V.d.F.P.); (R.C.B.); (V.C.Z.); (D.d.S.M.); (C.E.A.)
| | - Naira Caroline Godoy Pieri
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, Brazil; (A.S.J.); (L.V.d.F.P.); (R.C.B.); (V.C.Z.); (D.d.S.M.); (C.E.A.)
| |
Collapse
|
32
|
Zhou X, He Y, Li N, Bai G, Pan X, Zhang Z, Zhang H, Li J, Yuan X. DNA methylation mediated RSPO2 to promote follicular development in mammals. Cell Death Dis 2021; 12:653. [PMID: 34175894 PMCID: PMC8236063 DOI: 10.1038/s41419-021-03941-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/18/2022]
Abstract
In female mammals, the proliferation, apoptosis, and estradiol-17β (E2) secretion of granulosa cells (GCs) have come to decide the fate of follicles. DNA methylation and RSPO2 gene of Wnt signaling pathway have been reported to involve in the survival of GCs and follicular development. However, the molecular mechanisms for how DNA methylation regulates the expression of RSPO2 and participates in the follicular development are not clear. In this study, we found that the mRNA and protein levels of RSPO2 significantly increased during follicular development, but the DNA methylation level of RSPO2 promoter decreased gradually. Inhibition of DNA methylation or DNMT1 knockdown could decrease the methylation level of CpG island (CGI) in RSPO2 promoter and upregulate the expression level of RSPO2 in porcine GCs. The hypomethylation of -758/-749 and -563/-553 regions in RSPO2 promoter facilitated the occupancy of transcription factor E2F1 and promoted the transcriptional activity of RSPO2. Moreover, RSPO2 promoted the proliferation of GCs with increasing the expression level of PCNA, CDK1, and CCND1 and promoted the E2 secretion of GCs with increasing the expression level of CYP19A1 and HSD17B1 and inhibited the apoptosis of GCs with decreasing the expression level of Caspase3, cleaved Caspase3, cleaved Caspase8, cleaved Caspase9, cleaved PARP, and BAX. In addition, RSPO2 knockdown promoted the apoptosis of GCs, blocked the development of follicles, and delayed the onset of puberty with decreasing the expression level of Wnt signaling pathway-related genes (LGR4 and CTNNB1) in vivo. Taken together, the hypomethylation of -758/-749 and -563/-553 regions in RSPO2 promoter facilitated the occupancy of E2F1 and enhanced the transcription of RSPO2, which further promoted the proliferation and E2 secretion of GCs, inhibited the apoptosis of GCs, and ultimately ameliorated the development of follicles through Wnt signaling pathway. This study will provide useful information for further exploration on DNA-methylation-mediated RSPO2 pathway during follicular development.
Collapse
Affiliation(s)
- Xiaofeng Zhou
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Yingting He
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Nian Li
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Guofeng Bai
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Xiangchun Pan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Zhe Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Hao Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jiaqi Li
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.
| | - Xiaolong Yuan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Laboratory Animals, Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China.
| |
Collapse
|
33
|
Yu L, Zhai J, Wang Y, Geng Y, Chen X, Wen Y, Tang H, Yu R, Zhang Y, Liu X. Exposure to N-monoacetyl-p-phenylenediamine impaired ovarian function in mice. J Appl Toxicol 2021; 41:2031-2041. [PMID: 34014586 DOI: 10.1002/jat.4183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/14/2021] [Accepted: 04/29/2021] [Indexed: 01/14/2023]
Abstract
p-Phenylenediamine (PPD) is the main constituent of permanent hair dye and is also widely used in the photographic and rubber industries. PPD and its metabolites have been shown to increase the risk of cancer (especially ovarian cancer); however, their effect on female reproduction is unclear. We investigated the effects of the PPD metabolite N-monoacetyl-PPD (MAPPD) on mouse blastocyst development and ovarian function. Sixty 8-week-old female Kunming mice were administered at 0-, 100-, and 300-mg/kg/day MPPD by gavage for 28 days. KGN (human ovarian granulosa cells) were treated with MAPPD at concentrations of 0, 50, 100, and 300 μg/ml for 48 h. The number of abnormal blastocysts increased on gestation day 3.5 in all treatment groups. Compared with the control group, in MAPPD exposed group, the number of antral follicles decreased, the levels of E2 and P4 decreased in ovarian tissue, the serum levels of E2 , P4 , luteinizing hormone (LH), and T decreased, and follicle-stimulating hormone (FSH) increased. The expression of FSH receptor (FSHR) and LH receptor (LHR) was significantly downregulated, and the level of oxidative stress was significantly increased. In KGN cells, the level of reactive oxygen species increased in a dose-dependent manner, and the mRNA levels of FSHR, LHR, and aromatase increased. These results suggest that MAPPD inhibits FSH- and LH-induced aromatase activity by causing oxidative stress, which decrease hormone levels, leading to abnormal follicle development. Meanwhile, MAPPD exposure could affect early embryonic development abnormalities by affecting the quality of ovum.
Collapse
Affiliation(s)
- Liliang Yu
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Jingwei Zhai
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Yingxiong Wang
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Yanqing Geng
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Xuemei Chen
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Yixian Wen
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Hongyu Tang
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Rao Yu
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Yue Zhang
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Xueqing Liu
- Joint International Research Laboratory of Reproductive and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| |
Collapse
|
34
|
Jonas KC, Rivero Müller A, Oduwole O, Peltoketo H, Huhtaniemi I. The Luteinizing Hormone Receptor Knockout Mouse as a Tool to Probe the In Vivo Actions of Gonadotropic Hormones/Receptors in Females. Endocrinology 2021; 162:6144965. [PMID: 33605422 PMCID: PMC8171189 DOI: 10.1210/endocr/bqab035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Indexed: 11/25/2022]
Abstract
Mouse models with altered gonadotropin functions have provided invaluable insight into the functions of these hormones/receptors. Here we describe the repurposing of the infertile and hypogonadal luteinizing hormone receptor (LHR) knockout mouse model (LuRKO), to address outstanding questions in reproductive physiology. Using crossbreeding strategies and physiological and histological analyses, we first addressed the physiological relevance of forced LHR homomerization in female mice using BAC expression of 2 ligand-binding and signaling deficient mutant LHR, respectively, that have previously shown to undergo functional complementation and rescue the hypogonadal phenotype of male LuRKO mice. In female LuRKO mice, coexpression of signaling and binding deficient LHR mutants failed to rescue the hypogonadal and anovulatory phenotype. This was apparently due to the low-level expression of the 2 mutant LHR and potential lack of luteinizing hormone (LH)/LHR-dependent pleiotropic signaling that has previously been shown at high receptor densities to be essential for ovulation. Next, we utilized a mouse model overexpressing human chorionic gonadotropin (hCG) with increased circulating "LH/hCG"-like bioactivity to ~40 fold higher than WT females, to determine if high circulating hCG in the LuRKO background could reveal putative LHR-independent actions. No effects were found, thus, suggesting that LH/hCG mediate their gonadal and non-gonadal effects solely via LHR. Finally, targeted expression of a constitutively active follicle stimulating hormone receptor (FSHR) progressed antral follicles to preovulatory follicles and displayed phenotypic markers of enhanced estrogenic activity but failed to induce ovulation in LuRKO mice. This study highlights the critical importance and precise control of functional LHR and FSHR for mediating ovarian functions and of the potential repurposing of existing genetically modified mouse models in answering outstanding questions in reproductive physiology.
Collapse
Affiliation(s)
- Kim Carol Jonas
- Department of Women and Children’s Health, King’s College London,
London SE1 1UL, UK
- Institute of Reproductive and Developmental Biology, Department of Metabolism,
Digestion and Reproduction, Imperial College London, London W12
0NN, UK
- Correspondence: Dr Kim Jonas, Department of Women and Children’s Health, King’s College London,
London SE1 1UL, UK; Institute of Reproductive and Developmental Biology, Department of
Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK.
; or Prof. Ilpo Huhtaniemi, Institute of Reproductive and
Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial
College London, London, W12 0NN, UK; Institute for Biomedicine, Department of Physiology,
University of Turku, 20520 Turku, Finland.
| | - Adolfo Rivero Müller
- Institute for Biomedicine, Department of Physiology, University of
Turku, 20520 Turku, Finland
- Department of Biochemistry and Molecular Biology, Medical University of
Lublin, 20-093 Lublin, Poland
| | - Olayiwola Oduwole
- Institute of Reproductive and Developmental Biology, Department of Metabolism,
Digestion and Reproduction, Imperial College London, London W12
0NN, UK
| | - Hellevi Peltoketo
- Institute of Reproductive and Developmental Biology, Department of Metabolism,
Digestion and Reproduction, Imperial College London, London W12
0NN, UK
- Laboratory of Cancer Genetics and Tumour Biology, Cancer and Translational
Medicine Research Unit, Biocenter Oulu and University of Oulu,
90220 Oulu, Finland
| | - Ilpo Huhtaniemi
- Institute of Reproductive and Developmental Biology, Department of Metabolism,
Digestion and Reproduction, Imperial College London, London W12
0NN, UK
- Institute for Biomedicine, Department of Physiology, University of
Turku, 20520 Turku, Finland
- Correspondence: Dr Kim Jonas, Department of Women and Children’s Health, King’s College London,
London SE1 1UL, UK; Institute of Reproductive and Developmental Biology, Department of
Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK.
; or Prof. Ilpo Huhtaniemi, Institute of Reproductive and
Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial
College London, London, W12 0NN, UK; Institute for Biomedicine, Department of Physiology,
University of Turku, 20520 Turku, Finland.
| |
Collapse
|
35
|
Abstract
Gonadotropins are glycoprotein sex hormones regulating development and reproduction and bind to specific G protein–coupled receptors expressed in the gonads. Their effects on multiple signaling cascades and intracellular events have recently been characterized using novel technological and scientific tools. The impact of allosteric modulators on gonadotropin signaling, the role of sugars linked to the hormone backbone, the detection of endosomal compartments supporting signaling modules, and the dissection of different effects mediated by these molecules are areas that have advanced significantly in the last decade. The classic view providing the exclusive activation of the cAMP/protein kinase A (PKA) and the steroidogenic pathway by these hormones has been expanded with the addition of novel signaling cascades as determined by high-resolution imaging techniques. These new findings provided new potential therapeutic applications. Despite these improvements, unanswered issues of gonadotropin physiology, such as the intrinsic pro-apoptotic potential to these hormones, the existence of receptors assembled as heteromers, and their expression in extragonadal tissues, remain to be studied. Elucidating these issues is a challenge for future research.
Collapse
Affiliation(s)
- Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria di Modena, Via P. Giardini 1355, 41126 Modena, Italy
| |
Collapse
|
36
|
Orisaka M, Miyazaki Y, Shirafuji A, Tamamura C, Tsuyoshi H, Tsang BK, Yoshida Y. The role of pituitary gonadotropins and intraovarian regulators in follicle development: A mini-review. Reprod Med Biol 2021; 20:169-175. [PMID: 33850449 PMCID: PMC8022101 DOI: 10.1002/rmb2.12371] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/15/2021] [Accepted: 01/25/2021] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND The process of follicle development is tightly regulated by pituitary gonadotropins (follicle-stimulating hormone [FSH] and luteinizing hormone [LH]) and intraovarian regulators (eg, steroids, growth factors, and cytokines). METHODS This review outlines recent findings on the mechanisms of human follicle development, based on the research on animal models such as mice, rats, cows, and sheep. MAIN FINDINGS Phosphatidylinositol 3-kinase/protein kinase B signaling pathway and anti-Müllerian hormone are involved in primordial follicle activation during the gonadotropin-independent phase. The intraovarian regulators, such as androgen, insulin-like growth factor system, activin, oocyte-derived factors (growth differentiation factor-9 and bone morphogenetic protein 15), and gap junction membrane channel protein (connexin), play a central role in the acquisition of FSH dependence in preantral follicles during the gonadotropin-responsive phase. Antral follicle development can be divided into FSH-dependent growth and LH-dependent maturation. The indispensable tetralogy for follicle selection and final maturation of antral follicles involves (a) acquisition of LH dependence, (b) greater capacity for E2 production, (c) activation of the IGF system, and (d) an antiapoptotic follicular microenvironment. CONCLUSION We reproductive endocrinologists should accumulate further knowledge from animal model studies to develop methods that promote early folliculogenesis and connect to subsequent gonadotropin therapy in infertile women.
Collapse
Affiliation(s)
- Makoto Orisaka
- Department of Obstetrics and GynecologyUniversity of FukuiYoshida‐GunJapan
| | - Yumiko Miyazaki
- Department of Obstetrics and GynecologyUniversity of FukuiYoshida‐GunJapan
| | - Aya Shirafuji
- Department of Obstetrics and GynecologyUniversity of FukuiYoshida‐GunJapan
| | - Chiyo Tamamura
- Department of Obstetrics and GynecologyUniversity of FukuiYoshida‐GunJapan
| | - Hideaki Tsuyoshi
- Department of Obstetrics and GynecologyUniversity of FukuiYoshida‐GunJapan
| | - Benjamin K. Tsang
- Reproductive Biology UnitDepartments of Obstetrics & Gynecology and Cellular & Molecular MedicineUniversity of OttawaOttawaONCanada
- Chronic Disease ProgramOttawa Hospital Research InstituteCritical Care WingThe Ottawa Hospital ‐ General CampusOttawaONCanada
| | - Yoshio Yoshida
- Department of Obstetrics and GynecologyUniversity of FukuiYoshida‐GunJapan
| |
Collapse
|
37
|
The ratio of exogenous Luteinizing hormone to Follicle stimulating hormone administered for controlled ovarian stimulation is associated with oocytes' number and competence. Biosci Rep 2021; 40:221613. [PMID: 31850491 PMCID: PMC6944660 DOI: 10.1042/bsr20190811] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 12/01/2022] Open
Abstract
We performed a retrospective study aiming to study the relationship between the ratio of the exogenous luteinizing hormone to follicle stimulating hormone (LH/FSH) administrated for controlled ovarian stimulation (COS) and the number and competence of the oocytes retrieved for in vitro fertilization (IVF) or intracytoplasmic sperm injection (ICSI). Eight hundred sixty-eight consecutive infertile patients (mean age 34.54 ± 4.01 years, mean anti-Müllerian hormone (AMH) 2.94 ± 2.07 ng/ml) treated with long agonist protocol and a mixed gonadotropin protocol (human menopausal gonadotropin in association with recombinant FSH (recFSH)) who performed IVF/ICSI between January 2013 and February 2016, were included. Patients with severe male factor were excluded. LH/FSH was calculated based on total doses of the two gonadotropins. We found, after adjustment for confounders, a positive relationship between LH/FSH and the retrieved oocytes’ (β = 0.229, P<0.0001) and zygotes’ number (β = 0.144, P<0.0001) in the entire study group and in subgroups according to age (<35 and ≥35 years) and ovarian reserve (AMH < 1.1 and ≥ 1.1 ng/ml). The fertilization rate was positively associated with LH/FSH in patients with LH/FSH in the lowest three quartiles (below 0.77) (β = 0.096, P=0.034). However, patients in the fourth quartile of LH/FSH had a lower fertilization rate as compared with patients in quartiles 1–3 which, after adjustment for covariates, was only marginally negatively related with LH/FSH (β = −0.108, P=0.05). In conclusion, our results suggest that the adequate LH/FSH administrated during COS can improve the oocytes’ and zygotes’ number in IVF/ICSI cycles, but also the fertilization rate when a certain proportion of LH/FSH is not exceeded.
Collapse
|
38
|
Satué K, Fazio E, Muñoz A, Medica P. Endocrine and Electrolyte Balances during Periovulatory Period in Cycling Mares. Animals (Basel) 2021; 11:ani11020520. [PMID: 33671405 PMCID: PMC7922651 DOI: 10.3390/ani11020520] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/07/2021] [Accepted: 02/11/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary This study provides new evidence on the physiological mechanisms involved in the electrolyte balance during periovulatory period in cycling mares. The interrelationships among adrenocorticotropic hormone (ACTH), cortisol (CORT), aldosterone (ALD) and electrolytes (sodium—Na+, potassium—K+ and chloride—Cl−) were evaluated. The simultaneous increase in ACTH, CORT and ALD toward the time of ovulation could suggest the involvement of the adrenocortical pituitary axis in the ovulatory mechanisms, contributing at the same time to the maintenance of electrolyte homeostasis. Abstract In cycling females, the periovulatory period is characterized by stimulation of the hypothalamic pituitary adrenal (HPA) axis. The aim of present study was to analyze the pattern and interrelationships among adrenocorticotropic hormone (ACTH), cortisol (CORT), aldosterone (ALD) and electrolytes (sodium—Na+, potassium—K+ and chloride—Cl−) during periovulatory period in cycling mares. Venous blood samples were obtained daily from a total of 23 Purebred Spanish broodmares, aged 7.09 ± 2.5 years, from day −5 to day +5 of estrous cycle, considering day 0, the day of ovulation. Plasma ACTH was measured by a fluorescent immunoassay kit, serum CORT and ALD by means of a competitive ELISA immunoassay, and plasma Na+, K+ and Cl− were quantified by an analyzer with selective electrodes for the three ions. ACTH showed higher concentrations at day 0 compared to days −5 to −1 and +1 to +3 (p < 0.05). CORT showed higher concentrations at day 0 compared to days −5 to −2 and +1 to +5 (p < 0.05). ALD showed higher concentrations at day 0 compared to days −5 to −2 (p < 0.05) and +2 (p < 0.05). Na+ and Cl− showed higher concentrations at day 0, compared to day −5 and +5. K+ showed lower concentrations at day 0 compared to day +1 (p < 0.05). The significant correlations obtained between ACTH and CORT (r = 0.20) and between ACTH and ALD (r = 0.32) suggest that although ACTH may have an effect both on CORT and ALD, there are other very important determinants that could be considered. Hence, it is possible to presume that the pituitary adrenocortical response and ALD may be involved in the ovulatory mechanisms without a direct relation with electrolyte pattern.
Collapse
Affiliation(s)
- Katiuska Satué
- Department of Animal Medicine and Surgery, Faculty of Veterinary Medicine, CEU-Cardenal Herrera University, 46115 Valencia, Spain
- Correspondence: ; Tel.: +96-136-900066020
| | - Esterina Fazio
- Department of Veterinary Sciences, Veterinary Physiology Unit, Polo Universitario Annunziata, Viale Palatucci 13, 98168 Messina, Italy; (E.F.); (P.M.)
| | - Ana Muñoz
- Department of Animal Medicine and Surgery, School of Veterinary Medicine, University of Córdoba, 14014 Córdoba, Spain;
| | - Pietro Medica
- Department of Veterinary Sciences, Veterinary Physiology Unit, Polo Universitario Annunziata, Viale Palatucci 13, 98168 Messina, Italy; (E.F.); (P.M.)
| |
Collapse
|
39
|
Wang J, Wan C, Shuju Z, Yang Z, Celi P, Ding X, Bai S, Zeng Q, Mao X, Xu S, Zhang K, Li M. Differential analysis of gut microbiota and the effect of dietary Enterococcus faecium supplementation in broiler breeders with high or low laying performance. Poult Sci 2021; 100:1109-1119. [PMID: 33518070 PMCID: PMC7858034 DOI: 10.1016/j.psj.2020.10.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/20/2020] [Accepted: 10/01/2020] [Indexed: 02/07/2023] Open
Abstract
The difference in microbiota was examined for breeders with different egg-laying rates, and the impact of dietary Enterococcus faecium (EF) was also determined in the present study. A total of 256 Arbor Acres broiler breeders (48-wk-old) were used in a 2 × 2 factorial design, which encompassed 2 egg-laying rate levels [average (average egg laying: AP, 80.45 ± 0.91%) and low (lower egg laying: LP, 70.61 ± 1.16%)] and 2 different dietary groups [control (no additive), 6 × 108 cfu/kg EF]. The results showed that the AP breeders presented a lower egg weight, feed conversion ratio, abdominal fat rate, and serum leptin level (P(laying) ≤ 0.05) as well as a higher egg-laying rate (P(laying) < 0.01) than the LP breeders. Dietary supplementation with EF improved the egg weight (P(EF) = 0.03) and had a higher concentration of follicle-stimulating hormone (FSH) in the serum (P(EF) = 0.04). The relative expression of Caspase 9, Bax, AMHR, BMP15, and GATA4 in the ovary of AP breeders was lower, whereas the FSHR and BMPR1B expression was higher than that measured in LP breeders (P(laying) ≤ 0.05). LP increased the abundance of Bacteroidetes (phylum), Firmicutes (phylum), Bacteroidia (class), Clostridia (class), Bacteroidales (order), Clostridiales (order), and Lachnospiraceae (family), whereas the AP promoted the enrichment of Proteobacteria (phylum) and Gammaproteobacteria (class) (P(laying) < 0.05). The genera Bacillus, Rhodanobacter, and Streptomyces were positively correlated with the egg-laying rate and BMPR1B expression (P < 0.05) but negatively correlated with the abdominal fat rate (P < 0.05) and Caspase 9 (P < 0.05). These findings indicate that the low reproductive performance breeders had lower microbiota diversity and higher Firmicutes, which triggers the energy storage that led to higher fat deposition. Besides, increases in the abdominal fat rate, leptin level, and apoptosis (Caspase 9, Bax) and reproduction-related gene (BMP15, AMHR, BMPR1B, and GATA4) expression would possibly be the potential mechanisms under which breeders have different reproductive performance. Dietary EF increased the egg weight and serum FSH level and decreased the Bacteroidetes (phylum) in low reproductive breeders.
Collapse
Affiliation(s)
- Jianping Wang
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Chunpeng Wan
- Research Center of Tea and Tea Culture, College of Agronomy, Jiangxi Agricultural, University, Nanchang, 330045, P.R. China
| | - Zhao Shuju
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zengqiao Yang
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Pietro Celi
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville 3010, Australia
| | - Xuemei Ding
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shiping Bai
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Qiufeng Zeng
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiangbing Mao
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shengyu Xu
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Keying Zhang
- Animal Nutrition Institute, Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Mingxi Li
- Research Center of Tea and Tea Culture, College of Agronomy, Jiangxi Agricultural, University, Nanchang, 330045, P.R. China.
| |
Collapse
|
40
|
Hu H, Fu Y, Zhou B, Li Z, Liu Z, Jia Q. Long non-coding RNA TCONS_00814106 regulates porcine granulosa cell proliferation and apoptosis by sponging miR-1343. Mol Cell Endocrinol 2021; 520:111064. [PMID: 33091558 DOI: 10.1016/j.mce.2020.111064] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 10/14/2020] [Accepted: 10/18/2020] [Indexed: 12/14/2022]
Abstract
Recent evidence shows that long non-coding RNAs (lncRNAs), a class of non-coding RNAs, are involved in the regulation of reproductive processes. In this study, we identified a lncRNA, TCONS_00814106, that was upregulated in high-fecundity sow ovarian tissues and influenced by reproductive hormones. Bioinformatics analyses and luciferase reporter assays showed that TCONS_00814106 is a miR-1343 target. Cell counting kit (CCK)-8 and apoptosis assays showed that TCONS_00814106 promotes proliferation and inhibits apoptosis in porcine granulosa cells (GCs), and that this could be reversed by miR-1343. Also, we observed that transforming growth factor-β receptor type I (TGFBR1) is a functional target of miR-1343 in GCs. TCONS_00814106 serves as a competing endogenous RNA to regulate TGFBR1 expression by sponging miR-1343, thereby exerting regulatory functions in GCs. Overall, these results provide new insights into the biological function of the lncRNA TCONS_00814106.
Collapse
Affiliation(s)
- Huiyan Hu
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, 071000, China
| | - Yanfang Fu
- Hebei Provincial Animal Husbandry Station, Shijiazhuang, 050000, China
| | - Bo Zhou
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, 071000, China
| | - Zhiqiang Li
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, 071000, China
| | - Zhongwu Liu
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, 071000, China
| | - Qing Jia
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, 071000, China; Hebei Technology Innovation Center for Agriculture in Mountainous Areas, Baoding, 071000, China.
| |
Collapse
|
41
|
Liu S, Liu M, Li L, Li H, Qu D, Ren H, Su H, Zhang Y, Li Y. Patients With Deep Ovarian Suppression Following GnRH Agonist Long Protocol May Benefit From a Modified GnRH Antagonist Protocol: A Retrospective Cohort Study. Front Endocrinol (Lausanne) 2021; 12:618580. [PMID: 34326810 PMCID: PMC8314175 DOI: 10.3389/fendo.2021.618580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 06/29/2021] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE To verify if patients with deep ovarian suppression following gonadotropin releasing hormone (GnRH) agonist long protocol may benefit from a modified GnRH antagonist protocol based on luteinizing hormone (LH) levels. DESIGN Retrospective cohort study. SETTING University-based hospital. PATIENTS 110 patients exhibited ultra-low LH levels during ovarian stimulation using GnRH agonist long protocol. INTERVENTIONS As all the embryos in the first cycle were exhausted without being pregnant, these patients proposed to undergo a second cycle of ovarian stimulation. 74 of them were treated with a modified GnRH antagonist protocol based on LH levels. Other 36 patients were still stimulated following GnRH agonist long protocol. MAIN OUTCOME MEASURE The primary outcome was live birth rate (LBR). The second outcomes were biochemical pregnancy rate, clinical pregnancy rate (CPR), ongoing pregnancy rate (OPR) and cancellation rate. RESULTS Reproductive outcomes were much better in the modified GnRH antagonist protocol. The OPR and LBR were much higher in the GnRH antagonist protocol group than in the GnRH agonist long protocol group [odds ratio (OR) 3.82, 95% confidence interval (CI) 1.47, 10.61, P=0.018; OR 4.33, 95% CI 1.38, 13.60, P=0.008; respectively]. Meanwhile, the cancellation rate was much lower in the GnRH antagonist protocol group (OR 0.13, 95% CI 0.02, 0.72; P=0.014). Mean LH level during stimulation did not have a predictive value on live birth. However, it was independently associated with the occurrence of ongoing pregnancy (OR 2.70, 95% CI 1.25, 5.85; P=0.01). The results of sensitivity analyses were consistent with the data mentioned above. The patients got completely different and excellent clinical outcomes in their second cycles stimulated with the modified GnRH antagonist protocol. CONCLUSION Patients with deep ovarian suppression following GnRH agonist long protocol may benefit from a modified GnRH antagonist protocol based on LH levels.
Collapse
|
42
|
Ito M, Yoshino O, Ono Y, Yamaki‐Ushijima A, Tanaka T, Shima T, Orisaka M, Iwase A, Nakashima A, Saito S. Bone morphogenetic protein‐2 enhances gonadotropin‐independent follicular development via sphingosine kinase 1. Am J Reprod Immunol 2020; 85:e13374. [DOI: 10.1111/aji.13374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Affiliation(s)
- Masami Ito
- Department of Obstetrics and Gynecology University of Toyama Japan
| | - Osamu Yoshino
- Department of Obstetrics and Gynecology School of Medicine Kitasato University Kanagawa Japan
| | - Yosuke Ono
- Department of Obstetrics and Gynecology Teinekeijinkai Hospital Hokkaido Japan
| | | | - Tomoko Tanaka
- Department of Obstetrics and Gynecology University of Toyama Japan
| | - Tomoko Shima
- Department of Obstetrics and Gynecology University of Toyama Japan
| | - Makoto Orisaka
- Department of Obstetrics and Gynecology University of Fukui Fukui Japan
| | - Akira Iwase
- Department of Obstetrics and Gynecology University of Gunma Maebashi Japan
| | | | | |
Collapse
|
43
|
Arvis P, Massin N, Lehert P. Effect of recombinant LH supplementation on cumulative live birth rate compared with FSH alone in poor ovarian responders: a large, real-world study. Reprod Biomed Online 2020; 42:546-554. [PMID: 33431337 DOI: 10.1016/j.rbmo.2020.08.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 12/21/2022]
Abstract
RESEARCH QUESTION The benefit of LH supplementation (LHS) over sole use of FSH during controlled ovarian stimulation (COS) remains controversial. Meta-analyses have provided some evidence that the benefit of LHS is limited to women with poor ovarian response (POR). This study aimed to assess the effectiveness of LHS on cumulative live birth rate (CLBR) in POR using a large controlled study in a real-world context. DESIGN This retrospective multicentre controlled study used data from registries at 12 French ART centres. All instances of POR undergoing ovarian stimulation and treated with follitrophin-alfa (FSH-α) with or without lutrophin-α were selected following an intention-to-treat principle. POR was defined according to the ESHRE Bologna criteria, and classified into three categories (Mild, Moderate and Severe) according to the Poor Responder Outcome Prediction (PROsPeR) score. The primary end-point was the CLBR associated with fresh and frozen embryos originating from the same ovarian stimulation. RESULTS A total of 9787 instances of ovarian stimulation (5218 LHS, 4569 FSH-α only) were analysed, 33.0%, 52.4% and 14.6% being allocated to the Mild, Moderate and Severe PROsPeR categories, respectively. Using a mixed logistic model and adjusting for matched subclasses and baseline POR severity, it was found that the benefit of LHS compared with use of FSH alone differed between baseline severity categories (interaction test, P = 0.007): a significant benefit of LHS for CLBR was found for patients in the Moderate (14.3% versus 11.3%, odds ratio [OR] = 1.37, 95% confidence interval [CI] 1.07-1.75, risk ratio [RR] = 1.29, P = 0.013) and Severe (9.8% versus 4.4%, OR = 2.40, 95% CI- 1.48-3.89, RR = 1.89, P < 0.001) categories, but not for the Mild category (18.8% versus 19.6%, OR = 0.95, 95% CI 0.78-1.15, RR = 0.95, P = 0.60). CONCLUSION LHS has a significant effect on increasing CLBR in moderately and severely poor ovarian responders.
Collapse
Affiliation(s)
| | | | - Philippe Lehert
- Faculty of Economics, UCL Mons, Louvain, Belgium, Faculty of Medicine, University of Melbourne, Melbourne Victoria, Australia
| |
Collapse
|
44
|
Berker B, Şükür YE, Özdemir EÜ, Özmen B, Sönmezer M, Atabekoğlu CS, Aytaç R. Human Menopausal Gonadotropin Commenced on Early Follicular Period Increases Live Birth Rates in POSEIDON Group 3 and 4 Poor Responders. Reprod Sci 2020; 28:488-494. [PMID: 32833190 DOI: 10.1007/s43032-020-00300-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022]
Abstract
Human menopausal gonadotropin (hMG) has LH activity, and it may have beneficial effects in terms of oocyte quality and endometrial receptivity similar to recombinant LH supplementation. The aim of this study was to assess the effects of hMG, and its commencement time on the outcome of assisted reproductive technology (ART) cycles of POSEIDON group 3 and 4 poor responders. Data of 558 POSEIDON group 3 and 4 poor responders who underwent ART treatment following a GnRH antagonist cycle at a university-based infertility clinic between January 2014 and December 2019 were reviewed. hMG was commenced at the early follicular phase or mid-follicular phase in the study groups. The control group did not receive hMG stimulation. Live birth rate (LBR) was the main outcome measure. The mean duration of stimulation was significantly shorter in early follicular hMG group than in mid-follicular hMG group (11.9 ± 3.6 days vs. 12.8 ± 4 days, respectively; P = 0.027). The mean numbers of oocytes retrieved and MII oocytes were comparable between the groups. The LBRs per embryo transfer in early follicular hMG, mid-follicular hMG, and control groups were 21.9%, 11.7%, and 11.6%, respectively (P = 0.035). In conclusion, there is a significant association between the commencement time of hMG and live birth chance in ART cycles of POSEIDON group 3 and 4 poor responders. Early initiation of hMG together with rFSH seems to be beneficial in this specific population.
Collapse
Affiliation(s)
- Bülent Berker
- Department of Obstetrics and Gynecology, Ankara University School of Medicine, Cebeci Hospital, 06100, Dikimevi/Ankara, Turkey
| | - Yavuz Emre Şükür
- Department of Obstetrics and Gynecology, Ankara University School of Medicine, Cebeci Hospital, 06100, Dikimevi/Ankara, Turkey.
| | - Eda Üreyen Özdemir
- Department of Obstetrics and Gynecology, Halil Şıvgın Çubuk State Hospital, Ankara, Turkey
| | - Batuhan Özmen
- Department of Obstetrics and Gynecology, Ankara University School of Medicine, Cebeci Hospital, 06100, Dikimevi/Ankara, Turkey
| | - Murat Sönmezer
- Department of Obstetrics and Gynecology, Ankara University School of Medicine, Cebeci Hospital, 06100, Dikimevi/Ankara, Turkey
| | - Cem Somer Atabekoğlu
- Department of Obstetrics and Gynecology, Ankara University School of Medicine, Cebeci Hospital, 06100, Dikimevi/Ankara, Turkey
| | - Ruşen Aytaç
- Department of Obstetrics and Gynecology, Ankara University School of Medicine, Cebeci Hospital, 06100, Dikimevi/Ankara, Turkey
| |
Collapse
|
45
|
Lv X, He C, Huang C, Hua G, Chen X, Timm BK, Maclin VM, Haggerty AA, Aust SK, Golden DM, Dave BJ, Tseng YA, Chen L, Wang H, Chen P, Klinkebiel DL, Karpf AR, Dong J, Drapkin RI, Rueda BR, Davis JS, Wang C. Reprogramming of Ovarian Granulosa Cells by YAP1 Leads to Development of High-Grade Cancer with Mesenchymal Lineage and Serous Features. Sci Bull (Beijing) 2020; 65:1281-1296. [PMID: 34888112 PMCID: PMC8654108 DOI: 10.1016/j.scib.2020.03.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Understanding the cell-of-origin of ovarian high grade serous cancer (HGSC) is the prerequisite for efficient prevention and early diagnosis of this most lethal gynecological cancer. Recently, a mesenchymal type of ovarian HGSC with the poorest prognosis among ovarian cancers was identified by both TCGA and AOCS studies. The cell-of-origin of this subtype of ovarian cancer is unknown. While pursuing studies to understand the role of the Hippo pathway in ovarian granulosa cell physiology and pathology, we unexpectedly found that the Yes-associated protein 1 (YAP1), the major effector of the Hippo signaling pathway, induced dedifferentiation and reprogramming of the ovarian granulosa cells, a unique type of ovarian follicular cells with mesenchymal lineage and high plasticity, leading to the development of high grade ovarian cancer with serous features. Our research results unveil a potential cell-of-origin for a subtype of HGSC with mesenchymal features.
Collapse
Affiliation(s)
- Xiangmin Lv
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chunbo He
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan 47000, China
| | - Cong Huang
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Guohua Hua
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan 47000, China
| | - Xingcheng Chen
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Barbara K. Timm
- Heartland Center for Reproductive Medicine, Omaha, NE 68198, USA
| | | | - Abigail A Haggerty
- Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Shelly K Aust
- Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Denae M Golden
- Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bhavana J Dave
- Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yun-An Tseng
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Li Chen
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongbo Wang
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Peichao Chen
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - David L Klinkebiel
- Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Adam R Karpf
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jixin Dong
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ronny I Drapkin
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bo R Rueda
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - John S Davis
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Cheng Wang
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
46
|
The CC-chemokine receptor 2 is involved in the control of ovarian folliculogenesis and fertility lifespan in mice. J Reprod Immunol 2020; 141:103174. [PMID: 32615332 DOI: 10.1016/j.jri.2020.103174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/05/2020] [Accepted: 06/24/2020] [Indexed: 11/24/2022]
Abstract
The chemokine receptor 2 (CCR2) was first described as a chemotactic factor involved in immune responses, but it also plays an essential function in several biological processes. The chemokine (C-C motif) ligand 2 (CCL2) binds to CCR2 triggering G protein-coupled receptor (GPCR) signaling in leukocytes, including activation of PI3K/Akt/mTOR, a key pathway that is also related to follicular activation and survival. However, the potential role of CCR2 in ovarian follicular physiology remain unexplored. Thus, we investigated the role of CCR2 on follicular growth during adult life and aging. Ovaries and oocytes were collected from wild type (WT) mice at 1.5 months old (mo), and CCR2 expression was observed predominantly in oocytes included in growing follicles, as well as after ovulation. Follicle populations were assessed in WT and CCR2-/- mice at 1.5 mo, and CCR2-/- mice had more primordial and less primary and secondary follicles, while there were no differences in antral follicle numbers. Pro-apoptotic genes Bax and Casp3 were downregulated, while anti-apoptotic Bcl2 was upregulated in CCR2-/- mice. To further characterize the role of CCR2 in ovarian aging, follicle populations were assessed in WT and CCR2-/- mice at 1.5, 2.5, 6, 10, and 12 mo. A larger ovarian follicular reserve at 1.5-6 mo was observed in CCR2-/- mice. Finally, CCR2-/- aged mice (6-12 mo) ovulated more oocytes than WT mice. Altogether, these data suggest that CCR2 plays an important role in the regulation of murine folliculogenesis, potentially affecting the reproductive lifespan.
Collapse
|
47
|
Gao XY, Liu Y, Lv Y, Huang T, Lu G, Liu HB, Zhao SG. Role of Androgen Receptor for Reconsidering the "True" Polycystic Ovarian Morphology in PCOS. Sci Rep 2020; 10:8993. [PMID: 32488141 PMCID: PMC7265442 DOI: 10.1038/s41598-020-65890-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/12/2020] [Indexed: 11/21/2022] Open
Abstract
Purpose: Polycystic ovarian morphology (PCOM) is one of the key features of polycystic ovary syndrome (PCOS). The diagnosis of PCOM according to the Rotterdam criteria (≥12 antral follicles per ovary) is debated because of the high prevalence of PCOM in the general population. Androgen receptor (AR) is associated with the PCOS phenotype and might as well play a role during folliculogenesis. This study is aimed to investigate the expression of the AR in PCOS granulosa cells (GCs) and its relationship with the PCOM phenotype. Methods: 106 PCOS cases and 63 controls were included from the Center for Reproductive Medicine, Shandong University. The diagnosis of PCOS was following the Rotterdam criteria (2003). Total RNA was extracted from GCs retrieved from ovarian stimulation. The expression of AR was amplified by means of quantitative real-time polymerase chain reaction. Results: The AR expression was significantly decreased in PCOS cases, especially in the tPCOM subgroup (≥20 antral follicles per ovary). Correlation analyses showed that AR expression was significantly correlated with serum FSH levels in controls and non-tPCOM. In the tPCOM subgroup, the AR expression was significantly correlated with serum LH levels. Interestingly, the significance of these correlations gradually disappeared as the threshold of antral follicles increased above 24 for PCOM. Conclusions:AR was differently expressed in PCOS and especially in the tPCOM subtype. The correlation of AR expression with serum FSH and LH might be associated with the number of follicles in PCOM.
Collapse
Affiliation(s)
- Xue-Ying Gao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
| | - Yue Liu
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
| | - Yue Lv
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.,School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,CUHK-SDU Joint Laboratory on Reproductive Genetics, Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Tao Huang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
| | - Gang Lu
- CUHK-SDU Joint Laboratory on Reproductive Genetics, Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hong-Bin Liu
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
| | - Shi-Gang Zhao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China. .,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China. .,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
48
|
Shi B, Lu H, Zhang L, Zhang W. Nr5a1b promotes and Nr5a2 inhibits transcription of lhb in the orange-spotted grouper, Epinephelus coioides†. Biol Reprod 2019; 101:800-812. [PMID: 31317174 DOI: 10.1093/biolre/ioz121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/25/2019] [Accepted: 07/10/2019] [Indexed: 01/02/2023] Open
Abstract
Nr5a1 (Sf-1) up-regulates lhb expression across vertebrates; however, its regulatory roles on fshb remain to be defined. Moreover, the involvement of Nr5a2 in the regulation of gonadotropin expression is not clear either. In the present study, the involvement of Nr5a1b (a homologue of Nr5a1) and Nr5a2 in the regulation of lhb and fshb expression in the orange-spotted grouper was examined. Dual fluorescent immunohistochemistry using homologous antisera showed that in the pituitary of orange-spotted groupers, Lh cells contain both immunoreactive Nr5a1b and Nr5a2 signals, whereas Fsh cells contain neither of them. In LβT2 cells, Nr5a1b up-regulated basal activities of lhb and fshb promoters possibly via Nr5a sites, and synergistically (on lhb promoter) or additively (on fshb promoter) with forskolin. Surprisingly, Nr5a2 inhibited basal activities of lhb promoter possibly via Nr5a sites and attenuated the stimulatory effects of both forskolin and Nr5a1b. In contrast, Nr5a2 had no effects on fshb promoter. Chromatin immunoprecipitation analysis showed that both Nr5a1b and Nr5a2 bound to lhb promoter, but not fshb promoter in the pituitary of the orange-spotted grouper. The abundance of Nr5a1b bound to lhb promoter was significantly higher at the vitellogenic stage than the pre-vitellogenic stage, whereas that of Nr5a2 exhibited an opposite trend. Taken together, data of the present study demonstrated antagonistic effects of Nr5a1b and Nr5a2 on lhb transcription in the orange-spotted grouper and revealed novel regulatory mechanisms of differential expression of lhb and fshb genes through Nr5a homologues in vertebrates.
Collapse
Affiliation(s)
- Boyang Shi
- Institute of Aquatic Economic Animals, Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Huijie Lu
- Institute of Aquatic Economic Animals, Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Lihong Zhang
- Institute of Aquatic Economic Animals, Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Weimin Zhang
- Institute of Aquatic Economic Animals, Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
49
|
Kan O, Simsir C, Atabekoglu CS, Sonmezer M. The impact of adding hp-hMG in r-FSH started GnRH antagonist cycles on ART outcome. Gynecol Endocrinol 2019; 35:869-872. [PMID: 30973022 DOI: 10.1080/09513590.2019.1600667] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
While luteinizing hormone (LH) activity is believed to play a role in follicle maturation, human chorionic gonadotropin (hCG) might play an important role in implantation process. We aimed to investigate whether addition of human menopausal gonadotropin (hMG) in recombinant-follicle-stimulating hormone (r-FSH) started GnRH antagonist controlled ovarian hyperstimulation (COH) cycles might enhance implantation rate and improve in vitro fertilization (IVF) success. A total of 246 patients undergoing GnRH antagonist IVF cycles were analyzed. One hundred and twenty-three cycles (%50) were treated with only r-FSH and 123 cycles were treated with r-FSH plus hp-hMG combination. Total gonadotropin doses, total number of oocytes retrieved, metaphase 2 (MII) oocytes, top quality embryos, fertilization and implantation rates, clinical pregnancy rates (CPRs) and ovarian hyperstimulation syndrome (OHSS) rates were compared between the groups. Both groups were comparable in terms of demographic details and baseline characteristics. Peak estradiol and progesterone levels in hCG trigger day, number of retrieved oocytes and top quality embryo counts, fertilization rates were similar between the groups. In r-FSH + hp-hMG group, significantly higher implantation rates (35.3% vs 24.3%, p=.017), CPRs (51.2% vs 35.8%, p=.015) and lower OHSS rates (1.6% vs 7.4%, p = .03) were observed respectively compared to r-FSH only treated patients. In conclusion, addition of hp-hMG on the day of antagonist initiation might increase CPRs. A better endometrial receptivity associated with higher implantation rates might be achieved due to hCG component in hp-hMG.
Collapse
Affiliation(s)
- Ozgur Kan
- Department of Obstetrics and Gynecology, Faculty of Medicine, Hitit University, Corum, Turkey
| | - Coskun Simsir
- Department of Obstetrics and Gynecology, Liv Hospital, Ankara, Turkey
| | - Cem Somer Atabekoglu
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Murat Sonmezer
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
50
|
Hosein Rashidi B, Kabodmehri R, Shariat M, Shahrokh Tehraninejad E, Abdollahi A, Bagheri M, Hagholahi F. Luteinizing hormone changes in gonadotropin-releasing hormone antagonist protocol in in vitro fertilization cycles: A cross-sectional study. Int J Reprod Biomed 2019; 17. [PMID: 31435601 PMCID: PMC6661140 DOI: 10.18502/ijrm.v17i3.4520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 10/29/2018] [Accepted: 12/12/2018] [Indexed: 11/24/2022] Open
Abstract
Background Because of the unexpected and often dramatic inhibition of luteinizing hormone (LH) secretion related with the usage of gonadotropin-releasing hormone (GnRH)-antagonist, there has been a probable need for exogenous LH supplementation. There is a basic and clinical evidences that show late development of follicle needs an LH but there is a threshold for LH requirements during folliculogenesis. Objective The purpose of this study was to evaluate the changes in serum LH and the identification of patients who benefit from the addition of LH. Materials and Methods Seventy volunteers for antagonist protocol in IVF cycle were enrolled in this prospective cross-sectional study. The study was carried out in Reproductive Health Research Center, University of Medical Sciences between July 2016 and February 2016. Serum LH level was estimated 24 h before and after the first (GnRH) antagonist injection. The primary outcome was the serum level of LH and its change in the three groups and the secondary outcome was Egg and Embryo quality. Results LH changes above or below 50% had no effect on the number of follicle, the number of oocyte, Germinal vesicle oocyte, metaphase 1 oocyte, metaphase 2 oocyte, endometrial thickness, and chemical and clinical pregnancy. Conclusion We evaluated the changes of serum LH in the patients who were entered in the antagonist protocol. Our study showed no significant difference in LH levels 24 h before and after the injection of the antagonist between the three groups, and LH changes did not affect the outcome of pregnancy.
Collapse
Affiliation(s)
- Batool Hosein Rashidi
- Health Reproductive Research Center, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Roya Kabodmehri
- Health Reproductive Research Center, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mamak Shariat
- Health Reproductive Research Center, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ensieh Shahrokh Tehraninejad
- Health Reproductive Research Center, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Abdollahi
- Health Reproductive Research Center, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Bagheri
- Health Reproductive Research Center, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fedieh Hagholahi
- Health Reproductive Research Center, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|