Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 444
Filtrar
1.
Mar Environ Res ; 186: 105938, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36870161

RESUMEN

Progestins in aquatic environments are of increasing concern, as shown by the results of toxicological studies on adult invertebrates with external fertilization. However, their potential effects on the gametes and reproductive success of such animals remain largely unknown. Thus, the current study assessed the effect of in vitro exposure of environmentally relevant concentrations (10 ng/L and 1000 ng/L) of norgestrel (NGT) on the sperm of Pacific oyster Crassostrea gigas, analyzing sperm motility, ultrastructure, mitochondrial function, ATP status, characteristic enzyme activities, and DNA integrity underlying fertilization and hatching success. The results showed that NGT increased the percentage of motile sperm by elevating intracellular Ca2+ levels, Ca2+-ATPase activity, creatine kinase activity, and ATP content. Although superoxide dismutase activity was enhanced to eliminate reactive oxygen species generated by NGT, oxidative stress occurred, as indicated by the increase in malonaldehyde content and damage to plasma membranes and DNA. As a consequence, fertilization rates decreased. However, hatching rates did not alter significantly, possibly as a result of DNA repair processes. This study demonstrates oyster sperm as a useful, sensitive tool for toxicological research of progestins and provides ecologically relevant information on reproductive disturbance in oysters resulting from exposure to NGT.


Asunto(s)
Crassostrea , Animales , Masculino , Crassostrea/fisiología , Norgestrel/metabolismo , Norgestrel/farmacología , Progestinas/metabolismo , Progestinas/farmacología , Motilidad Espermática/fisiología , Semen , Espermatozoides/fisiología , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología
2.
Expert Rev Clin Pharmacol ; 14(2): 211-224, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33464138

RESUMEN

INTRODUCTION: Norgestimate (NGM) is a testosterone derivative with peculiar receptor activities. AREAS COVERED: This is a narrative review of the available data on the pharmacotherapy of NGM in combined hormonal contraceptives (CHCs) in terms of contraceptive efficacy, venous thromboembolism (VTE) risk, safety, tolerability and bleeding patterns. A comprehensive literature review was conducted in August 2020 using PubMed with the keyword 'norgestimate'. EXPERT OPINION: NGM shows a mild estrogenic activity associated with anti-mineralocorticoid and anti-androgenic properties, largely responsible for the cardiovascular safety profile. The anti-androgenic property depends on the androgen receptor (AR) nuclear translocation (AR trafficking and its subnuclear distribution), the inhibition of 5α-reductase activity (it possesses higher activity compared to other available progestins), and the increase on sexual hormone binding globulin (SHBG) levels if combined with an estrogenic counterpart. NGM is one of the molecules that best modulates the power of ethinyl-estradiol on the thromboembolic risk, being associated with the lowest VTE risk between different CHCs. NGM has the advantage of retaining peripheral anti-androgenic activity, demonstrated by the impact on lipid and glucose metabolism, and it should be preferred if compared with other similar progestins of the same class of risk which are much more androgenic, such as levonorgestrel.


Asunto(s)
Agentes Anticonceptivos Hormonales/administración & dosificación , Norgestrel/análogos & derivados , Tromboembolia Venosa/inducido químicamente , Animales , Agentes Anticonceptivos Hormonales/efectos adversos , Agentes Anticonceptivos Hormonales/farmacología , Femenino , Humanos , Levonorgestrel/administración & dosificación , Levonorgestrel/efectos adversos , Levonorgestrel/farmacología , Norgestrel/administración & dosificación , Norgestrel/efectos adversos , Norgestrel/farmacología , Riesgo , Tromboembolia Venosa/epidemiología
3.
Invest Ophthalmol Vis Sci ; 60(8): 3221-3235, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31335948

RESUMEN

Purpose: Retinitis pigmentosa (RP) refers to a group of inherited blinding retinal diseases, whereby the death of mutated rod photoreceptors is followed closely by the death of cone photoreceptors. Cone cell death can be hugely debilitating as color/daytime vision becomes impaired. Thus, treatments that are effective against cone cell death are urgently needed. Our research has been working toward development of a neuroprotective treatment for RP. We have previously demonstrated significant neuroprotective properties of norgestrel, a progesterone analogue, in the mouse retina. The current study further investigates the potential of norgestrel as a treatment for RP, with a focus on long-term preservation of cone photoreceptors. Methods: Using the well-established rd10 mouse model of RP, we administered a norgestrel-supplemented diet at postnatal day (P)30, following widespread loss of rod photoreceptors and at the outset of cone degeneration. We subsequently assessed cone cell morphology and retinal function at P50, P60, and P80, using immunohistochemistry, electroretinograph recordings, and optomotor testing. Results: While cone cell degeneration was widespread in the untreated rd10 retina, we observed profound preservation of cone photoreceptor morphology in the norgestrel-treated mice for at least 50 days, out to P80. This was demonstrated by up to 28-fold more cone arrestin-positive photoreceptors. This protection transpired to functional preservation at all ages. Conclusions: This work presents norgestrel as an incredibly promising long-term neuroprotective compound for the treatment of RP. Crucially, norgestrel could be used in the mid-late stages of the disease to protect remaining cone cells and help preserve color/daytime vision.


Asunto(s)
Neuroprotección/efectos de los fármacos , Norgestrel/farmacología , Progesterona/farmacología , Células Fotorreceptoras Retinianas Conos/efectos de los fármacos , Retinitis Pigmentosa/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Electrorretinografía , Femenino , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Progestinas/farmacología , Células Fotorreceptoras Retinianas Conos/patología , Retinitis Pigmentosa/patología , Retinitis Pigmentosa/fisiopatología , Transducción de Señal
4.
CNS Drugs ; 33(5): 513-522, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30963506

RESUMEN

BACKGROUND: Erenumab is a human anti-calcitonin gene-related peptide monoclonal antibody developed for migraine prevention. Migraine predominately affects women of childbearing age; thus, it is important to determine potential drug-drug interactions between a common oral contraceptive and drugs used to treat migraine. OBJECTIVES: We sought to evaluate potential drug-drug interactions between erenumab and a common oral contraceptive. METHODS: Healthy women received three cycles of a norgestimate/ethinyl estradiol-containing oral contraceptive with a single 140-mg subcutaneous dose of erenumab during cycle three. Norgestimate metabolites (norgestrel and norelgestromin) and ethinyl estradiol pharmacokinetics were evaluated in the absence and presence of erenumab. Primary endpoint was peak plasma concentration (Cmax) and area under concentration-time curve from time 0 to 24 h (AUCtau). Luteinizing hormone, follicle-stimulating hormone, and progesterone concentrations were evaluated as pharmacodynamic markers. RESULTS: Erenumab did not influence the pharmacokinetics of norelgestromin, norgestrel, or ethinyl estradiol. Least-squares mean estimates (90% confidence interval) for Cmax ratios were 1.05 (0.90-1.23), 1.06 (0.97-1.16), and 1.04 (0.88-1.22) for norelgestromin, norgestrel, and ethinyl estradiol, respectively. Respective AUCtau ratios were 1.02 (0.94-1.12), 1.03 (0.96-1.10), and 1.02 (0.91-1.14). Luteinizing hormone, follicle-stimulating hormone, and progesterone concentrations were similar after exposure to oral contraceptive alone and with erenumab. CONCLUSION: Erenumab did not alter the pharmacokinetics of the active components of an estrogen/progestin combination oral contraceptive. Thus, no change in contraceptive efficacy is expected with erenumab. TRIAL REGISTRATION: ClinicalTrials.gov NCT02792517.


Asunto(s)
Anticuerpos Monoclonales Humanizados/sangre , Antagonistas del Receptor Peptídico Relacionado con el Gen de la Calcitonina/sangre , Anticonceptivos Orales Combinados/sangre , Etinilestradiol/sangre , Norgestrel/análogos & derivados , Adolescente , Adulto , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacología , Área Bajo la Curva , Antagonistas del Receptor Peptídico Relacionado con el Gen de la Calcitonina/administración & dosificación , Antagonistas del Receptor Peptídico Relacionado con el Gen de la Calcitonina/efectos adversos , Antagonistas del Receptor Peptídico Relacionado con el Gen de la Calcitonina/farmacología , Anticonceptivos Orales Combinados/administración & dosificación , Anticonceptivos Orales Combinados/efectos adversos , Anticonceptivos Orales Combinados/farmacología , Combinación de Medicamentos , Interacciones Farmacológicas , Etinilestradiol/administración & dosificación , Etinilestradiol/efectos adversos , Etinilestradiol/farmacología , Femenino , Hormona Folículo Estimulante/sangre , Voluntarios Sanos , Humanos , Hormona Luteinizante/sangre , Tasa de Depuración Metabólica , Persona de Mediana Edad , Norgestrel/administración & dosificación , Norgestrel/efectos adversos , Norgestrel/sangre , Norgestrel/farmacología , Progesterona/sangre , Adulto Joven
5.
Glia ; 66(2): 295-310, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29034506

RESUMEN

Norgestrel, a progesterone analogue, has demonstrated neuroprotective effects in a mouse model of retinitis pigmentosa. Neuroprotection is achieved in part through Norgestrels anti-inflammatory properties, alleviating detrimental microglial activity. Gliosis is a feature of many neurodegenerative diseases of the retina, including retinitis pigmentosa. Müller glia, a type of macroglia found in the retina, are major contributors of gliosis, characterized by the upregulation of glial fibrillary acidic protein (GFAP). Microglia-Müller glia crosstalk has been implicated in the initiation of gliosis. In the rd10 retina, increased microglial activity and gliotic events are observed prior to the onset of photoreceptor loss. We hypothesized that Norgestrels dampening effects on harmful microglial activity would consequently impact on gliosis. In the current study, we explore the role of microglia-Müller glia crosstalk in degeneration and Norgestrel-mediated neuroprotection in the rd10 retina. Norgestrels neuroprotective effects in the rd10 retina coincide with significant decreases in both microglial activity and Müller cell gliosis. Using a Müller glial cell line, rMC-1, and isolated microglia, we show that rd10 microglia stimulate GFAP production in rMC-1 cells. Norgestrel attenuates gliosis through direct actions on both microglia and Müller glia. Norgestrel reduces the release of harmful stimuli from microglia, such as interferon-γ, which might otherwise signal to Müller glia and stimulate gliosis. We propose that Norgestrel also targets Müller cell gliosis directly, by limiting the availability of pSTAT3, a known transcription factor for GFAP. These findings highlight an important aspect to Norgestrels neuroprotective effects in the diseased retina, in combating Müller cell gliosis.


Asunto(s)
Modelos Animales de Enfermedad , Células Ependimogliales/efectos de los fármacos , Gliosis/prevención & control , Microglía/efectos de los fármacos , Progesterona/uso terapéutico , Retinitis Pigmentosa/tratamiento farmacológico , Animales , Línea Celular , Células Cultivadas , Células Ependimogliales/metabolismo , Femenino , Gliosis/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Norgestrel/farmacología , Norgestrel/uso terapéutico , Progesterona/farmacología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo
6.
Chemosphere ; 190: 17-24, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28972919

RESUMEN

The aim of this study was to assess the adverse effects of synthetic progestin norgestrel (NGT) on the reproduction of zebrafish by measuring the egg production, histology and transcriptional expression profiles along the hypothalamic-pituitary-gonadal (HPG) axis in adult zebrafish. After a pre-exposure period of 7 days, adult zebrafish were exposed to 6, 29 and 69 ng L-1 NGT for 21 days. The results showed that exposure to 69 ng L-1 NGT led to a significant up-regulation of follicle stimulating hormone, beta polypeptide (fshb), luteinizing hormone, beta polypeptide (lhb), progesterone receptor (pgr), estrogen receptor 1 (esr1) and androgen receptor (ar) genes in the brains, as well as significant up-regulation of hydroxysteroid 20-beta dehydrogenase (hsd20b) and hydroxysteroid 11-beta dehydrogenase 2 (hsd11b2) genes and down-regulation of 11-beta-hydroxylase (cyp11b) gene in the ovaries of females. In the testes of males, an overall down-regulation of steroidogenic acute regulatory protein (star), cytochrome P450-mediated side-chain cleavage enzyme (cyp11a1), cyp11b, hsd20b, hydroxysteroid 17-beta dehydrogenase type 3 (hsd17b3), hsd11b2 and ar genes were observed following exposure to different treatments of NGT. These transcriptional alterations imply that NGT could exhibit the potent progestogenic and androgenic activities in zebrafish. Egg production as well as histology in the ovaries and testes was not affected by NGT. Taken together, the overall results demonstrated that NGT could significantly affect transcriptional expression levels of genes related to HPG axis in zebrafish, and whether that change translates to additional physiological effects is needed further research.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Norgestrel/farmacología , Reproducción/efectos de los fármacos , Pez Cebra/fisiología , Animales , Anticonceptivos Sintéticos Orales/farmacología , Femenino , Gonadotropinas Hipofisarias/genética , Hormonas Hipotalámicas/genética , Masculino , Norgestrel/metabolismo , Progestinas/fisiología , Receptores de Progesterona/genética , Pez Cebra/metabolismo
7.
Eur J Neurosci ; 46(1): 1663-1672, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28493650

RESUMEN

Retinitis pigmentosa (RP) is a group of hereditary retinal diseases, characterised by photoreceptor cell loss. Despite a substantial understanding of the mechanisms leading to cell death, an effective therapeutic strategy is sought. Our laboratory has previously demonstrated the neuroprotective properties of Norgestrel, a progesterone analogue, in the degenerating retina, mediated in part by the neurotrophic factor basic fibroblast growth factor (bFGF). In other retinal studies, we have also presented a pro-survival role for reactive oxygen species (ROS), downstream of bFGF. Thus, we hypothesized that Norgestrel utilises bFGF-driven ROS production to promote photoreceptor survival. Using the 661W photoreceptor-like cell line, we now show that Norgestrel, working through progesterone receptor membrane complex 1 (PGRMC1); generates an early burst of pro-survival bFGF-induced ROS. Using the rd10 mouse model of RP, we confirm that Norgestrel induces a similar early pro-survival increase in retinal ROS. Norgestrel-driven protection in the rd10 retina was attenuated in the presence of antioxidants. This study therefore presents an essential role for ROS signalling in Norgestrel-mediated neuroprotection in vitro and demonstrates that Norgestrel employs a similar pro-survival mechanism in the degenerating retina.


Asunto(s)
Fármacos Neuroprotectores/farmacología , Norgestrel/farmacología , Células Fotorreceptoras/metabolismo , Progesterona/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Retinitis Pigmentosa/metabolismo , Animales , Línea Celular , Femenino , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Fotorreceptoras/efectos de los fármacos , Receptores de Progesterona/metabolismo , Transducción de Señal
8.
Sci Rep ; 7: 43067, 2017 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-28216676

RESUMEN

Retinitis pigmentosa (RP) encompasses a group of retinal diseases resulting in photoreceptor loss and blindness. We have previously shown in the rd10 mouse model of RP, that rd10 microglia drive degeneration of viable neurons. Norgestrel, a progesterone analogue, primes viable neurons against potential microglial damage. In the current study we wished to investigate this neuroprotective effect further. We were particularly interested in the role of fractalkine-CX3CR1 signaling, previously shown to mediate photoreceptor-microglia crosstalk and promote survival in the rd10 retina. Norgestrel upregulates fractalkine-CX3CR1 signaling in the rd10 retina, coinciding with photoreceptor survival. We show that Norgestrel-treated photoreceptor-like cells, 661Ws, and C57 explants modulate rd10 microglial activity in co-culture, resulting in increased photoreceptor survival. Assessment of Norgestrel's neuroprotective effects when fractalkine was knocked-down in 661 W cells and release of fractalkine was reduced in rd10 explants confirms a crucial role for fractalkine-CX3CR1 signaling in Norgestrel-mediated neuroprotection. To further understand the role of fractalkine in neuroprotection, we assessed the release of 40 cytokines in fractalkine-treated rd10 microglia and explants. In both cases, treatment with fractalkine reduced a variety of pro-inflammatory cytokines. These findings further our understanding of Norgestrel's neuroprotective properties, capable of modulating harmful microglial activity indirectly through photoreceptors, leading to increased neuroprotection.


Asunto(s)
Receptor 1 de Quimiocinas CX3C/metabolismo , Quimiocina CX3CL1/metabolismo , Neuroprotección , Norgestrel/farmacología , Retina/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Citocinas , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Células Fotorreceptoras/efectos de los fármacos , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/fisiología , Progesterona/farmacología , Retina/efectos de los fármacos , Retina/fisiología
9.
Artículo en Inglés | MEDLINE | ID: mdl-28219785

RESUMEN

Synthetic estrogens and progestins are commonly used in human and veterinary medicine. After use, they reach aquatic environments via discharge of wastewaters from human and animals, thus posing potential risks to organisms. So far, very little is known about their combined effects in aquatic organisms. The aim of this study was to investigate the effects of binary mixtures of ethinylestradiol (EE2) and norgestrel (NGT) on embryonic zebrafish (Danio rerio) by measuring transcriptional alterations. Zebrafish embryos were exposed to EE2 and NGT alone or in combination at concentrations between 36 and 5513ngL-1 for 96h post-fertilization (hpf). The results showed that most of gene transcriptions of hypothalamic-pituitary-gonadal axis (e.g., Pgr, Mprα, Esr1, Esr2a, Vtg1, Ar, Cyp11b, Star, Gnrh3 and Fshb) and circadian rhythm signaling (e.g., Cry1a, Cry2a, Cry2b, Per3, Arntl1b, Arntl2, Clock1a, Cry3 and Cry4) displayed most pronounced alterations in the mixtures as compared to single EE2 and NGT exposures. This finding suggests exposure to the binary mixtures of EE2 and NGT produced significantly enhanced effects in fish as compared to single chemical exposures, and their coexistence could have significant environmental implications.


Asunto(s)
Etinilestradiol/farmacología , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Norgestrel/farmacología , Pez Cebra/genética , Análisis de Varianza , Animales , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Estrógenos/farmacología , Femenino , Masculino , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Pez Cebra/embriología , Proteínas de Pez Cebra/genética
10.
Artículo en Inglés | MEDLINE | ID: mdl-27174800

RESUMEN

Microsomal prostaglandin E synthase-1 (mPGES-1) is primarily expressed in granulosa cells (GCs) in the preovulatory follicle. Both prostaglandin E2 (PGE2) and progesterone (P4) are implicated in various reproductive functions. Here, we demonstrate that mPges-1 may be a direct downstream target gene of the P4 receptor and P4-stimulated PGE2 secretion can stimulate P4 production in a newly generated mouse GC line (GtsT). Treatment of GtsT cells with a P4 receptor agonist, norgestrel, markedly increased mPGES-1 expression detected by RT-PCR analysis. PGE2 secretion measured by an enzyme-linked immunosorbent assay was enhanced by P4 treatment. Luciferase assays revealed that the proximal promoter region of the mPges-1 gene was responsible for the effects of P4 treatment. Conversely, PGE2 treatment stimulated P4 secretion, which coordinated with mRNA expression of steroidogenic acute regulatory protein. Taken together, P4 may regulate mPGES-1 expression to increase PGE2 secretion and in turn P4 production. An autocrine loop between P4 and PGE2 might function to maintain the increased levels of both in GCs.


Asunto(s)
Dinoprostona/metabolismo , Retroalimentación Fisiológica , Células de la Granulosa/metabolismo , Progesterona/metabolismo , Prostaglandina-E Sintasas/genética , Receptores de Progesterona/genética , Animales , Comunicación Autocrina , Dinoprostona/farmacología , Femenino , Regulación de la Expresión Génica , Genes Reporteros , Células de la Granulosa/citología , Células de la Granulosa/efectos de los fármacos , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Microsomas/metabolismo , Norgestrel/farmacología , Cultivo Primario de Células , Progesterona/farmacología , Regiones Promotoras Genéticas , Prostaglandina-E Sintasas/metabolismo , Receptores de Progesterona/agonistas , Receptores de Progesterona/metabolismo , Transducción de Señal , Acetato de Tetradecanoilforbol/farmacología
11.
Mol Vis ; 22: 264-74, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27081297

RESUMEN

PURPOSE: Retinal degenerative conditions affect thousands of people worldwide. Retinitis pigmentosa (RP) is among the most common, but it is currently incurable. It is characterized by the progressive death of photoreceptor cells, eventually leading to blindness. Neurotrophic factors play an important role in such retinopathies, and much research has been performed on their use as treatments. Our group previously demonstrated the ability of the synthetic progestin norgestrel to rescue photoreceptors from cell death, the mechanism of which is believed to include upregulation of the neurotrophic factor basic fibroblast growth factor (bFGF). The objective of the present study was to investigate whether the protection provided by norgestrel is likely to be mediated by other neurotrophins. METHODS: The 661W photoreceptor cells and retinal explants from P30 to P40 wild-type (wt) C57BL/6 mice were treated with norgestrel over time. Homozygous rd10/rd10 mice that mimic the human form of RP were fed either a control or a norgestrel-containing diet. Changes in neurotrophic factor expression in response to norgestrel were detected with real-time PCR, western blotting, or immunofluorescence staining. Using specific siRNA, leukemia inhibitory factor (Lif) expression was knocked down in 661W photoreceptor cells that were stressed by serum starvation. Cells were treated with norgestrel followed by measurement of cell viability with (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) (MTS) assay. RESULTS: LIF, a potent neuroprotective cytokine, was found to be upregulated in response to norgestrel in vitro and in vivo. Upregulation of LIF in degenerating rd10 retinas coincided with preservation of the photoreceptor layer. We also found LIF was necessary for the norgestrel-mediated rescue of stressed photoreceptor cells from cell death in vitro. CONCLUSIONS: LIF was upregulated in response to norgestrel in all models studied and is necessary for the protective effects of norgestrel in vitro. The increase in LIF expression in rd10 mice undergoing retinal degeneration was concurrent with rescue of the photoreceptor cell layer. These results highlight the ability of norgestrel to induce prosurvival molecules in the compromised retina, underlining norgestrel's potential as a viable drug for treatment of RP.


Asunto(s)
Anticonceptivos Sintéticos Orales/farmacología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/fisiología , Factor Inhibidor de Leucemia/genética , Norgestrel/farmacología , Retinitis Pigmentosa/tratamiento farmacológico , Animales , Western Blotting , Supervivencia Celular , Células Cultivadas , Anticonceptivos Sintéticos Orales/síntesis química , Dieta , Técnica del Anticuerpo Fluorescente Indirecta , Factor Inhibidor de Leucemia/metabolismo , Ratones , Ratones Endogámicos C57BL , Norgestrel/síntesis química , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Transfección
12.
J Womens Health (Larchmt) ; 25(6): 638-45, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26871978

RESUMEN

BACKGROUND: Data on combined hormonal oral contraceptives' (OCs) effects on metabolic changes in women with polycystic ovary syndrome (PCOS) have been conflicting and were predominantly based on OCs with cyproterone acetate (unavailable in the United States) Most studies did not include normal women as controls. We compared metabolic changes before and after an OC commonly used in the United States between women with and without PCOS. METHODS: Ten PCOS and 20 control women took ethinyl estradiol 35 µg and norgestimate 0.18/0.215/0.25 mg. Fasting glucose and insulin, area-under-the-curve (AUC) glucose and insulin, insulin sensitivity (homeostatic model assessment of insulin sensitivity index [HOMA-ISI] and Matsuda index), insulinogenic index (Δinsulin0-30 minutes/Δglucose0-30 minutes), blood pressure, and lipids were evaluated at baseline and after three cycles of OC. RESULTS: At baseline, PCOS women had lower insulin sensitivity (Matsuda index p = 0.0093, HOMA-ISI p = 0.0397), higher fasting insulin (p = 0.0495), fasting glucose (p = 0.0393), AUC insulin (p = 0.0023), and triglycerides (p = 0.0044) versus controls. Baseline AUC glucose did not differ between PCOS women and controls. After 3 months of OC use, glucose tolerance worsened in PCOS women versus controls (p = 0.0468). Higher baseline androgens were predictive of worsened glucose tolerance, and a reduction of AUC insulin during OC use. The insulinogenic index significantly decreased in PCOS women (p < 0.01), while fasting insulin and insulin resistance significantly worsened in control women. CONCLUSION: Women with PCOS exhibited worsened glucose tolerance (demonstrated by AUC glucose) after 3 months of a commonly used OC compared with control women. Larger studies with longer follow-up should confirm these findings.


Asunto(s)
Glucemia/metabolismo , Anticonceptivos Orales Combinados/farmacología , Anticonceptivos Hormonales Orales/farmacología , Anticonceptivos Orales/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Norgestrel/análogos & derivados , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Síndrome del Ovario Poliquístico/metabolismo , Adulto , Glucemia/efectos de los fármacos , Carbohidratos/sangre , Estudios de Casos y Controles , Anticonceptivos Orales/administración & dosificación , Anticonceptivos Orales Combinados/administración & dosificación , Anticonceptivos Hormonales Orales/administración & dosificación , Relación Dosis-Respuesta a Droga , Estrógenos/administración & dosificación , Etinilestradiol/administración & dosificación , Etinilestradiol/farmacología , Femenino , Prueba de Tolerancia a la Glucosa , Hormonas Esteroides Gonadales/sangre , Humanos , Insulina/sangre , Lípidos/sangre , Norgestrel/administración & dosificación , Norgestrel/farmacología , Síndrome del Ovario Poliquístico/fisiopatología
13.
Eur J Neurosci ; 43(7): 899-911, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26750157

RESUMEN

The synthetic progesterone Norgestrel has been shown to have proven neuroprotective efficacy in two distinct models of retinitis pigmentosa: the rd10/rd10 (B6.CXBI-Pde6b(rd10)/J) mouse model and the Balb/c light-damage model. However, the cellular mechanism underlying this neuroprotection is still largely unknown. Therefore, this study aimed to examine the downstream signalling pathways associated with Norgestrel both in vitro and ex vivo. In this work, we identify the potential of Norgestrel to rescue stressed 661W photoreceptor-like cells and ex vivo retinal explants from cell death over 24 h. Norgestel is thought to work through an upregulation of neuroprotective basic fibroblast growth factor (bFGF). Analysis of 661W cells in vitro by real-time polymerase chain reaction (rt-PCR), enzyme-linked immunosorbent assay (ELISA) and Western blotting revealed an upregulation of bFGF in response to Norgestrel over 6 h. Specific siRNA knockdown of bFGF abrogated the protective properties of Norgestrel on damaged photoreceptors, thus highlighting the crucial importance of bFGF in Norgestrel-mediated protection. Furthermore, Norgestrel initiated a bFGF-dependent inactivation of glycogen synthase kinase 3ß (GSK3ß) through phosphorylation at serine 9. The effects of Norgestrel on GSK3ß were dependent on protein kinase A (PKA) pathway activation. Specific inhibition of both the PKA and GSK3ß pathways prevented Norgestrel-mediated neuroprotection of stressed photoreceptor cells in vitro. Involvement of the PKA pathway following Norgestrel treatment was also confirmed ex vivo. Therefore, these results indicate that the protective efficacy of Norgestrel is, at least in part, due to the bFGF-mediated activation of the PKA pathway, with subsequent inactivation of GSK3ß.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Fármacos Neuroprotectores/farmacología , Norgestrel/farmacología , Células Fotorreceptoras/metabolismo , Animales , Línea Celular , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Células Fotorreceptoras/efectos de los fármacos , Transducción de Señal
14.
Eur J Pharmacol ; 769: 48-54, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26506558

RESUMEN

Clinical studies have shown that the use of combined oral contraceptive in pre-menopausal women is associated with fluid retention. However, the molecular mechanism is still elusive. We hypothesized that combined oral contraceptive (COC) ethinyl estradiol (EE) and norgestrel (N) synergistically activates mineralocorticoid receptor (MR) through histone code modifications. Twelve-week-old female Sprague-Dawley rats were treated with olive oil (control), a combination of 0.1µg EE and 1.0µg N (low COC) or 1.0µg EE and 10.0µg N (high COC) as well as 0.1 or 1.0µg EE and 1.0 or 10.0µg N daily for 6 weeks. Expression of MR target genes in kidney cortex was determined by quantitative real-time polymerase chain reaction. MR was quantified by western blot. Recruitment of MR and RNA polymerase II (Pol II) on promoters of target genes as well as histone code modifications was analyzed by chromatin immunoprecipitation assay. Treatment with COC increased renal cortical expression of MR target genes such as serum and glucocorticoid-regulated kinase 1 (Sgk-1), glucocorticoid-induced leucine zipper (Gilz), epithelial Na(+)channel (Enac) and Na(+)-K(+)-ATPase subunit α1 (Atp1a1). Although COC increased neither serum aldosterone nor MR expression in kidney cortex, it increased recruitment of MR and Pol II in parallel with increased H3Ac and H3K4me3 on the promoter regions of MR target genes. However, treatment with EE or N alone did not affect renal cortical expression of Sgk-1, Gilz, Enac or Atp1a1. These results indicate that COC synergistically activates MR through histone code modifications.


Asunto(s)
Anticonceptivos Orales Combinados/farmacología , Código de Histonas/efectos de los fármacos , Receptores de Mineralocorticoides/química , Receptores de Mineralocorticoides/metabolismo , Animales , Secuencia de Bases , Sinergismo Farmacológico , Etinilestradiol/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Inmediatas-Precoces/genética , Corteza Renal/efectos de los fármacos , Corteza Renal/metabolismo , Norgestrel/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas/efectos de los fármacos , ARN Polimerasa II/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Transcripción/genética
15.
Gynecol Endocrinol ; 31(6): 487-90, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25970022

RESUMEN

BACKGROUND: Acne occurs because the sebaceous glands are overstimulated by high levels of androgens or are hypersensitive to normal levels of testosterone. In women with mild or moderate acne, the association of norgestimate (NG), and ethinyl estradiol (EE) is an effective treatment. This is related to the effect of oral contraceptives on androgen production and transport and the antiandrogenic properties of NG itself. DESIGN: The present work was undertaken to find out whether NG and its derivative, 17-deacetylnorgestimate(dNG), present steroid activities other than antiandrogen activities, using human progesterone receptor(PR), estrogen receptor α(ERα) and ß(ERß), glucocorticoid receptor(GR) and mineralocorticoid receptor(MR)-responsive cell lines. RESULTS: We confirmed that NG and its metabolite were progestogen partial agonists (EC50 of 13 and 11.1 nM) and ERα selective agonists (EC50 of 30.4 and 43.4 nM), as well as full antagonists of low affinity for GR (IC50 of 325 and 255 nM) and moderate affinity for MR (IC50 of 81.2 and 83.7). CONCLUSION: We demonstrated that NG and dNG have full progestogen and weak estrogenic (through ERα) properties, which could explain in part the efficacy of NG in association with EE for the treatment of moderate acne in women. Moreover, their antagonist MR activity might have a favorable impact on cardiovascular risk, atherosclerosis and lipid profiles.


Asunto(s)
Anticonceptivos Sintéticos Orales/farmacología , Receptor alfa de Estrógeno/agonistas , Antagonistas de Receptores de Mineralocorticoides/farmacología , Norgestrel/análogos & derivados , Progestinas/agonistas , Receptores de Glucocorticoides/antagonistas & inhibidores , Línea Celular , Humanos , Norgestrel/farmacología
16.
Artículo en Inglés | MEDLINE | ID: mdl-25277675

RESUMEN

The aim of this study was to investigate the effects of progestins on the hypothalamic-pituitary-thyroid (HPT) axis in the early stage of zebrafish. Zebrafish embryos were exposed to progesterone (P4) or norgestrel (NGT) at 5, 50 and 100 ng L(-1) for 144 h post fertilization (hpf), and the transcriptional levels of target genes along the hypothalamic-pituitary-thyroid axis were determined daily. The results showed that P4 had only minor effects on the mRNA expression of thyroglobulin (Tg), iodothyronine deiodinase type Ι (Dio1) and thyroid hormone receptor ß (Thrb) genes. Similarly, the effects of NGT on transcripts of thyrotropin-releasing hormone (Trh), Dio1, iodothyronine deiodinase type II (Dio2) and thyroid hormone receptor α (Thra) genes were generally low. In addition, NGT resulted in some alterations of Tg and Thrb transcripts at different time points. However, a strong induction of Nis mRNA by P4 and NGT was observed in zebrafish embryos-larvae. The overall results showed that besides Nis no effects on the hypothalamic-pituitary-thyroid (HPT) axis are observed following exposure to P4 and NGT, which imply that both P4 and NGT have potential effects on the thyroid endocrine system by inducing transcript of Nis gene during the early stage of zebrafish.


Asunto(s)
Embrión no Mamífero/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Norgestrel/farmacología , Progesterona/farmacología , Pez Cebra/embriología , Animales , Larva/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Glándula Tiroides/efectos de los fármacos , Transcripción Genética
17.
Eur J Contracept Reprod Health Care ; 18(4): 274-83, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23638631

RESUMEN

OBJECTIVES: To determine the effect of oestradiol valerate/dienogest (E2V/DNG) versus ethinylestradiol/norgestimate (EE/NGM) on hormone-withdrawal associated symptoms (HWAS) in otherwise healthy women who had experienced at least one of these symptoms when using 21/7-day combined oral contraceptives (COCs). METHODS: This phase III, parallel-group study randomised 409 women aged 18 to 50 years to E2V/DNG or EE/NGM. The primary efficacy variable was the change from baseline to cycle 6 in the average of the three highest visual analogue scale values for headache and/or pelvic pain during cycle days 22 to 28. RESULTS: In total, 395 were included in the full analysis set (E2V/DNG, n = 191; EE/NGM, n = 204). E2V/DNG reduced the symptoms of headache or pelvic pain during cycle days 22 to 28 from baseline to cycle 6 to a significantly greater extent than EE/NGM (mean decrease 43.6 vs. 35.5 mm; p = 0.0024). Both treatments were well tolerated with a similar proportion of women experiencing adverse events that were considered at least possibly related to treatment (35% E2V/DNG vs. 34% EE/NGM). CONCLUSIONS: E2V/DNG reduces the frequency and intensity of headache and pelvic pain to a greater extent than EE/NGM, and may be a good option for women susceptible to HWAS with conventional 21/7-day COCs.


Asunto(s)
Anticonceptivos Orales/efectos adversos , Estradiol/análogos & derivados , Etinilestradiol/efectos adversos , Cefalea/etiología , Nandrolona/análogos & derivados , Norgestrel/análogos & derivados , Dolor Pélvico/etiología , Síndrome de Abstinencia a Sustancias/etiología , Adulto , Anticonceptivos Orales/farmacología , Método Doble Ciego , Combinación de Medicamentos , Estradiol/efectos adversos , Estradiol/farmacología , Etinilestradiol/farmacología , Femenino , Humanos , Menstruación/efectos de los fármacos , Nandrolona/efectos adversos , Nandrolona/farmacología , Norgestrel/efectos adversos , Norgestrel/farmacología , Resultado del Tratamiento , Adulto Joven
18.
PLoS One ; 7(4): e35393, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22530015

RESUMEN

TRPC channels are a family of nonselective cation channels that regulate ion homeostasis and intracellular Ca(2+) signaling in numerous cell types. Important physiological functions such as vasoregulation, neuronal growth, and pheromone recognition have been assigned to this class of ion channels. Despite their physiological relevance, few selective pharmacological tools are available to study TRPC channel function. We, therefore, screened a selection of pharmacologically active compounds for TRPC modulating activity. We found that the synthetic gestagen norgestimate inhibited diacylglycerol-sensitive TRPC3 and TRPC6 with IC(50)s of 3-5 µM, while half-maximal inhibition of TRPC5 required significantly higher compound concentrations (>10 µM). Norgestimate blocked TRPC-mediated vasopressin-induced cation currents in A7r5 smooth muscle cells and caused vasorelaxation of isolated rat aorta, indicating that norgestimate could be an interesting tool for the investigation of TRP channel function in native cells and tissues. The steroid hormone progesterone, which is structurally related to norgestimate, also inhibited TRPC channel activity with IC(50)s ranging from 6 to 18 µM but showed little subtype selectivity. Thus, TRPC channel inhibition by high gestational levels of progesterone may contribute to the physiological decrease of uterine contractility and immunosuppression during pregnancy.


Asunto(s)
Diglicéridos/metabolismo , Esteroides/farmacología , Canales Catiónicos TRPC/antagonistas & inhibidores , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Calcio/metabolismo , Humanos , Masculino , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Norgestrel/análogos & derivados , Norgestrel/farmacología , Progesterona/farmacología , Ratas , Ratas Wistar , Receptores de Vasopresinas/metabolismo , Esteroides/síntesis química , Canales Catiónicos TRPC/metabolismo , Canal Catiónico TRPC6 , Vasopresinas/farmacología
19.
Biochem J ; 442(2): 345-56, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22115192

RESUMEN

PFKFB (6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase) catalyses the synthesis and degradation of Fru-2,6-P2 (fructose-2,6-bisphosphate), a key modulator of glycolysis and gluconeogenesis. The PFKFB3 gene is extensively involved in cell proliferation owing to its key role in carbohydrate metabolism. In the present study we analyse its mechanism of regulation by progestins in breast cancer cells. We report that exposure of T47D cells to synthetic progestins (ORG2058 or norgestrel) leads to a rapid increase in Fru-2,6-P2 concentration. Our Western blot results are compatible with a short-term activation due to PFKFB3 isoenzyme phosphorylation and a long-term sustained action due to increased PFKFB3 protein levels. Transient transfection of T47D cells with deleted gene promoter constructs allowed us to identify a PRE (progesterone-response element) to which PR (progesterone receptor) binds and thus transactivates PFKFB3 gene transcription. PR expression in the PR-negative cell line MDA-MB-231 induces endogenous PFKFB3 expression in response to norgestrel. Direct binding of PR to the PRE box (-3490 nt) was confirmed by ChIP (chromatin immunoprecipiation) experiments. A dual mechanism affecting PFKFB3 protein and gene regulation operates in order to assure glycolysis in breast cancer cells. An immediate early response through the ERK (extracellular-signal-regulated kinase)/RSK (ribosomal S6 kinase) pathway leading to phosphorylation of PFKFB3 on Ser461 is followed by activation of mRNA transcription via cis-acting sequences on the PFKFB3 promoter.


Asunto(s)
Neoplasias de la Mama/metabolismo , Fosfofructoquinasa-2/metabolismo , Congéneres de la Progesterona/farmacología , Secuencia de Bases , Neoplasias de la Mama/genética , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas , Norgestrel/farmacología , Fosfofructoquinasa-2/genética , Pregnenodionas/farmacología , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Receptores de Progesterona/metabolismo
20.
J Neurochem ; 118(5): 915-27, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21689103

RESUMEN

Retinal degenerations such as Retinitis Pigmentosa remain difficult to treat given the diverse array of genes responsible for their aetiology. Rather than concentrate on specific genes, our focus is on identifying therapeutic avenues for the treatment of retinal disease that target general survival mechanisms or pathways. Norgestrel is a synthetic progestin commonly used in hormonal contraception. Here, we report a novel anti-apoptotic role for Norgestrel in diseased mouse retinas in vivo. Dosing with Norgestrel protects photoreceptor cells from undergoing apoptosis in two distinct models of retinal degeneration; the light damage model and the Pde6b(rd10) model. Photoreceptor rescue was assessed by analysis of cell number, structural integrity and function. Improvements in cell survival of up to 70% were achieved in both disease models, indicating that apoptosis had been halted or at least delayed. A speculative mechanism of action for Norgestrel involves activation of survival pathways in the retina. Indeed, Norgestrel increases the expression of basic fibroblast growth factor which is known to both promote cell survival and inhibit apoptosis. In summary, our results demonstrate significant protection of photoreceptor cells which may be attributed to Norgestrel mediated activation of endogenous survival pathways within the retina.


Asunto(s)
Apoptosis/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Norgestrel/farmacología , Células Fotorreceptoras/efectos de los fármacos , Retina/citología , Degeneración Retiniana/tratamiento farmacológico , Animales , Animales Recién Nacidos , Recuento de Células/métodos , Modelos Animales de Enfermedad , Electrorretinografía , Factores de Crecimiento de Fibroblastos/metabolismo , Etiquetado Corte-Fin in Situ/métodos , Técnicas In Vitro , Luz/efectos adversos , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Norgestrel/uso terapéutico , Congéneres de la Progesterona/farmacología , Congéneres de la Progesterona/uso terapéutico , Degeneración Retiniana/etiología , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA