Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(5)2021 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-33806348

RESUMEN

There is substantial genetic variation for common traits associated with reproductive lifespan and for common diseases influencing female fertility. Progress in high-throughput sequencing and genome-wide association studies (GWAS) have transformed our understanding of common genetic risk factors for complex traits and diseases influencing reproductive lifespan and fertility. The data emerging from GWAS demonstrate the utility of genetics to explain epidemiological observations, revealing shared biological pathways linking puberty timing, fertility, reproductive ageing and health outcomes. The observations also identify unique genetic risk factors specific to different reproductive diseases impacting on female fertility. Sequencing in patients with primary ovarian insufficiency (POI) have identified mutations in a large number of genes while GWAS have revealed shared genetic risk factors for POI and ovarian ageing. Studies on age at menopause implicate DNA damage/repair genes with implications for follicle health and ageing. In addition to the discovery of individual genes and pathways, the increasingly powerful studies on common genetic risk factors help interpret the underlying relationships and direction of causation in the regulation of reproductive lifespan, fertility and related traits.


Asunto(s)
Fertilidad/genética , Reproducción/genética , Envejecimiento/genética , Envejecimiento/fisiología , Femenino , Fertilidad/fisiología , Hormona Folículo Estimulante Humana/genética , Hormona Folículo Estimulante Humana/fisiología , Variación Genética , Estudio de Asociación del Genoma Completo , Humanos , Longevidad/genética , Longevidad/fisiología , Hormona Luteinizante/genética , Hormona Luteinizante/fisiología , Menopausia/genética , Menopausia/fisiología , Polimorfismo de Nucleótido Simple , Reproducción/fisiología , Factores de Riesgo
2.
Hum Reprod ; 32(7): 1402-1409, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28472321

RESUMEN

STUDY QUESTION: Do live birth rates (LBRs) differ between fresh embryo transfer (fresh ET) cycles and their subsequent paired frozen ET (FET) cycles, when comparing cycles based on the total FSH dose used during the fresh cycle? SUMMARY ANSWER: When compared to the paired frozen embryo transfer cycles, the LBR in the fresh cycle of the highest total FSH dose group (>2500 IU) was reduced by 38%. WHAT IS KNOWN ALREADY: There may be a negative association with high gonadotropin doses and LBR after fresh ET. It is unknown whether a similar effect is seen in FET cycles, which are done with increasing frequency. STUDY DESIGN, SIZE, DURATION: In this retrospective observational paired study, we studied IVF cycles between 10 January 2005 and 19 September 2015, for all patients who underwent a fresh, autologous IVF cycle that resulted in at least one fresh ET and at least one FET. PARTICIPANTS/MATERIALS, SETTING, METHODS: The study included 862 women, treated in our academic medical centre, who underwent 935 fresh ET and 1274 FET cycles. Cycles were allocated into three groups based on the total gonadotropin dose they received during their fresh IVF cycle: Group 1 (≤1800 IU FSH), Group 2 (1801-2500 IU), Group 3 (>2500 IU). The primary outcome was LBR after fresh ET and its subsequent paired FET(s), as well as LBR among fresh ETs and FETs as independent samples, based on the total FSH dose used. Implantation rates obtained from fresh and FET cycles were also compared. MAIN RESULTS AND THE ROLE OF CHANCE: The unadjusted fresh LBR was similar between Groups 1 and 2 (46.0% [95% CI: 40.4-51.6] versus 43.8% [38.3-49.4], respectively) but significantly lower in Group 3 (34.4% [29.5-39.8]). The unadjusted frozen transfer LBR was similar among all groups (51.4% [46.7-56.1] versus 46.3% [41.3-51.4] versus 47.5% [42.5-52.4], respectively). When logistic regression analysis with generalized estimating equations was used to control for confounders, the adjusted LBR was found to be similar between the groups both for fresh (odds ratio [OR] = 0.97 [95% CI: 0.61-1.56] Group 2 versus Group 1, OR = 0.69 [0.39-1.21] Group 3 versus Group 1) and FET cycles (OR = 0.87 [0.58-1.31] Group 2 versus Group 1, OR = 0.95 [0.58-1.55] Group 3 versus Group 1). However, for Group 3, the adjusted fresh LBR was 38% lower than its subsequent frozen transfer LBR (OR = 0.62 [0.41-0.93]); this was a statistically significant difference, which was not observed in Group 1 (OR = 0.85 [0.56-1.27]) or Group 2 (OR = 0.95 [0.64-1.41]). LIMITATIONS, REASONS FOR CAUTION: This study is a retrospective cohort, with all of the associated inherent biases. WIDER IMPLICATIONS OF THE FINDINGS: Fresh LBR is negatively impacted by a high dose of total FSH used, as compared to the LBR in subsequent paired FET cycles. Frozen transfer LBR seems unaffected by the total FSH dose used in the fresh cycle, suggesting that the endometrium may be adversely affected, probably indirectly, by high dose gonadotropin use in the fresh IVF cycle only. STUDY FUNDING/COMPETING INTEREST(S): No funding source was used for the completion of this project. There are no conflicts of interest.


Asunto(s)
Transferencia de Embrión/efectos adversos , Fármacos para la Fertilidad Femenina/administración & dosificación , Hormona Folículo Estimulante Humana/administración & dosificación , Infertilidad Femenina/terapia , Inducción de la Ovulación/efectos adversos , Centros Médicos Académicos , Adulto , Tasa de Natalidad , Estudios de Cohortes , Criopreservación , Relación Dosis-Respuesta a Droga , Femenino , Fármacos para la Fertilidad Femenina/efectos adversos , Fármacos para la Fertilidad Femenina/uso terapéutico , Fertilización In Vitro/efectos adversos , Hormona Folículo Estimulante Humana/efectos adversos , Hormona Folículo Estimulante Humana/genética , Hormona Folículo Estimulante Humana/uso terapéutico , Humanos , Iowa/epidemiología , Embarazo , Índice de Embarazo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico , Reproducibilidad de los Resultados , Estudios Retrospectivos
3.
Reprod Fertil Dev ; 29(7): 1426-1434, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27328025

RESUMEN

Androgens synergise with FSH in female reproduction but the nature of their interaction in ovarian function and fertility is not clear. In the present study, we investigated this interaction, notably whether higher endogenous FSH can overcome defective androgen actions in androgen receptor (AR)-knockout (ARKO) mice. We generated and investigated the reproductive function of mutant mice exhibiting AR resistance with or without expression of human transgenic FSH (Tg-FSH). On the background of inactivated AR signalling, which alone resulted in irregular oestrous cycles and reduced pups per litter, ovulation rates and antral follicle health, Tg-FSH expression restored follicle health, ovulation rates and litter size to wild-type levels. However, Tg-FSH was only able to partially rectify the abnormal oestrous cycles observed in ARKO females. Hence, elevated endogenous FSH rescued the intraovarian defects, and partially rescued the extraovarian defects due to androgen insensitivity. In addition, the observed increase in litter size in Tg-FSH females was not observed in the presence of AR signalling inactivation. In summary, the findings of the present study reveal that FSH can rescue impaired female fertility and ovarian function due to androgen insensitivity in female ARKO mice by maintaining follicle health and ovulation rates, and thereby optimal female fertility.


Asunto(s)
Hormona Folículo Estimulante Humana/genética , Hormona Folículo Estimulante Humana/fisiología , Infertilidad Femenina/terapia , Receptores Androgénicos/deficiencia , Animales , Modelos Animales de Enfermedad , Estro , Femenino , Fertilidad/genética , Fertilidad/fisiología , Humanos , Infertilidad Femenina/genética , Infertilidad Femenina/fisiopatología , Tamaño de la Camada , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ovario/patología , Ovario/fisiopatología , Embarazo , Receptores Androgénicos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
4.
Zhonghua Nan Ke Xue ; 23(12): 1121-1126, 2017 Dec.
Artículo en Zh | MEDLINE | ID: mdl-29738186

RESUMEN

OBJECTIVE: To assess the association of the FSHR Thr307Ala-Asn680Ser gene polymorphism with male infertility. METHODS: We searched Pubmed, EMBASE, Web of Science, CNKI, and WANFANG databases for literature on the correlation of the FSHR Thr307Ala-Asn680Ser gene polymorphism with male infertility published from 2005 to the present time. According to the inclusion criteria, we included 12 epidemiological case-control studies and subjected them to a comprehensive analysis with the Stata11.0 software. RESULTS: A total of 2 893 male infertility patients and 3 312 controls were involved in the 12 studies. The Thr307Ala (rs6165) gene polymorphism was shown to be a risk factor for male infertility among the three comparison models (homozygous comparison model, hybrid comparison model and dominant comparison model), with the pooled odds ratios (OR) of 1.26 (95% CI: 1.03-1.54, P = 0.023), 1.18 (95% CI: 1.03-1.36, P = 0.018), and 1.20 (95% CI: 1.05-1.37, P = 0.006), respectively. And the Asn680Ser(rs6166) polymorphism was a risk factor for male infertility in the homozygous comparison and recessive comparison models, with the pooled ORs of 1.24, (95% CI: 1.05-1.45, P = 0.009) and 1.20 (95% CI: 1.04-1.39, P = 0.013), respectively. Layered meta-analysis showed that in the homozygous comparison model, the Thr307Ala-Asn680Ser polymorphism is a risk factor for male infertility in the white population, with the OR of 1.37 (95% CI: 1.03-1.82, P = 0.003) and 1.21 (95% CI: 1.00-1.47, P = 0.048), respectively. CONCLUSIONS: In the homozygous model (GG vs AA), the FSHRThr307Ala-Asn680Ser gene polymorphism might be a protective factor against male infertility.


Asunto(s)
Hormona Folículo Estimulante Humana/genética , Infertilidad Masculina/genética , Polimorfismo Genético , Estudios de Casos y Controles , Homocigoto , Humanos , Masculino , Factores de Riesgo
5.
Reproduction ; 149(1): R35-48, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25269615

RESUMEN

Despite extensive research, genetic basis of premature ovarian failure (POF) and ovarian cancer still remains elusive. It is indeed paradoxical that scientists searched for mutations in FSH receptor (FSHR) expressed on granulosa cells, whereas more than 90% of cancers arise in ovary surface epithelium (OSE). Two distinct populations of stem cells including very small embryonic-like stem cells (VSELs) and ovarian stem cells (OSCs) exist in OSE, are responsible for neo-oogenesis and primordial follicle assembly in adult life, and are modulated by FSH via its alternatively spliced receptor variant FSHR3 (growth factor type 1 receptor acting via calcium signaling and the ERK/MAPK pathway). Any defect in FSH-FSHR3-stem cell interaction in OSE may affect folliculogenesis and thus result in POF. Ovarian aging is associated with a compromised microenvironment that does not support stem cell differentiation into oocytes and further folliculogenesis. FSH exerts a mitogenic effect on OSE and elevated FSH levels associated with advanced age may provide a continuous trigger for stem cells to proliferate resulting in cancer, thus supporting gonadotropin theory for ovarian cancer. Present review is an attempt to put adult ovarian biology, POF, aging, and cancer in the perspective of FSH-FSHR3-stem cell network that functions in OSE. This hypothesis is further supported by the recent understanding that: i) cancer is a stem cell disease and OSE is the niche for ovarian cancer stem cells; ii) ovarian OCT4-positive stem cells are regulated by FSH; and iii) OCT4 along with LIN28 and BMP4 are highly expressed in ovarian cancers.


Asunto(s)
Envejecimiento , Epitelio/patología , Mutación/genética , Neoplasias Ováricas/patología , Ovario/patología , Insuficiencia Ovárica Primaria/patología , Células Madre/patología , Adulto , Epitelio/metabolismo , Femenino , Hormona Folículo Estimulante Humana/genética , Hormona Folículo Estimulante Humana/metabolismo , Humanos , Neoplasias Ováricas/metabolismo , Ovario/metabolismo , Insuficiencia Ovárica Primaria/metabolismo , Receptores de HFE/genética , Receptores de HFE/metabolismo , Células Madre/metabolismo
6.
Reprod Biol Endocrinol ; 12: 52, 2014 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-24942155

RESUMEN

BACKGROUND: This Phase IV, open-label, multicentre, randomized study (MEnTOR) compared two low-dose recombinant human follicle-stimulating hormone (r-hFSH) protocols for ovulation induction. METHODS: This study was conducted in six Middle Eastern countries between March 2009 and March 2011. Eligible women (18-37 years), with World Health Organization Group II anovulatory infertility, were randomized to receive r-hFSH (starting daily dose: 75 IU) as a chronic low-dose (CLD) (37.5 IU dose increase on Day 14) or low-dose (LD) (37.5 IU dose increase on Day 7) protocol if no follicles were ≥ 10 mm. The maximum r-hFSH daily dose permitted was 225 IU/day. The total length of ovarian stimulation could not exceed 35 days, unless ultrasound assessment suggested imminent follicular growth and maturation. Patients underwent only one treatment cycle. Primary endpoint: incidence of mono-follicular development. Secondary endpoints included: stimulation duration and rates of bi-follicular development; human chorionic gonadotrophin administration rate; clinical pregnancy; and cycle cancellation (owing to inadequate response). Adverse events (AEs) were recorded. The primary efficacy analysis was performed using data from all patients who received at least one dose of correct study medication, had at least one efficacy assessment, and no protocol violations at treatment start (CLD group, n=122; LD group, n=125). RESULTS: Mono-follicular development rates (primary endpoint) were similar in both groups (CLD: 56.6% [69/122] versus LD: 55.2% [69/125], p=0.93; primary efficacy analysis population). Similarly, there were no significant differences between groups in bi-follicular development, clinical pregnancy or cycle cancellation (inadequate response) rates. In patients who received human chorionic gonadotrophin injections, the mean duration of stimulation was 13.7 days in the CLD group and 12.9 days in the LD group. Clinical pregnancy rates for those patients who received an hCG injection were similar in both groups (CLD: 20.2% [19/94] versus LD: 19.8% [18/91], p=0.94; primary efficacy analysis population). Most AEs were mild in severity. Only one case of ovarian hyperstimulation syndrome was reported (mild; CLD group). CONCLUSIONS: Efficacy and safety outcomes were similar for the two protocols.


Asunto(s)
Anovulación/tratamiento farmacológico , Fármacos para la Fertilidad Femenina/administración & dosificación , Hormona Folículo Estimulante Humana/administración & dosificación , Infertilidad Femenina/prevención & control , Folículo Ovárico/efectos de los fármacos , Inducción de la Ovulación , Adolescente , Adulto , Anovulación/diagnóstico por imagen , Anovulación/patología , Anovulación/fisiopatología , Esquema de Medicación , Monitoreo de Drogas , Femenino , Fármacos para la Fertilidad Femenina/efectos adversos , Fármacos para la Fertilidad Femenina/uso terapéutico , Hormona Folículo Estimulante Humana/efectos adversos , Hormona Folículo Estimulante Humana/genética , Hormona Folículo Estimulante Humana/uso terapéutico , Humanos , Infertilidad Femenina/etiología , Perdida de Seguimiento , Medio Oriente/epidemiología , Folículo Ovárico/diagnóstico por imagen , Folículo Ovárico/patología , Síndrome de Hiperestimulación Ovárica/prevención & control , Pacientes Desistentes del Tratamiento , Embarazo , Índice de Embarazo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapéutico , Ultrasonografía , Adulto Joven
7.
Reprod Biol Endocrinol ; 12: 128, 2014 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-25543693

RESUMEN

In the menstrual cycle, the mid-cycle surge of gonadotropins (both luteinising hormone [LH] and follicle-stimulating hormone [FSH]) signals the initiation of the periovulatory interval, during which the follicle augments progesterone production and begins to luteinise, ultimately leading to the rupture of the follicle wall and the release of an oocyte. The administration of gonadotropins in controlled ovarian stimulation (COS) leads to supraphysiological steroid concentrations of a very different profile compared with those seen during natural cycles. It has been suggested that these high steroid concentrations cause alterations in endometrial development, affecting oocyte viability in assisted reproductive technology. Furthermore, it has been proposed that elevated progesterone levels have a negative effect on the reproductive outcome of COS. This may arise from an asynchrony between embryo stage and endometrium status at the window of implantation. The regulation of progesterone production by the developing follicles during COS is a complicated interplay of hormonal systems involving the theca and granulosa cells, and the effect of the actions of both LH and FSH. The present paper reviews current knowledge of the regulation of progesterone in the human ovary during the follicular phase and highlights areas where knowledge remains limited. In this review, we provide in-depth information outlining the regulation and function of gonadotropins in the complicated area of steroidogenesis. Based on current evidence, it is not clear whether the high levels of progesterone produced during COS have detrimental effects on fertility.


Asunto(s)
Medicina Basada en la Evidencia , Infertilidad Femenina/terapia , Modelos Biológicos , Ovario/efectos de los fármacos , Inducción de la Ovulación/métodos , Progesterona/metabolismo , Animales , Femenino , Hormona Folículo Estimulante Humana/efectos adversos , Hormona Folículo Estimulante Humana/genética , Hormona Folículo Estimulante Humana/metabolismo , Hormona Folículo Estimulante Humana/farmacología , Humanos , Infertilidad Femenina/metabolismo , Hormona Luteinizante/efectos adversos , Hormona Luteinizante/genética , Hormona Luteinizante/metabolismo , Hormona Luteinizante/farmacología , Ovario/enzimología , Ovario/metabolismo , Ovulación/efectos de los fármacos , Ovulación/metabolismo , Inducción de la Ovulación/efectos adversos , Progesterona/agonistas , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología
8.
Gynecol Endocrinol ; 30(6): 444-50, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24576226

RESUMEN

The aim was to compare ovarian response and clinical outcome of potential high-responders after stimulation with highly purified menotropin (HP-hMG) or recombinant follicle-stimulating hormone (rFSH) for in vitro fertilisation/intracytoplasmic sperm injection. Retrospective analysis was performed on data collected in two randomized controlled trials, one conducted following a long GnRH agonist protocol and the other with an antagonist protocol. Potential high-responders (n = 155 and n = 188 in the agonist and antagonist protocol, respectively) were defined as having an initial anti-Müllerian hormone (AMH) value >75th percentile (5.2 ng/ml). In both protocols, HP-hMG stimulation in women in the high AMH category was associated with a significantly lower occurrence of high response (≥15 oocytes retrieved) than rFSH stimulation; 33% versus 51% (p = 0.025) and 31% versus 49% (p = 0.015) in the long agonist and antagonist protocol, respectively. In the potential high-responder women, trends for improved live birth rate were observed with HP-hMG compared with rFSH (long agonist protocol: 33% versus 20%, p = 0.074; antagonist protocol: 34% versus 23%, p = 0.075; overall population: 34% versus 22%, p = 0.012). In conclusion, the type of gonadotropin used for ovarian stimulation influences high-response rates and potentially clinical outcome in women identified as potential high-responders.


Asunto(s)
Hormona Antimülleriana/sangre , Fármacos para la Fertilidad Femenina/efectos adversos , Hormona Folículo Estimulante Humana/efectos adversos , Menotropinas/efectos adversos , Síndrome de Hiperestimulación Ovárica/epidemiología , Ovario/efectos de los fármacos , Inducción de la Ovulación/efectos adversos , Adulto , Biomarcadores/sangre , Transferencia de Embrión , Femenino , Fertilización In Vitro , Hormona Folículo Estimulante Humana/genética , Hormona Liberadora de Gonadotropina/agonistas , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Humanos , Infertilidad Femenina/terapia , Nacimiento Vivo , Síndrome de Hiperestimulación Ovárica/sangre , Ovario/metabolismo , Ovario/fisiopatología , Embarazo , Proteínas Recombinantes/efectos adversos , Estudios Retrospectivos , Riesgo , Inyecciones de Esperma Intracitoplasmáticas , Regulación hacia Arriba
9.
J Assist Reprod Genet ; 31(6): 657-65, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24639041

RESUMEN

FSH is a key hormone in the regulation of follicular development. Together with the EGF network, these molecules mediate oocyte maturation and competence in preparation for the action of LH. FSH isoforms regulate distinct biological pathways and have specific effects on granulosa cell function and maturation of the ovarian follicle. Their dynamic interactions occur during the follicular cycle; short-living forms are predominant in the pre-ovulatory phase, whereas long-acting molecules characterize the luteal-follicular transition. Recombinant FSH (rFSH) molecules have a reduced number of isoforms and are less acidic, with a shorter half-life. We have investigated sequential stimulation, comparing hFSH + rFSH, vs. rFSH alone and hFSH alone for the entire stimulation phase. Sequential stimulation leads to an E2 per MII oocyte ratio that is much lower than is seen during treatment with the two drugs individually. Although there is a positive tendency in favor of the sequential treatment, there was no significant difference in pregnancy rates, even taking frozen embryos into consideration. The cumulus cell transcriptome varies considerably between the treatments, although with no clear significance. When comparing pregnant vs. non-pregnant patients, in general a decrease in mRNA expression can be observed in the pregnant patients, especially in expression of folic acid receptor 1 and ovostatin 2. This indicates that material has been transferred from CC to the oocyte. However, a common observation in the literature is that variations in the transcriptome of the cumulus cells are highly dependent upon the patient genotype; the potential for applying this strategy as a basis for selecting embryos is, at the very least, questionable.


Asunto(s)
Hormona Folículo Estimulante Humana/administración & dosificación , Folículo Ovárico/crecimiento & desarrollo , Inducción de la Ovulación/métodos , Células del Cúmulo/efectos de los fármacos , Células del Cúmulo/metabolismo , Femenino , Fertilización In Vitro/métodos , Hormona Folículo Estimulante Humana/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Técnicas de Maduración In Vitro de los Oocitos/métodos , Oocitos/efectos de los fármacos , Oocitos/crecimiento & desarrollo , Oocitos/metabolismo , Folículo Ovárico/efectos de los fármacos , Embarazo , Índice de Embarazo , Proteínas Recombinantes/administración & dosificación
10.
Liver Int ; 33(6): 914-25, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23617956

RESUMEN

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is a common genetic disorder characterized by the progressive development of renal and hepatic cysts. Follicle-stimulating hormone (FSH) has been demonstrated to be a trophic factor for biliary cells in normal rats and experimental cholestasis induced by bile duct ligation (BDL). AIMS: To assess the effect of FSH on cholangiocyte proliferation during ADPKD using both in vivo and in vitro models. METHODS: Evaluation of FSH receptor (FSHR), FSH, phospho-extracellular-regulated kinase (pERK) and c-myc expression in liver fragments from normal patients and patients with ADPKD. In vitro, we studied proliferating cell nuclear antigen (PCNA) and cAMP levels in a human immortalized, non-malignant cholangiocyte cell line (H69) and in an immortalized cell line obtained from the epithelium lining the hepatic cysts from the patients with ADPKD (LCDE) with or without transient silencing of the FSH gene. RESULTS: Follicle-stimulating hormone is linked to the active proliferation of the cystic wall and to the localization of p-ERK and c-myc. This hormone sustains the biliary growth by activation of the cAMP/ERK signalling pathway. CONCLUSION: These results showed that FSH has an important function in cystic growth acting on the cAMP pathway, demonstrating that it provides a target for medical therapy of hepatic cysts during ADPKD.


Asunto(s)
Proliferación Celular , Quiste del Colédoco/metabolismo , Quistes/metabolismo , Hormona Folículo Estimulante Humana/metabolismo , Hepatopatías/metabolismo , Riñón Poliquístico Autosómico Dominante/metabolismo , Anciano , Animales , Estudios de Casos y Controles , Línea Celular , Quiste del Colédoco/genética , Quiste del Colédoco/patología , AMP Cíclico/metabolismo , Quistes/genética , Quistes/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Hormona Folículo Estimulante Humana/genética , Humanos , Hepatopatías/genética , Hepatopatías/patología , Masculino , Persona de Mediana Edad , Fosforilación , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Interferencia de ARN , Receptores de HFE/metabolismo , Transducción de Señal , Transfección
11.
Coll Antropol ; 37(2): 653-6, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23941020

RESUMEN

Spontaneous forms of the ovarian hyperstimulation syndrome (sOHSS) are nearly always reported between 8 and 14 weeks of pregnancy and also with follicle-stimulating hormone (FSH) producing pituitary adenoma. The syndrome has been previously reported in rare instances of increased production of human chorionic gonadotrophin (hCG) such as multiple pregnancies, hydatiforme mole, polycystic ovary disease and elevated concentrations of thyroid-stimulating hormone (TSH) in hypothyreoidism. High levels of these hormones are able to stimulate by natural promiscuous activation the wild-type FSHr, resulting in sporadic presentations of the syndrome. Since 2003, only six different activating FSHr gene mutations have been reported in cases of familial or habitual sOHSS. In addition to five mutations which have been found in the transmembrane helices (Asp567Asn, Asp567Gly, Thr449Ile, Thr449Ala, Ile545Thr), the first germline mutation (c.383C > A, p. Ser 128 Tyr) in the extracelullar domain was identified. All five mutants were abnormally activated by TSH and normal levels of hCG while displaying constitutive activity. In contrast to these mutations, the p.Ser128Tyr mutant displayed an increase in sensitivity only toward hCG. Accordingly, the mutated FSHrs, may be hyperstimulated by the pregnancy-derived hCG or TSH, inducing the occurrence of the syndrome. In the differential diagnosis, malignancy, pregnancy luteoma and hyperreactio luteinalis would have to be excluded. In almost all of the cases the disease regresses spontaneously and could be managed expectantly or conservatively, but with termination of pregnancy or surgery in cases of complications.


Asunto(s)
Hormona Folículo Estimulante Humana/genética , Síndrome de Hiperestimulación Ovárica/genética , Síndrome de Hiperestimulación Ovárica/fisiopatología , Diagnóstico Diferencial , Femenino , Hormona Folículo Estimulante Humana/metabolismo , Humanos , Síndrome de Hiperestimulación Ovárica/diagnóstico , Mutación Puntual , Embarazo
12.
Yao Xue Xue Bao ; 47(4): 421-6, 2012 Apr.
Artículo en Zh | MEDLINE | ID: mdl-22799021

RESUMEN

Follicle-stimulating hormone (FSH) is a glycoprotein which regulates the development, growth, pubertal maturation and reproductive processes of the body. Exogenous FSH has been used to promote ovarian follicular growth and maturation in female and spermatogenesis in male. The relative short elimination half life and rapid metabolic clearance of current versions of FSH require a daily or twice-daily scheduled subcutaneous injection to maintain stable FSH level being not below the threshold during ovarian stimulation. The development of recombinant long-acting FSH with enhanced biological activities may be helpful for less injection therefore to improve patient compliance, while reducing patient stress and error rates. A number of technological strategies have been explored to develop recombinant longer-acting FSH. For examples, attachment of the C-terminal peptide (CTP) of the human chorionic gonadotropin beta subunit or a sequence containing potential glycosylation sites to either subunit of FSH, creation of a single chain containing the alpha and beta subunits of FSH combined with CTP or N-linked glycosylation signal sequence as a linker, or fusion of the Fc domain of IgGi to FSH. Based on the modifiable molecular structure and pharmacokinetic and pharmacodynamic properties of recombinant FSH, it is hopeful that more FSH drugs with prolonged half-life and increased bioactivity will be developed to meet the modern clinical demands.


Asunto(s)
Hormona Folículo Estimulante Humana/farmacología , Animales , Hormona Folículo Estimulante Humana/química , Hormona Folículo Estimulante Humana/genética , Hormona Folículo Estimulante Humana/metabolismo , Glicosilación , Semivida , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inducción de la Ovulación/métodos , Receptores de HFE/química , Receptores de HFE/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Reproducción/efectos de los fármacos
13.
J Med Primatol ; 40(2): 111-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21044091

RESUMEN

BACKGROUND: Common marmoset monkeys (Callithrix jacchus) are readily used in biomedical research. However, superovulation for embryonic stem cell production and developmental research still remain difficult. Inexplicably, follicle-stimulating hormone (FSH) as key player in superovulation has to be administered in extremely high dosages in this non-human primate compared to human. METHODS: To evaluate whether marmoset FSH (cjFSH) is functionally more competent than its human homologue on the marmoset FSH receptor (cjFSHR), we established in vitro a homologous system characterizing homologous and recombinantly produced cjFSH. RESULTS: Upon stimulation of two cell lines stably expressing either the marmoset or the human FSH receptor (cj/hFSHR), respectively, the second messenger signaling of the activated receptor displayed no significant difference in ED(50) values. Thermostability of cjFSH was significantly prolonged by roughly 20% on both FSHRs. CONCLUSION: High FSH dosage in marmoset superovulation cannot be explained by enhanced biopotency of the natural animal's gonadotropin.


Asunto(s)
Callithrix/genética , Clonación Molecular , Hormona Folículo Estimulante/genética , Receptores de HFE/metabolismo , Secuencia de Aminoácidos , Animales , Callithrix/metabolismo , Femenino , Hormona Folículo Estimulante/química , Hormona Folículo Estimulante/metabolismo , Hormona Folículo Estimulante Humana/química , Hormona Folículo Estimulante Humana/genética , Hormona Folículo Estimulante Humana/metabolismo , Humanos , Receptores de HFE/química , Receptores de HFE/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Superovulación
14.
Hum Reprod ; 25(9): 2290-7, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20627979

RESUMEN

BACKGROUND: Ovarian stimulation regimens for in vitro fertilization seem to have a deleterious effect on oocyte quality and embryo aneuploidy in a dose-dependent manner. This study aims to test the influence of gonadotrophin doses on embryo aneuploidy rates. METHODS: A total of 32 young oocyte donors with a high response to ovarian stimulation, were included in the study. Two subsequent stimulation treatments were performed in each donor: first, a standard dose cycle using a 225 IU starting dose of recombinant FSH (r-FSH) and secondly, a reduced dose cycle with a starting dose of 150 IU r-FSH. In both cycles, GnRH agonist co-treatment was used for down-regulation. Ovarian response, embryo development and aneuploidy for chromosomes 13, 15, 16, 17, 18, 21, 22, X and Y were the main outcomes of the study. RESULTS: A total of 22 donors completed both treatments with different gonadotrophin doses. In the remaining 10 donors, the reduced dose cycle was cancelled due to low ovarian response. In those donors who completed both regimens, significant increases in rates of fertilization and chromosomally normal blastocysts were observed in the reduced dose cycle. No differences were observed in pregnancy and implantation rates in recipients who received oocytes from standard and reduced doses cycles. CONCLUSIONS: Despite the limited numbers in our study, we can conclude that in high responder donors, a decrease in the gonadotrophin dose could improve fertilization rates and embryo quality. However, due to the reduced oocyte numbers with lower doses, a similar reproductive outcome in terms of live births would be expected.


Asunto(s)
Aneugénicos/efectos adversos , Selección de Donante , Embrión de Mamíferos/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Gonadotropinas/efectos adversos , Donación de Oocito , Inducción de la Ovulación/métodos , Adolescente , Adulto , Aneugénicos/administración & dosificación , Aneugénicos/uso terapéutico , Aneuploidia , Blastocisto/efectos de los fármacos , Estudios Cruzados , Relación Dosis-Respuesta a Droga , Implantación del Embrión/efectos de los fármacos , Femenino , Hormona Folículo Estimulante Humana/administración & dosificación , Hormona Folículo Estimulante Humana/efectos adversos , Hormona Folículo Estimulante Humana/genética , Hormona Folículo Estimulante Humana/uso terapéutico , Hormona Liberadora de Gonadotropina/agonistas , Gonadotropinas/administración & dosificación , Gonadotropinas/uso terapéutico , Humanos , Persona de Mediana Edad , Embarazo , Índice de Embarazo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico , Adulto Joven
15.
J Clin Endocrinol Metab ; 105(7)2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32374828

RESUMEN

CONTEXT: Despite the new opportunities provided by assisted reproductive technology (ART), male infertility treatment is far from being optimized. One possibility, based on pathophysiological evidence, is to stimulate spermatogenesis with gonadotropins. EVIDENCE ACQUISITION: We conducted a comprehensive systematic PubMed literature review, up to January 2020, of studies evaluating the genetic basis of follicle-stimulating hormone (FSH) action, the role of FSH in spermatogenesis, and the effects of its administration in male infertility. Manuscripts evaluating the role of genetic polymorphisms and FSH administration in women undergoing ART were considered whenever relevant. EVIDENCE SYNTHESIS: FSH treatment has been successfully used in hypogonadotropic hypogonadism, but with questionable results in idiopathic male infertility. A limitation of this approach is that treatment plans for male infertility have been borrowed from hypogonadism, without daring to overstimulate, as is done in women undergoing ART. FSH effectiveness depends not only on its serum levels, but also on individual genetic variants able to determine hormonal levels, activity, and receptor response. Single-nucleotide polymorphisms in the follicle-stimulating hormone subunit beta (FSHB) and follicle-stimulating hormone receptor (FSHR) genes have been described, with some of them affecting testicular volume and sperm output. The FSHR p.N680S and the FSHB -211G>T variants could be genetic markers to predict FSH response. CONCLUSIONS: FSH may be helpful to increase sperm production in infertile men, even if the evidence to recommend the use of FSH in this setting is weak. Placebo-controlled clinical trials, considering the FSHB-FSHR haplotype, are needed to define the most effective dosage, the best treatment length, and the criteria to select candidate responder patients.


Asunto(s)
Hormona Folículo Estimulante Humana/administración & dosificación , Hormona Folículo Estimulante Humana/fisiología , Infertilidad Masculina/tratamiento farmacológico , Técnicas Reproductivas Asistidas , Espermatogénesis , Hormona Folículo Estimulante Humana/genética , Humanos , Infertilidad Masculina/fisiopatología , Masculino , Polimorfismo de Nucleótido Simple , Espermatogénesis/efectos de los fármacos , Resultado del Tratamiento
16.
PLoS One ; 14(7): e0219434, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31276557

RESUMEN

Recombinant human follicle stimulating hormone (FSH), produced in Chinese hamster ovary (CHO) cells, is widely used for treatment of fertility disorders and is subject to biosimilars development. Cell lines with high specific productivities may simplify the FSH production process. Here, we used our previously established expression system based on vector p1.1 to create new cell lines secreting heterodimeric FSH protein. To this end, we linked open reading frames of both FSH subunits by the wild-type internal ribosome entry site from the encephalomyocarditis virus (EMCV IRES). Intact and double-negative for the dihydrofolate reductase CHO cells were stably transfected by the FSH-coding plasmids. Stably transfected intact cells showed higher level of the FSH secretion and were utilized for subsequent methotrexate-driven transgene amplification, which doubled their productivity. The excess of the free α-subunit was corrected by transfecting the cells by the additional p1.1-based plasmid encoding the ß-subunit of the FSH. Clonal cell lines obtained secreted mostly the heterodimeric FSH and possessed specific productivities up to 12.3±1.7 pg/cell/day. Candidate clonal cell line C-P1.3-FSH-G4 maintained a constant specific productivity for at least 2 months of culturing without the section pressure. The resulting FSH protein conformed to the international pharmaceutical quality criteria as evidenced by the receptor binding kinetics, distribution pattern of hormone isoforms and biological activity. In conclusion, our expression system offers a simple and cost-effective approach to production of FSH.


Asunto(s)
Hormona Folículo Estimulante Humana/genética , Hormona Folículo Estimulante Humana/metabolismo , Expresión Génica , Vectores Genéticos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Animales , Células CHO , Línea Celular , Cricetulus , Ensayo de Inmunoadsorción Enzimática , Orden Génico , Humanos , Hibridación Fluorescente in Situ , Plásmidos/genética , Reacción en Cadena de la Polimerasa , Polisacáridos , Sensibilidad y Especificidad
17.
Mol Cell Endocrinol ; 247(1-2): 103-15, 2006 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-16414183

RESUMEN

A paucity of in vitro models has hampered studies of molecular mechanisms of FSH subunit gene expression. Consequently, we used an in vivo transgenic strategy to map the location of regulatory elements in the cloned 10 kb human FSHbeta gene. Analyses of transgenic mouse lines revealed that successive 5' truncations of the hFSHbeta promoter region to -350 bp relative to the transcriptional initiation site retained gonadotroph-specific expression and the sexually dimorphic pattern of male greater than female FSHbeta mRNA levels found normally in rodent pituitary. Truncation of the 3' flanking sequences from positions +3142 to +2138 bp relative to the translational stop codon in exon 3 resulted in a complete loss of transgene expression, suggesting the presence of critical regulatory elements mapping to the 1 kb genomic segment downstream of position +2138, in addition to the proximal 5' promoter elements. In silico phylogenetic comparisons of mammalian FSHbeta genes revealed five islands of highly conserved sequence homology corresponding precisely to the proximal 5' promoter region, exon 2, the 5' translated region of exon 3, and two regions at the 3' untranslated end of exon 3 that include putative polyadenylation and transcriptional termination signals. Sequence analyses of the 5' proximal promoter revealed the presence of several putative homeodomain binding sites as well as GATA, SMAD, AP-1, NF-1, NF-Y and steroid hormone transcription factor binding sites within the highly conserved -350 bp promoter region. Notably absent from these 5' sequences, however, are consensus binding sites for either Egr-1 or Lim-2 transcription factors known to be critical for the gonadotroph-specific expression of the LHbeta gene. These findings support the hypothesis that one of the mechanisms underlying the differential regulation of the LHbeta, FSHbeta, and common alpha-gonadotropin subunits within pituitary gonadotrophs may be differences in sequence-specific binding requirements for distinct combinations of transcription factors.


Asunto(s)
Hormona Folículo Estimulante Humana/biosíntesis , Hormona Folículo Estimulante de Subunidad beta/biosíntesis , Hipófisis/metabolismo , Región de Flanqueo 3' , Animales , Secuencia de Bases , Codón de Terminación , Femenino , Hormona Folículo Estimulante Humana/genética , Hormona Folículo Estimulante de Subunidad beta/genética , Humanos , Masculino , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Especificidad de Órganos , Filogenia , Regiones Promotoras Genéticas , ARN Mensajero/biosíntesis , Homología de Secuencia de Ácido Nucleico , Factores Sexuales
18.
Fertil Steril ; 83 Suppl 1: 1303-9, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15831306

RESUMEN

OBJECTIVE: To evaluate the efficacy of two novel long-acting rhFSH analogs, rhFSH-N2 and rhFSH-N4, in stimulating murine folliculogenesis. DESIGN: Experimental study. SETTING: Academic research environment. ANIMAL(S): Immature female mice. INTERVENTION(S): Recombinant hFSH-N2 and -N4 were administered via single IP injection to 3-week-old female mice (n = 10) who were killed 48 hours later for dissection and histologic examination of reproductive organs and serum inhibin A. Results were compared with other groups of mice who received either single or q 12 hour injections for 48 hours of commercial rhFSH, or a single injection of pregnant mare serum gonadotropin (PMSG). A subgroup of the mice receiving rhFSH-N4 was supplemented with daily injections of small doses of hCG to simulate LH add-back. MAIN OUTCOME MEASURE(S): Serum inhibin A levels, ovarian and uterine weights, and ovarian antral follicle counts. RESULTS(S): Recombinant human FSH-N2 and -N4 administration induced a statistically significant increase in ovarian weights, uterine weights, and inhibin A levels compared with single and twice-daily injection of rhFSH. PMSG induced the greatest increases in all three measured parameters. There was no statistical difference between rhFSH-N2 and rhFSH-N4 for any parameter analyzed. A single injection of rhFSH-N2 or -N4 induced a greater number of antral follicles than did either single or q 12 hour injections of rhFSH. The addition of small doses of hCG to rhFSH-N4 increased inhibin A levels and antral follicle number to reach statistical equivalence to PMSG treatment. CONCLUSION(S): Addition of a synthetic polypeptide containing two or four N-linked glycosylation sites to rhFSH increases in vivo bioactivity of the hormone compared to commercial rhFSH. After a single injection, both rhFSH-N2 and rhFSH-N4 effectively induced a greater follicular response in the mouse than did rhFSH.


Asunto(s)
Hormona Folículo Estimulante Humana/análogos & derivados , Hormona Folículo Estimulante Humana/farmacología , Oligosacáridos/farmacología , Folículo Ovárico/efectos de los fármacos , Inducción de la Ovulación/métodos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Femenino , Hormona Folículo Estimulante Humana/genética , Glicosilación , Gonadotropinas Equinas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Datos de Secuencia Molecular , Oligosacáridos/metabolismo , Folículo Ovárico/citología , Folículo Ovárico/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
19.
Endokrynol Pol ; 56(3): 359-61, 2005.
Artículo en Polaco | MEDLINE | ID: mdl-16350732

RESUMEN

Among the causes of premature ovarian failure (POF) two groups of factors are reported: factors which lead to decrease of follicular number and factors which stimulate follicular atresia. In the first group genetic factors are the most important whereas in the second: enzymatic autoimmunological, iatrogenic, toxins and infections are reported. In 1986 familiar POF on the background of long arm of chromosome X deletion was reported. Other chromosomes which are important for normal ovarian function are: chromosome 21 (AIRE gene), chromosome 11 (gene of beta FSH, ATM gene), chromosome 3 (gene responsible for BEPS syndrome) and chromosome 2 (genes of FSH and LH receptors). In this review the role of these genes and results of several epidemiological studies are reported.


Asunto(s)
Hormona Folículo Estimulante Humana/genética , Infertilidad Femenina/genética , Insuficiencia Ovárica Primaria/genética , Femenino , Humanos , Masculino
20.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 26(2): 178-81, 2004 Apr.
Artículo en Zh | MEDLINE | ID: mdl-15171557

RESUMEN

OBJECTIVE: To evaluate the effect of superovulation with recombinant follicle stimulating hormone (r-FSH) therapy and intrauterine insemination in the treatment of idiopathic infertility. METHODS: Superovulation with r-FSH therapy and intrauterine insemination were used in 202 cycles of 88 couples in the Department of Obstetrics and Gynecology of Monash Medical Centre. RESULTS: The per cycle ovulation rate and in-ovulation rate were 95.7% and 4.3% respectively, and the per cycle pregnancy rate was 11.6% with no cases of hyperstimulation. The cancelling rate was 7.4% because of the development of multiple follicles. The overall cumulative conception rate was 22.7% per patient, with 15% of twin pregnancies. There were no differences between pregnancy group and non-pregnancy group in age, BMI, treatment days, number of mature follicles, endometrial thickness and number of treatment cycles. The only significant parameter observed between the two groups was infertility time (P < 0.05), which was longer in non-pregnancy group [(30.52 +/- 13.08) months] than in pregnancy group [(24.25 +/- 6.45) months]. CONCLUSIONS: Superovulation and intrauterine insemination is a safe and more cost-effective method in treatment of idiopathic infertility.


Asunto(s)
Hormona Folículo Estimulante Humana/uso terapéutico , Infertilidad Femenina/terapia , Inseminación Artificial Homóloga , Superovulación/efectos de los fármacos , Adulto , Femenino , Hormona Folículo Estimulante Humana/biosíntesis , Hormona Folículo Estimulante Humana/genética , Humanos , Inseminación Artificial Homóloga/métodos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA