Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31.149
Filter
1.
Reprod Domest Anim ; 59(9): e14722, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39295165

ABSTRACT

The objective of the study was to characterise the expression patterns of the two key components of cortisol action namely HSD11B1 (11-beta-hydroxysteroid dehydrogenase type 1) and NR3C1 (nuclear receptor subfamily 3, group C, member 1, also known as the glucocorticoid receptor) in superovulation induced bovine follicles during the periovulation and subsequent corpus luteum (CL) formation. Bovine ovaries containing preovulatory follicles or CL were timely defined during induced ovulation as follows: 0 h before GnRH (Gonadotropin-releasing hormone) application, and 4, 10, 20, 25 (follicles) and 60 h (early CL) after GnRH. The low mRNA expression of HSD11B1 and NR3C1 in the follicle group before the GnRH application increased significantly in the follicle group 20 h after GnRH and remained high afterward also in the early CL group. In contrast, the high NR3C1 mRNA decreased in follicles 25 h after GnRH (close to ovulation) and significantly increased again after ovulation (early CL). Our results indicated the involvement of HSD11B1 and NR3C1 as the two key components of cortisol action in the local mechanisms coordinating final follicle maturation, ovulation, follicular-luteal transition and CL development in the cow.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1 , Corpus Luteum , Gonadotropin-Releasing Hormone , Ovarian Follicle , Receptors, Glucocorticoid , Animals , Female , Cattle/physiology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , Gonadotropin-Releasing Hormone/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics , Ovulation Induction/veterinary , Ovulation/physiology , Gene Expression Regulation
2.
N Z Med J ; 137(1603): 79-88, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39326021

ABSTRACT

AIM: To investigate use of puberty-blocking hormones (gonadotropin-releasing hormone analogues [GnRHa]) for gender dysphoria in New Zealand. Specifically, to describe demographic characteristics and time trends in the prevalence and incidence of prescribing, and to calculate cumulative incidence (proportion) of first prescribing of GnRHa for gender dysphoria in order to make valid international comparisons. METHOD: The national Pharmaceutical Collection was used to identify all dispensing from 2006 to 2023 to those aged <18, by sex/gender and age. Cumulative incidence of first prescriptions between ages 12 and 17 (which largely excludes prescribing for other indications) was calculated and compared with the Netherlands and England and Wales. RESULTS: In New Zealand, prescription of GnRHa for gender dysphoria started around 2011; prevalence of use increased to 2014, then more steeply to 2022, followed by a decline. Incidence data show the decline started from 2021. New Zealand, compared to the Netherlands (which started prescribing in the 1990s), had 1.7 times the cumulative incidence of first prescriptions by 2018. Compared to England and Wales up to 2020, New Zealand had 3.5-6.9 times the cumulative incidence. CONCLUSION: The high rate of prescribing for probable gender dysphoria in New Zealand, and the decline after 2021, require further investigation.


Subject(s)
Gender Dysphoria , Gonadotropin-Releasing Hormone , Humans , New Zealand/epidemiology , Male , Gender Dysphoria/drug therapy , Gender Dysphoria/epidemiology , Adolescent , Female , Child , Gonadotropin-Releasing Hormone/agonists , Gonadotropin-Releasing Hormone/antagonists & inhibitors , England/epidemiology , Netherlands/epidemiology , Wales/epidemiology , Incidence , Puberty/drug effects , Practice Patterns, Physicians'/statistics & numerical data
3.
Endocrinology ; 165(10)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39248143

ABSTRACT

Luteinizing hormone (LH), a heterodimeric glycoprotein produced by pituitary gonadotrope cells, regulates gonadal function. Hypothalamic gonadotropin-releasing hormone (GnRH) stimulates LH synthesis and secretion. GnRH induces LHß subunit (Lhb) expression via the transcription factor, early growth response 1 (EGR1), acting on the Lhb promoter. In contrast, overexpression of zinc finger E-box binding homeobox 1 (ZEB1) represses LH production in mice, but the underlying mechanism was not previously elucidated. Here, we observed that ZEB1 inhibited GnRH-stimulated but not basal Lhb mRNA expression in homologous murine LßT2 cells. Moreover, ZEB1 blocked GnRH and/or EGR1 induction of murine Lhb but not human LHB promoter-reporter activity in these cells. Using chimeric reporters, we mapped the species-specific ZEB1 sensitivity to sequence differences, including in Z- and E-boxes, in the proximal Lhb/LHB promoters, immediately upstream of the transcription start sites. ZEB1 bound to the murine Lhb promoter with higher affinity than to the human LHB promoter in this region. To examine ZEB1's physiological role in LH synthesis, we characterized gonadotrope-specific Zeb1 knockout mice. Loss of ZEB1 in gonadotropes did not affect LH production or secretion. Collectively, the data suggest that ZEB1, when overexpressed, can inhibit GnRH/EGR1 induction of murine Lhb transcription but does not play a necessary role in LH synthesis in mice.


Subject(s)
Gonadotropin-Releasing Hormone , Luteinizing Hormone, beta Subunit , Luteinizing Hormone , Promoter Regions, Genetic , Zinc Finger E-box-Binding Homeobox 1 , Animals , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , Mice , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/genetics , Luteinizing Hormone, beta Subunit/genetics , Luteinizing Hormone, beta Subunit/metabolism , Luteinizing Hormone/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Humans , Transcription, Genetic , Mice, Knockout , Cell Line , Gene Expression Regulation , Male
4.
Open Vet J ; 14(8): 2079-2084, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39308740

ABSTRACT

Background: The outbreak of foot and mouth disease (FMD) in Indonesia induces reproductive disorders in dairy cows that lead to economic losses to smallholder dairy farms. Aim: The study was to assess the influence of FMD on reproductive traits and evaluate the effect of gonadotropin hormone-releasing hormone (GnRH) administrations on the reproductive performance in FMD-infected dairy cows. Methods: The study was conducted in Jemowo village, Taman Sari sub-district, Boyolali district, Central Java, Indonesia. A total of 155 cows were used to identify the reproductive disorders on FMD-infected dairy cows aged 2-10 years old. Cows were raised in similar conditions and fed diets. A single dose of 2 ml GnRH was injected intramuscularly into 96 ovarian disorder cows. Reproductive performance was measured by service per conception (S/C), conception rate (CR), and pregnancy rate (PR). A descriptive study was conducted to demonstrate the results. Results: The study showed that 61.9% of FMD-infected cows had reproductive disorders, whereby 53.5% ovarian hypofunction, 4.52% silent heat, 1.94% repeat breeder, 1.29% ovarian atrophy, and 0.65% endometritis. FMD-infected cows injected with GnRH had a 98% reproductive recovery rate. Moreover, the S/C, CR, and PR of cows injected with GnRH were 2.02%, 51%, and 85%. Conclusion: GnRH administrations enhanced the reproductive traits of FMD-infected dairy cows indicated by the improvement of CR and PR.


Subject(s)
Cattle Diseases , Foot-and-Mouth Disease , Gonadotropin-Releasing Hormone , Ovarian Diseases , Animals , Cattle , Female , Gonadotropin-Releasing Hormone/administration & dosage , Cattle Diseases/drug therapy , Indonesia , Ovarian Diseases/veterinary , Ovarian Diseases/drug therapy , Dairying , Pregnancy , Reproduction/drug effects
5.
Int J Hyperthermia ; 41(1): 2384471, 2024.
Article in English | MEDLINE | ID: mdl-39251216

ABSTRACT

OBJECTIVES: To observe the therapeutic efficacy of high-intensity focused ultrasound (HIFU) combined with different pharmacological treatments for adenomyosis. MATERIALS AND METHODS: A total of 126 patients with adenomyosis who underwent HIFU combined with pharmacological treatment were retrospectively reviewed. Patients were treated with either dienogest (DNG) (Group A, N = 38) or GnRH-a (Group B, N = 88) for three months after HIFU, and received levonorgestrel-releasing intrauterine systems (LNG-IUS) at the end of the third month. Visual Analog Scale (VAS) and Pictorial Blood Loss Assessment Chart (PBAC) scores were used for evaluating symptom improvement. RESULTS: After propensity score matching (1:2), 38 patients were included in Group A and 76 in Group B. All patients showed significant improvement in VAS and PBAC scores after HIFU, but the PBAC score of Group A was significantly higher than that of patients in Group B at 18 months [11.50 (1.00, 29.50) vs. 0.00 (0.00, 16.50), p < 0.01] and 24 months [4.00 (0.25, 27.75) vs. 0.00 (0.00, 12.75), p = 0.04] after HIFU. Furthermore, patients in Group B had a greater uterine volume reduction at 24 months after HIFU than that of patients in Group A [51.00 (27.00, 62.00) vs. 30.00 (17.00, 42.75, p = 0.02)]. However, the adverse effects in Group A were lower than those in Group B [7 (15.79) vs. 35 (46.05), p < 0.01]. No significant difference was observed in the recurrence rate between the two groups. CONCLUSIONS: HIFU combined with DNG and LNG-IUS is a safe and effective treatment for patients with adenomyosis.


Subject(s)
Adenomyosis , High-Intensity Focused Ultrasound Ablation , Humans , Female , Adenomyosis/therapy , Adenomyosis/drug therapy , Adenomyosis/surgery , High-Intensity Focused Ultrasound Ablation/methods , Adult , Middle Aged , Gonadotropin-Releasing Hormone/therapeutic use , Retrospective Studies , Nandrolone/analogs & derivatives , Nandrolone/therapeutic use , Nandrolone/pharmacology , Combined Modality Therapy/methods , Levonorgestrel/therapeutic use , Levonorgestrel/administration & dosage , Treatment Outcome
6.
Theriogenology ; 229: 202-213, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39217649

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.


Subject(s)
Gonadotropin-Releasing Hormone , Sexual Maturation , Animals , Female , Gonadotropin-Releasing Hormone/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Mice , Sexual Maturation/drug effects , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Polyamines/metabolism , Aging , Ovary/drug effects , Ovary/metabolism , Gene Expression Regulation/drug effects
7.
Trials ; 25(1): 578, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223633

ABSTRACT

BACKGROUND: Around 4% of women receive an endometrial cancer diagnosis before turning 40, mainly those without prior childbirth experience and a strong desire to preserve their ability to conceive. Consequently, for young patients diagnosed with atypical endometrial hyperplasia (AEH) or early endometrial carcinoma (EC), a fertility-preserving approach employing high-dose oral progesterone has been adopted. However, previous research has shown a notable relapse rate. Furthermore, the extended use of substantial oral progesterone doses may hinder ovarian function and raise the risk of weight gain, liver issues, blood clotting, and breast cancer. We previously assessed the clinical effectiveness and pregnancy outcomes of gonadotropin-releasing hormone agonist (GnRH-a) based re-treatment for women with EC and AEH who did not respond to oral progestin therapy but achieved favorable treatment results and reproductive outcomes. METHODS: This study will be an open-label, two-armed, randomized, investigator-initiated multicenter trial evaluating the combination of GnRH-a with the levonorgestrel-releasing intrauterine system or the combination of GnRH-a with an aromatase inhibitor (comprising a subcutaneous GnRH-a injection every 4 weeks and daily oral letrozole 2.5 mg). A total of 226 participants will be randomly allocated to one of the two treatment groups in a 1:1 ratio. The primary objective is to determine the effectiveness of GnRH-a-based re-treatment in achieving a complete response (CR) at 24 weeks for patients with AEH or EC. Secondary objectives include assessing the pregnancy rate 12 weeks after treatment, as well as post-treatment pregnancy outcomes and the rate of recurrence. ETHICS AND DISSEMINATION: The protocol received approval from the Institutional Review Board of Peking Union Medical College Hospital and from boards at five other institutions. The trial will adhere to the principles outlined in the World Medical Association's Declaration of Helsinki and follow Good Clinical Practice standards. The trial results will be disseminated through publication in a peer-reviewed journal. CONCLUSIONS: Prospective evidence supporting conservative treatment for EC and AEH is limited. There is a need for new approaches that can achieve higher CR rates with fewer side effects. This trial will assess the effectiveness of GnRH-a-based fertility-sparing treatment in obese women and recurrent patients, offering a promising alternative for patients with EC and AEH. TRIAL REGISTRATION NUMBER: Chinese Clinical Trial Registry ChiCTR2200067099. Registered on December 27, 2022.


Subject(s)
Endometrial Hyperplasia , Endometrial Neoplasms , Fertility Preservation , Gonadotropin-Releasing Hormone , Levonorgestrel , Multicenter Studies as Topic , Randomized Controlled Trials as Topic , Humans , Female , Gonadotropin-Releasing Hormone/agonists , Endometrial Hyperplasia/drug therapy , Endometrial Hyperplasia/complications , Endometrial Neoplasms/drug therapy , Fertility Preservation/methods , Pregnancy , Levonorgestrel/administration & dosage , Levonorgestrel/adverse effects , Levonorgestrel/therapeutic use , Aromatase Inhibitors/therapeutic use , Aromatase Inhibitors/adverse effects , Aromatase Inhibitors/administration & dosage , Intrauterine Devices, Medicated , Treatment Outcome , Adult , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Agents, Hormonal/adverse effects , Antineoplastic Agents, Hormonal/administration & dosage , Letrozole/administration & dosage , Letrozole/therapeutic use , China , Pregnancy Rate
8.
Front Endocrinol (Lausanne) ; 15: 1461317, 2024.
Article in English | MEDLINE | ID: mdl-39229374

ABSTRACT

Introduction: This study compared, in high responders undergoing IVF treatment, GnRH agonist-only trigger and dual trigger on oocyte retrieval rate and cumulative live birth rate (LBR). The aim was to determine if the GnRH agonist-only triggers had provided outcomes comparable to dual trigger, while minimizing the risk of ovarian hyperstimulation syndrome (OHSS). Materials and methods: A retrospective, matched case-control study was conducted at Taichung Veterans General Hospital, Taiwan, including women who underwent IVF/ICSI between January 1, 2014, and December 31, 2022. Inclusion criteria were: GnRH antagonist protocol and estrogen level >3,000 pg/ml on trigger day. Exclusion criteria were: immune/metabolic diseases, donated oocytes, and mixed stimulation cycles. Propensity score matching was applied to balance age, AMH level, and oocyte number between the GnRH agonist-only and dual trigger groups. Outcomes were analyzed for patients who had complete treatment cycles, focusing on oocyte retrieval rate and cumulative LBR. Results: We analyzed 116 cycles in the agonist-only group, and 232 cycles in the dual trigger group. No inter-group difference was found in their age, BMI, and AMH levels. The dual trigger group had a higher oocyte retrieval rate (93% vs. 80%; p <0.05), while fertilization rates, blastocyst formation rates, and cumulative LBR were comparable. Notably, no OHSS cases had been reported in the GnRH agonist-only group, compared with 7 cases in the dual trigger group. Conclusion: GnRH agonist-only triggers resulted in a lower oocyte retrieval rate compared to dual triggers but did not significantly affect cumulative LBR in high responders. This approach effectively reduces OHSS risk without compromising pregnancy outcomes, making it a preferable option in freeze-all strategies, despite a longer oocyte pick-up duration and a medium cost. GnRH agonist-only trigger, however, may not be suitable for fresh embryo transfers or patients with low serum LH levels on trigger day.


Subject(s)
Birth Rate , Fertilization in Vitro , Gonadotropin-Releasing Hormone , Oocyte Retrieval , Ovarian Hyperstimulation Syndrome , Ovulation Induction , Humans , Female , Gonadotropin-Releasing Hormone/agonists , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Adult , Oocyte Retrieval/methods , Ovulation Induction/methods , Retrospective Studies , Pregnancy , Case-Control Studies , Fertilization in Vitro/methods , Ovarian Hyperstimulation Syndrome/prevention & control , Ovarian Hyperstimulation Syndrome/epidemiology , Live Birth/epidemiology , Pregnancy Rate , Fertility Agents, Female/therapeutic use , Fertility Agents, Female/administration & dosage , Taiwan/epidemiology , Sperm Injections, Intracytoplasmic/methods
9.
Medicine (Baltimore) ; 103(36): e39553, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39252290

ABSTRACT

To investigate the effects of pretreatment with long-acting gonadotropin-releasing hormone agonist (GnRH-a) before frozen-thawed embryo transfer (FET) on pregnancy outcomes in patients after minimal-mild (stages I-II) peritoneal endometriosis surgery. A retrospective cohort study was performed from March 2018 to May 2019. Overall, 274 patients met inclusion criteria of undergoing FET after minimal/mild peritoneal endometriosis surgery. For the FET protocol, patients were divided into 2 groups: GnRH-a plus hormone replacement therapy (HRT) (group A, n = 154) and HRT-only (group B, n = 120), with the former divided into 2 subgroups receiving 1 (group A1, n = 80) or 2 doses (group A2, n = 74) of GnRH-a. Basic characteristics and pregnancy outcomes of groups A and B and groups A1 and A2 were compared. Clinical pregnancy rate (CPR) and live birth rate (LBR) were the primary outcomes and logistic regression was used to analyze independent correlation factors. The CPR and LBR in group A were 58.4% and 50.0%, respectively, and were not significantly higher than in group B (49.2% and 40.0%; respectively, χ2 = 2.339, P = .126 and χ2 = 2.719, P = .099, respectively). CPR and LBR in group A1 were not significantly lower than those in group A2 (52.5% and 45.0% vs 64.9% and 55.4%, respectively; χ2 = 2.420, P = .120 and χ2 = 1.665, P = .197, respectively). However, group A2's CPR and LBR were significantly higher than group B's (64.9% and 55.4% vs 49.2% and 40.0%, respectively; χ2 = 4.560, P = .023 and χ2 = 4.375, P = .026, respectively). Logistic regression analysis showed that GnRH-a pretreatment (1 or 2 doses) had no significant effect on CPR and LBR compared with the HRT-only group. Patients with minimal-mild (stages I-II) peritoneal endometriosis surgery may not require GnRH-a pretreatment before FET.


Subject(s)
Embryo Transfer , Endometriosis , Gonadotropin-Releasing Hormone , Pregnancy Outcome , Humans , Female , Endometriosis/drug therapy , Endometriosis/surgery , Pregnancy , Retrospective Studies , Adult , Embryo Transfer/methods , Gonadotropin-Releasing Hormone/agonists , Pregnancy Rate , Hormone Replacement Therapy/methods , Peritoneal Diseases
11.
BMC Res Notes ; 17(1): 260, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39267194

ABSTRACT

This study hypothesized that SCFA, acetate impacts positively on hypothalamic pyroptosis and its related abnormalities in experimentally induced PCOS rat model, possibly through NrF2/HIF1-α modulation. Eight-week-old female Wister rats were divided into groups (n = 5), namely control, PCOS, acetate and PCOS + acetate groups. Induction of PCOS was performed by administering 1 mg/kg body weight of letrozole for 21 days. After PCOS confirmation, the animals were treated with 200 mg/kg of acetate for 6 weeks. Rats with PCOS were characterized with insulin resistance, leptin resistance, increased plasma testosterone as well as degenerated ovarian follicles. There was also a significant increase in hypothalamic triglyceride level, triglyceride-glucose index, inflammatory biomarkers (SDF-1 and NF-kB) and caspase-6 as well as plasma LH and triglyceride. A decrease was observed in plasma adiponectin, GnRH, FSH, and hypothalamic GABA with severe inflammasome expression in PCOS rats. These were accompanied by decreased level of NrF2/HIF1-α, and the alterations were reversed when treated with acetate. Collectively, the present results suggest the therapeutic impact of acetate on hypothalamic pyroptosis and its related comorbidity in PCOS, a beneficial effect that is accompanied by modulation of NrF2/HIF1-α.


Subject(s)
Hypothalamus , Hypoxia-Inducible Factor 1, alpha Subunit , Polycystic Ovary Syndrome , Pyroptosis , Rats, Wistar , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/pathology , Female , Animals , Hypothalamus/metabolism , Hypothalamus/drug effects , Hypothalamus/pathology , Pyroptosis/drug effects , Rats , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin Resistance , NF-E2-Related Factor 2/metabolism , Disease Models, Animal , Letrozole/pharmacology , Triglycerides/blood , Triglycerides/metabolism , Luteinizing Hormone/blood , Follicle Stimulating Hormone/blood , Adiponectin/metabolism , Adiponectin/blood , Testosterone/blood , Leptin/blood , Leptin/metabolism , Gonadotropin-Releasing Hormone/metabolism , gamma-Aminobutyric Acid/metabolism
12.
Molecules ; 29(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39274973

ABSTRACT

The gonadotropin-releasing hormone (GnRH) receptor (GnRH-R) is highly expressed in ovarian cancer cells (OCC), and it is an important molecular target for cancer therapeutics. To develop a new class of drugs targeting OCC, we designed and synthesized Con-3 and Con-7 which are novel high-affinity GnRH-R agonists, covalently coupled through a disulfide bond to the DNA synthesis inhibitor mitoxantrone. We hypothesized that Con-3 and Con-7 binding to the GnRH-R of OCC would expose the conjugated mitoxantrone to the cellular thioredoxin, which reduces the disulfide bond of Con-3 and Con-7. The subsequent release of mitoxantrone leads to its intracellular accumulation, thus exerting its cytotoxic effects. To test this hypothesis, we determined the cytotoxic effects of Con-3 and Con-7 using the SKOV-3 human OCC. Treatment with Con-3 and Con-7, but not with their unconjugated GnRH counterparts, resulted in the accumulation of mitoxantrone within the SKOV-3 cells, increased their apoptosis, and reduced their proliferation, in a dose- and time-dependent manner, with half-maximal inhibitory concentrations of 0.6-0.9 µM. It is concluded that Con-3 and Con-7 act as cytotoxic "prodrugs" in which mitoxantrone is delivered in a GnRH-R-specific manner and constitute a new class of lead compounds for use as anticancer drugs targeting ovarian tumors.


Subject(s)
Apoptosis , Cell Proliferation , Gonadotropin-Releasing Hormone , Mitoxantrone , Ovarian Neoplasms , Receptors, LHRH , Humans , Mitoxantrone/pharmacology , Mitoxantrone/chemistry , Female , Gonadotropin-Releasing Hormone/pharmacology , Gonadotropin-Releasing Hormone/chemistry , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Apoptosis/drug effects , Receptors, LHRH/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Survival/drug effects
13.
Reprod Fertil Dev ; 362024 Sep.
Article in English | MEDLINE | ID: mdl-39283977

ABSTRACT

Context Understanding of central nervous system mechanisms related to age-related infertility remains limited. Fibril α-synuclein, distinct from its monomer form, is implicated in age-related diseases and propagates among neurons akin to prions. Aims We compared α-synuclein expression in gonadotropin-releasing hormone-expressing neurons (GnRH neurons) in the pre-optic area, arcuate nucleus, and median eminence of healthy heifers and aged cows to determine its role in age-related infertility. Methods We analysed mRNA and protein expression, along with fluorescent immunohistochemistry for GnRH and α-synuclein, followed by Congo red staining to detect amyloid deposits, and confocal microscopy. Key results Both mRNA and protein expressions of α-synuclein were confirmed by reverse transcription-polymerase chain reaction (RT-PCR) and western blots in bovine cortex, hippocampus, and anterior and posterior hypothalamus tissues. Significant differences in α-synuclein mRNA expression were observed in the cortex and hippocampus between young and old cows. Western blots showed five bands of α-synuclein, probably reflecting monomer, dimer, and oligomers, in the cortex, hippocampus, hypothalamus tissues, and there were significant differences in some bands between young and old cows. Bright-field and polarised light microscopy did not detect obvious amyloid deposition in aged hypothalami; however, higher-sensitive confocal microscopy unveiled strong positive signal of Congo red and α-synuclein in GnRH neurons in aged hypothalami. Additionally, α-synuclein expression was detected in immortalised GnRH neurons, GT1-7 cells. Conclusion Alpha-synuclein was expressed in GnRH neurons, and some differences were observed between young and old hypothalami. Implications Alpha-synuclein may play an important role in aging-related infertility.


Subject(s)
Aging , Gonadotropin-Releasing Hormone , Hypothalamus , Neurons , alpha-Synuclein , Animals , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/genetics , Cattle , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Neurons/metabolism , Hypothalamus/metabolism , Female , Aging/metabolism , RNA, Messenger/metabolism
14.
Endocrinology ; 165(10)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39253941

ABSTRACT

Reproductive function in mammals depends on the ability of progesterone (P4) to suppress pulsatile gonadotrophin-releasing hormone (GnRH) and luteinizing hormone (LH) secretion in a homeostatic-negative feedback loop. Previous research identified that cells upstream from GnRH neurons expressing the nuclear progesterone receptor (PGR) are required for P4-negative feedback. However, the identity of these cells and the mechanism by which they reduce GnRH/LH pulsatile secretion is unknown. We aimed to address the hypothesis that PGR expressed by a neural population in the arcuate nucleus recently identified as the GnRH pulse generator, cells expressing kisspeptin, neurokinin B, and dynorphin (KNDy cells), mediate P4-negative feedback. To achieve this, we used female mice with the PGR gene conditionally deleted from kisspeptin cells (KPRKO mice) and observed a substantial decrease in the percentage of KNDy neurons coexpressing PGR messenger RNA (mRNA) (11% in KPRKO mice vs 86% in wild-type [WT] mice). However, KPRKO mice did not display changes in the frequency or amplitude of LH pulses in diestrus or estrus, nor in the ability of exogenous P4 to blunt a postcastration increase in LH. Further, mRNA expression of arcuate kisspeptin and dynorphin, which are excitatory and inhibitory to GnRH secretion, respectively, remained unaltered in KPRKO mice compared to WT controls. Together, these findings show that the near-complete loss of PGR signaling from KNDy cells does not affect negative feedback regulation of GnRH pulse generation in mice, suggesting that feedback through this receptor can occur via a small number of KNDy cells or a yet unidentified cell population.


Subject(s)
Arcuate Nucleus of Hypothalamus , Feedback, Physiological , Gonadotropin-Releasing Hormone , Kisspeptins , Luteinizing Hormone , Mice, Knockout , Progesterone , Receptors, Progesterone , Animals , Female , Kisspeptins/metabolism , Kisspeptins/genetics , Receptors, Progesterone/metabolism , Receptors, Progesterone/genetics , Luteinizing Hormone/metabolism , Mice , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/genetics , Arcuate Nucleus of Hypothalamus/metabolism , Progesterone/metabolism , Dynorphins/metabolism , Dynorphins/genetics , Neurons/metabolism , Neurokinin B/genetics , Neurokinin B/metabolism
15.
Endocrinology ; 165(10)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39254333

ABSTRACT

There has been an alarming trend toward earlier puberty in girls, suggesting the influence of an environmental factor(s). As the reactivation of the reproductive axis during puberty is thought to be mediated by the hypothalamic neuropeptides kisspeptin and gonadotropin-releasing hormone (GnRH), we asked whether an environmental compound might activate the kisspeptin (KISS1R) or GnRH receptor (GnRHR). We used GnRHR or KISS1R-expressing HEK293 cells to screen the Tox21 10K compound library, a compendium of pharmaceuticals and environmental compounds, for GnRHR and KISS1R activation. Agonists were identified using Ca2+ flux and phosphorylated extracellularly regulated kinase (p-ERK) detection assays. Follow-up studies included measurement of genes known to be upregulated upon receptor activation using relevant murine or human cell lines and molecular docking simulation. Musk ambrette was identified as a KISS1R agonist, and treatment with musk ambrette led to increased expression of Gnrh1 in murine and human hypothalamic cells and expansion of GnRH neuronal area in developing zebrafish larvae. Molecular docking demonstrated that musk ambrette interacts with the His309, Gln122, and Gln123 residues of the KISS1R. A group of cholinergic agonists with structures similar to methacholine was identified as GnRHR agonists. When applied to murine gonadotrope cells, these agonists upregulated Fos, Jun, and/or Egr1. Molecular docking revealed a potential interaction between GnRHR and 5 agonists, with Asn305 constituting the most conservative GnRHR binding site. In summary, using a Tox21 10K compound library screen combined with cellular, molecular, and structural biology techniques, we have identified novel environmental agents that may activate the human KISS1R or GnRHR.


Subject(s)
Receptors, Kisspeptin-1 , Receptors, LHRH , Humans , Female , Animals , Receptors, Kisspeptin-1/metabolism , Receptors, Kisspeptin-1/genetics , Receptors, LHRH/metabolism , Receptors, LHRH/genetics , Mice , HEK293 Cells , Zebrafish , Gonadotropin-Releasing Hormone/metabolism , Puberty/drug effects , Hypothalamus/metabolism , Hypothalamus/drug effects , Molecular Docking Simulation , Sexual Maturation/drug effects , Sexual Maturation/physiology , Kisspeptins/metabolism , Kisspeptins/genetics , Environmental Pollutants/toxicity , Environmental Pollutants/pharmacology
16.
Bull Exp Biol Med ; 177(4): 436-441, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39264556

ABSTRACT

We studied the effect of a high-fat, high-carbohydrate diet (HFHCD) on basal testosterone levels in the blood and testosterone, its precursors, and expression of steroidogenic genes in the testes of rats treated with human chorionic gonadotropin (hCG, 10 IU/rat, subcutaneously, once), gonadotropin-releasing hormone receptor antagonist cetrorelix (75 µg/kg, subcutaneously, 3 days), and their combination. In HFHCD rats, no obvious signs of androgen deficiency were observed and the response of the testes to hCG stimulation was preserved. Unlike control rats (normal diet), the expression of the luteinizing hormone receptor gene in these rats did not change in response to hCG stimulation and cetrorelix administration; they also showed a paradoxical, more pronounced response to hCG administration under conditions of suppression of the gonadotropin secretion by cetrorelix. This suggests that the etiology and pathogenesis of obesity may have different effects on the hormonal status of the male reproductive system.


Subject(s)
Chorionic Gonadotropin , Gonadotropin-Releasing Hormone , Obesity , Testis , Testosterone , Male , Animals , Chorionic Gonadotropin/pharmacology , Obesity/metabolism , Obesity/drug therapy , Rats , Testosterone/blood , Testis/drug effects , Testis/metabolism , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/analogs & derivatives , Receptors, LHRH/metabolism , Receptors, LHRH/antagonists & inhibitors , Receptors, LHRH/genetics , Diet, High-Fat/adverse effects , Hormone Antagonists/pharmacology , Humans , Rats, Wistar
17.
Sci Rep ; 14(1): 22481, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39341875

ABSTRACT

To evaluate if a preoperative medical treatment with the GnRH-antagonist relugolix in combination therapy in a series of patients with abnormal uterine bleeding associated with uterine myomas may correct the anemia before scheduled surgery for myoma-associated AUB. Thirty-one patients scheduled for surgery underwent a pre-operative three-month course with a daily oral tablet of 40 mg relugolix, 1 mg estradiol, and 0.5 mg norethindrone acetate. Hemoglobin levels, uterine volumes, largest myoma diameter, and VAS score for dysmenorrhea, pelvic pressure and bleeding discomfort, and indication to surgery were evaluated at study enrollment and at the end of therapy. Mean hemoglobin levels increased by 25%, from 9.3 ± 1.1 to 11.6 ± 1.7 g/dL after three months (p < 0.001). Uterine volume decreased from 380.7 ± 273.4 mL to 281.7 ± 198.7 mL (p < 0.001), whereas the diameter of the largest myoma decreased from 6.4 ± 2.8 cm to 5.5 ± 2.2 cm (p < 0.001). Four patients (13%), initially planned for a laparotomy procedure, were converted to a minimally-access procedure, whereas in eight patients (26%) surgery was avoided after medical therapy. Dysmenorrhea score improved from 4.7 ± 3.2 to 0.6 ± 1.1 (p < 0.0001). Pelvic pressure score decreased from 5.9 ± 2.1 to 3.1 ± 2.3 (p < 0.0001), whereas bleeding discomfort decreased from 7.4 ± 3.0 to 0.4 ± 1.6 (p < 0.0001). Preoperative GnRH-antagonist therapy may enhance hemoglobin levels, decrease uterine and myoma size, and alleviate symptoms, potentially enabling safe surgical procedures.


Subject(s)
Gonadotropin-Releasing Hormone , Leiomyoma , Uterine Neoplasms , Humans , Female , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Adult , Uterine Neoplasms/drug therapy , Uterine Neoplasms/surgery , Leiomyoma/drug therapy , Leiomyoma/surgery , Middle Aged , Preoperative Care , Treatment Outcome , Estradiol/administration & dosage , Estradiol/blood , Uterine Hemorrhage , Hemoglobins/metabolism , Drug Therapy, Combination , Phenylurea Compounds , Pyrimidinones
18.
Int J Mol Sci ; 25(18)2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39337542

ABSTRACT

Sexual maturation in goats is a dynamic process regulated precisely by the hypothalamic-pituitary-gonadal axis and is essential for reproduction. The hypothalamus plays a crucial role in this process and is the control center of the reproductive activity. It is significant to study the molecular mechanisms in the hypothalamus regulating sexual maturation in goats. We analyzed the serum hormone profiles and hypothalamic mRNA expression profiles of female goats during sexual development (1 day old (neonatal, D1, n = 5), 2 months old (prepuberty, M2, n = 5), 4 months old (sexual maturity, M4, n = 5), and 6 months old (breeding period, M6, n = 5)). The results indicated that from D1 to M6, serum hormone levels, including FSH, LH, progesterone, estradiol, IGF1, and leptin, exhibited an initial increase followed by a decline, peaking at M4. Furthermore, we identified a total of 508 differentially expressed genes in the hypothalamus, with a total of four distinct expression patterns. Nuclear receptor subfamily 1, group D, member 1 (NR1D1), glucagon-like peptide 1 receptor (GLP1R), and gonadotropin-releasing hormone 1 (GnRH-1) may contribute to hormone secretion, energy metabolism, and signal transduction during goat sexual maturation via circadian rhythm regulation, ECM receptor interactions, neuroactive ligand-receptor interactions, and Wnt signaling pathways. This investigation offers novel insights into the molecular mechanisms governing the hypothalamic regulation of goat sexual maturation.


Subject(s)
Goats , Hypothalamus , Sexual Maturation , Transcriptome , Animals , Goats/genetics , Goats/growth & development , Hypothalamus/metabolism , Sexual Maturation/genetics , Female , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/genetics , Gene Expression Profiling , Luteinizing Hormone/blood , Luteinizing Hormone/metabolism
19.
Article in English | MEDLINE | ID: mdl-39089445

ABSTRACT

Temperature is a preeminent factor in the regulation of fish reproduction and hinders gonadal development beyond a specific threshold. To comprehend the molecular mechanism responsible for reproductive suppression at different temperature, expression of the genes encoding kisspeptin (kiss2), gonadotropin-releasing hormone (gnrh1) and their receptors (gpr54, gnrh1r) in the brain, and the gonadotropin (GTH) subunits (fshb and lhb) in the pituitary were studied in juvenile Nile tilapia (Oreochromis niloticus) along with gonadal histology. Fish were acclimatized to three distinct temperatures, including 31 °C, 34 °C and 37 °C for 14 days. The mRNA levels of kiss2, gpr54, gnrh1, and gnrh1r were significantly decreased at 37 °C compared to 31 °C and 34 °C in the both sexes. In parallel, the expression level of fshb in the both sexes and lhb in the female were significantly lower at 37 °C in the pituitary. Histologically, the gonads of both sexes had normal growth of gametes at control temperature (31 °C), whereas the spermatogenesis and oocyte maturation were slowed down and atretic oocytes were found in the ovary at 37 °C acclimation temperature. Taken together, the results imply that elevated temperature beyond the specific threshold may have a negative impact on reproduction by suppressing the gene expressions of kisspeptin/GnRH1/GTH system and eventually restrains normal growth and maturation of gametes in the both sexes of Nile tilapia.


Subject(s)
Cichlids , Gonadotropin-Releasing Hormone , Gonads , Kisspeptins , Animals , Kisspeptins/genetics , Kisspeptins/metabolism , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Cichlids/genetics , Cichlids/growth & development , Cichlids/metabolism , Female , Male , Gonads/metabolism , Gonads/growth & development , Temperature , Fish Proteins/genetics , Fish Proteins/metabolism , Pituitary Gland/metabolism , Ovary/metabolism , Ovary/growth & development , Gonadotropins/metabolism , Gene Expression Regulation, Developmental
20.
Eur J Obstet Gynecol Reprod Biol ; 301: 87-94, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39116480

ABSTRACT

IMPORTANCE: Adenomyosis can reduce the chance of clinical pregnancy in women undergoing assisted conception. Treatment with prolonged gonadotrophin-releasing hormone analogue (GnRHa) downregulation prior to IVF/ICSI has been postulated to improve pregnancy outcomes. OBJECTIVE: We aimed to evaluate the effectiveness and safety of prolonged GnRHa treatment (minimum one month) versus no pre-treatment in women with adenomyosis undergoing IVF/ICSI using a systematic review and meta-analysis. DATA SOURCES: We searched electronic databases: Embase (OVID), MEDLINE® (OVID), APA PsycInfo (OVID), Maternity & Infant Care Database (MIDIRS (OVID), HMIC Health Management Information Consortium (OVID) and ClinicalTrials.gov from inception until 27th of March 2023. STUDY SELECTION AND SYNTHESIS: We included studies that reported on women with adenomyosis receiving GnRHa to down-regulate the hypothalamic-pituitary-ovarian axis for one to six months before IVF/ICSI. We pooled data using the Haensel-Mantel method and reported using Odds Ratio (OR) with 95 % confidence intervals (CI). We assessed the quality of included studies using the Newcastle-Ottowa Scale and confidence in evidence using the GRADE criteria. Bias analysis was conducted via the Cochrane recommended tool (RevMan Web, Academic License). MAIN OUTCOMES AND RESULTS: We screened 365 citations and eight retrospective studies were included in the meta-analysis (n = 2422 women). The median age was 34 years [IQR 31.95-35.05], median BMI 21.30 kg/m2 [IQR 21.05-23.55] and median duration of GnRHa downregulation was 2.5 months [Range 1-4; IQR 1.37-3]. Women with adenomyosis receiving prolonged GnRHa treatment had a higher implantation rate 1/OR 1.69 [95 % CI 1.09, 2.56], I2 = 81 %, (P = 0.02) and clinical pregnancy rate 1/OR 1.42 [95 % CI 1.03, 2.0], I2 70 %, P = 0.03. There was no overall difference in live birth rate 1/OR 1.12 [95 % CI 0.70, 1.79], I2 = 78 %, p = 0.63), miscarriage rate 1/OR 0.92 [95 % CI 0.63, 1.28, P = 0.61, I2 0 % or mean number of oocytes retrieved (10 oocytes [IQR 8.95; 11.15] vs. 9.28 [IQR 8; 10.20], p = 0.22) between groups. CONCLUSIONS AND RELEVANCE: The benefit of prolonged GnRHa treatment in women with adenomyosis undergoing assisted conception treatment is uncertain based on existing retrospective studies. Implantation and clinical pregnancy rates were higher following prolonged downregulation in this population, though there was no statistically significant difference in live birth and miscarriage rates. Given the limited, low-quality existing data, there is a need for a well-designed, prospective randomised controlled trial to precisely evaluate the effectiveness of prolonged GnRHa treatment in this population.


Subject(s)
Adenomyosis , Gonadotropin-Releasing Hormone , Sperm Injections, Intracytoplasmic , Humans , Female , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/agonists , Pregnancy , Adenomyosis/drug therapy , Fertilization in Vitro/methods , Pregnancy Rate , Down-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL