1
|
He J, Wang Z, Yang L, Jiang Y, Yan G, Pan Y, Gao F, Yuan J, Gao Y. Unveiling the role of FOXL2 in female differentiation and disease: a comprehensive review†. Biol Reprod 2025; 112:600-613. [PMID: 39976382 DOI: 10.1093/biolre/ioaf013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/30/2024] [Accepted: 02/20/2025] [Indexed: 02/21/2025] Open
Abstract
Ovarian differentiation relies on the accurate and orderly expression of numerous related genes. Forkhead box protein L2 (FOXL2) is one of the earliest ovarian differentiation markers and transcription factors. In sex determination, FOXL2 maintains the differentiation of the female pathway by inhibiting male differentiation genes, including SOX9 and SF1. In addition, FOXL2 promotes the synthesis of follicle-stimulating hormone and anti-Müllerian hormone to support follicle development. Mutations in FOXL2 are associated with numerous female reproductive diseases. A comprehensive and in-depth study of FOXL2 provides novel strategies for the diagnosis and treatment of such diseases. This review discusses the mechanism of FOXL2 in female sex differentiation and maintenance, hormone synthesis, and disease occurrence and reveals the role of FOXL2 as a central factor in female sex development and fertility maintenance. This review will serve as a reference for identifying novel targets of other regulatory factors interacting with FOXL2 in female sex determination and follicle development and for the diagnosis and treatment of female reproductive diseases.
Collapse
Affiliation(s)
- Jia He
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Zican Wang
- College of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Lici Yang
- College of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Yongjian Jiang
- College of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Ge Yan
- College of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Yongwei Pan
- College of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Fei Gao
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, Shandong, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jinxiang Yuan
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, Shandong, China
| | - Yang Gao
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
2
|
Heidarzadehpilehrood R, Pirhoushiaran M, Osman MB, Ling KH, Hamid HA. Identifying Genetic Profiles in Peripheral Blood Mononuclear Cells in Women with Polycystic Ovary Syndrome: An Observational Case-Control Study. Arch Med Res 2025; 56:103129. [PMID: 39647252 DOI: 10.1016/j.arcmed.2024.103129] [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: 05/30/2024] [Revised: 09/15/2024] [Accepted: 11/07/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is an endocrine and reproductive condition affecting women of reproductive age, although its expression profiles and molecular pathways are not fully understood. AIMS To identify the transcriptome expression profiles of peripheral blood mononuclear cells (PBMCs) in women with PCOS and controls. To investigate noninvasive diagnostic biomarkers and potential treatment targets to improve women's fertility. METHODS RNA sequencing (RNA-Seq) was conducted on PBMC samples from six patients with PCOS and six healthy controls. qRT-PCR validation was carried out in 68 subjects. Multivariate logistic regression was performed to assess the combined impact of biomarkers. RESULTS A total of 186 differentially expressed genes (DEG) were found between patients and controls (log2FC >1, p <0.05). Enrichment analysis revealed cytokine-mediated signaling pathways, cytokine activity, and cytokine-cytokine receptor interaction. RNA sequencing showed consistency with qRT-PCR. Women with PCOS had significantly higher levels of AQP9 (p <0.001), PROK2 (p = 0.001), and S100A12 (p <0.001) expression compared to controls. AQP9 (AUC = 0.77), PROK2 (AUC = 0.71), and S100A12 (AUC = 0.82) adequately discriminated women with PCOS from healthy controls. In addition, multiple logistic regression on biomarkers resulted in a significant diagnostic power with an AUC = 0.89, 95% CI: 0.81-0.97, p <0.0001. Further associations were analyzed between relative gene expression and clinical, anthropometric, hormonal, and ultrasonographic data. CONCLUSIONS Dysregulated RNA expression in PBMCs may contribute to an increased risk of PCOS and serve as a potential diagnostic biomarker. The involvement of inflammatory and cytokine-related pathways supports the notion that PCOS is a chronic inflammatory condition.
Collapse
Affiliation(s)
- Roozbeh Heidarzadehpilehrood
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Maryam Pirhoushiaran
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran
| | - Malina Binti Osman
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - King-Hwa Ling
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia; Malaysian Research Institution on Ageing, Universiti Putra Malaysia, 43400 Serdang, Selangor Darul Ehsan, Malaysia
| | - Habibah Abdul Hamid
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| |
Collapse
|
3
|
Zhao H, Yu F, Wu W. The Mechanism by Which Estrogen Level Affects Knee Osteoarthritis Pain in Perimenopause and Non-Pharmacological Measures. Int J Mol Sci 2025; 26:2391. [PMID: 40141035 PMCID: PMC11942494 DOI: 10.3390/ijms26062391] [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: 02/04/2025] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
Perimenopausal women have fluctuating estrogen levels, which often trigger a range of symptoms of perimenopausal syndromes as estrogen levels decrease. Changes in perimenopausal estrogen levels are closely related to pain in knee osteoarthritis (KOA), which has long been a research area of great interest in perimenopausal women. In recent years, it has been found that perimenopausal estrogen levels have an important role in KOA pain, namely, that estrogen can affect KOA pain through the regulation of inflammatory responses, inhibition of cellular senescence and apoptosis, and modulation of neurotransmitters, which may provide new ideas for KOA treatment. This study aims to describe the mechanism of estrogen level on knee osteoarthritis pain in perimenopause and related non-pharmacological measures, such as physical therapy, physical factor therapy, traditional Chinese medicine, and diet, which can provide a reference for the study and treatment of pain in perimenopausal women with KOA.
Collapse
Affiliation(s)
- Huiying Zhao
- School of Exercise and Health, Shanghai University of Sports, Shanghai 200438, China; (H.Z.); (F.Y.)
| | - Fan Yu
- School of Exercise and Health, Shanghai University of Sports, Shanghai 200438, China; (H.Z.); (F.Y.)
| | - Wei Wu
- School of Athletic Performance, Shanghai University of Sports, Shanghai 200438, China
| |
Collapse
|
4
|
Kawada T, Aoyama M, Matsubara S, Osugi T, Sakai T, Kirimoto S, Nakaoka S, Sugiura Y, Yasuda K, Satake H. Ovarian tachykinin signaling system induces the growth of secondary follicles during the gonadotropin-independent process. J Biol Chem 2025; 301:108375. [PMID: 40043951 PMCID: PMC11999619 DOI: 10.1016/j.jbc.2025.108375] [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: 04/18/2024] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Mammalian follicle growth development is mainly regulated by the hypothalamus-pituitary-gonadal axis after puberty. Although pituitary hormones, gonadotropins, are involved in hypothalamus-pituitary-gonadal axis signaling, they are not responsible for the growth of early stage follicles, namely, primordial follicles, primary follicles, and secondary follicles, in both sexually immature and mature individuals. Unlike those of gonadotropin-dependent follicle growth, the specific regulatory factors of gonadotropin-independent follicle growth have yet to be identified. Here, we identified tachykinins (TKs) as inducers of gonadotropin-independent secondary follicle growth. TKs play various roles as neuropeptides or hormones in a wide variety of biological events both in the central nervous system and in peripheral tissues, but a direct effect of TKs on ovarian follicles has yet to investigated. Follicle development was suppressed in sexually immature 3-week-old KO mice of Tac1 gene encoding TKs (substance P and neurokinin A), which is independent of gonadotropins. TKs and their receptors are specifically localized to granulosa cells in mouse secondary follicles. Furthermore, TKs upregulate the prostaglandin (PG) synthase cyclooxygenase 2 via the Janus kinase 1-signal transducers and activators of transcription protein 3 signaling cascade. We also demonstrated that PGE2 and PGF2α are major PGs in the immature ovary, and the secondary follicle growth was enhanced by interaction between PGE2-PGF2α and their receptors, PGE2 receptor localized in the oocyte membrane and PGF2α receptor localized in the oocyte membrane, granulosa cells, and theca cells. Consequently, this study paves the way for exploring gonadotropin-independent early stage follicle growth systems and relevant dysfunctions, including pediatric endocrinological diseases.
Collapse
Affiliation(s)
- Tsuyoshi Kawada
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Masato Aoyama
- Department of Chemistry, Biology, and Environmental Science, Faculty of Science, Nara Women's University, Nara, Japan
| | - Shin Matsubara
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Tomohiro Osugi
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Tsubasa Sakai
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Shinji Kirimoto
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Satsuki Nakaoka
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan
| | - Yuki Sugiura
- Graduate School of Medicine and Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Keiko Yasuda
- Department of Chemistry, Biology, and Environmental Science, Faculty of Science, Nara Women's University, Nara, Japan
| | - Honoo Satake
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, Kyoto, Japan.
| |
Collapse
|
5
|
Hong P, Lu Y, Li H, Liu Z, Ou J, Li T, Shu Y. Predicting the risk of a high proportion of three/multiple pronuclei (3PN/MPN) zygotes in individual IVF cycles using comparative machine learning algorithms. Eur J Obstet Gynecol Reprod Biol 2025; 306:139-146. [PMID: 39826276 DOI: 10.1016/j.ejogrb.2025.01.023] [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: 11/20/2024] [Revised: 01/06/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND The majority of machine learning applications in assisted reproduction have been focused on predicting the likelihood of pregnancy. In the present study, we aim to investigate which machine learning models are most effective in predicting the occurrence of a high proportion (>30 %) of 3PN/MPN zygotes in individual IVF cycles. METHODS Eight machine learning algorithms were trained and compared, including the AdaBoost and Gaussian NB. Data from IVF cycles carried out from September 2015 to September 2019 were used as a training set. Cycle data from October 2019 to June 2020 were used as a validation set to verify the training model. Cycles with a 3PN/MPN zygote proportion higher than 30 % were classified as high 3PN/MPN zygote proportion cycles. RESULTS The AdaBoost algorithm was the best model for model construction and external validation. In both the training and validation sets, age, basal FSH, FSH and E2 level on the day of Gonadotrophin (GN) stimulation, and FSH and LH levels on the day of HCG were statistically higher in patients with 3PN/MPN > 30 % than in patients with 3PN/MPN ≤ 30 %; AFC, AMH, E2 level on HCG day and total number of oocytes were lower in patients with 3PN/MPN > 30 % than in patients with 3PN/MPN ≤ 30 %. The top five predictors were the number of oocytes retrieved, age, male factor infertility, AFC, and total days of GN stimulation. CONCLUSION By applying a suitable machine learning algorithm, we can potentially predict the risk of a high proportion of 3PN/MPN zygotes in individual IVF cycles before insemination and avoid polyspermy fertilization by ICSI fertilization method.
Collapse
Affiliation(s)
- Pingping Hong
- Reproductive Medicine Center, The Third Affiliated Hospital of Sun Yat-sen University, 6# East Longkou Road, Gangding, Tianhe District, Guangzhou 510630 China.
| | - Yaxin Lu
- Centre for Big Data and Artificial Intelligence, The third Affiliated Hospital of Sun Yat-sen University, No.600, Tianhe Road, Tianhe District, Guangzhou, Guangdong, China.
| | - Haiyang Li
- Reproductive Medicine Center, The Third Affiliated Hospital of Sun Yat-sen University, 6# East Longkou Road, Gangding, Tianhe District, Guangzhou 510630 China.
| | - Zifeng Liu
- Centre for Big Data and Artificial Intelligence, The third Affiliated Hospital of Sun Yat-sen University, No.600, Tianhe Road, Tianhe District, Guangzhou, Guangdong, China.
| | - Jianpin Ou
- Reproductive Medicine Center, The Third Affiliated Hospital of Sun Yat-sen University, 6# East Longkou Road, Gangding, Tianhe District, Guangzhou 510630 China.
| | - Tao Li
- Reproductive Medicine Center, The Third Affiliated Hospital of Sun Yat-sen University, 6# East Longkou Road, Gangding, Tianhe District, Guangzhou 510630 China.
| | - Yimin Shu
- Center for Advanced Reproductive Medicine, Department of Obstetrics & Gynecology, University of Kansas Medical Center, Overland Park, KS 66211, USA.
| |
Collapse
|
6
|
Guimarães-Ervilha LO, Assis MQ, Bento IPDS, Lopes IDS, Iasbik-Lima T, Carvalho RPR, Machado-Neves M. Exploring the endocrine-disrupting potential of atrazine for male reproduction: A systematic review and meta-analysis. Reprod Biol 2025; 25:100989. [PMID: 39708576 DOI: 10.1016/j.repbio.2024.100989] [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: 09/16/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024]
Abstract
Atrazine is an herbicide widely used on plantations worldwide. Experimental studies suggest that the herbicide impairs male reproductive function in mammals. This systematic review and meta-analysis aimed to evaluate the impact of atrazine exposure on the levels of hormones from the hypothalamic-pituitary-testicular axis using murine as the animal model. After an extensive literature search, we selected 25 articles for the systematic review. Bias analysis and methodological quality assessments were examined using the SYRCLE Risk of Bias tool. Moreover, 20 out of the 25 studies were eligible for performing a meta-analysis to evaluate the intensity of atrazine damage on the levels of intratesticular testosterone and serum follicle-stimulating hormone (FSH), luteinizing hormone (LH), testosterone, estradiol, and progesterone. The meta-analysis revealed that atrazine exposure decreased serum FSH, LH, and testosterone levels, besides increased serum estradiol and progesterone levels. Atrazine also caused a reduction in intratesticular testosterone levels. Exposure to atrazine in high concentrations (≥ 100 mg Kg-1) was the main cause of endocrine disruption, regardless of the exposure time. None of the studies have tested doses relevant to human health risk. Oxidative stress and inflammation are involved in atrazine toxicity, impairing the gonadotropin release by the pituitary, disturbing steroidogenesis, and affecting the male hormone regulatory system. We may conclude that hormone disturbances lead to a failure in testicular steroidogenesis, with possible implications for male reproductive function. The registration number on the Prospero platform is CRD42024495626.
Collapse
Affiliation(s)
| | | | | | | | - Thainá Iasbik-Lima
- Universidade Federal de Viçosa, Departamento de Biologia Geral, Vicosa, Brasil
| | | | | |
Collapse
|
7
|
Everaere H, Simon V, Bachelot A, Leroy M, Decanter C, Dewailly D, Catteau-Jonard S, Robin G. Pulsatile gonadotropin-releasing hormone therapy: comparison of efficacy between functional hypothalamic amenorrhea and congenital hypogonadotropic hypogonadism. Fertil Steril 2025; 123:270-279. [PMID: 39233038 DOI: 10.1016/j.fertnstert.2024.08.354] [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: 02/29/2024] [Revised: 08/10/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
OBJECTIVE To compare the ongoing pregnancy rate per initiated cycle between patients with functional hypothalamic amenorrhea (FHA) and patients with congenital hypogonadotropic hypogonadism (CHH) treated with pulsatile gonadotropin-releasing hormone (GnRH) administration. DESIGN Retrospective monocentric cohort study conducted at the University Hospital of Lille from 2004 to 2022. SUBJECTS A total of 141 patients diagnosed with central suprapituitary amenorrhea during infertility evaluation and subsequently treated with pulsatile GnRH therapy. 111 and 30 patients were diagnosed with FHA or CHH, respectively. EXPOSURE Pulsatile GnRH administration. MAIN OUTCOME MEASURE(S) Ongoing pregnancy rate per initiated cycle. RESULT(S) Ongoing pregnancy rates per initiated cycle were comparable between groups: 21.5% in the FHA group vs. 22% in the CHH group. Comparison of baseline characteristics showed a more pronounced follicle-stimulating hormone (FSH) deficiency in patients with CHH than in those with FHA: 2.55 (0.6-4.92) vs. 4.80 (3.90-5.70) UI/L. Within the CHH group, basal FSH level was positively associated with the occurrence of ongoing pregnancies (odds ratio, 1.57; 95% confidence interval, 1.11-2.22). In the CHH group, the duration of treatment was higher than in the FHA group: 23.59 (± 8.02) vs. 18.16 (± 7.66) days. CONCLUSION(S) The baseline FSH level is lower in patients with CHH than in patients with FHA. The lower the FSH, the lower the chance of pregnancy in patients with CHH. These patients also require more days of GnRH administration. However, the rate of ongoing pregnancies is comparable between the two groups.
Collapse
Affiliation(s)
- Hortense Everaere
- Department of Endocrine Gynecology, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France; Department of Assisted Reproductive Technologies and Fertility Preservation, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France.
| | - Virginie Simon
- Department of Endocrine Gynecology, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France; Department of Assisted Reproductive Technologies and Fertility Preservation, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France; Department of Development and Plasticity of the Neuroendocrine Brain, U1172-Lille Neurosciences and Cognition (Jean-Pierre Aubert Research Center)-Lille Neurosciences and Cognition, Université Lille, Lille, France
| | - Anne Bachelot
- Endocrinology and Reproductive Medicine Department, Reference Center for Rare Growth and Development Endocrine Diseases, Assistance Publique-Hôpitaux de Paris, Sorbonne University, Pitié Salpêtrière University Hospital, Paris, France
| | - Maxime Leroy
- Biostatistics Department, Centre Hospitalier Universitaire Lille, Lille, France
| | - Christine Decanter
- Department of Assisted Reproductive Technologies and Fertility Preservation, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France
| | | | - Sophie Catteau-Jonard
- Department of Endocrine Gynecology, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France; Department of Assisted Reproductive Technologies and Fertility Preservation, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France; Department of Development and Plasticity of the Neuroendocrine Brain, U1172-Lille Neurosciences and Cognition (Jean-Pierre Aubert Research Center)-Lille Neurosciences and Cognition, Université Lille, Lille, France
| | - Geoffroy Robin
- Department of Endocrine Gynecology, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France; Department of Assisted Reproductive Technologies and Fertility Preservation, Lille University Hospital, Hospital Jeanne de Flandre, Lille, France; Department of Development and Plasticity of the Neuroendocrine Brain, U1172-Lille Neurosciences and Cognition (Jean-Pierre Aubert Research Center)-Lille Neurosciences and Cognition, Université Lille, Lille, France
| |
Collapse
|
8
|
Li X, Hou M, Zhang F, Ji Z, Cai Y, Shi Y. Per- and Polyfluoroalkyl Substances and Female Health Concern: Gender-based Accumulation Differences, Adverse Outcomes, and Mechanisms. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:1469-1486. [PMID: 39803974 DOI: 10.1021/acs.est.4c08701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
The deleterious health implications of perfluoroalkyl and polyfluoroalkyl substances (PFAS) are widely recognized. Females, in contrast to males, exhibit unique pathways for PFAS exposure and excretion, leading to complex health outcomes. The health status of females is largely influenced by hormone-related processes. PFAS have been reported to be associated with various aspects of female health, including reproductive system disorders and pregnancy-related diseases. In this article, we provide insights into the correlations between PFAS and female-prevalent diseases. Current epidemiological and toxicological evidence has demonstrated that the adverse effects of PFAS on the health of the female reproductive system are primarily attributed to the disruption of the hypothalamic-pituitary-gonadal (HPG) axis and hormonal homeostasis. However, these findings do not sufficiently elucidate the intricate associations between PFAS and specific diseases. Furthermore, autoimmune disorders, another category that is more prevalent in women compared to men, require additional investigation. Immune biomarkers pertinent to autoimmune disorders have been observed to be influenced by PFAS exposure, although epidemiological evidence is insufficient to substantiate these relations. Further thorough exploration encompassing epidemiological and toxicological studies is essential to elucidating the inherent influence of PFAS on human pathologies. Additionally, comprehensive investigations into female health issues beyond their reproductive functions is essential.
Collapse
Affiliation(s)
- Xin Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Minmin Hou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Feng Zhang
- Environmental Science Research & Design Institute of Zhejiang Province and Key Laboratory of Environmental Pollution Control Technology of Zhejiang Province, HangzhouZhejiang310007, China
| | - Zhengquan Ji
- Environmental Science Research & Design Institute of Zhejiang Province and Key Laboratory of Environmental Pollution Control Technology of Zhejiang Province, HangzhouZhejiang310007, China
| | - Yaqi Cai
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yali Shi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
9
|
You Y, Huo K, He L, Wang T, Zhao L, Li R, Cheng X, Ma X, Yue Z, Siwko S, Wang N, Liao L, Liu M, Luo J. GnIH secreted by green light exposure, regulates bone mass through the activation of Gpr147. Bone Res 2025; 13:13. [PMID: 39837853 PMCID: PMC11751147 DOI: 10.1038/s41413-024-00389-7] [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: 03/08/2024] [Revised: 10/30/2024] [Accepted: 11/13/2024] [Indexed: 01/23/2025] Open
Abstract
Reproductive hormones associated with the hypothalamic-pituitary-gonadal (HPG) axis are closely linked to bone homeostasis. In this study, we demonstrate that Gonadotropin inhibitory hormone (GnIH, one of the key reproductive hormones upstream of the HPG axis) plays an indispensable role in regulating bone homeostasis and maintaining bone mass. We find that deficiency of GnIH or its receptor Gpr147 leads to a significant reduction in bone mineral density (BMD) in mice primarily by enhancement of osteoclast activation in vivo and in vitro. Mechanistically, GnIH/Gpr147 inhibits osteoclastogenesis by the PI3K/AKT, MAPK, NF-κB and Nfatc1 signaling pathways. Furthermore, GnIH treatment was able to alleviate bone loss in aging, ovariectomy (OVX) or LPS-induced mice. Moreover, the therapy using green light promotes the release of GnIH and rescues OVX-induced bone loss. In humans, serum GnIH increases and bone resorption markers decrease after green light exposure. Therefore, our study elucidates that GnIH plays an important role in maintaining bone homeostasis via modulating osteoclast differentiation and demonstrates the potential of GnIH therapy or green light therapy in preventing osteoporosis.
Collapse
Affiliation(s)
- Yu You
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China
| | - Konglin Huo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China
| | - Liang He
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China
| | - Tongyue Wang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China
| | - Lei Zhao
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China
| | - Rong Li
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China
| | - Xiaoqing Cheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Xuebin Ma
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China
| | - Zhiying Yue
- Precision Research Center for Refractory Diseases, Shanghai General Hospital,Shanghai Jiaotong University, School of Medicine, Shanghai, PR China
| | - Stefan Siwko
- Department of Translational Medical Sciences, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, 77030, USA
| | - Ning Wang
- Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Lujian Liao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China.
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China.
| | - Jian Luo
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China.
| |
Collapse
|
10
|
Maclean A, Tipple L, Newton E, Hapangama DK. Hormone receptor profile of ectopic and eutopic endometrium in adenomyosis: a systematic review. Hum Reprod Open 2025; 2025:hoaf002. [PMID: 39935764 PMCID: PMC11810641 DOI: 10.1093/hropen/hoaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/03/2024] [Indexed: 02/13/2025] Open
Abstract
STUDY QUESTION What is the hormone receptor profile of adenomyosis lesions in comparison to correctly located endometrium? SUMMARY ANSWER Adenomyosis lesions exhibit increased oestrogen receptor (ER) expression compared to the eutopic endometrium; there are conflicting results regarding progesterone receptor (PR) expression and a lack of studies on androgen receptor (AR) expression. WHAT IS KNOWN ALREADY Adenomyosis lesions express hormone receptors indicating an influence from ovarian steroid hormones. However, hormone treatments are often ineffective in controlling adenomyosis symptoms, which suggests alternate hormonal responses and, potentially, a distinct hormone receptor expression profile within adenomyosis lesions compared to the eutopic endometrium. STUDY DESIGN SIZE DURATION This systematic review with a thematic analysis retrieved studies from the PubMed, Ovid Medline, Embase, Scopus, and Cochrane Library databases, and searches were conducted from inception through to May 2024. Human studies were included and identified using a combination of exploded MeSH terms ('adenomyosis') and free-text search terms ('oestrogen receptor', 'progesterone receptor', 'androgen receptor', 'hormone receptor'). PARTICIPANTS/MATERIALS SETTING METHODS This review was reported in accordance with the PRISMA guidelines. All studies reporting original data concerning hormone receptors in adenomyosis lesions compared to eutopic endometrium in adenomyosis were included. Studies that did not report original data or provide a review of the field were excluded. Bias analysis was completed for each study using the Newcastle-Ottawa scoring system. MAIN RESULTS AND THE ROLE OF CHANCE There were 1905 studies identified, which were screened to include 12 studies that met the eligibility criteria, including 11 proteomic studies and one transcriptional study, with a total of 555 individual participants. ER expression was consistently increased in adenomyosis lesions compared to the eutopic endometrium, specifically in the secretory phase. When endometrial subregion was considered, this difference was specific to the endometrial functionalis only. When different isoforms were considered, this increase in ER expression was specific to ERα rather than ERβ. There were conflicting results on PR expression, with most studies showing no significant difference or reduced levels in adenomyosis lesions compared to the eutopic endometrium. There is a paucity of data on AR expression in adenomyosis lesions, with only one study of small sample size included. LIMITATIONS REASONS FOR CAUTION A high risk of bias arose from studies grouping endometrial samples across different menstrual cycle phases for analysis. The coexistence of gynecological conditions like endometriosis may also confound the hormone receptor profile of the eutopic endometrium. Most studies employing immunostaining did not comment on region-specific differences in the endometrium. Given the well-documented cyclical variations in hormone receptor expression within the endometrium, the need for more attention to region-specific differences represents a notable limitation in the current body of literature. WIDER IMPLICATIONS OF THE FINDINGS The systematic review highlights oestrogen dominance through elevated ERα levels in adenomyosis lesions, which agrees with the literature suggesting local hyper-oestrogenism in adenomyosis lesions. Heterogeneity in menstrual cycle timing and lack of endometrial region specificity prevent conclusions on progesterone resistance within adenomyosis lesions in this study. Future investigations should minimize the bias through well-defined cohorts, leading to robust exploration of hormone receptor profiles in adenomyosis lesions to identify therapeutic targets and deepen our understanding of adenomyosis pathogenesis. STUDY FUNDING/COMPETING INTERESTS This work was supported by Wellbeing of Women Research Project grants RG1073 and RG2137 (D.K.H.), a Wellbeing of Women Entry-Level Scholarship ELS706 and a Medical Research Council grant MR/V007238/1 (A.M. and D.K.H.), as well as the University of Liverpool (L.T.). There are no conflicts of interest. HROPEN-24-0294R2 The review protocol was published in the PROSPERO Register of Systematic Reviews on 27 September 2023, registration number CRD4202346.
Collapse
Affiliation(s)
- Alison Maclean
- Department of Women’s and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Centre for Women’s Health Research, Liverpool Women’s Hospital, Liverpool, UK
| | - Laura Tipple
- School of Medicine, University of Liverpool, Liverpool, UK
| | - Emily Newton
- The Hewitt Fertility Centre, Liverpool Women’s NHS Foundation Trust, Knutsford, UK
| | - Dharani K Hapangama
- Department of Women’s and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Centre for Women’s Health Research, Liverpool Women’s Hospital, Liverpool, UK
| |
Collapse
|
11
|
Dai R, Sun Y. Altered GnRH neuron-glia networks close to interface of polycystic ovary syndrome: Molecular mechanism and clinical perspectives. Life Sci 2025; 361:123318. [PMID: 39719166 DOI: 10.1016/j.lfs.2024.123318] [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: 08/27/2024] [Revised: 11/24/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024]
Abstract
Polycystic ovary syndrome (PCOS) has been noticed as a neuroendocrine syndrome manifested by reproductive hormone dysregulation involving increased luteinizing hormone (LH) pulse frequency and an increased LH to follicle-stimulating hormone ratio, yet theory is just beginning to be established. Neuroglia located in the arcuate nucleus and median eminence (ARC-ME) that are close to gonadotropin-releasing hormone (GnRH) axon terminals, comprise the blood-brain barrier and fenestrated vessels implying their putative roles in the modulation of the abnormal GnRH pulse in PCOS. This review outlines the disturbances of neuron-glia networks that underlie hypothetically the deregulation of GnRH-LH release and impaired sex hormone negative feedback in PCOS. We then discuss chronic and low-grade inflammatory status together with gut dysbiosis and how the detriments may intrude the hypothalamus by virtue of violating interfaces between the brain and periphery, which might contribute to the etiology of the impaired neural circuits in the ARC-ME to induce PCOS.
Collapse
Affiliation(s)
- Ruoxi Dai
- Hospital & Institute of Obstetrics and Gynecology, Fudan University, Shanghai 200081, China
| | - Yan Sun
- Hospital & Institute of Obstetrics and Gynecology, Fudan University, Shanghai 200081, China; The Academy of Integrative Medicine, Fudan University, Shanghai 200081, China; Shanghai Key Laboratory of Female Reproductive Endocrine-related Disease, Shanghai 200081, China.
| |
Collapse
|
12
|
Patel S, Saxena B, Mehta P, Niazi SK. GnRH Peptide Antagonist: Comparative Analysis of Chemistry and Formulation with Implications for Clinical Safety and Efficacy. Pharmaceuticals (Basel) 2024; 18:36. [PMID: 39861098 PMCID: PMC11768417 DOI: 10.3390/ph18010036] [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: 12/12/2024] [Revised: 12/25/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
Overexpression of the gonadotropin-releasing hormone receptor (GnRH-R) plays a vital role in the advancement of reproductive malignancies such as ovarian, endometrial, and prostate cancer. Peptidomimetic GnRH antagonists are a substantial therapeutic development, providing fast and reversible suppression of gonadotropins by directly blocking GnRH-R. Unlike typical GnRH agonists, these antagonists prevent the early hormonal flare, have a faster onset of action, and have a lower risk of cardiovascular problems. These characteristics qualify GnRH antagonists as revolutionary therapy for diseases such as advanced prostate cancer, endometriosis, uterine fibroids, and in vitro fertilization procedures. Key GnRH peptide antagonists authorized by the regulatory agencies include Cetrorelix, Ganirelix, Abarelix, Degarelix, and Teverelix. Assisted reproductive technologies (ART) are dominated by Cetrorelix and Ganirelix, while Degarelix and Abarelix have shown significant promise in treating advanced prostate cancer. Teverelix appears as a next-generation GnRH antagonist with an ideal mix of efficacy and safety, showing promise in a variety of reproductive and hormone-dependent illnesses. This review investigates the pharmacological role of GnRH in reproductive physiology and its consequences in disease, emphasizing structural advances in third- and fourth-generation GnRH antagonists. All GnRH peptide-based antagonists were analyzed in detail for formulation strategy, pharmacokinetics, effectiveness, and safety. This review also emphasizes GnRH antagonists' clinical promise, providing insights into their evolution and the possibility for future research in developing safer, more effective treatments for complicated hormonal diseases.
Collapse
Affiliation(s)
- Shikha Patel
- Department of Pharmaceutical Analysis, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India; (S.P.); (P.M.)
| | - Bhagawati Saxena
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India;
| | - Priti Mehta
- Department of Pharmaceutical Analysis, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India; (S.P.); (P.M.)
| | | |
Collapse
|
13
|
▼Ryeqo for treatment of uterine fibroids. Drug Ther Bull 2024; 63:6-11. [PMID: 39725459 DOI: 10.1136/dtb.2024.000042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
|
14
|
Barretta M, Vignali M, La Marca A, Grandi G. The oral GnRH antagonists, a new class of drugs in gynecology: from pharmacokinetics to possible clinical applications. Expert Opin Drug Metab Toxicol 2024:1-13. [PMID: 39666595 DOI: 10.1080/17425255.2024.2441981] [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: 08/20/2024] [Revised: 11/29/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION In ovarian steroid-dependent diseases such as uterine fibroids, endometriosis and adenomyosis, oral GnRH antagonists have emerged as new therapeutic alternatives. These oral GnRH antagonists offer key advantages, including oral administration, dose-dependent estrogen suppression and rapid reversibility. AREAS COVERED This review examines the pharmacological, clinical and therapeutic profiles of the latest non-peptide oral GnRH antagonists, through an analysis of clinical evidence and randomized clinical trials, to provide a comprehensive and up-to-date overview of their clinical applications and potential benefits. EXPERT OPINION The clinical trials examined demonstrated significant efficacy in reducing heavy menstrual bleeding in women with fibroids and pelvic pain in women with endometriosis, with more than 70% of patients achieving primary endpoints. The use of add-back therapy minimized bone mass density loss, ensuring long-term safety. Adverse events were dose-dependent but generally well tolerated. In our opinion, the strength of oral GnRH antagonists lies in their pharmacological properties. Oral administration increases convenience, allows adjustable dosing and ensures a dose-dependent effect. These drugs provide an immediate antagonistic effect without the flare-up phenomenon. Furthermore, they are expected to act on ectopic endometrial and smooth muscle cell receptors, potentially providing additional anti-proliferative effects. However, further research is needed: long term clinical trials must compare them with existing treatments.
Collapse
Affiliation(s)
- Marta Barretta
- Department of Clinical Sciences and Community Health, University of Milan, Macedonio Melloni Hospital, Milan, Italy
| | - Michele Vignali
- Department of Clinical Sciences and Community Health, University of Milan, Macedonio Melloni Hospital, Milan, Italy
| | - Antonio La Marca
- Department of Medical and Surgical Sciences for Mother, Child and Adult, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Giovanni Grandi
- Department of Medical and Surgical Sciences for Mother, Child and Adult, University of Modena and Reggio Emilia, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| |
Collapse
|
15
|
Xu YP, Fu JC, Hong ZL, Zeng DF, Guo CQ, Li P, Wu JX. Psychological stressors involved in the pathogenesis of premature ovarian insufficiency and potential intervention measures. Gynecol Endocrinol 2024; 40:2360085. [PMID: 38813955 DOI: 10.1080/09513590.2024.2360085] [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: 12/12/2023] [Accepted: 05/14/2024] [Indexed: 05/31/2024] Open
Abstract
Premature ovarian insufficiency (POI) is a common gynecological endocrine disease, which seriously affects women's physical and mental health and fertility, and its incidence is increasing year by year. With the development of social economy and technology, psychological stressors such as anxiety and depression caused by social, life and environmental factors may be one of the risk factors for POI. We used PubMed to search peer-reviewed original English manuscripts published over the last 10 years to identify established and experimental studies on the relationship between various types of stress and decreased ovarian function. Oxidative stress, follicular atresia, and excessive activation of oocytes, caused by Stress-associated factors may be the main causes of ovarian function damage. This article reviews the relationship between psychological stressors and hypoovarian function and the possible early intervention measures in order to provide new ideas for future clinical treatment and intervention.
Collapse
Affiliation(s)
- Ying-Pei Xu
- Department of Reproductive Medicine, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Ji-Chun Fu
- Department of Reproductive Medicine, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Zhi-Lin Hong
- Clinical Laboratory Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - De-Fei Zeng
- Department of Reproductive Medicine, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Chao-Qin Guo
- Department of Reproductive Medicine, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Ping Li
- Department of Reproductive Medicine, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Jin-Xiang Wu
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Department of Reproductive Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| |
Collapse
|
16
|
Mbiydzenyuy NE, Qulu LA. Stress, hypothalamic-pituitary-adrenal axis, hypothalamic-pituitary-gonadal axis, and aggression. Metab Brain Dis 2024; 39:1613-1636. [PMID: 39083184 PMCID: PMC11535056 DOI: 10.1007/s11011-024-01393-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 07/08/2024] [Indexed: 11/05/2024]
Abstract
This comprehensive review explores the intricate relationship between the hypothalamic-pituitary-adrenal (HPA) axis, the hypothalamic-pituitary-gonadal (HPG) axis, and aggression. It provides a detailed overview of the physiology and functioning of these axes, as well as the implications for aggressive behavior. The HPA axis, responsible for the stress response, is activated in response to various stressors and can influence aggressive behavior. Glucocorticoids, such as cortisol, play a crucial role in stress-induced activation of the HPA axis and have been implicated in aggressive tendencies. Chronic stress can dysregulate the HPA axis, leading to alterations in cortisol levels and potentially contributing to aggressive behavior. The HPG axis, particularly the androgen hormone testosterone, is also closely linked to aggression. Animal and human studies have consistently shown a positive association between testosterone levels and aggression. The androgen receptors in the brain's neural circuitry play a critical role in modulating aggressive behavior. Interactions between the HPA and HPG axes further contribute to the regulation of aggression. Feedback mechanisms and crosstalk between these axes provide a complex system for the modulation of both stress and reproductive functions, which can impact aggressive behavior. Additionally,the influence of stress on reproductive functions, particularly the role of androgens in stress-induced aggression, adds further complexity to this relationship. The review also discusses the future directions and implications for clinical interventions. Understanding the neurobiological mechanisms underlying aggression requires integrating molecular, cellular, and circuit-level approaches. Translational perspectives, including animal models and human studies, can bridge the gap between basic research and clinical applications. Finally, therapeutic strategies for aggression-related disorders are explored, highlighting the importance of targeted interventions based on a comprehensive understanding of the interactions between the HPA and HPG axes. In conclusion, this review provides a comprehensive overview of the physiological and neurobiological mechanisms underlying aggression, with a specific focus on the interplay between the HPA and HPG axes. By elucidating the complex interactions between stress, hormones, and aggressive behavior, this research paves the way for future investigations and potential therapeutic interventions for aggression-related disorders.
Collapse
Affiliation(s)
- Ngala Elvis Mbiydzenyuy
- Basic Science Department, School of Medicine, Copperbelt University, P.O Box 71191, Ndola, Zambia
- Division of Medical Physiology, Biomedical Science Research Institute, Stellenbosch University, Private Bag X1, Matieland, 7602, Cape Town, South Africa
| | - Lihle-Appiah Qulu
- Division of Medical Physiology, Biomedical Science Research Institute, Stellenbosch University, Private Bag X1, Matieland, 7602, Cape Town, South Africa.
| |
Collapse
|
17
|
Karadağ MA, Gram A, Schäfer-Somi S, Aslan S, Kaya D. Expression of GnRH, Kisspeptin, and Their Specific Receptors in the Ovary and Uterus in Deslorelin-Treated Late-Prepubertal Bitches. Vet Sci 2024; 11:591. [PMID: 39728931 DOI: 10.3390/vetsci11120591] [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: 10/17/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
In this study, the expression and localization of gonadotropin-releasing hormone (GnRH1) and kisspeptin (KISS1) and their specific receptors in canine ovarian and uterine tissues were investigated after the application of deslorelin acetate (Suprelorin®, 4.7 mg, Virbac, France) in the late prepubertal period. We hypothesized that prolonged treatment of prepubertal dogs with deslorelin would alter the expression of GnRH and kisspeptin genes in the uterus and ovaries. Ovarian and uterine samples of 25 dogs with an average age of 7.8 ± 0.2 months and from mixed breeds were used. Following implant insertion, dogs entered estrus (EST; n = 6); dogs without estrus (N-EST; n = 10) comprised the experimental groups. Nine dogs with placebo implants served as a control (CONT). Ovarian and uterine tissues were investigated for expression of GnRH1, GnRHR, KISS1, and KISS1R/GPR54 mRNA and protein by using IHC and RT-qPCR. In the uterus, expression of GnRH1 significantly decreased in response to deslorelin treatment in the N-EST, compared with the control group. Compared with CONT, KISS1R expression in ovarian samples was significantly lower in the EST group. Uterine protein expression of GnRH1 appeared weaker in N-EST than in CONT. While GnRH1-system members and KISS1 protein were localized in the follicles at various stages and stroma, no or only weak signals were detected for KISS1R in the ovarian samples. Deslorelin-mediated induction of puberty by changing the expression of some of the GnRH and KISS1-system members seems to have an effect on ovarian and uterine functionality. Deslorelin implants can, therefore, not be considered a valuable alternative to induce fertile estrus in late-prepubertal bitches. However, further studies with a larger number of animals are needed to clarify the effect of deslorelin-mediated induction of puberty.
Collapse
Affiliation(s)
- Muhammet Ali Karadağ
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Kafkas University, Kars 36100, Türkiye
| | - Aykut Gram
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Erciyes University, Kayseri 38039, Türkiye
| | | | - Selim Aslan
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Near East University, Nicosia 99138, Cyprus
| | - Duygu Kaya
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Dokuz Eylül University, İzmir 35890, Türkiye
| |
Collapse
|
18
|
Lv W, An R, Li X, Zhang Z, Geri W, Xiong X, Yin S, Fu W, Liu W, Lin Y, Li J, Xiong Y. Multi-Omics Approaches Uncovered Critical mRNA-miRNA-lncRNA Networks Regulating Multiple Birth Traits in Goat Ovaries. Int J Mol Sci 2024; 25:12466. [PMID: 39596531 PMCID: PMC11595133 DOI: 10.3390/ijms252212466] [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: 09/19/2024] [Revised: 11/10/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
The goat breeding industry on the Tibetan Plateau faces strong selection pressure to enhance fertility. Consequently, there is an urgent need to develop goat lines with higher fertility and adaptability. The ovary, as a key organ determining reproductive performance, is regulated by a complex transcriptional network involving numerous protein-coding and non-coding genes. However, the molecular mechanisms of the key mRNA-miRNA-lncRNA regulatory network in goat ovaries remain largely unknown. This study focused on the histology and differential mRNA/miRNA/lncRNA between Chuanzhong black goat (CBG, high productivity, multiple births) and Tibetan goat (TG, strong adaptability, single birth) ovaries. Histomorphological analysis showed that the medulla proportion in CBG ovaries was significantly reduced compared to TG. RNA-Seq and small RNA-Seq analysis identified 1218 differentially expressed (DE) mRNAs, 100 DE miRNAs, and 326 DE lncRNAs, which were mainly enriched in ovarian steroidogenesis, oocyte meiosis, biosynthesis of amino acids and protein digestion, and absorption signaling pathways. Additionally, five key mRNA-miRNA-lncRNA interaction networks regulating goat reproductive performance were identified, including TCL1B-novel68_mature-ENSCHIT00000010023, AKAP6-novel475_mature-ENSCHIT00000003176, GLI2-novel68_mature-XR_001919123.1, ITGB5-novel65_star-TCONS_00013850, and VWA2-novel71_mature-XR_001919911.1. Further analyses showed that these networks mainly affected ovarian function and reproductive performance by regulating biological processes such as germ cell development and oocyte development, which also affected the plateau adaptive capacity of the ovary by participating in the individual immune and metabolic capacities. In conclusion, we identified numerous mRNA-miRNA-lncRNA interaction networks involved in regulating ovarian function and reproductive performance in goats. This discovery offers new insights into the molecular breeding of Tibetan Plateau goats and provides a theoretical foundation for developing new goat lines with high reproductive capacity and strong adaptability to the plateau environment.
Collapse
Affiliation(s)
- Weibing Lv
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China; (W.L.); (R.A.); (X.L.); (Z.Z.); (W.G.); (X.X.); (S.Y.); (W.F.); (W.L.); (Y.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu 610041, China
| | - Ren An
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China; (W.L.); (R.A.); (X.L.); (Z.Z.); (W.G.); (X.X.); (S.Y.); (W.F.); (W.L.); (Y.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu 610041, China
| | - Xinmiao Li
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China; (W.L.); (R.A.); (X.L.); (Z.Z.); (W.G.); (X.X.); (S.Y.); (W.F.); (W.L.); (Y.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu 610041, China
| | - Zengdi Zhang
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China; (W.L.); (R.A.); (X.L.); (Z.Z.); (W.G.); (X.X.); (S.Y.); (W.F.); (W.L.); (Y.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu 610041, China
| | - Wanma Geri
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China; (W.L.); (R.A.); (X.L.); (Z.Z.); (W.G.); (X.X.); (S.Y.); (W.F.); (W.L.); (Y.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu 610041, China
| | - Xianrong Xiong
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China; (W.L.); (R.A.); (X.L.); (Z.Z.); (W.G.); (X.X.); (S.Y.); (W.F.); (W.L.); (Y.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu 610041, China
| | - Shi Yin
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China; (W.L.); (R.A.); (X.L.); (Z.Z.); (W.G.); (X.X.); (S.Y.); (W.F.); (W.L.); (Y.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu 610041, China
| | - Wei Fu
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China; (W.L.); (R.A.); (X.L.); (Z.Z.); (W.G.); (X.X.); (S.Y.); (W.F.); (W.L.); (Y.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu 610041, China
| | - Wei Liu
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China; (W.L.); (R.A.); (X.L.); (Z.Z.); (W.G.); (X.X.); (S.Y.); (W.F.); (W.L.); (Y.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province, Southwest Minzu University, Chengdu 610041, China
| | - Yaqiu Lin
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China; (W.L.); (R.A.); (X.L.); (Z.Z.); (W.G.); (X.X.); (S.Y.); (W.F.); (W.L.); (Y.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province, Southwest Minzu University, Chengdu 610041, China
| | - Jian Li
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China; (W.L.); (R.A.); (X.L.); (Z.Z.); (W.G.); (X.X.); (S.Y.); (W.F.); (W.L.); (Y.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu 610041, China
| | - Yan Xiong
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China; (W.L.); (R.A.); (X.L.); (Z.Z.); (W.G.); (X.X.); (S.Y.); (W.F.); (W.L.); (Y.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu 610041, China
| |
Collapse
|
19
|
Mate NA, Wadhwa G, Taliyan R, Banerjee A. Impact of polyamine supplementation on GnRH expression, folliculogenesis, and puberty onset in young mice. Theriogenology 2024; 229:202-213. [PMID: 39217649 DOI: 10.1016/j.theriogenology.2024.07.028] [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: 04/16/2024] [Revised: 07/10/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The hypothalamic-pituitary-gonadal (HPG) axis is pivotal in regulating reproductive functions, with gonadotropin-releasing hormone (GnRH) acting as a central regulator. Recently, polyamines have been shown to regulate the HPG axis, including GnRH expression and ovarian biology in old and adult rodents. The present study firstly highlights the age-specific variation in the polyamine and their corresponding biosynthetic enzymes in the ovary during aging, and further, the study focuses on the effect of polyamines, putrescine, and agmatine, in young female mice. METHOD AND RESULT Immunofluorescence analysis revealed age-related differences in the expression of ornithine decarboxylase 1 (ODC1), spermine (SPM), and spermidine (SPD) in the ovaries, with adult mice exhibiting significantly higher expression levels compared to young and old mice. Likewise, qPCR analysis showed the mRNA levels of Odc1, Spermidine synthase (Srm), and Spermine synthase (Sms) show a significant increase in adult ovaries, which is then followed by a significant decline in old age. Histological examination demonstrated morphological alterations in the ovaries with age, including decreased follicle numbers and increased stromal cells in old mice. Furthermore, treatment with putrescine, a polyamine, in young mice resulted in larger ovaries and increased follicle numbers compared to controls. Additionally, serum levels of gonadotropin-releasing hormone (GnRH) and progesterone (P4) were measured, showing elevated levels in polyamine-treated mice. GnRH mRNA expression also increased significantly. Gene expression analysis revealed upregulation of genes associated with folliculogenesis such as Fshr, Bmp15, Gdf9, Amh, Star, Hsdb3, and Plaur in the ovaries and onset of puberty such as Tac2, and Kiss1, and a decrease in Mkrn3 in the hypothalamus of polyamine-treated mice. CONCLUSION This study investigates the effect of polyamines in young immature female mice, shedding light on their role in upregulating GnRH, and enhancing folliculogenesis. Overall, these findings suggest that polyamines play a crucial role in ovarian aging and HPG axis regulation, offering potential therapeutics to reinstate fertility in reproductively challenged individuals.
Collapse
Affiliation(s)
- Nayan Anand Mate
- Department of Biological Sciences, Birla Institute of Technology and Science, K K Birla Goa Campus, Zuarinagar, Goa, India
| | - Geetika Wadhwa
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, 333031, India
| | - Rajeev Taliyan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, Rajasthan, 333031, India
| | - Arnab Banerjee
- Department of Biological Sciences, Birla Institute of Technology and Science, K K Birla Goa Campus, Zuarinagar, Goa, India.
| |
Collapse
|
20
|
Hu R, Geng Y, Huang Y, Liu Z, Li F, Song K, Ma W, Dong H, Zhang M, Lei T, Song Y, Zhang Z. Jiawei Buzhong Yiqi Decoction attenuates polycystic ovary syndrome through regulating kisspeptin-GPR54-AKT-SHBG system. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155931. [PMID: 39116604 DOI: 10.1016/j.phymed.2024.155931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 07/27/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is one of the most common reproductive endocrine disorders. Accumulated evidence has suggested the indispensable role of kisspeptin-G protein-coupled receptor (GPR54) system and SHBG in development of PCOS. However, potential mechanisms and their relationship are unclear. Jiawei Buzhong Yiqi Decoction (JWBZYQ) has been reported to ameliorate obese PCOS. Whereas, potential mechanisms remain elusive. PURPOSE To determine whether JWBZYQ attenuates PCOS by regulating the kisspeptin-GPR54 system and SHBG production. And to explore potential mechanisms. METHODS An overweight PCOS rat model was developed with testosterone propionate (TP) and high-fat diet (HFD). The efficacy of JWBZYQ was assessed by tracking changes in weight, estrous cycle, ovarian morphology, and serum sex hormone levels. Additionally, kisspeptin-GPR54 system expression in multiple organs and PI3K-AKT pathway activity in liver of different rats were detected. Modifications in SHBG production were also measured. Kisspeptin54 was administered to establish a cellular model. The levels of AKT phosphorylation and SHBG protein within HepG2 cells were analyzed. Finally, confirmatory studies were performed using AKT phosphorylation activator and inhibitor. RESULTS JWBZYQ effectively attenuated the overweight, disrupted estrous cycle, altered sex hormone levels, and aberrant ovarian morphology in PCOS rats. Meanwhile, PCOS rats exhibited elevated levels of kisspeptin and GPR54, along with reduced SHBG levels, which could be reversed by JWBZYQ. These alterations might be connected with the activation of AKT phosphorylation. In vitro experiment identified that JWBZYQ could rectify the hyperactivated AKT phosphorylation and deficient production of SHBG caused by kisspeptin54. CONCLUSIONS Overexpressed kisspeptin-GPR54 system inhibited SHBG synthesis in PCOS. JWBZYQ curtailed the exorbitant expression of kisspeptin and GPR54, which moderated the rise in AKT phosphorylation and subsequently promoted the production of SHBG.
Collapse
Affiliation(s)
- Runan Hu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuli Geng
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yanjing Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhuo Liu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fan Li
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kunkun Song
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wenwen Ma
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Haoxu Dong
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mingmin Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ting Lei
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yufan Song
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhuo Zhang
- Sino-German Neuro-Oncology Molecular Laboratory, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
21
|
Kokori E, Olatunji G, Komolafe R, Ogieuhi IJ, Ukoaka B, Ajayi I, Aderinto N. Serum kisspeptin as a promising biomarker for PCOS: a mini review of current evidence and future prospects. Clin Diabetes Endocrinol 2024; 10:27. [PMID: 39343941 PMCID: PMC11440685 DOI: 10.1186/s40842-024-00190-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/06/2024] [Indexed: 10/01/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder affecting women of reproductive age, characterised by its multifactorial nature and intricate interplay of genetic, hormonal, and environmental factors. As the search for reliable biomarkers intensifies, serum kisspeptin emerges as a promising candidate due to its central role in regulating the hypothalamic-pituitary-gonadal (HPG) axis. This review aims to consolidate the evolving understanding of kisspeptin as a potential PCOS biomarker, comprehensively exploring its physiological basis, diagnostic challenges in PCOS, and clinical implications. Diagnostic challenges in PCOS are addressed, underscoring the limitations of current criteria and the need for objective and standardised biomarkers. Kisspeptin's introduction as a potential biomarker brings forth both promises and challenges in terms of its diagnostic utility. The review recognises the importance of standardisation in research methodologies and emphasises the exploration of genetic polymorphisms to enhance kisspeptin's robustness as a diagnostic tool.
Collapse
Affiliation(s)
- Emmanuel Kokori
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Gbolahan Olatunji
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Rosemary Komolafe
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | | | - Bonaventure Ukoaka
- Department of Internal Medicine, Asokoro District Hospital, Abuja, Nigeria
| | - Irene Ajayi
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Nicholas Aderinto
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Old Oyo/ Ilorin Rd, P.M.B 4000, Ogbomosho, Oyo-State, 210214, Nigeria.
| |
Collapse
|
22
|
Leng D, Zeng B, Wang T, Chen BL, Li DY, Li ZJ. Single nucleus/cell RNA-seq of the chicken hypothalamic-pituitary-ovarian axis offers new insights into the molecular regulatory mechanisms of ovarian development. Zool Res 2024; 45:1088-1107. [PMID: 39245652 PMCID: PMC11491784 DOI: 10.24272/j.issn.2095-8137.2024.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 06/17/2024] [Indexed: 09/10/2024] Open
Abstract
The hypothalamic-pituitary-ovarian (HPO) axis represents a central neuroendocrine network essential for reproductive function. Despite its critical role, the intrinsic heterogeneity within the HPO axis across vertebrates and the complex intercellular interactions remain poorly defined. This study provides the first comprehensive, unbiased, cell type-specific molecular profiling of all three components of the HPO axis in adult Lohmann layers and Liangshan Yanying chickens. Within the hypothalamus, pituitary, and ovary, seven, 12, and 13 distinct cell types were identified, respectively. Results indicated that the pituitary adenylate cyclase activating polypeptide (PACAP), follicle-stimulating hormone (FSH), and prolactin (PRL) signaling pathways may modulate the synthesis and secretion of gonadotropin-releasing hormone (GnRH), FSH, and luteinizing hormone (LH) within the hypothalamus and pituitary. In the ovary, interactions between granulosa cells and oocytes involved the KIT, CD99, LIFR, FN1, and ANGPTL signaling pathways, which collectively regulate follicular maturation. The SEMA4 signaling pathway emerged as a critical mediator across all three tissues of the HPO axis. Additionally, gene expression analysis revealed that relaxin 3 (RLN3), gastrin-releasing peptide (GRP), and cocaine- and amphetamine regulated transcripts (CART, also known as CARTPT) may function as novel endocrine hormones, influencing the HPO axis through autocrine, paracrine, and endocrine pathways. Comparative analyses between Lohmann layers and Liangshan Yanying chickens demonstrated higher expression levels of GRP, RLN3, CARTPT, LHCGR, FSHR, and GRPR in the ovaries of Lohmann layers, potentially contributing to their superior reproductive performance. In conclusion, this study provides a detailed molecular characterization of the HPO axis, offering novel insights into the regulatory mechanisms underlying reproductive biology.
Collapse
Affiliation(s)
- Dong Leng
- School of Pharmacy, Chengdu University, Chengdu, Sichuan 610106, China
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Bo Zeng
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Tao Wang
- School of Pharmacy, Chengdu University, Chengdu, Sichuan 610106, China
| | - Bin-Long Chen
- College of Animal Science, Xichang University, Xichang, Sichuan 615000, China. E-mail:
| | - Di-Yan Li
- School of Pharmacy, Chengdu University, Chengdu, Sichuan 610106, China. E-mail:
| | - Zhuan-Jian Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China. E-mail:
| |
Collapse
|
23
|
Shpakov AO. Hormonal and Allosteric Regulation of the Luteinizing Hormone/Chorionic Gonadotropin Receptor. FRONT BIOSCI-LANDMRK 2024; 29:313. [PMID: 39344322 DOI: 10.31083/j.fbl2909313] [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: 05/20/2024] [Revised: 06/20/2024] [Accepted: 07/10/2024] [Indexed: 10/01/2024]
Abstract
Luteinizing hormone (LH) and human chorionic gonadotropin (CG), like follicle-stimulating hormone, are the most important regulators of the reproductive system. They exert their effect on the cell through the LH/CG receptor (LHCGR), which belongs to the family of G protein-coupled receptors. Binding to gonadotropin induces the interaction of LHCGR with various types of heterotrimeric G proteins (Gs, Gq/11, Gi) and β-arrestins, which leads to stimulation (Gs) or inhibition (Gi) of cyclic adenosine monophosphate-dependent cascades, activation of the phospholipase pathway (Gq/11), and also to the formation of signalosomes that mediate the stimulation of mitogen-activated protein kinases (β-arrestins). The efficiency and selectivity of activation of intracellular cascades by different gonadotropins varies, which is due to differences in their interaction with the ligand-binding site of LHCGR. Gonadotropin signaling largely depends on the status of N- and O-glycosylation of LH and CG, on the formation of homo- and heterodimeric receptor complexes, on the cell-specific microenvironment of LHCGR and the presence of autoantibodies to it, and allosteric mechanisms are important in the implementation of these influences, which is due to the multiplicity of allosteric sites in different loci of the LHCGR. The development of low-molecular-weight allosteric regulators of LHCGR with different profiles of pharmacological activity, which can be used in medicine for the correction of reproductive disorders and in assisted reproductive technologies, is promising. These and other issues regarding the hormonal and allosteric regulation of LHCGR are summarized and discussed in this review.
Collapse
Affiliation(s)
- Alexander O Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| |
Collapse
|
24
|
Di Nisio V, Daponte N, Messini C, Anifandis G, Antonouli S. Oncofertility and Fertility Preservation for Women with Gynecological Malignancies: Where Do We Stand Today? Biomolecules 2024; 14:943. [PMID: 39199331 PMCID: PMC11353009 DOI: 10.3390/biom14080943] [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: 05/19/2024] [Revised: 07/19/2024] [Accepted: 08/01/2024] [Indexed: 09/01/2024] Open
Abstract
Oncofertility is a growing medical and research field that includes two main areas: oncology and reproductive medicine. Nowadays, the percentage of patients surviving cancer has exponentially increased, leading to the need for intervention for fertility preservation in both men and women. Specifically, gynecological malignancies in women pose an additional layer of complexity due to the reproductive organs being affected. In the present review, we report fertility preservation options with a cancer- and stage-specific focus. We explore the drawbacks and the necessity for planning fertility preservation applications during emergency statuses (i.e., the COVID-19 pandemic) and comment on the importance of repro-counseling for multifaceted patients during their oncological and reproductive journey.
Collapse
Affiliation(s)
- Valentina Di Nisio
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Huddinge, 14186 Stockholm, Sweden;
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, 14186 Stockholm, Sweden
| | - Nikoletta Daponte
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece; (N.D.); (C.M.); (G.A.)
| | - Christina Messini
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece; (N.D.); (C.M.); (G.A.)
| | - George Anifandis
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece; (N.D.); (C.M.); (G.A.)
| | - Sevastiani Antonouli
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece; (N.D.); (C.M.); (G.A.)
| |
Collapse
|
25
|
Longobardi S, Klinger FG, Zheng W, Campitiello MR, D’Hooghe T, La Marca A. Gonadotropin Activity during Early Folliculogenesis and Implications for Polycystic Ovarian Syndrome and Premature Ovarian Insufficiency: A Narrative Review. Int J Mol Sci 2024; 25:7520. [PMID: 39062762 PMCID: PMC11277126 DOI: 10.3390/ijms25147520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Female fertility depends on the ovarian reserve of follicles, which is determined at birth. Primordial follicle development and oocyte maturation are regulated by multiple factors and pathways and classified into gonadotropin-independent and gonadotropin-dependent phases, according to the response to gonadotropins. Folliculogenesis has always been considered to be gonadotropin-dependent only from the antral stage, but evidence from the literature highlights the role of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) during early folliculogenesis with a potential role in the progression of the pool of primordial follicles. Hormonal and molecular pathway alterations during the very earliest stages of folliculogenesis may be the root cause of anovulation in polycystic ovary syndrome (PCOS) and in PCOS-like phenotypes related to antiepileptic treatment. Excessive induction of primordial follicle activation can also lead to premature ovarian insufficiency (POI), a condition characterized by menopause in women before 40 years of age. Future treatments aiming to suppress initial recruitment or prevent the growth of resting follicles could help in prolonging female fertility, especially in women with PCOS or POI. This review will briefly introduce the impact of gonadotropins on early folliculogenesis. We will discuss the influence of LH on ovarian reserve and its potential role in PCOS and POI infertility.
Collapse
Affiliation(s)
| | - Francesca Gioia Klinger
- Department of Histology and Embryology, University of Health Sciences, Saint Camillus International, Via di Sant’Alessandro 8, 00131 Rome, Italy
| | | | - Maria Rosaria Campitiello
- Department of Obstetrics and Gynecology and Physiopathology of Human Reproduction, ASL Salerno, 84124 Salerno, Italy
| | - Thomas D’Hooghe
- Merck KGaA, 64293 Darmstadt, Germany (T.D.)
- Department of Development and Regeneration, Biomedical Sciences Group, KU Leuven (University of Leuven), 3000 Leuven, Belgium
| | - Antonio La Marca
- Department of Maternal-Child and Adult Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
26
|
Tu B, Zhang H, Chen L, Yang R, Liu P, Li R, Qiao J. Co-administration of GnRH-agonist and hCG (double trigger) for final oocyte maturation increases the number of top-quality embryos in patients undergoing IVF/ICSI cycles. J Ovarian Res 2024; 17:137. [PMID: 38961417 PMCID: PMC11223314 DOI: 10.1186/s13048-024-01465-6] [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: 04/20/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND The utilization of a double trigger, involving the co-administration of gonadotropin-releasing hormone agonist (GnRH-a) and human chorionic gonadotropin (hCG) for final oocyte maturation, is emerging as a novel approach in gonadotropin-releasing hormone antagonist (GnRH-ant) protocols during controlled ovarian hyperstimulation (COH). This protocol involves administering GnRH-a and hCG 40 and 34 h prior to ovum pick-up (OPU), respectively. This treatment modality has been implemented in patients with low/poor oocytes yield. This study aimed to determine whether the double trigger could improve the number of top-quality embryos (TQEs) in patients with fewer than three TQEs. METHODS The stimulation characteristics of 35 in vitro fertilization (IVF) cycles were analyzed. These cycles were triggered by the combination of hCG and GnRHa (double trigger cycles) and compared to the same patients' previous IVF attempt, which utilized the hCG trigger (hCG trigger control cycles). The analysis involved cases who were admitted to our reproductive center between January 2018 and December 2022. In the hCG trigger control cycles, all 35 patients had fewer than three TQEs. RESULTS Patients who received the double trigger cycles yielded a significantly higher number of 2PN cleavage embryos (3.54 ± 3.37 vs. 2.11 ± 2.15, P = 0.025), TQEs ( 2.23 ± 2.05 vs. 0.89 ± 0.99, P < 0.001), and a simultaneously higher proportion of the number of cleavage stage embryos (53.87% ± 31.38% vs. 39.80% ± 29.60%, P = 0.043), 2PN cleavage stage embryos (43.89% ± 33.01% vs. 27.22% ± 27.13%, P = 0.014), and TQEs (27.05% ± 26.26% vs. 14.19% ± 19.76%, P = 0.019) to the number of oocytes retrieved compared with the hCG trigger control cycles, respectively. The double trigger cycles achieved higher rates of cumulative clinical pregnancy (20.00% vs. 2.86%, P = 0.031), cumulative persistent pregnancy (14.29% vs. 0%, P < 0.001), and cumulative live birth (14.29% vs. 0%, P < 0.001) per stimulation cycle compared with the hCG trigger control cycles. CONCLUSION Co-administration of GnRH-agonist and hCG for final oocyte maturation, 40 and 34 h prior to OPU, respectively (double trigger) may be suggested as a valuable new regimen for treating patients with low TQE yield in previous hCG trigger IVF/intracytoplasmic sperm injection (ICSI) cycles.
Collapse
Affiliation(s)
- Binbin Tu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China
| | - Hua Zhang
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, China
| | - Lixue Chen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China
| | - Rui Yang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China.
| | - Ping Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China
| | - Rong Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
27
|
Bedoschi G, Ingold C, Navarro PA. Fertility Preservation and Ovarian Hyperstimulation Syndrome Management in Cancer Care: A Pathophysiological Perspective on Gonadotropin-Releasing Hormone Agonists and Antagonists. PATHOPHYSIOLOGY 2024; 31:288-297. [PMID: 38921726 PMCID: PMC11206524 DOI: 10.3390/pathophysiology31020021] [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: 12/31/2023] [Revised: 03/02/2024] [Accepted: 06/04/2024] [Indexed: 06/27/2024] Open
Abstract
This narrative review delves into the evolving landscape of fertility preservation techniques, with a particular focus on their use in patients undergoing oncology treatment that carries a risk of ovarian insufficiency. Advances in established methods such as cryopreservation of oocytes and embryos are highlighted, and the increasing use of gonadotropin-releasing hormone (GnRH) agonists is discussed. The review also addresses the complexities and controversies associated with these approaches, such as the 'flare-up' effect associated with GnRH agonists and the potential of GnRH antagonists to reduce the risk of ovarian hyperstimulation syndrome. Despite advances in fertility preservation, the report highlights the challenges we face, including the need for personalized treatment protocols and the management of associated risks. It calls for continued research and collaboration between healthcare professionals to refine these techniques and ultimately improve reproductive outcomes for patients facing the prospect of fertility-impairing treatment.
Collapse
Affiliation(s)
- Giuliano Bedoschi
- Department of Gynecology and Obstetrics, Reproductive Medicine Division, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14048-900, Brazil;
| | - Caroline Ingold
- Department of Collective Health, Sexual, Reproductive Health and Population Genetics, Faculdade de Medicina do ABC, Santo André 09060-870, Brazil;
| | - Paula Andrea Navarro
- Department of Gynecology and Obstetrics, Reproductive Medicine Division, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14048-900, Brazil;
| |
Collapse
|
28
|
Liao T, Gao Y, Yang X, Tang Y, Wang B, Yang Q, Gao X, Tang Y, He K, Shen J, Bao S, Pan G, Zhu P, Tao F, Shao S. Preconception depression reduces fertility: a couple-based prospective preconception cohort. Hum Reprod Open 2024; 2024:hoae032. [PMID: 38840940 PMCID: PMC11150884 DOI: 10.1093/hropen/hoae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 05/06/2024] [Indexed: 06/07/2024] Open
Abstract
STUDY QUESTION Is preconception depression associated with time to pregnancy (TTP) and infertility? SUMMARY ANSWER Couples with preconception depression needed a longer time to become pregnant and exhibited an increased risk of infertility. WHAT IS KNOWN ALREADY Preconception depression in women contributes to impaired fertility in clinical populations. However, evidence from the general population-especially based on couples-is relatively scant. STUDY DESIGN SIZE DURATION A couple-based prospective preconception cohort study was performed in 16 premarital examination centers between April 2019 and June 2021. The final analysis included 16 521 couples who tried to conceive for ≤6 months at enrollment. Patients with infertility were defined as those with a TTP ≥12 months and those who conceived through ART. PARTICIPANTS/MATERIALS SETTING METHODS Couples' depression was assessed using the Patient Health Questionnaire-9 at baseline. Reproductive outcomes were obtained via telephone at 6 and 12 months after enrollment. Fertility odds ratios (FORs) and infertility risk ratios (RRs) in different preconception depression groups were analyzed using the Cox proportional-hazard models and logistic regression, respectively. MAIN RESULTS AND THE ROLE OF CHANCE Of the 16 521 couples analyzed, 10 834 (65.6%) and 746 (4.5%) couples achieved pregnancy within the first 6 months and between the 6th and 12th months, respectively. The median (P25, P75) TTP was 3.0 (2.0, 6.0) months. The infertility rate was 13.01%. After adjusting for potential confounders, in the individual-specific analyses, we found that preconception depression in women was significantly related to reduced odds of fertility (FOR = 0.947, 95% CI: 0.908-0.988), and preconception depression in either men or women was associated with an increased risk of infertility (women: RR = 1.212, 95% CI: 1.076-1.366; men: RR = 1.214, 95% CI: 1.068-1.381); in the couple-based analyses, we found that-compared to couples where neither partner had depression-the couples where both partners had depression exhibited reduced fertility (adjusted FOR = 0.904, 95% CI: 0.838-0.975). The risk of infertility in the group where only the woman had depression and both partners had depression increased by 17.8% (RR = 1.178, 95% CI: 1.026-1.353) and 46.9% (RR = 1.469, 95% CI: 1.203-1.793), respectively. LIMITATIONS REASONS FOR CAUTION Reporting and recall bias were unavoidable in this large epidemiological study. Some residual confounding factors-such as the use of anti-depressants and other medications, sexual habits, and prior depressive and anxiety symptoms-remain unaddressed. We used a cut-off score of 5 to define depression, which is lower than prior studies. Finally, we assessed depression only at baseline, therefore we could not detect effects of temporal changes in depression on fertility. WIDER IMPLICATIONS OF THE FINDINGS This couple-based study indicated that preconception depression in individuals and couples negatively impacts couples' fertility. Early detection and intervention of depression to improve fertility should focus on both sexes. STUDY FUNDING/COMPETING INTERESTS This work was supported by grants from the National Natural Science Foundation of China (No. 82273638) and the National Key Research and Development Program of China (No. 2018YFC1004201). All authors declare no conflicts of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Tierong Liao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
| | - Yaya Gao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
| | - Xinliu Yang
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, China
| | - Yanlan Tang
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Hefei, China
| | - Baolin Wang
- Key Laboratory of Population Health Across Life Cycle, Ministry of Education of the People’s Republic of China, Anhui Medical University, Hefei, China
| | - Qianhui Yang
- Key Laboratory of Population Health Across Life Cycle, Ministry of Education of the People’s Republic of China, Anhui Medical University, Hefei, China
| | - Xin Gao
- Key Laboratory of Population Health Across Life Cycle, Ministry of Education of the People’s Republic of China, Anhui Medical University, Hefei, China
| | - Ying Tang
- Key Laboratory of Population Health Across Life Cycle, Ministry of Education of the People’s Republic of China, Anhui Medical University, Hefei, China
| | - Kunjing He
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
| | - Jing Shen
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
| | - Shuangshuang Bao
- Key Laboratory of Population Health Across Life Cycle, Ministry of Education of the People’s Republic of China, Anhui Medical University, Hefei, China
| | - Guixia Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Peng Zhu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
| | - Fangbiao Tao
- Key Laboratory of Population Health Across Life Cycle, Ministry of Education of the People’s Republic of China, Anhui Medical University, Hefei, China
| | - Shanshan Shao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
| |
Collapse
|
29
|
Wang M, Luo J, Dai L, Feng M, Cao X, Zhang J, Wan Y, Yang X, Wang Y. Foxp2 deficiency impairs reproduction by modulating the hypothalamic-pituitary-gonadal axis in zebrafish†. Biol Reprod 2024; 110:908-923. [PMID: 38288660 DOI: 10.1093/biolre/ioae019] [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: 10/07/2023] [Revised: 01/20/2024] [Accepted: 01/23/2024] [Indexed: 02/29/2024] Open
Abstract
FOXP2 was initially characterized as a transcription factor linked to speech and language disorders. Single-cell RNA sequencing reveals that Foxp2 is enriched in the gonadotrope cluster of the pituitary gland and colocalized with the hormones LHB and FSHB in chickens and mice, implying that FOXP2 might be associated with reproduction in vertebrates. Herein, we investigated the roles of foxp2 in reproduction in a Foxp2-deficient zebrafish model. The results indicated that the loss of Foxp2 inhibits courtship behavior in adult male zebrafish. Notably, Foxp2 deficiency disrupts gonad development, leading to retardation of follicle development and a decrease in oocytes in females at the full-growth stage, among other phenotypes. The transcriptome analysis (RNA-seq) also revealed that differentially expressed genes clustered into the estrogen signaling and ovarian steroidogenesis-related signaling pathways. In addition, we found that Foxp2 deficiency could modulate the hypothalamic-pituitary-gonadal axis, especially the regulation of lhb and fshb expression, in zebrafish. In contrast, the injection of human chorionic gonadotropin, a specific LH agonist, partially rescues Foxp2-impaired reproduction in zebrafish, suggesting that Foxp2 plays an important role in the regulation of reproduction via the hypothalamic-pituitary-gonadal axis in zebrafish. Thus, our findings reveal a new role for Foxp2 in the regulation of reproduction in vertebrates.
Collapse
Affiliation(s)
- Maya Wang
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Juanjuan Luo
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Lu Dai
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Meilan Feng
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Xiaoqian Cao
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Jiannan Zhang
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Yiping Wan
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Xiaojun Yang
- Shantou University Medical College, Shantou, China
| | - Yajun Wang
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| |
Collapse
|
30
|
Vallée A, Carbonnel M, Ceccaldi PF, Feki A, Ayoubi JM. Postmenopausal endometriosis: a challenging condition beyond menopause. Menopause 2024; 31:447-456. [PMID: 38531006 DOI: 10.1097/gme.0000000000002338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
IMPORTANCE AND OBJECTIVE Postmenopausal endometriosis is a complex condition that challenges the conventional belief that endometriosis resolves with menopause. Despite the cessation of menstruation, a subset of women continues to experience or develop endometriosis-related symptoms during the postmenopausal period. Thus, this review aimed to shed light on postmenopausal endometriosis, exploring its clinical features, diagnostic considerations, management approaches, and the potential impact on women's health. METHODS PubMed/Medline, Scopus, and Web of Science databases were used for the research, with only articles in English language, using the following terms: "postmenopausal endometriosis," "menopause," "management," "treatment," and "quality of life," from inception to 2023. DISCUSSION AND CONCLUSION The clinical features of postmenopausal endometriosis include persistent or recurrent pelvic pain, dyspareunia, bowel, or urinary symptoms and, occasionally, abnormal vaginal bleeding. The absence of menstrual cycles presents a diagnostic challenge, as the traditional diagnostic criteria for endometriosis rely on menstrual patterns. Visual cues may be less evident, and the symptoms often overlap with other gynecological conditions, necessitating a thorough evaluation to differentiate postmenopausal endometriosis from other potential causes. Management approaches for postmenopausal endometriosis encompass surgical intervention, hormonal therapies, pain management, and individualized care. Postmenopausal endometriosis significantly impacts the quality of life, sexual health, and long-term well-being of women. Understanding the clinical features, diagnostic challenges, and management approaches of postmenopausal endometriosis is crucial for healthcare professionals to provide effective care and to improve the quality of life of women affected by this condition.
Collapse
Affiliation(s)
- Alexandre Vallée
- From the Department of Epidemiology and Public Health, Foch Hospital, Suresnes, France
| | | | | | - Anis Feki
- Department of Gynecology and Obstetrics, University Hospital of Fribourg, Fribourg, Switzerland
| | | |
Collapse
|
31
|
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
|
32
|
Moon S, Yun B, Lee M, Seok E, Ha J, Yang H. Gonadotropins Regulate the mRNA Expression of Gonadotropin-Releasing
Hormone and Its Receptors in the Mouse Ovary and Uterus. Dev Reprod 2024; 28:1-12. [PMID: 38654976 PMCID: PMC11034991 DOI: 10.12717/dr.2024.28.1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/31/2024] [Accepted: 02/27/2024] [Indexed: 04/26/2024]
Abstract
Gonadotropin-releasing hormone (GnRH), a critical hormone produced in the hypothalamus, is essential for regulating reproductive processes. It has also been demonstrated the presence of GnRH and its receptors (GnRHR) in ovarian and uterine tissues, but little was known about the regulation mechanism of their expression in these organs and ovarian aging. Therefore, the aim of this study was to investigate the expression of GnRHR in the ovary and uterus of mice, particularly after high-dose gonadotropin treatments and in relation to aging. Quantitative real-time-PCR (qRT-PCR) revealed that pituitary gland had the highest GnRHR expression in both young and aged mice. In addition, liver expression was higher in young mice, whereas thymus expression was higher in aged mice. GnRHR mRNA was present in the ovaries of both young and aged mice but nearly undetectable in the uterus of aged mice. We next examined the expression of GnRHR in the ovary and uterus in response to high-dose administration of pregnant mare serum gonadotropin (PMSG). After PMSG administration, GnRH mRNA levels were significantly decreased in the ovary but increased in the uterus. The expression of GnRH mRNA in these organs showed opposite trends to that of GnRHR expression. These results suggest the involvement of GnRH in age-related reproductive decline and the potential effects of high-dose gonadotropin treatments on reproductive organ function.
Collapse
Affiliation(s)
- Soeun Moon
- Department of Bioenvironmental
Technology, College of Sciences Technology Convergence, Seoul Women’s
University, Seoul 01797, Korea
| | - Bokyeong Yun
- Department of Bioenvironmental
Technology, College of Sciences Technology Convergence, Seoul Women’s
University, Seoul 01797, Korea
| | - Minju Lee
- Department of Bioenvironmental
Technology, College of Sciences Technology Convergence, Seoul Women’s
University, Seoul 01797, Korea
| | - Eunji Seok
- Department of Biohealth Convergence,
College of Sciences Technology Convergence, Seoul Women’s
University, Seoul 01797, Korea
| | - Jinah Ha
- Department of Biohealth Convergence,
College of Sciences Technology Convergence, Seoul Women’s
University, Seoul 01797, Korea
| | - Hyunwon Yang
- Department of Biohealth Convergence,
College of Sciences Technology Convergence, Seoul Women’s
University, Seoul 01797, Korea
| |
Collapse
|
33
|
Axiak CJ, Pleven A, Attard R, Borg Carbott F, Ebejer JP, Brincat I, Cassar K, Gruppetta M, Vassallo J, Bezzina Wettinger S, Farrugia R. High Population Frequency of GNRHR p.Q106R in Malta: An Evaluation of Fertility and Hormone Profiles in Heterozygotes. J Endocr Soc 2024; 8:bvad172. [PMID: 38196663 PMCID: PMC10775685 DOI: 10.1210/jendso/bvad172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Indexed: 01/11/2024] Open
Abstract
Context The gonadotropin-releasing hormone receptor variant GNRHR p.Q106R (rs104893836) in homozygosity, compound heterozygosity, or single heterozygosity is often reported as the causative variant in idiopathic hypogonadotropic hypogonadism (IHH) patients with GnRH deficiency. Genotyping of a Maltese newborn cord-blood collection yielded a minor allele frequency (MAF) 10 times higher (MAF = 0.029; n = 493) than that of the global population (MAF = 0.003). Objective To determine whether GNRHR p.Q106R in heterozygosity influences profiles of endogenous hormones belonging to the hypothalamic-pituitary axis and the onset of puberty and fertility in adult men (n = 739) and women (n = 239). Design Setting and Participants Analysis of questionnaire data relating to puberty and fertility, genotyping of the GNRHR p.Q106R variant, and hormone profiling of a highly phenotyped Maltese adult cohort from the Maltese Acute Myocardial Infarction Study. Main Outcome and Results Out of 978 adults, 43 GNRHR p.Q106R heterozygotes (26 men and 17 women) were identified. Hormone levels and fertility for all heterozygotes are within normal parameters except for TSH, which was lower in men 50 years or older. Conclusion Hormone data and baseline fertility characteristics of GNRHR p.Q106R heterozygotes are comparable to those of homozygous wild-type individuals who have no reproductive problems. The heterozygous genotype alone does not impair the levels of investigated gonadotropins and sex steroid hormones or affect fertility. GNRHR p.Q106R heterozygotes who exhibit IHH characteristics must have at least another variant, probably in a different IHH gene, that drives pathogenicity. We also conclude that GNRHR p.Q106R is likely a founder variant due to its overrepresentation and prevalence in the island population of Malta.
Collapse
Affiliation(s)
- Clayton John Axiak
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, MSD 2080, Malta
| | - Adrian Pleven
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, MSD 2080, Malta
- Clinical Chemistry Section, Department of Pathology, Mater Dei Hospital, Msida, MSD 2080, Malta
| | - Ritienne Attard
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, MSD 2080, Malta
| | - Francesca Borg Carbott
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, MSD 2080, Malta
| | - Jean-Paul Ebejer
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, MSD 2080, Malta
| | - Ian Brincat
- Clinical Chemistry Section, Department of Pathology, Mater Dei Hospital, Msida, MSD 2080, Malta
| | - Karen Cassar
- Department of Medicine, Faculty of Medicine and Surgery, University of Malta, Msida, MSD 2080, Malta
| | - Mark Gruppetta
- Department of Medicine, Faculty of Medicine and Surgery, University of Malta, Msida, MSD 2080, Malta
- Division of Endocrinology and Diabetes, Department of Medicine, Mater Dei Hospital, Msida, MSD 2080, Malta
| | - Josanne Vassallo
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, MSD 2080, Malta
- Department of Medicine, Faculty of Medicine and Surgery, University of Malta, Msida, MSD 2080, Malta
- Division of Endocrinology and Diabetes, Department of Medicine, Mater Dei Hospital, Msida, MSD 2080, Malta
| | - Stephanie Bezzina Wettinger
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, MSD 2080, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, MSD 2080, Malta
| | - Rosienne Farrugia
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, MSD 2080, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, MSD 2080, Malta
| |
Collapse
|
34
|
Khatun M, Lundin K, Naillat F, Loog L, Saarela U, Tuuri T, Salumets A, Piltonen TT, Tapanainen JS. Induced Pluripotent Stem Cells as a Possible Approach for Exploring the Pathophysiology of Polycystic Ovary Syndrome (PCOS). Stem Cell Rev Rep 2024; 20:67-87. [PMID: 37768523 PMCID: PMC10799779 DOI: 10.1007/s12015-023-10627-w] [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] [Accepted: 09/05/2023] [Indexed: 09/29/2023]
Abstract
Polycystic ovary syndrome (PCOS) is the most prevalent endocrine condition among women with pleiotropic sequelae possessing reproductive, metabolic, and psychological characteristics. Although the exact origin of PCOS is elusive, it is known to be a complex multigenic disorder with a genetic, epigenetic, and environmental background. However, the pathogenesis of PCOS, and the role of genetic variants in increasing the risk of the condition, are still unknown due to the lack of an appropriate study model. Since the debut of induced pluripotent stem cell (iPSC) technology, the ability of reprogrammed somatic cells to self-renew and their potential for multidirectional differentiation have made them excellent tools to study different disease mechanisms. Recently, researchers have succeeded in establishing human in vitro PCOS disease models utilizing iPSC lines from heterogeneous PCOS patient groups (iPSCPCOS). The current review sets out to summarize, for the first time, our current knowledge of the implications and challenges of iPSC technology in comprehending PCOS pathogenesis and tissue-specific disease mechanisms. Additionally, we suggest that the analysis of polygenic risk prediction based on genome-wide association studies (GWAS) could, theoretically, be utilized when creating iPSC lines as an additional research tool to identify women who are genetically susceptible to PCOS. Taken together, iPSCPCOS may provide a new paradigm for the exploration of PCOS tissue-specific disease mechanisms.
Collapse
Affiliation(s)
- Masuma Khatun
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki University Central Hospital, Haartmaninkatu 8, Helsinki, 00029 HUS, Finland.
| | - Karolina Lundin
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki University Central Hospital, Haartmaninkatu 8, Helsinki, 00029 HUS, Finland
| | - Florence Naillat
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Liisa Loog
- Institute of Genomics, University of Tartu, Tartu, 51010, Estonia
- Department of Genetics, University of Cambridge, Cambridge, CB2 3EH, UK
| | - Ulla Saarela
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Timo Tuuri
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki University Central Hospital, Haartmaninkatu 8, Helsinki, 00029 HUS, Finland
| | - Andres Salumets
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, 50406, Estonia
- Competence Centre of Health Technologies, Tartu, 50411, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, Huddinge, Stockholm, 14186, Sweden
| | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Juha S Tapanainen
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki University Central Hospital, Haartmaninkatu 8, Helsinki, 00029 HUS, Finland
- Department of Obstetrics and Gynecology, HFR - Cantonal Hospital of Fribourg and University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
35
|
Bhattacharya K, Dey R, Sen D, Paul N, Basak AK, Purkait MP, Shukla N, Chaudhuri GR, Bhattacharya A, Maiti R, Adhikary K, Chatterjee P, Karak P, Syamal AK. Polycystic ovary syndrome and its management: In view of oxidative stress. Biomol Concepts 2024; 15:bmc-2022-0038. [PMID: 38242137 DOI: 10.1515/bmc-2022-0038] [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: 10/30/2023] [Accepted: 12/11/2023] [Indexed: 01/21/2024] Open
Abstract
In the past two decades, oxidative stress (OS) has drawn a lot of interest due to the revelation that individuals with many persistent disorders including diabetes, polycystic ovarian syndrome (PCOS), cardiovascular, and other disorders often have aberrant oxidation statuses. OS has a close interplay with PCOS features such as insulin resistance, hyperandrogenism, and chronic inflammation; there is a belief that OS might contribute to the development of PCOS. PCOS is currently recognized as not only one of the most prevalent endocrine disorders but also a significant contributor to female infertility, affecting a considerable proportion of women globally. Therefore, the understanding of the relationship between OS and PCOS is crucial to the development of therapeutic and preventive strategies for PCOS. Moreover, the mechanistic study of intracellular reactive oxygen species/ reactive nitrogen species formation and its possible interaction with women's reproductive health is required, which includes complex enzymatic and non-enzymatic antioxidant systems. Apart from that, our current review includes possible regulation of the pathogenesis of OS. A change in lifestyle, including physical activity, various supplements that boost antioxidant levels, particularly vitamins, and the usage of medicinal herbs, is thought to be the best way to combat this occurrence of OS and improve the pathophysiologic conditions associated with PCOS.
Collapse
Affiliation(s)
- Koushik Bhattacharya
- School of Paramedics and Allied Health Sciences, Centurion University of Technology and Management, Khurda Road, Bhubaneswar, Odisha, India
| | - Rajen Dey
- Department of Medical Laboratory Technology, Swami Vivekananda University, Barrackpore, West Bengal, India
| | - Debanjana Sen
- Post-Graduate Department of Physiology, Hooghly Mohsin College, Chinsurah, West-Bengal, India
| | - Nimisha Paul
- Department of General Human Physiology and Biochemistry, Hitkarini Dental College and Hospital, Jabalpur, Madhya Pradesh, India
| | - Asim Kumar Basak
- School of Allied Health Sciences, Brainware University, Barasat, West-Bengal, India
| | | | - Nandini Shukla
- Department of Anatomy, Pt. J.N.M. Medical College, Raipur, Chhattisgarh, India
| | - Gargi Ray Chaudhuri
- Department of Physiotherapy, Nopany Institute of Health Care Studies, Kolkata, West-Bengal, India
| | - Aniruddha Bhattacharya
- Department of Physiology, International Medical School, Management and Science University, Selangor, Malaysia
| | - Rajkumar Maiti
- Department of Physiology, Bankura Christian College, Bankura, West Bengal, India
| | - Krishnendu Adhikary
- Department of Interdisciplinary Science, Centurion University of Technology and Management, Khurda Road, Bhubaneswar, Odisha, India
| | - Prity Chatterjee
- Department of Biotechnology, Paramedical College, Durgapur, West Bengal, India
| | - Prithviraj Karak
- Department of Physiology, Bankura Christian College, Bankura, West Bengal, India
| | - Alak Kumar Syamal
- Post-Graduate Department of Physiology, Hooghly Mohsin College, Chinsurah, West-Bengal, India
| |
Collapse
|
36
|
Wang J, Liu Z, Cao D, Liu J, Li F, Han H, Han H, Lei Q, Liu W, Li D, Wang J, Zhou Y. Elucidation of the genetic determination of clutch traits in Chinese local chickens of the Laiwu Black breed. BMC Genomics 2023; 24:686. [PMID: 37968610 PMCID: PMC10652520 DOI: 10.1186/s12864-023-09798-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/08/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND Egg laying rate (LR) is associated with a clutch, which is defined as consecutive days of oviposition. The clutch trait can be used as a selection indicator to improve egg production in poultry breeding. However, little is known about the genetic basis of clutch traits. In this study, our aim was to estimate genetic parameters and identify quantitative trait single nucleotide polymorphisms for clutch traits in 399 purebred Laiwu Black chickens (a native Chinese breed) using a genome-wide association study (GWAS). METHODS In this work, after estimating the genetic parameters of age at first egg, body weight at first egg, LR, longest clutch until 52 week of age, first week when the longest clutch starts, last week when the longest clutch ends, number of clutches, and longest number of days without egg-laying until 52 week of age, we identified single nucleotide polymorphisms (SNPs) and potential candidate genes associated with clutch traits in Laiwu Black chickens. The restricted maximum likelihood method was used to estimate genetic parameters of clutch pattern in 399 Laiwu Black hens, using the GCTA software. RESULTS The results showed that SNP-based heritability estimates of clutch traits ranged from 0.06 to 0.59. Genotyping data were obtained from whole genome re-sequencing data. After quality control, a total of 10,810,544 SNPs remained to be analyzed. The GWAS revealed that 421 significant SNPs responsible for clutch traits were scattered on chicken chromosomes 1-14, 17-19, 21-25, 28 and Z. Among the annotated genes, NELL2, SMYD9, SPTLC2, SMYD3 and PLCL1 were the most promising candidates for clutch traits in Laiwu Black chickens. CONCLUSION The findings of this research provide critical insight into the genetic basis of clutch traits. These results contribute to the identification of candidate genes and variants. Genes and SNPs potentially provide new avenues for further research and would help to establish a framework for new methods of genomic prediction, and increase the accuracy of estimated genetic merit for egg production and clutch traits.
Collapse
Affiliation(s)
- Jie Wang
- Poultry Breeding Engineering Technology Center of Shandong Province, Poultry Institute, Shandong Academy of Agricultural Sciences, Jinan, 250023, Shandong, China
| | - Zhansheng Liu
- Shandong Animal Husbandry General Station, Jinan, 250023, China
| | - Dingguo Cao
- Poultry Breeding Engineering Technology Center of Shandong Province, Poultry Institute, Shandong Academy of Agricultural Sciences, Jinan, 250023, Shandong, China
| | - Jie Liu
- Poultry Breeding Engineering Technology Center of Shandong Province, Poultry Institute, Shandong Academy of Agricultural Sciences, Jinan, 250023, Shandong, China
| | - Fuwei Li
- Poultry Breeding Engineering Technology Center of Shandong Province, Poultry Institute, Shandong Academy of Agricultural Sciences, Jinan, 250023, Shandong, China
| | - Heguo Han
- Lijin County Center for Animal Disease Control, Lijin, 257400, China
| | - Haixia Han
- Poultry Breeding Engineering Technology Center of Shandong Province, Poultry Institute, Shandong Academy of Agricultural Sciences, Jinan, 250023, Shandong, China
| | - Qiuxia Lei
- Poultry Breeding Engineering Technology Center of Shandong Province, Poultry Institute, Shandong Academy of Agricultural Sciences, Jinan, 250023, Shandong, China
| | - Wei Liu
- Poultry Breeding Engineering Technology Center of Shandong Province, Poultry Institute, Shandong Academy of Agricultural Sciences, Jinan, 250023, Shandong, China
| | - Dapeng Li
- Poultry Breeding Engineering Technology Center of Shandong Province, Poultry Institute, Shandong Academy of Agricultural Sciences, Jinan, 250023, Shandong, China
| | - Jianxia Wang
- Administrative Examination and Approval Service Bureau of Lijin County, Lijin, 257400, China
| | - Yan Zhou
- Poultry Breeding Engineering Technology Center of Shandong Province, Poultry Institute, Shandong Academy of Agricultural Sciences, Jinan, 250023, Shandong, China.
| |
Collapse
|
37
|
Fanis P, Neocleous V, Papapetrou I, Phylactou LA, Skordis N. Gonadotropin-Releasing Hormone Receptor (GnRHR) and Hypogonadotropic Hypogonadism. Int J Mol Sci 2023; 24:15965. [PMID: 37958948 PMCID: PMC10650312 DOI: 10.3390/ijms242115965] [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: 10/04/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
Human sexual and reproductive development is regulated by the hypothalamic-pituitary-gonadal (HPG) axis, which is primarily controlled by the gonadotropin-releasing hormone (GnRH) acting on its receptor (GnRHR). Dysregulation of the axis leads to conditions such as congenital hypogonadotropic hypogonadism (CHH) and delayed puberty. The pathophysiology of GnRHR makes it a potential target for treatments in several reproductive diseases and in congenital adrenal hyperplasia. GnRHR belongs to the G protein-coupled receptor family and its GnRH ligand, when bound, activates several complex and tissue-specific signaling pathways. In the pituitary gonadotrope cells, it triggers the G protein subunit dissociation and initiates a cascade of events that lead to the production and secretion of the luteinizing hormone (LH) and follicle-stimulating hormone (FSH) accompanied with the phospholipase C, inositol phosphate production, and protein kinase C activation. Pharmacologically, GnRHR can be modulated by synthetic analogues. Such analogues include the agonists, antagonists, and the pharmacoperones. The agonists stimulate the gonadotropin release and lead to receptor desensitization with prolonged use while the antagonists directly block the GnRHR and rapidly reduce the sex hormone production. Pharmacoperones include the most recent GnRHR therapeutic approaches that directly correct the misfolded GnRHRs, which are caused by genetic mutations and hold serious promise for CHH treatment. Understanding of the GnRHR's genomic and protein structure is crucial for the most appropriate assessing of the mutation impact. Such mutations in the GNRHR are linked to normosmic hypogonadotropic hypogonadism and lead to various clinical symptoms, including delayed puberty, infertility, and impaired sexual development. These mutations vary regarding their mode of inheritance and can be found in the homozygous, compound heterozygous, or in the digenic state. GnRHR expression extends beyond the pituitary gland, and is found in reproductive tissues such as ovaries, uterus, and prostate and non-reproductive tissues such as heart, muscles, liver and melanoma cells. This comprehensive review explores GnRHR's multifaceted role in human reproduction and its clinical implications for reproductive disorders.
Collapse
Affiliation(s)
- Pavlos Fanis
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (P.F.); (V.N.)
| | - Vassos Neocleous
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (P.F.); (V.N.)
| | - Irene Papapetrou
- School of Medicine, University of Nicosia, Nicosia 1678, Cyprus;
| | - Leonidas A. Phylactou
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (P.F.); (V.N.)
| | - Nicos Skordis
- School of Medicine, University of Nicosia, Nicosia 1678, Cyprus;
- Division of Paediatric Endocrinology, Paedi Center for Specialized Paediatrics, Nicosia 2024, Cyprus
| |
Collapse
|
38
|
Ndinguri M, Middleton L, Unrine J, Lui S, Rollins J, Nienaber E, Spease C, Williams A, Cormier L. Therapeutic dosing and targeting efficacy of Pt-Mal-LHRH towards triple negative breast cancer. PLoS One 2023; 18:e0287151. [PMID: 37816015 PMCID: PMC10564129 DOI: 10.1371/journal.pone.0287151] [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: 12/31/2022] [Accepted: 05/31/2023] [Indexed: 10/12/2023] Open
Abstract
OBJECTIVE Pt-Mal-LHRH is a newly synthesized chemotherapeutic agent that was designed to selectively target the luteinizing hormone-releasing hormone (LHRH) receptor expressed by triple negative breast cancer (TNBC). The aim of this study was to evaluate the therapeutic dosing, tumor reduction efficacy, and selective distribution of Pt-Mal-LHRH in-vivo. METHODS AND RESULTS LHRH tissue expression levels in-vivo were investigated using western blotting and LHRH was found to be increased in reproductive tissues (mammary, ovary, uterus). Further, Pt-Mal-LHRH was found to have increased TNBC tumor tissue platinum accumulation compared to carboplatin by inductively coupled plasma mass spectrometry analysis. The platinum family, compound carboplatin, was selected for comparison due to its similar chemical structure and molar equivalent doses were evaluated. Moreover, in-vivo distribution data indicated selective targeting of Pt-Mal-LHRH by enhanced reproductive tissue accumulation compared to carboplatin. Further, TNBC tumor growth was found to be significantly attenuated by Pt-Mal-LHRH compared to carboplatin in both the 4T1 and MDA-MB-231 tumor models. There was a significant reduction in tumor volume in the 4T1 tumor across Pt-Mal-LHRH doses (2.5-20 mg/kg/wk) and in the MDA-MB-231 tumor at the dose of 10 mg/kg/wk in models conducted by an independent contract testing laboratory. CONCLUSION Our data indicates Pt-Mal-LHRH is a targeting chemotherapeutic agent towards the LHRH receptor and reduces TNBC tumor growth in-vivo. This study supports drug conjugation design models using the LHRH hormone for chemotherapeutic delivery as Pt-Mal-LHRH was found to be a more selective and efficacious than carboplatin. Further examination of Pt-Mal-LHRH is warranted for its clinical use in TNBCs, along with, other reproductive cancers overexpressing the LHRH receptor.
Collapse
Affiliation(s)
- Margaret Ndinguri
- Department of Chemistry, Eastern Kentucky University, Richmond, Kentucky, United States of America
| | - Lisa Middleton
- Department of Biological Sciences, Eastern Kentucky University, Richmond, Kentucky, United States of America
| | - Jason Unrine
- Department of Plant and Soil Sciences, University of Kentucky, Lexington, Kentucky, United States of America
| | - Shu Lui
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Joseph Rollins
- Department of Biological Sciences, Eastern Kentucky University, Richmond, Kentucky, United States of America
| | - Emma Nienaber
- Department of Biological Sciences, Eastern Kentucky University, Richmond, Kentucky, United States of America
| | - Cassidy Spease
- Department of Biological Sciences, Eastern Kentucky University, Richmond, Kentucky, United States of America
| | - Aggie Williams
- Department of Biological Sciences, Eastern Kentucky University, Richmond, Kentucky, United States of America
| | - Lindsay Cormier
- Department of Biological Sciences, Eastern Kentucky University, Richmond, Kentucky, United States of America
| |
Collapse
|
39
|
He FF, Hu W, Yong L, Li YM. Triggering of ovulation for GnRH-antagonist cycles in normal and low ovarian responders undergoing IVF/ICSI: A systematic review and meta-analysis of randomized trials. Eur J Obstet Gynecol Reprod Biol 2023; 289:65-73. [PMID: 37639817 DOI: 10.1016/j.ejogrb.2023.08.014] [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: 02/24/2023] [Revised: 06/01/2023] [Accepted: 08/15/2023] [Indexed: 08/31/2023]
Abstract
OBJECTIVE To conduct a systematic review andmeta-analysis of all randomized controlled trials (RCTs) that investigated whether dual triggering [a combination of gonadotropin-releasing hormone (GnRH) agonist and human chorionic gonadotropin (hCG)] of final oocyte maturation can improve the number of oocytes retrieved and clinical pregnancy rate in low or normal responders undergoing in-vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) cycles using a GnRH-antagonist protocol. STUDY DESIGN Studies up to October 2022 were identified from PubMed, Scopus, Cochrane Library and Web of Science. The risk of bias of included studies was assessed. Dichotomous outcomes were reported as relative risks (RR), and continuous outcomes were reported as weighted mean differences (WMD) with 95% confidence intervals (CI). The primary outcomes were number of oocytes retrieved, number of mature [metaphase II (MII)] oocytes, clinical pregnancy rate and ongoing pregnancy rate; other IVF outcomes were considered as secondary outcomes. RESULTS Seven studies were identified, and 898 patients were eligible for inclusion in this meta-analysis. The results showed that the number of oocytes retrieved [WMD = 1.38 (95% CI 0.47-2.28), I2 = 66%, p = 0.003, low evidence], number of MII oocytes [WMD = 0.7 (95% CI 0.35-1.05), I2 = 42%, p < 0.0001, moderate evidence], number of embryos [WMD = 0.68 (95% CI 0.07-1.3), I2 = 67%, p = 0.03, low evidence] and number of good-quality embryos [WMD = 1.14 (95% CI 0.35-1.93), I2 = 0%, p = 0.005, moderate evidence] in the dual trigger group were significantly higher than in the hCG trigger group. The results of the ovarian response subgroup analysis showed significant differences in all of these outcomes in normal responders, and no differences in any of the outcomes in low responders, except for the number of MII oocytes. In low responders, clinical pregnancy rates may be improved in the dual trigger group [RR = 2.2 (95% CI 1.05-4.61), I2 = 28%, p = 0.04, low evidence]. CONCLUSION Dual triggering by GnRH agonist and hCG improved oocyte maturity and embryo grading for normal responders in GnRH-antagonist cycles. Dual triggering for final oocyte maturation may improve clinical pregnancy rates in low responders.
Collapse
Affiliation(s)
- Fang-Fang He
- Reproductive Center of Chengdu Jinjiang District Maternal and Child Health Hospital, Chengdu, People's Republic of China
| | - Wenhui Hu
- Reproductive Center of Chengdu Jinjiang District Maternal and Child Health Hospital, Chengdu, People's Republic of China
| | - Lin Yong
- Reproductive Center of Chengdu Jinjiang District Maternal and Child Health Hospital, Chengdu, People's Republic of China
| | - Yu-Mei Li
- Department of Assisted Reproduction, Xiangya Hospital, Central South University, Changsha, People's Republic of China.
| |
Collapse
|
40
|
Xu B, Hou Z, Liu N, Zhao J, Li Y. Pretreatment with a long-acting GnRH agonist for frozen-thawed embryo transfer cycles: how to improve live birth? J Ovarian Res 2023; 16:197. [PMID: 37743479 PMCID: PMC10518919 DOI: 10.1186/s13048-023-01277-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/12/2023] [Indexed: 09/26/2023] Open
Abstract
BACKGROUND Whether pretreatment with gonadotropin-releasing hormone agonist (GnRHa) can improve the pregnancy outcomes in frozen-thawed embryo transfer (FET) cycles is controversial. The inconsistencies in the results of different studies would be related to the characteristics of the included patients and the protocol of GnRHa use. In this study, we investigated the efficacy of pretreatment with a long-acting GnRH agonist in the early follicular phase of FET cycles and determined which population was suitable for the protocol. RESULTS We retrospectively included 630 and 1141 patients in the GnRHa FET and hormone replacement treatment (HRT) FET without GnRHa groups respectively, between October 2017 and March 2019 at a university-affiliated in vitro fertilization center. On the second or third day of menstruation, 3.75 mg of leuprorelin was administered. After 14 days, HRT was initiated for endometrial preparation. No significant differences were observed between the two groups in terms of patient characteristics. However, the GnRHa FET group showed a higher percentage of endometrium with a triple line pattern (94.8% vs 89.6%, p < 0.001) on the day of progesterone administration, with increased implantation (35.6% vs 29.8%, p = 0.005), clinical pregnancy (49.8% vs 43.3%, p = 0.008), and live birth rate (39.4% vs 33.7%, p = 0.016), than the HRT FET cycles with similar endometrial thickness, ectopic pregnancy and early miscarriage rates. Binary logistic regression analysis showed the GnRHa FET group to be associated with an increased chance of clinical pregnancy (P=0.028, odds ratio [OR] 1.32, 95% confidence interval [CI] 1.03-1.70) and live birth (P=0.013, odds ratio [OR] 1.34, 95% confidence interval [CI] 1.06-1.70) compared to the HRT FET without GnRHa group. After subgroup analysis, we found that the GnRHa FET group showed a significantly higher live birth rate in the subgroups of age < 40 years, primary infertility, with polycystic ovary syndrome (PCOS), and irregular menstruation. CONCLUSIONS Pretreatment with a long-acting GnRHa during the early follicular phase improved the live birth rate in FET cycles. Age < 40 years, primary infertility, PCOS, and irregular menstruation are effective indications for endometrial preparation with GnRHa pretreatment in FET cycles. However, further randomized controlled trials are required to verify these results.
Collapse
Affiliation(s)
- Bin Xu
- Reproductive Medicine Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha City, Hunan Province, People's Republic of China, 410008
- Clinical Research Center For Woman's Reproductive Health in Hunan Province, Changsha City, China
| | - Zhaojuan Hou
- Reproductive Medicine Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha City, Hunan Province, People's Republic of China, 410008
- Clinical Research Center For Woman's Reproductive Health in Hunan Province, Changsha City, China
| | - Nenghui Liu
- Reproductive Medicine Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha City, Hunan Province, People's Republic of China, 410008
- Clinical Research Center For Woman's Reproductive Health in Hunan Province, Changsha City, China
| | - Jing Zhao
- Reproductive Medicine Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha City, Hunan Province, People's Republic of China, 410008.
- Clinical Research Center For Woman's Reproductive Health in Hunan Province, Changsha City, China.
| | - Yanping Li
- Reproductive Medicine Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha City, Hunan Province, People's Republic of China, 410008.
- Clinical Research Center For Woman's Reproductive Health in Hunan Province, Changsha City, China.
| |
Collapse
|
41
|
Kumari R, Muneshwar KN, Pathade AG, Yelne S. Unveiling the Effects of Triptorelin on Endocrine Profiles: Insights From Healthy, Polycystic Ovary Syndrome, and Hypothalamic Amenorrhea Women. Cureus 2023; 15:e44752. [PMID: 37809244 PMCID: PMC10556375 DOI: 10.7759/cureus.44752] [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: 08/19/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Triptorelin, a synthetic gonadotropin-releasing hormone (GnRH) agonist, has garnered increasing attention for its profound effects on endocrine profiles across diverse populations. This review article explores triptorelin's impact on women's health by examining its effects on healthy individuals, those with polycystic ovary syndrome (PCOS), and those experiencing hypothalamic amenorrhea (HA). The mechanism of triptorelin involves a transient surge in gonadotropin release, followed by receptor desensitization, leading to downregulation of the hypothalamus-pituitary-gonadal (HPG) axis. In healthy women, triptorelin's controlled modulation of the HPG axis is a foundation for assisted reproduction techniques. In PCOS, it offers promise in restoring ovulatory function and mitigating hyperandrogenism. For HA individuals, triptorelin's potential to restore proper GnRH pulsatility emerges as a therapeutic avenue. This review emphasizes the importance of personalized approaches based on specific health conditions, highlighting triptorelin's versatility and potential applications beyond its current scope. As research progresses, triptorelin's role in endocrine management is poised to reshape women's health by optimizing hormonal equilibrium and overall well-being.
Collapse
Affiliation(s)
- Riya Kumari
- Community Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Komal N Muneshwar
- Community Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Aniket G Pathade
- Research and Development, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Seema Yelne
- Nursing, Shalinitai Meghe College of Nursing, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
42
|
Ciebiera M, Madueke-Laveaux OS, Feduniw S, Ulin M, Spaczyński R, Zgliczyńska M, Bączkowska M, Zarychta E, Łoziński T, Ali M, Al-Hendy A. GnRH agonists and antagonists in therapy of symptomatic uterine fibroids - current roles and future perspectives. Expert Opin Pharmacother 2023; 24:1799-1809. [PMID: 37602464 DOI: 10.1080/14656566.2023.2248890] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/12/2023] [Accepted: 08/14/2023] [Indexed: 08/22/2023]
Abstract
INTRODUCTION Uterine fibroids are the most common noncancerous tumors in women of childbearing age. This review was developed to evaluate the current role of gonadotropin-releasing hormone (GnRH) agonists and antagonists in the therapy of symptomatic uterine fibroids. AREAS COVERED There is a great need for alternative methods for surgical treatment of uterine fibroids. Hormonal therapy remains the first-line treatment option for most patients. GnRH analogs (agonists and antagonists) modulate the pulsatile release of GnRH. This review summarizes the available literature concerning pharmacologic principles underlying the mechanism of action of GnRH and its analogs, as well as individual therapeutic applications to which these drugs have been applied. EXPERT OPINION In many cases, it is possible to try to treat uterine fibroids pharmacologically. Both groups of GnRH analogs are used in therapy, agonists instead as a preparation for surgery, and antagonists as a drug for long-term use. It is essential to develop this path further and look for at least long-term-release systems or new methods of administering these drugs. It is also important from the patient's perspective to search for possible drugs that may have an additive effect of decreasing side effects when combined with GnRH analogs.
Collapse
Affiliation(s)
- Michał Ciebiera
- Second Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Warsaw Institute of Women's Health, Warsaw, Poland
- Development and Research Center of Non-Invasive Therapies, Pro-Familia Hospital, Rzeszów, Poland
| | | | - Stepan Feduniw
- Department of Gynecology, University Hospital Zurich, Zurich, Switzerland
| | - Mara Ulin
- Department of Obstetrics and Gynecology, Mount Sinai Hospital, Chicago, IL, USA
| | - Robert Spaczyński
- Obstetrics and Infertility Treatment Pastelova, Center for Gynecology, Poznan, Poland
| | - Magdalena Zgliczyńska
- Department of Obstetrics, Perinatology and Neonatology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Monika Bączkowska
- Second Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Elżbieta Zarychta
- Second Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Tomasz Łoziński
- Department of Obstetrics and Gynecology, Pro-Familia Hospital, Rzeszow, Poland
- Department of Gynecology and Obstetrics, College of Medical Sciences, Institute of Medical Sciences, University of Rzeszow, Rzeszow, Poland
| | - Mohamed Ali
- Department of Obstetrics and Gynecology, University of Chicago Medicine, Chicago, IL, USA
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago Medicine, Chicago, IL, USA
| |
Collapse
|
43
|
Concepción-Zavaleta MJ, Coronado-Arroyo JC, Quiroz-Aldave JE, Durand-Vásquez MDC, Ildefonso-Najarro SP, Rafael-Robles LDP, Concepción-Urteaga LA, Gamarra-Osorio ER, Suárez-Rojas J, Paz-Ibarra J. Endocrine factors associated with infertility in women: an updated review. Expert Rev Endocrinol Metab 2023; 18:399-417. [PMID: 37702309 DOI: 10.1080/17446651.2023.2256405] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/09/2023] [Accepted: 09/04/2023] [Indexed: 09/14/2023]
Abstract
INTRODUCTION Infertility is defined as the inability to conceive after unprotected sexual intercourse for at least 12 consecutive months. Our objective is to present an updated narrative review on the endocrine causes of infertility in women. AREAS COVERED A comprehensive review was conducted using Scielo, Scopus, and EMBASE databases, comprising 245 articles. The pathophysiology of infertility in women was described, including endocrinopathies such as hypothalamic amenorrhea, hyperprolactinemia, polycystic ovary syndrome, primary ovarian insufficiency, obesity, thyroid dysfunction, and adrenal disorders. The diagnostic approach was outlined, emphasizing the necessity of hormonal studies and ovarian response assessments. Additionally, the treatment plan was presented, commencing with non-pharmacological interventions, encompassing the adoption of a Mediterranean diet, vitamin supplementation, moderate exercise, and maintaining a healthy weight. Subsequently, pharmacological treatment was discussed, focusing on the management of associated endocrine disorders and ovulatory dysfunction. EXPERT OPINION This comprehensive review highlights the impact of endocrine disorders on fertility in women, providing diagnostic and therapeutic algorithms. Despite remaining knowledge gaps that hinder more effective treatments, ongoing research and advancements show promise for improved fertility success rates within the next five years. Enhanced comprehension of the pathophysiology behind endocrine causes and the progress in genetic research will facilitate the delivery of personalized treatments, thus enhancing fertility rates.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - José Paz-Ibarra
- Department of Medicine, School of Medicine, Universidad Nacional Mayor de San Marcos, Lima, Perú
| |
Collapse
|
44
|
Song Y, Qin C, Zhang C, Peng Y, Yang W, Du Y, Xu T. GNRH family genes contributed to gender-specific disparity of bladder cancer prognosis through exerting opposite regulatory roles between males and females. J Cancer Res Clin Oncol 2023; 149:6827-6840. [PMID: 36806614 DOI: 10.1007/s00432-023-04640-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 02/07/2023] [Indexed: 02/23/2023]
Abstract
PURPOSE There is gender-specific disparity in bladder cancer (BlCa) prognosis. Female BlCa patients present with more advanced tumor and have higher risks of disease recurrence, progression, and mortality than males. Since gonadotropin-releasing hormone (GNRH) family genes were critical genes in gender-related biological activity and could be detected in BlCa specimens, this study aimed to explore potential roles of GNRH1 and GNRHR in gender disparity of BlCa. METHODS RNA-sequencing data from The Cancer Genome Atlas Bladder Urothelial Carcinoma dataset, IMvigor210 immunotherapy cohort and Cancer Cell Line Encyclopedia database were used to compare potential roles of GNRH1 and GNRHR in males and females, respectively. Gene set enrichment analysis was used to analyze the biological functions. RESULTS Males with higher GNRH1 and GNRHR have better overall survival (P < 0.05, HR < 1), while females with higher expression have a trend toward worse overall survival (P < 0.05, HR > 1). Gene set enrichment analysis identified GNRH1 and GNRHR exert opposite regulatory roles in myogenesis (M5909), interferon-α response (M5911), interferon-γ response (M5913), inflammatory response (M5932) and TNF-α signaling via NF-κβ (M5890) between males and females. The five functions are up-regulated in females (NES > 0), while down-regulated in males (NES < 0). GNRH1 in females was positively correlated with CD3D (R-value > 0 and P < 0.05), while GNRHR in males was negatively correlated with CD247, CD3D and CD3E (R-value < 0 and P < 0.05). CONCLUSION GNRH1 and GNRHR have opposite effects on overall survival in different genders, and exert opposite roles in immune-related functions between different genders, which could emerge as a contributor to gender disparity of BlCa prognosis.
Collapse
Affiliation(s)
- Yuxuan Song
- Department of Urology, Peking University People's Hospital, Beijing, 100044, China
| | - Caipeng Qin
- Department of Urology, Peking University People's Hospital, Beijing, 100044, China
| | - Chunlong Zhang
- Department of Urology, Peking University People's Hospital, Beijing, 100044, China
| | - Yun Peng
- Department of Urology, Peking University People's Hospital, Beijing, 100044, China
| | - Wenbo Yang
- Department of Urology, Peking University People's Hospital, Beijing, 100044, China
| | - Yiqing Du
- Department of Urology, Peking University People's Hospital, Beijing, 100044, China.
| | - Tao Xu
- Department of Urology, Peking University People's Hospital, Beijing, 100044, China.
| |
Collapse
|
45
|
Liu Y, Huang K, Chen C, Wen L, Lei M, Guo Y, Tang B. Effect of luteal-phase GnRH agonist on frozen-thawed embryo transfer during artificial cycles: a randomised clinical pilot study. Front Endocrinol (Lausanne) 2023; 14:1098576. [PMID: 37361538 PMCID: PMC10289255 DOI: 10.3389/fendo.2023.1098576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 05/15/2023] [Indexed: 06/28/2023] Open
Abstract
Purpose This randomised clinical pilot study evaluated the effect of the mid-luteal additional single dose of gonadotropin-releasing hormone agonist (GnRH-a) on the clinical outcome of the females subjected to artificial cycle frozen-thawed embryo transfer (AC-FET). Methods A total of 129 females were randomised into two groups (70 in the control group and 59 in the intervention group). Both groups received standard luteal support. The intervention group was given an extra dose of 0.1 mg GnRH-a in the luteal phase. The live birth rate served as the primary endpoint. The secondary endpoints were the positivity of pregnancy tests, the clinical pregnancy rate, the miscarriage rate, the implantation rate, and the multiple pregnancy rate. Results There were more positive pregnancy tests, clinical pregnancies, live births, and twinning pregnancies, and fewer miscarriages observed in the intervention arm compared to the controls, though no statistical significance was concluded. No difference was found in the number of macrosomia in the two groups. There was no congenital abnormality newborn. Conclusion Overall, the difference of 12.1 percentage points in the live births rate (40.7% vs 28.6%) between the two groups, however, is statistically insignificant. the improvement of the pregnancy outcome supports the non-inferiority of GnRH-a added during the luteal phase in AC-FET. Larger-scale clinical trials are required to further establish the positive benefits.
Collapse
Affiliation(s)
- Yanghong Liu
- Reproductive Medicine Center, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Kaishu Huang
- Reproductive Medicine Center, The First People’s Hospital of Changde City, Changde, Hunan, China
| | - Cheng Chen
- Reproductive Medicine Center, The First People’s Hospital of Changde City, Changde, Hunan, China
| | - Li Wen
- Reproductive Medicine Center, The First People’s Hospital of Changde City, Changde, Hunan, China
| | - Min Lei
- Reproductive Medicine Center, The First People’s Hospital of Changde City, Changde, Hunan, China
| | - Yabin Guo
- Reproductive Medicine Center, The First People’s Hospital of Changde City, Changde, Hunan, China
| | - Bin Tang
- Reproductive Medicine Center, The First People’s Hospital of Changde City, Changde, Hunan, China
| |
Collapse
|
46
|
Day JR, Flanagan CL, David A, Hartigan-O'Connor DJ, Garcia de Mattos Barbosa M, Martinez ML, Lee C, Barnes J, Farkash E, Zelinski M, Tarantal A, Cascalho M, Shikanov A. Encapsulated Allografts Preclude Host Sensitization and Promote Ovarian Endocrine Function in Ovariectomized Young Rhesus Monkeys and Sensitized Mice. Bioengineering (Basel) 2023; 10:bioengineering10050550. [PMID: 37237620 DOI: 10.3390/bioengineering10050550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 05/28/2023] Open
Abstract
Transplantation of allogeneic donor ovarian tissue holds great potential for female cancer survivors who often experience premature ovarian insufficiency. To avoid complications associated with immune suppression and to protect transplanted ovarian allografts from immune-mediated injury, we have developed an immunoisolating hydrogel-based capsule that supports the function of ovarian allografts without triggering an immune response. Encapsulated ovarian allografts implanted in naïve ovariectomized BALB/c mice responded to the circulating gonadotropins and maintained function for 4 months, as evident by regular estrous cycles and the presence of antral follicles in the retrieved grafts. In contrast to non-encapsulated controls, repeated implantations of encapsulated mouse ovarian allografts did not sensitize naïve BALB/c mice, which was confirmed with undetectable levels of alloantibodies. Further, encapsulated allografts implanted in hosts previously sensitized by the implantation of non-encapsulated allografts restored estrous cycles similarly to our results in naïve recipients. Next, we tested the translational potential and efficiency of the immune-isolating capsule in a rhesus monkey model by implanting encapsulated ovarian auto- and allografts in young ovariectomized animals. The encapsulated ovarian grafts survived and restored basal levels of urinary estrone conjugate and pregnanediol 3-glucuronide during the 4- and 5-month observation periods. We demonstrate, for the first time, that encapsulated ovarian allografts functioned for months in young rhesus monkeys and sensitized mice, while the immunoisolating capsule prevented sensitization and protected the allograft from rejection.
Collapse
Affiliation(s)
- James R Day
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Colleen L Flanagan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anu David
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dennis J Hartigan-O'Connor
- Department of Medical Microbiology and Immunology, University of California, Davis, CA 95616, USA
- California National Primate Research Center, University of California, Davis, CA 95616, USA
| | | | - Michele L Martinez
- California National Primate Research Center, University of California, Davis, CA 95616, USA
- Department of Pediatrics, University of California, Davis, CA 95616, USA
| | - Charles Lee
- California National Primate Research Center, University of California, Davis, CA 95616, USA
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA 95616, USA
| | - Jenna Barnes
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Evan Farkash
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mary Zelinski
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR 97006, USA
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Alice Tarantal
- California National Primate Research Center, University of California, Davis, CA 95616, USA
- Department of Pediatrics, University of California, Davis, CA 95616, USA
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA 95616, USA
| | - Marilia Cascalho
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ariella Shikanov
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Macromolecular Science & Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
47
|
Baby M, Ilkowitz J, Cheema Brar P. Impacts of the COVID-19 pandemic on the diagnosis of idiopathic central precocious puberty in pediatric females in New York City. J Pediatr Endocrinol Metab 2023:jpem-2022-0628. [PMID: 37114861 DOI: 10.1515/jpem-2022-0628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/04/2023] [Indexed: 04/29/2023]
Abstract
BACKGROUND The COVID-19 pandemic had profound effect on physical and mental health. Stress was due physical inactivity, increased screen time, social isolation, fear of illness/death, as well as relative lack of resources including healthy food and finances. These stressors may be associated with an increase in idiopathic central precocious puberty (ICPP). OBJECTIVES To assess the incidence of ICPP in females during the COVID-19 pandemic and compare biochemical and radiological parameters of females diagnosed in the previous two years, looking at associations among BMI, screen time, isolation, and stress in relation to the development of early puberty. METHODS A retrospective chart review was performed of females diagnosed with ICPP. We divided subjects into a pandemic group and pre-pandemic group based on time of diagnosis. We compared anthropometric, serologic and radiologic data between the two groups. To assess psychosocial stress, we reviewed a COVID-19 impact survey which was administered to families at our endocrine clinic. RESULTS There were a total of 56 subjects in the study; 23 subjects in the pre-pandemic group and 33 in the pandemic group. The pandemic cohort had significantly higher estradiol and LH levels and larger ovarian volumes. Survey results showed parental report of stress was moderate in 38 % of subjects and severe in 25 % of parents. In children, reported stress was moderate in 46 % of subjects. CONCLUSIONS As puberty is influenced by exogenous factors including weight gain and psychosocial stress, we suspect that the environmental stress surrounding the pandemic influenced the increase in ICPP.
Collapse
Affiliation(s)
- Merilyn Baby
- Pediatric Endocrine Fellow, Division of Pediatric Endocrinology and Diabetes, NYU, New York, USA
| | - Jeniece Ilkowitz
- Clinical Research Nurse, Division of Pediatric Endocrinology and Diabetes, NYU, New York, USA
| | - Preneet Cheema Brar
- Pediatric Endocrine Attending, Associate Professor, Division of Pediatric Endocrinology and Diabetes, NYU, New York, USA
| |
Collapse
|
48
|
Neblett MF, Stewart EA. Oral Gonadotropin-Releasing Hormone Antagonists for the Treatment of Uterine Leiomyomas. Obstet Gynecol 2023; 141:901-910. [PMID: 37103532 DOI: 10.1097/aog.0000000000005145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/27/2022] [Indexed: 04/28/2023]
Abstract
Uterine leiomyomas are common hormone-responsive neoplasms that frequently cause heavy menstrual bleeding, anemia, pelvic pressure, pain, and adverse reproductive outcomes. In this overview, the efficacy and safety of oral gonadotropin-releasing hormone (GnRH) antagonists, co-administered with menopausal replacement-level steroid hormones or used at doses to avoid complete hypothalamic suppression, are reviewed for the management of uterine leiomyomas. Oral GnRH antagonists provide rapid suppression of sex steroids and avoid the initial steroidal flare and resultant temporary worsening of symptoms typically seen with parenteral GnRH agonists. Oral GnRH antagonists are effective in reducing leiomyoma-associated heavy menstrual bleeding, with high rates of amenorrhea and improved anemia and leiomyoma-associated pain, and providing modest reduction in uterine volume when used in combination with menopausal replacement-level steroid hormones. This add-back therapy can reduce hypogonadal side effects, including hot flushes and bone mineral density loss, close to levels seen with placebo therapy. Currently, both elagolix 300 mg twice daily with once-daily estradiol (1 mg) and norethindrone (0.5 mg) and relugolix 40 mg once daily with estradiol (1 mg) and norethindrone (0.5 mg) combination therapy are approved for leiomyoma treatment by the U.S. Food and Drug Administration. Linzagolix is under investigation in the United States but approved at two does with and without steroid hormones in the European Union. The efficacy of these agents appears to be robust over a wide spectrum of clinical presentations, demonstrating that worse disease parameters at baseline do not appear to inhibit efficacy. Across clinical trials, participants largely reflected the population of individuals affected by uterine leiomyomas.
Collapse
Affiliation(s)
- Michael F Neblett
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, the Department of Physiology and Biomedical Engineering, the Division of Endocrinology, Department of Medicine, the Department of Surgery, and the Women's Health Research Center, Mayo Clinic, and Mayo Clinic Alix School of Medicine, Rochester, Minnesota
| | | |
Collapse
|
49
|
Buhbut E, Nabulsi R, Avigdor G, Ben-Ami I. Comparison of pregnancy rates in antagonist cycles after luteal support with GnRH-agonist versus progesterone: prospective randomized study. Arch Gynecol Obstet 2023; 308:255-263. [PMID: 37186265 DOI: 10.1007/s00404-023-07017-5] [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: 12/22/2022] [Accepted: 03/20/2023] [Indexed: 05/17/2023]
Abstract
PURPOSE To compare pregnancy rates in GnRH-antagonist cycles triggered with hCG after luteal phase support with intranasal GnRH-agonist as sole luteal phase support versus standard vaginal progesterone preparation. METHODS Prospective randomized controlled study of patients who underwent antagonist-based IVF cycles triggered with hCG at university-affiliated tertiary medical center between 2020 and 2022. Patients meeting the inclusion criteria were randomly assigned to either intranasal GnRH-agonist or vaginal progesterone for luteal phase support. Pregnancy rates were the main outcome compared between the two study groups. RESULTS A total of 150 patients underwent 164 cycles, 127 cycles of which were included in the study cohort. Of them, 64 (50.4%) and 63 (49.6%) cycles were treated with GnRH-agonist or progesterone, respectively, as sole luteal phase support. A significantly higher pregnancy rate was demonstrated in the GnRH-agonist group compared with the progesterone group. After adjustment of several potential confounders such as age, body mass index, past obstetric history, number of IVF cycles, oocyte retrieved and embryos transferred, GnRH-agonist was still associated with a higher pregnancy rate (odds ratio 3.4, 95% confidence interval 1.4-8.3). Ovarian hyperstimulation syndrome rates were similar between the groups. CONCLUSIONS This prospective study suggests that nasal GnRH-agonist for luteal phase support is associated with higher pregnancy rates compared with standard progesterone support in an antagonist-based protocol triggered with hCG, while maintaining a similar safety profile. TRIAL REGISTRATION Clinicaltrials.gov NCT05484193. Date of registration: August 02 2022. The trial was retrospectively registered.
Collapse
Affiliation(s)
- Eadit Buhbut
- IVF and Infertility Unit, Department of Obstetrics and Gynecology, Shaare Zedek Medical Center, Jerusalem, affiliated with the Faculty of Medicine, Hebrew University of Jerusalem, Shmu'el Bait St 12, 9103102, Jerusalem, Israel.
| | - Rinad Nabulsi
- IVF and Infertility Unit, Department of Obstetrics and Gynecology, Shaare Zedek Medical Center, Jerusalem, affiliated with the Faculty of Medicine, Hebrew University of Jerusalem, Shmu'el Bait St 12, 9103102, Jerusalem, Israel
| | - Gilad Avigdor
- IVF and Infertility Unit, Department of Obstetrics and Gynecology, Shaare Zedek Medical Center, Jerusalem, affiliated with the Faculty of Medicine, Hebrew University of Jerusalem, Shmu'el Bait St 12, 9103102, Jerusalem, Israel
| | - Ido Ben-Ami
- IVF and Infertility Unit, Department of Obstetrics and Gynecology, Shaare Zedek Medical Center, Jerusalem, affiliated with the Faculty of Medicine, Hebrew University of Jerusalem, Shmu'el Bait St 12, 9103102, Jerusalem, Israel
| |
Collapse
|
50
|
Casati L, Ciceri S, Maggi R, Bottai D. Physiological and Pharmacological overview of the Gonadotropin Releasing Hormone. Biochem Pharmacol 2023; 212:115553. [PMID: 37075816 DOI: 10.1016/j.bcp.2023.115553] [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: 02/22/2023] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 04/21/2023]
Abstract
Gonadotropin-releasing Hormone (GnRH) is a decapeptide responsible for the control of the reproductive functions. It shows C- and N-terminal aminoacid modifications and two other distinct isoforms have been so far identified. The biological effects of GnRH are mediated by binding to high-affinity G-protein couple receptors (GnRHR), showing characteristic very short C tail. In mammals, including humans, GnRH-producing neurons originate in the embryonic nasal compartment and during early embryogenesis they undergo rapid migration towards the hypothalamus; the increasing knowledge of such mechanisms improved diagnostic and therapeutic approaches to infertility. The pharmacological use of GnRH, or its synthetic peptide and non-peptide agonists or antagonists, provides a valid tool for reproductive disorders and assisted reproduction technology (ART). The presence of GnRHR in several organs and tissues indicates additional functions of the peptide. The identification of a GnRH/GnRHR system in the human endometrium, ovary, and prostate has extended the functions of the peptide to the physiology and tumor transformation of such tissues. Likely, the activity of a GnRH/GnRHR system at the level of the hippocampus, as well as its decreased expression in mice brain aging, raised interest in its possible involvement in neurogenesis and neuronal functions. In conclusion, GnRH/GnRHR appears to be a fascinating biological system that exerts several possibly integrated pleiotropic actions in the complex control of reproductive functions, tumor growth, neurogenesis, and neuroprotection. This review aims to provide an overview of the physiology of GnRH and the pharmacological applications of its synthetic analogs in the management of reproductive and non-reproductive diseases.
Collapse
Affiliation(s)
- Lavinia Casati
- Department of Health Sciences, Università degli Studi di Milano, Milano, Italy
| | - Samuele Ciceri
- Dept. of Pharmaceutical Sciences (DISFARM), Università degli Studi di Milano, Milano Italy
| | - Roberto Maggi
- Dept. of Pharmaceutical Sciences (DISFARM), Università degli Studi di Milano, Milano Italy.
| | - Daniele Bottai
- Dept. of Pharmaceutical Sciences (DISFARM), Università degli Studi di Milano, Milano Italy
| |
Collapse
|