Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
1.
J Chem Inf Model ; 62(20): 5009-5022, 2022 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-36184840

RESUMO

The gonadotrophin-releasing hormone (GnRH) is a central regulator of the human reproductive system and exerts physiological effects by binding to GnRH1R. The GnRH-GnRH1R system is a promising therapeutic target for the maintenance of reproductive function. There are several GnRH1R agonists on the market, but like GnRH, they are all peptide compounds and are limited by their way of administration (subcutaneous or intramuscular injection). To date, no published GnRH1R small molecule agonists have been reported. In this paper, the HTRF-based screening method has been used to screen our in-house chemical library, and we found and confirmed CD304 as a hit compound. Subsequently, structure optimization led to the discovery of compound 6d, exhibited with a certain GnRH1R activation activity (EC50: 1.59 ± 0.38 µM). Further molecular dynamics simulation experiments showed that 6d can well bind to the orthosteric site of GnRH1R through forming a hydrogen-bonding interaction with Y2836.51. Binding of 6d further induces conformational changes in TM6 and TM7, promoting the formation of a continuous water channel in GnRH1R, thereby promoting GnRH1R activation. This well-characterized hit compound will facilitate the further development of novel small molecule agonists of GnRH1R.


Assuntos
Hormônio Liberador de Gonadotropina , Receptores LHRH , Humanos , Hormônio Liberador de Gonadotropina/farmacologia , Receptores LHRH/agonistas , Receptores LHRH/química , Bibliotecas de Moléculas Pequenas/farmacologia , Ligação de Hidrogênio
2.
Genomics ; 113(1 Pt 2): 1265-1271, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32971214

RESUMO

In this study, RNA sequencing was used to identify the hepatic gene expression profile in grass carp associated with luteinizing hormone-releasing hormone agonist (LHRH-A) treatment. A total of 93,912,172 reads were generated by HiSeq 4000 sequencing platform. After filtering, 83,450,860 clean reads were mapped to the reference genome. By calculating the FPKM of genes, 1475 differentially expressed genes were identified. PPAR signaling pathway was enriched with upregulated genes in LHRH-A injection group showing the regulation of the lipid metabolism by LHRH-A. The expression of eight key genes in PPAR signaling pathway was confirmed by qPCR and the results suggested that ACSL4A, ACSL4B, ANGPTL4, LPL, RXRBA and SLC27A1B were significantly stimulated by LHRH-A injection. This investigation provides the evidence that LHRH-A could play a role in lipid metabolism.


Assuntos
Carpas/metabolismo , Hormônio Liberador de Gonadotropina/análogos & derivados , Metabolismo dos Lipídeos , Transcriptoma , Animais , Carpas/genética , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Fígado/metabolismo , Receptores LHRH/agonistas , Receptores LHRH/metabolismo
3.
Eur J Med Chem ; 211: 113018, 2021 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-33223264

RESUMO

Peptide-drug conjugates (PDCs) are gaining considerable attention as anti-neoplastic agents. However, their development is often laborious and time-consuming. Herein, we have developed and preclinically evaluated three PDCs with gemcitabine as the anticancer cytotoxic unit and D-Lys6-GnRH (gonadotropin-releasing hormone; GnRH) as the cancer-targeting unit. These units were tethered via acid-labile programmable linkers to guide a differential drug release rate from the PDC through a combination of ester or amide and "click" type oxime ligations. The pro-drugs were designed to enable the selective targeting of malignant tumor cells with linker guided differential drug release rates. We exploited the oxime bond responsiveness against the acidic pH of the tumor microenvironment and the GnRH endocytosis via the GnRH-R GPCR which is overexpressed on cancer cells. The challenging metabolic properties of gemcitabine were addressed during design of the PDCs. We developed a rapid (1 hour) and cost-effective "click" oxime bond ligation platform to assemble in one-pot the 3 desired PDCs that does not require purification, surpassing traditional time-ineffective and low yield methods. The internalization of the tumor-homing peptide unit in cancer cells, overexpressing the GnRH-R, was first validated through confocal laser microscopy and flow cytometry analysis. Subsequently, the three PDCs were evaluated for their in vitro antiproliferative effect in prostate cancer cells. Their stability and the release of gemcitabine over time were monitored in vitro in cell culture and in human plasma using LC-MS/MS. We then assessed the ability of the developed PDCs to internalize in prostate cancer cells and to release gemcitabine. The most potent analog, designated GOXG1, was used for pharmacokinetic studies in mice. The metabolism of GOXG1 was examined in liver microsomes, as well as in buffers mimicking the pH of intracellular organelles, resulting in the identification of two metabolites. The major metabolite at low pH emanated from the cleavage of the pH-labile oxime bond, validating our design approach. NMR spectroscopy and in vitro radioligand binding assays were exploited for GOXG1 to validate that upon conjugating the drug to the peptide, the peptide microenvironment responsible for its GnRH-R binding is not perturbed and to confirm its high binding potency to the GnRH-R. Finally, the binding of GOXG1 to the GnRH-R and the associated elicitation of testosterone release in mice were also determined. The facile platform established herein for the rapid assembly of PDCs with linker controllable characteristics from aldehyde and aminooxy units through rapid "click" oxime ligation, that does not require purification steps, could pave the way for a new generation of potent cancer therapeutics, diagnostics and theranostics.


Assuntos
Desoxicitidina/análogos & derivados , Hormônio Liberador de Gonadotropina/farmacologia , Oximas/farmacologia , Pró-Fármacos/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Receptores LHRH/agonistas , Animais , Proliferação de Células/efeitos dos fármacos , Desoxicitidina/administração & dosagem , Desoxicitidina/química , Desoxicitidina/farmacologia , Relação Dose-Resposta a Droga , Desenvolvimento de Medicamentos , Hormônio Liberador de Gonadotropina/administração & dosagem , Hormônio Liberador de Gonadotropina/química , Humanos , Concentração de Íons de Hidrogênio , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Molecular , Oximas/administração & dosagem , Oximas/química , Pró-Fármacos/administração & dosagem , Pró-Fármacos/química , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptores LHRH/genética , Receptores LHRH/metabolismo , Relação Estrutura-Atividade , Células Tumorais Cultivadas , Gencitabina
5.
Sci Rep ; 10(1): 10579, 2020 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-32601341

RESUMO

Molecules that correct the folding of protein mutants, restoring their functional trafficking, are called pharmacoperones. Most are clinically irrelevant and possess intrinsic antagonist or agonist activity. Here, we identify compounds capable of rescuing the activity of mutant gonadotropin-releasing hormone receptor or GnRHR which, is sequestered within the cell and if dysfunctional leads to Hypogonadotropic Hypogonadism. To do this we screened the E90K GnRHR mutant vs. a library of 645,000 compounds using a cell-based calcium detection system. Ultimately, we identified 399 compounds with EC50 ≤ 5 µM with no effect in counterscreen assays. Medicinal chemistry efforts confirmed activity of 70 pure samples and mode of action studies, including radioligand binding, inositol phosphate, and toxicity assays, proved that we have a series of tractable compounds that can be categorized into structural clusters. These early lead molecules rescue mutant GnRHR function and are neither agonist nor antagonists of the GnRHR cognate receptor, a feature required for potential clinical utility.


Assuntos
Receptores LHRH/agonistas , Receptores LHRH/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Cálcio/metabolismo , Avaliação Pré-Clínica de Medicamentos , Hormônio Liberador de Gonadotropina/agonistas , Hormônio Liberador de Gonadotropina/metabolismo , Células HeLa , Ensaios de Triagem em Larga Escala , Humanos , Fosfatos de Inositol/metabolismo , Mutação , Dobramento de Proteína , Transporte Proteico , Receptores LHRH/genética
6.
Biochim Biophys Acta Rev Cancer ; 1874(1): 188383, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32535158

RESUMO

Androgen deprivation therapy (ADT) is the primary systemic therapy for treating locally advanced or metastatic prostate cancer (PCa). Despite its positive effect on PCa patient survival, ADT causes various adverse effects, including increased cardiovascular risk factors and cardiotoxicity. Lifespans extension, early use of ADT, and second-line treatment with next-generation androgen receptor pathway inhibitors would further extend the duration of ADT and possibly increase the risk of ADT-induced cardiotoxicity. Meanwhile, information on the molecular mechanisms underlying ADT-induced cardiotoxicity and measures to prevent it is limited, mainly due to the lack of specifically designed preclinical studies and clinical trials. This review article compiles up-to-date evidence obtained from observational studies and clinical trials, in order to gain new insights for deciphering the association between ADT use and cardiotoxicity. In addition, potential cardioprotective strategies involving GnRH receptors and second messenger cGMP are discussed.


Assuntos
Antagonistas de Androgênios/efeitos adversos , Antineoplásicos Hormonais/efeitos adversos , Doenças Cardiovasculares/epidemiologia , Neoplasias da Próstata/tratamento farmacológico , Antagonistas de Androgênios/administração & dosagem , Androgênios/metabolismo , Antineoplásicos Hormonais/administração & dosagem , Cardiotoxicidade/epidemiologia , Cardiotoxicidade/etiologia , Cardiotoxicidade/fisiopatologia , Cardiotoxicidade/prevenção & controle , Doenças Cardiovasculares/induzido quimicamente , Doenças Cardiovasculares/fisiopatologia , Doenças Cardiovasculares/prevenção & controle , Ensaios Clínicos como Assunto , GMP Cíclico/metabolismo , Hormônio Liberador de Gonadotropina/agonistas , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Longevidade/fisiologia , Masculino , Estudos Observacionais como Assunto , Inibidores da Fosfodiesterase 5/farmacologia , Inibidores da Fosfodiesterase 5/uso terapêutico , Receptores LHRH/agonistas , Receptores LHRH/antagonistas & inibidores , Receptores LHRH/metabolismo , Fatores de Risco , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Resultado do Tratamento
7.
Hum Reprod ; 34(5): 813-823, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31067329

RESUMO

STUDY QUESTION: Does the GnRH antagonist, ASP1707, reduce endometriosis-associated pelvic pain? SUMMARY ANSWER: ASP1707 significantly reduced endometriosis-associated pelvic pain in a dose-related manner. WHAT IS KNOWN ALREADY: GnRH agonists are an effective therapeutic option for endometriosis that is refractory to non-steroidal anti-inflammatory drugs, oral contraceptives, and progestins. However, GnRH agonists cause complete suppression of estradiol (E2), resulting in hypoestrogenic side-effects such as bone loss that may increase the future risk of osteoporotic fractures. STUDY DESIGN, SIZE, DURATION: This was a Phase II, multicenter, double-blind, randomized, parallel-group, placebo-controlled study conducted in 540 women from 04 December 2012 to 30 July 2015 in Europe and Japan. A sample size of 504 (84 subjects per group) was calculated to provide ≥80% power to detect a dose-related treatment effect among placebo and ASP1707 doses in change from baseline in pelvic pain, assuming different dose-response curves after 12 weeks of treatment. PARTICIPANTS/MATERIALS, SETTING, METHODS: Of 912 women with endometriosis-associated pelvic pain screened, 540 were enrolled, and 532 received ≥1 dose of study drug (placebo, n = 88; ASP1707 3 mg, n = 86; ASP1707 5 mg, n = 91; ASP1707 10 mg, n = 90; ASP1707 15 mg, n = 88; leuprorelin, n = 89) for 24 weeks. MAIN RESULTS AND THE ROLE OF CHANCE: After 12 weeks of treatment with ASP1707, the mean (95% CI) changes in numeric rating score (NRS) for overall pelvic pain (OPP) were -1.56 (-1.91, -1.21), -1.63 (-1.99, -1.27), -1.93 (-2.27, -1.60), -2.29 (-2.64, -1.94), and -2.13 (-2.47, -1.79) for placebo, ASP1707 3 mg, ASP1707 5 mg, ASP1707 10 mg, and ASP1707 15 mg, respectively. Mean (95% CI) changes in NRS for dysmenorrhea were -1.50 (-2.00, -1.00), -2.72 (-3.22, -2.21), -2.85 (-3.33, -2.38), -3.97 (-4.46, -3.48), and -4.18 (-4.66, -3.70), respectively. Mean (95% CI) changes in NRS for non-menstrual pelvic pain (NMPP) were -1.53 (-1.88, -1.19), -1.51 (-1.87, -1.16), -1.80 (-2.14, -1.47), -2.03 (-2.37, -1.68), and -1.86 (-2.20, -1.52), respectively. Statistically significant dose-related treatment effects in reduction in NRS for OPP (P = 0.001), dysmenorrhea (P < 0.001), and NMPP (P = 0.029) were observed after 12 weeks among ASP1707 doses and were maintained through 24 weeks. Serum estradiol and bone mineral density decreased dose dependently with ASP1707 through 24 weeks, however, to a lesser extent than with leuprorelin. LIMITATIONS, REASON FOR CAUTION: This study was not powered for pairwise comparison of each ASP1707 group versus placebo. WIDER IMPLICATIONS OF THE FINDINGS: All doses of ASP1707 reduced serum E2 levels to within the target range and to a lesser extent than leuprorelin. ASP1707 is a potential alternative treatment to leuprorelin for endometriosis-associated pelvic pain with lower impact on bone health. STUDY FUNDING/COMPETING INTEREST(S): This study was funded by Astellas Pharma Inc. T.D'.H is Vice President and Head of Global Medical Affairs Fertility at Merck, Darmstadt, Germany since October 1, 2015. At the time that the TERRA study was conducted, he served as Principal Investigator in his role as Coordinator of the Leuven University Fertility Center. Since October 2015, T.D'.H has left Leuven University Hospital Gasthuisberg, but continues to serve as Professor in Reproductive Medicine and Biology at KU Leuven (University of Leuven) Belgium and at the Dept of Obstetrics, Gynecology and Reproduction at Yale University, New Haven, USA. T. Fukaya and Y. Osuga report personal consulting fees from Astellas Pharma Inc. during the conduct of the study and outside the submitted work. G.M. Holtkamp, and L. Skillern are employed by Astellas Pharma Europe B.V.; K. Miyazaki is employed by Astellas Pharma Inc.; B. López, was a biostatistician for Astellas Pharma Europe B.V. during conduct of the study; R. Besuyen was a contract Associate Director of Medical Science for Astellas during conduct of the study. TRIAL REGISTRATION NUMBER: ClinicalTrials.gov, www.clinicaltrials.gov, NCT01767090. EudraCT number 2012-002791-14. TRIAL REGISTRATION DATE: 18 December 2012. DATE OF FIRST SUBJECT'S ENROLLMENT: One subject signed informed consent on 04 December 2012; the first subject was randomized on 16 April 2013.


Assuntos
Endometriose/complicações , Antagonistas de Hormônios/administração & dosagem , Imidazóis/administração & dosagem , Dor Pélvica/tratamento farmacológico , Receptores LHRH/antagonistas & inibidores , Sulfonas/administração & dosagem , Administração Oral , Adolescente , Adulto , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Antagonistas de Hormônios/efeitos adversos , Humanos , Imidazóis/efeitos adversos , Leuprolida/administração & dosagem , Leuprolida/efeitos adversos , Pessoa de Meia-Idade , Medição da Dor , Dor Pélvica/diagnóstico , Dor Pélvica/etiologia , Receptores LHRH/agonistas , Sulfonas/efeitos adversos , Resultado do Tratamento , Adulto Jovem
8.
Asian J Androl ; 21(2): 115-120, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30604695

RESUMO

We aimed to evaluate the current nationwide trend, efficacy, safety, and quality of life (QoL) profiles of hormone treatment in real-world practice settings for prostate cancer (PCa) patients in Korea. A total of 292 men with any biopsy-proven PCa (TanyNanyMany) from 12 institutions in Korea were included in this multi-institutional, observational study of prospectively collected data. All luteinizing hormone-releasing hormone (LHRH) agonists were allowed to be investigational drugs. Efficacy was defined as (1) the rate of castration (serum testosterone ≤50 ng dl-1) at 4-week visit and (2) breakthrough (serum testosterone >50 ng dl-1 after castration). Safety assessments included routine examinations for potential adverse events, laboratory tests, blood pressure, body weight, and bone mineral density (BMD, at baseline and at the last follow-up visit). QoL was assessed using the Expanded Prostate Cancer Index Composite-26 (EPIC-26). The most common initial therapeutic regimen was LHRH agonist with anti-androgen (78.0%), and the most commonly used LHRH agonist for combination and monotherapy was leuprolide (64.0% for combination and 58.0% for monotherapy). The castration and breakthrough rates were 78.4% and 6.6%, respectively. The laboratory results related to dyslipidemia worsened after 4 weeks of hormone treatment. In addition, the mean BMD T-score was significantly lower at the last follow-up (mean: -1.950) compared to baseline (mean: -0.195). The mean total EPIC-26 score decreased from 84.8 (standard deviation [s.d.]: 12.2) to 78.3 (s.d.: 8.1), with significant deterioration only in the urinary domain (mean: 23.5 at baseline and 21.9 at the 4-week visit). These findings demonstrate the nationwide trend of current practice settings in hormone treatment for PCa in Korea.


Assuntos
Antagonistas de Androgênios/uso terapêutico , Antineoplásicos Hormonais/uso terapêutico , Leuprolida/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Receptores LHRH/agonistas , Idoso , Colesterol/sangue , Quimioterapia Combinada , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias da Próstata/sangue , Neoplasias da Próstata/patologia , Qualidade de Vida , República da Coreia , Testosterona/sangue , Resultado do Tratamento , Triglicerídeos/sangue
9.
Med J Aust ; 209(9): 412-416, 2018 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-30376664

RESUMO

Alkylating chemotherapy is often used to treat pre-menopausal women for various malignancies and autoimmune diseases. Chemotherapy-associated ovarian failure is a potential consequence of this treatment which can cause infertility, and increases the risk of other long term adverse health sequelae. Randomised trials, predominantly of women undergoing alkylating chemotherapy for breast cancer, have shown evidence for the efficacy of gonadotropin-releasing hormone agonists (GnRHa) in preventing chemotherapy-associated ovarian failure. The European St Gallen and United States National Comprehensive Cancer Network guidelines recommend the use of concurrent GnRHa to reduce the risk of ovarian failure for pre-menopausal women undergoing chemotherapy for breast cancer. The GnRHa goserelin, a monthly 3.6 mg depot subcutaneous injection, has recently been listed on the Australian Pharmaceutical Benefits Scheme to reduce risk of ovarian failure for pre-menopausal women receiving alkylating therapies for malignancy or autoimmune disease. The first dose of goserelin should ideally be administered at least 1 week before commencement of alkylating treatment and continued 4-weekly during chemotherapy. Concurrent goserelin use should now be considered for all pre-menopausal women due to commence alkylating chemotherapy (except those with incurable cancer), regardless of their childbearing status, in an effort to preserve their ovarian function. For women who have not completed childbearing, consideration of other fertility preservation options, such as cryopreservation of embryos or oocytes, is also important.


Assuntos
Antineoplásicos Alquilantes/efeitos adversos , Neoplasias da Mama/tratamento farmacológico , Hormônio Liberador de Gonadotropina/uso terapêutico , Infertilidade Feminina/prevenção & controle , Ovário/efeitos dos fármacos , Neoplasias da Mama/patologia , Quimioterapia Adjuvante , Feminino , Preservação da Fertilidade/métodos , Gosserrelina/uso terapêutico , Humanos , Infertilidade Feminina/induzido quimicamente , Ovário/fisiopatologia , Gravidez , Taxa de Gravidez , Pré-Menopausa , Ensaios Clínicos Controlados Aleatórios como Assunto , Receptores LHRH/agonistas
10.
Neurourol Urodyn ; 37(5): 1574-1582, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-30133853

RESUMO

AIM: To evaluate the effects of a treatment with leuprolide acetate (LA) on bladder overactivity as well as the expression of gonadotropin releasing hormone receptor (GnRH-R), and neurofilaments NF68 and NF200 in female rats with overactive bladder induced by castration. METHODS: Changes in the urodynamic parameters were determined in SHAM, ovariectomized (OVX) and ovariectomized rats treated with LA (OVX-LA). A semi-quantitative analysis for the expression pattern of GnRH-R and neurofilaments NF68 and NF200 were determined. RESULTS: Forty-three days after ovariectomy, rats from the OVX group have significant lower values for intercontractile interval (ICI) and compliance (C); as well as higher values for basal bladder pressure (BP) and frequency of non-voiding contractions (NVC). The systemic application of LA increased voiding volume (Vv) and pressure threshold (ThP) in the OVX-LA animals. The application of LA reduced the high frequency of NVC in the OVX rats. No significant differences were found for Vv and NVCs between the OVX-LA vs SHAM groups. At the mid part of the bladder, the presence of GnRH-R was evidenced in the urothelium of the SHAM group. The OVX animals showed different pattern of immunolabeling for GnRH-R as well as for neurofilaments NF200 and NF68, whereas in the OVX-LA group the immunofluorescence pattern was similar to the one seen in SHAM bladders (P < 0.05 for OVX vs OVX + LA). CONCLUSIONS: the results suggest that systemic application of LA can improve bladder dysfunction in castrated rats, and perhaps considered as a treatment for overactive bladder conditions secondary to menopause.


Assuntos
Leuprolida/farmacologia , Ovariectomia , Receptores LHRH/agonistas , Urodinâmica/efeitos dos fármacos , Animais , Complacência (Medida de Distensibilidade)/efeitos dos fármacos , Feminino , Contração Muscular/efeitos dos fármacos , Proteínas de Neurofilamentos/biossíntese , Proteínas de Neurofilamentos/genética , Ratos , Ratos Wistar , Receptores LHRH/biossíntese , Bexiga Urinária/efeitos dos fármacos , Bexiga Urinária/metabolismo , Urotélio/efeitos dos fármacos , Urotélio/metabolismo
12.
Vitam Horm ; 107: 27-66, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29544634

RESUMO

Gonadotropin-releasing hormone (GnRH) is recognized as the central regulator of the functions of the pituitary-gonadal axis. The increasing knowledge on the mechanisms controlling the development and the function of GnRH-producing neurons is leading to a better diagnostic and therapeutic approach for hypogonadotropic hypogonadisms and for alterations of the puberty onset. During female life span, the function of the GnRH pulse generator may be affected by a number of inputs from other neuronal systems, offering alternative strategies for diagnostic and therapeutic interventions. Moreover, the identification of a GnRH/GnRH receptor system in both human ovary and endometrium has widened the spectrum of action of the peptide outside its hypothalamic functions. The pharmacological use of GnRH itself or its synthetic analogs (agonists and antagonists) provides a valid tool to either stimulate or block gonadotropin secretion and to modulate the female fertility in several reproductive disorders and in assisted reproduction technology. The use of GnRH agonists in young female patients undergoing chemotherapy is also considered a promising therapeutic approach to counteract iatrogenic ovarian failure.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Sistema Hipotálamo-Hipofisário/fisiologia , Modelos Biológicos , Células Neuroendócrinas/fisiologia , Ovário/fisiologia , Precursores de Proteínas/metabolismo , Receptores LHRH/agonistas , Reprodução , Animais , Endométrio/efeitos dos fármacos , Endométrio/crescimento & desenvolvimento , Endométrio/fisiologia , Endométrio/fisiopatologia , Feminino , Fármacos para a Fertilidade Feminina/farmacologia , Fármacos para a Fertilidade Feminina/uso terapêutico , Preservação da Fertilidade/tendências , Hormônio Liberador de Gonadotropina/química , Humanos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/crescimento & desenvolvimento , Sistema Hipotálamo-Hipofisário/fisiopatologia , Infertilidade Feminina/tratamento farmacológico , Infertilidade Feminina/patologia , Infertilidade Feminina/fisiopatologia , Infertilidade Feminina/terapia , Ciclo Menstrual/efeitos dos fármacos , Células Neuroendócrinas/citologia , Células Neuroendócrinas/metabolismo , Células Neuroendócrinas/patologia , Ovário/efeitos dos fármacos , Ovário/crescimento & desenvolvimento , Ovário/fisiopatologia , Gravidez , Insuficiência Ovariana Primária/tratamento farmacológico , Insuficiência Ovariana Primária/patologia , Insuficiência Ovariana Primária/fisiopatologia , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/metabolismo , Precursores de Proteínas/química , Puberdade/efeitos dos fármacos , Receptores LHRH/antagonistas & inibidores , Receptores LHRH/metabolismo , Receptores LHRH/uso terapêutico , Reprodução/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
13.
J Biol Chem ; 292(50): 20720-20731, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29054929

RESUMO

Pituitary gonadotropin hormones are regulated by gonadotropin-releasing hormone (GnRH) via MAPK signaling pathways that stimulate gene transcription of the common α-subunit (Cga) and the hormone-specific ß-subunits of gonadotropin. We have reported previously that GnRH-induced activities at these genes include various histone modifications, but we did not examine histone phosphorylation. This modification adds a negative charge to residues of the histone tails that interact with the negatively charged DNA, is associated with closed chromatin during mitosis, but is increased at certain genes for transcriptional activation. Thus, the functions of this modification are unclear. We initially hypothesized that GnRH might induce phosphorylation of Ser-10 in histone 3 (H3S10p) as part of its regulation of gonadotropin gene expression, possibly involving cross-talk with H3K9 acetylation. We found that GnRH increases the levels of both modifications around the Cga gene transcriptional start site and that JNK inhibition dramatically reduces H3S10p levels. However, this modification had only a minor effect on Cga expression and no effect on H3K9ac. GnRH also increased H3S28p and H3K27ac levels and also those of activated mitogen- and stress-activated protein kinase 1 (MSK1). MSK1 inhibition dramatically reduced H3S28p levels in untreated and GnRH-treated cells and also affected H3K27ac levels. Although not affecting basal Cga expression, MSK1/2 inhibition repressed GnRH activation of Cga expression. Moreover, ChIP analysis revealed that GnRH-activated MSK1 targets the first nucleosome just downstream from the TSS. Given that the elongating RNA polymerase II (RNAPII) stalls at this well positioned nucleosome, GnRH-induced H3S28p, possibly in association with H3K27ac, would facilitate the progression of RNAPII.


Assuntos
Regulação da Expressão Gênica , Subunidade alfa de Hormônios Glicoproteicos/agonistas , Gonadotrofos/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Nucleossomos/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Sítio de Iniciação de Transcrição , Acetilação/efeitos dos fármacos , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , Regulação da Expressão Gênica/efeitos dos fármacos , Subunidade alfa de Hormônios Glicoproteicos/genética , Subunidade alfa de Hormônios Glicoproteicos/metabolismo , Gonadotrofos/efeitos dos fármacos , Gonadotrofos/enzimologia , Histonas/metabolismo , Lisina/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Nucleossomos/enzimologia , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Receptores LHRH/agonistas , Receptores LHRH/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Serina/metabolismo , Sítio de Iniciação de Transcrição/efeitos dos fármacos
14.
J Eval Clin Pract ; 23(4): 866-869, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28585354

RESUMO

OBJECTIVE: To determine whether hormonal treatments for frequent clinical cases (short stature, delayed and precocious puberty) are prescribed strictly according to clinical guidelines or based on personal tendencies, and whether the decisions correlate with physician's personal demographics (age, sex, and place of practice). METHODS: Cross-sectional survey, with made-up clinical cases, distributed to pediatric endocrinologists using 2 web-based professional forums, Israeli and an international. The questionnaire included 8 clinical cases and 5 demographic questions regarding the physician. Differences in practice between Israeli and international endocrinologists were assessed, and correlation between the physician's gender and their decisions regarding treatment. RESULTS: One hundred fifty-five physicians responded, 28% Israeli and 72% international. In girls with early puberty, 60% of international and 26% of Israeli physicians chose not to treat with a gonadotropin-releasing hormone agonist. In girls with short stature, 79% of Israeli and 34% of international physicians offered growth hormone treatment. In girls with early puberty, both male and female physicians responded similarly in the international group, but in the Israeli group 47% of male and 15% of female doctors would not treat. In girls with constitutional growth delay, 67% of Israeli male doctors would not treat with growth hormone compared to 30% of Israeli female physicians. CONCLUSIONS: Our study demonstrated significant practice differences between Israeli and international pediatric endocrinologists. Within the Israeli group, significant practice differences were seen between male and female physicians. Given that Israeli physicians follow the same clinical guidelines it is clear that a large "grey zone" of clinical cases exist and much of the decisions on treatment are personal and influenced by personal beliefs or gender.


Assuntos
Endocrinologistas/estatística & dados numéricos , Fidelidade a Diretrizes/estatística & dados numéricos , Pediatria/estatística & dados numéricos , Guias de Prática Clínica como Assunto , Padrões de Prática Médica/estatística & dados numéricos , Características de Residência/estatística & dados numéricos , Adulto , Fatores Etários , Estatura , Estudos Transversais , Feminino , Transtornos do Crescimento/tratamento farmacológico , Hormônio do Crescimento Humano/administração & dosagem , Humanos , Israel , Masculino , Pessoa de Meia-Idade , Puberdade Precoce , Receptores LHRH/agonistas , Proteínas Recombinantes , Fatores Sexuais
15.
Psychoneuroendocrinology ; 82: 164-172, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28550793

RESUMO

Leuprolide acetate (LEU), also known as Lupron, is commonly used to treat prostate cancer in men. As a gonadotropin-releasing hormone (GnRH) receptor agonist, it initially stimulates the release of gonadal hormones, testosterone (T) and estradiol. This surge eventually suppresses these hormones, preventing the further growth and spread of cancer cells. Individuals receiving this treatment often report anxiety and cognitive changes, but LEU's effects on the neural mechanisms that are involved in anxiety during the trajectory of treatment are not well known. In this study, we examined the acute effects of LEU on fear extinction, hypothesizing that increased T levels following a single administration of LEU will facilitate extinction recall by altering neuronal activity within the fear extinction circuitry. Two groups of naïve adult male rats underwent a 3-day fear conditioning, extinction, and recall experiment. The delayed group (n=15) received a single injection of vehicle or LEU (1.2mg/kg) 3weeks before behavioral testing. The acute group (n=25) received an injection one day after fear conditioning, 30min prior to extinction training. Following recall, the brains for all animals were collected for c-fos immunohistochemistry. Blood samples were also collected and assayed for T levels. Acute administration of LEU increased serum T levels during extinction training and enhanced extinction recall 24h later. This enhanced extinction memory was correlated with increased c-fos activity within the infralimbic cortex and amygdala, which was not observed in the delayed group. These results suggest that the elevation in T induced by acute administration of LEU can influence extinction memory consolidation, perhaps through modification of neuronal activity within the infralimbic cortex and amygdala. This may be an important consideration in clinical applications of LEU and its effects on anxiety and cognition.


Assuntos
Extinção Psicológica/efeitos dos fármacos , Leuprolida/farmacologia , Memória/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Encéfalo/metabolismo , Córtex Cerebral/metabolismo , Condicionamento Clássico/fisiologia , Condicionamento Psicológico/efeitos dos fármacos , Extinção Psicológica/fisiologia , Medo/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Sistema Hipotálamo-Hipofisário , Leuprolida/metabolismo , Masculino , Memória/fisiologia , Neurônios/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores LHRH/agonistas , Receptores LHRH/uso terapêutico , Testosterona/sangue
16.
Urol Oncol ; 35(5): 183-191, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28325650

RESUMO

PURPOSE: To explore how follicle-stimulating hormone (FSH) may contribute to cardiovascular, metabolic, skeletal, and cognitive events in men treated for prostate cancer, with various forms of androgen deprivation therapy (ADT). MATERIALS AND METHODS: A colloquium of prostate cancer experts was convened in May 2015, to discuss the role of FSH in the development of unwanted effects associated with ADT. Subsequently, a literature review (Medline, PubMed, and relevant congress abstract databases) was performed to further explore and evaluate the collected evidence. RESULTS: It has become evident that, in the setting of ADT, FSH can promote the development of atherosclerotic plaque formation, metabolic syndrome, and insulin resistance. Data also suggest that FSH is an important mediator of bone remodeling, particularly bone resorption, and thereby increases the risk for bone fracture. Additional evidence implicates a role for FSH in bone metastasis as well. The influence of FSH on ADT-induced cognitive deficits awaits further elucidation; however, the possibility that FSH may be involved therein cannot be ruled out. CONCLUSIONS: The widespread molecular and physiological consequences of FSH system activation in normal and pathological conditions are becoming better understood. Progress in this area has been achieved by the development of additional investigative and clinical measures to better evaluate specific adverse effects. More research is needed on FSH function in the development of cancer as well as its association with cardiovascular, metabolic, musculoskeletal, and cognitive effects in ADT.


Assuntos
Aterosclerose/metabolismo , Neoplasias Ósseas/secundário , Hormônio Foliculoestimulante/metabolismo , Neoplasias da Próstata/terapia , Receptores LHRH/agonistas , Receptores LHRH/antagonistas & inibidores , Animais , Neoplasias Ósseas/metabolismo , Reabsorção Óssea/metabolismo , Disfunção Cognitiva/metabolismo , Humanos , Resistência à Insulina , Masculino , Orquiectomia , Neoplasias da Próstata/patologia
17.
Reprod Biol ; 17(1): 1-8, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28215488

RESUMO

GnRH agonist (GnRHa) trigger for final oocyte maturation in GnRH antagonist co-treated IVF/ICSI cycles significantly reduces the risk of ovarian hyperstimulation syndrome (OHSS). GnRHa trigger followed by modifications of the standard luteal phase support (modified luteal phase support) secures fresh transfer in the majority of patients with excellent reproductive outcomes. In freeze all cycles (segmented cycles) GnRHa trigger allows oocyte retrieval with a minimal risk of early onset OHSS and good reproductive outcomes in subsequent frozen thaw cycles. Overall, two different luteal phase support strategies have been proposed when a fresh transfer is performed after GnRHa trigger. These involve either boosting the endogenous steroid production or adding exogenous steroids. The present review discusses the advancement of GnRHa trigger in fresh and segmented cycles and how a modified luteal phase support policy in fresh transfer cycles results in good reproductive outcomes as well as a high safety in terms of OHSS reduction. Finally, the new concept of an individualized luteal phase support policy taking the number of pre-ovulatory follicles into account when planning a fresh transfer in GnRHa triggered IVF/ICSI cycle is discussed.


Assuntos
Manutenção do Corpo Lúteo/efeitos dos fármacos , Fármacos para a Fertilidade Feminina/uso terapêutico , Hormônio Liberador de Gonadotropina/agonistas , Síndrome de Hiperestimulação Ovariana/prevenção & controle , Medicina de Precisão , Receptores LHRH/agonistas , Transdução de Sinais/efeitos dos fármacos , Animais , Feminino , Fármacos para a Fertilidade Feminina/efeitos adversos , Fertilização In Vitro , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Técnicas de Maturação in Vitro de Oócitos , Infertilidade Feminina/terapia , Nascido Vivo , Recuperação de Oócitos/efeitos adversos , Síndrome de Hiperestimulação Ovariana/epidemiologia , Reserva Ovariana/efeitos dos fármacos , Indução da Ovulação/efeitos adversos , Gravidez , Taxa de Gravidez , Receptores LHRH/antagonistas & inibidores , Receptores LHRH/metabolismo , Risco
18.
Sci Rep ; 6: 26220, 2016 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-27189011

RESUMO

Androgen-deprivation therapy (ADT) for prostate cancer has been associated with increased risk for development of cardiovascular events and recent pooled analyses of randomized intervention trials suggest that this primarily is the case for patients with pre-existing cardiovascular disease treated with gonadotropin-releasing hormone receptor (GnRH-R) agonists. In the present study we investigated the effects of the GnRH-R agonist leuprolide and the GnRH-R antagonist degarelix on established atherosclerotic plaques in ApoE(-/-) mice. A shear stress modifier was used to produce both advanced and more stable plaques in the carotid artery. After 4 weeks of ADT, increased areas of necrosis was observed in stable plaques from leuprolide-treated mice (median and IQR plaque necrotic area in control, degarelix and leuprolide-treated mice were 0.6% (IQR 0-3.1), 0.2% (IQR 0-4.4) and 11.0% (IQR 1.0-19.8), respectively). There was also evidence of increased inflammation as assessed by macrophage immunohistochemistry in the plaques from leuprolide-treated mice, but we found no evidence of such changes in plaques from control mice or mice treated with degarelix. Necrosis destabilizes plaques and increases the risk for rupture and development of acute cardiovascular events. Destabilization of pre-existing atherosclerotic plaques could explain the increased cardiovascular risk in prostate cancer patients treated with GnRH-R agonists.


Assuntos
Apolipoproteínas E/deficiência , Leuprolida/administração & dosagem , Oligopeptídeos/administração & dosagem , Placa Aterosclerótica/patologia , Receptores LHRH/agonistas , Receptores LHRH/antagonistas & inibidores , Animais , Artérias Carótidas/patologia , Modelos Animais de Doenças , Imuno-Histoquímica , Macrófagos/imunologia , Camundongos , Camundongos Knockout
19.
J Steroid Biochem Mol Biol ; 163: 35-44, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27036999

RESUMO

PCOS is a major cause of anovulatory infertility in women in their reproductive age. However, its etiology and pathophysiology remain uncertain. The immature mice chronically injected with DHEA, termed as PCO-mice, develop numerous large cystic follicles, high circulating androgen and anovulation similar to PCOS in women. Although PCO-mice show decreased ovarian GnRH I-receptor in immunoblot but show increased immunostaining for GnRH I-receptor in oocytes of cystic follicles. PCO-mice show reduced ovarian LH receptor expression, circulating estradiol and progesterone level compared to normal mice injected with vehicle only. The treatment with low dose of GnRH-Agonist in PCO-mice restores ovarian LH receptor expression to the level of normal mice and promote ovulation and formation of functional corpus luteum. GnRH-Antagonist although cause ovulation in PCO-mice but does not restore LH receptor expression to the level of normal mice, and they show low circulating progesterone and hypertrophied vacuolated corpus luteum. Our study suggests that GnRH-agonist restores ovulation in PCO-mice and produces biphasic and beneficial effect over the use of GnRH-Antagonist.


Assuntos
Corpo Lúteo/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/farmacologia , Síndrome do Ovário Policístico/tratamento farmacológico , Receptores LHRH/genética , Animais , Corpo Lúteo/metabolismo , Corpo Lúteo/patologia , Desidroepiandrosterona/administração & dosagem , Modelos Animais de Doenças , Estradiol/sangue , Feminino , Regulação da Expressão Gênica , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Hormônio Luteinizante/genética , Hormônio Luteinizante/metabolismo , Camundongos , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/patologia , Progesterona/sangue , Receptores LHRH/agonistas , Receptores LHRH/antagonistas & inibidores , Receptores LHRH/metabolismo
20.
Biosens Bioelectron ; 79: 721-7, 2016 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-26774084

RESUMO

The gonadotropin-releasing hormone (GnRH) receptor is a drug target for certain hormone-dependent diseases such as prostate cancer. In this study, we examined the activation profiles of the endogenous ligand, GnRH and a well-known marketed analog, buserelin using a label-free assay in pituitary αT3-1 cells with endogenous GnRH receptor expression. This whole cell impedance-based technology allows for the real-time measurement of morphological cellular changes. Both agonists dose-dependently decreased the impedance as a result of GnRH receptor activation with potencies of 9.3 ± 0.1 (pEC50 value, buserelin) and 7.8 ± 0.06 (pEC50 value, GnRH). Subsequently, GnRH receptor activation was completely abolished with a selective Gαq inhibitor, thereby confirming the Gαq-coupling of the GnRH receptor in pituitary αT3-1 cells. Additionally, we observed continued responses after agonist stimulation of αT3-1 cells indicating long-lasting cellular effects. Wash-out experiments demonstrated that the long-lasting effects induced by GnRH were most likely caused by rebinding since over 70% of the original response was abolished after wash-out. In contrast, a long receptor residence time was responsible for the prolonged effects caused by buserelin, with over 70% of the original response remaining after wash-out. In summary, we validated that impedance-based label-free technology is suited for studying receptor-mediated activation in cell lines endogenously expressing the target of interest. Moreover, this real-time monitoring allows the examination of binding kinetics and its influence on receptor activation at a cellular level.


Assuntos
Hipófise/metabolismo , Receptores LHRH/metabolismo , Técnicas Biossensoriais , Busserrelina/farmacologia , Linhagem Celular , Impedância Elétrica , Humanos , Fosfatos de Inositol/metabolismo , Hipófise/citologia , Hipófise/efeitos dos fármacos , Receptores LHRH/agonistas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...