Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Eur J Obstet Gynecol Reprod Biol ; 263: 72-78, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34167037

RESUMEN

OBJECTIVE: The purpose of this study was to evaluate the vitamin B status related to the homocysteine pathway and the prevalence of polymorphisms of the MTHFR gene in infertile women programming homologous or heterologous ART. STUDY DESIGN: We investigated 393 consecutive Caucasian women, referred to the Internal Medicine Clinic at the Center for Assisted Reproductive Technology, in order to be framed for their vascular risk before starting homologous or heterologous (oocyte donation) procedures. Total homocysteine, Vitamin B12, folate and vitamin B6 were measured. The women were divided into quartiles of serum concentration of folate, vitamin B12 and vitamin B6. The C677T and A1298C polymorphisms of the MTHFR gene were genotyped by an electronic microchip technology. RESULTS: Sixty-one women (15.5%) had hyperhomocysteinemia, 22.9% had reduced levels of vitamin B12, 4.1% had reduced levels of serum folate and 0.1% had a deficiency of vitamin B6. Women in the highest quartile of vitamin B12 and folates had lower homocysteine ​​levels than women in the first and second quartiles (p < 0.0001). The homozygosity for MTHFR C677T polymorphism was detected in 33.3% (131), and heterozygosity for MTHFR C677T polymorphism in 45.3% (178) of women. We observed a significant association between hyperhomocysteinemia and 677T allele, but not 1298C, of the MTHFR polymorphisms (p = 0.04). CONCLUSIONS: We found inadequate vitamin B status related to the homocysteine ​​pathway in women planning Assisted Reproductive Technology. Moreover, interesting association was found regarding hyperhomocysteinemia in women carrying T allele of the C677T MTHFR polymorphism. A specific supplementation with 5-MTHF and adequate vitamin B12 concentrations before Assisted Reproductive Technology warrant serious consideration, in particular in women carrying T allele of the C677T MTHFR polymorphism.


Asunto(s)
Infertilidad Femenina , Complejo Vitamínico B , Femenino , Ácido Fólico , Genotipo , Homocisteína , Humanos , Infertilidad Femenina/genética , Italia/epidemiología , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Vitamina B 12
2.
Nutrients ; 13(4)2021 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-33923969

RESUMEN

Methylenetetrahydrofolate reductase (MTHFR) has various polymorphisms, and the effects of periconceptional folic acid supplementation for decreasing neural tube defects (NTDs) risk differ depending on the genotypes. This study analyzed the effectiveness of multivitamin supplementation on folate insufficiency and hyperhomocysteinemia, depending on MTHFR polymorphisms. Of 205 women, 72 (35.1%), 100 (48.8%) and 33 (16.1%) had MTHFR CC, CT and TT, respectively. Serum folate and homocysteine levels in women with homozygous mutant TT were significantly lower and higher, respectively, than those in women with CC and CT. In 54 women (26.3% of all women) with a risk of NTDs, multivitamin supplementation containing folic acid and vitamin D for one month increased folate level (5.8 ± 0.9 to 19.2 ± 4.0 ng/mL, p < 0.0001) and decreased the homocysteine level (8.2 ± 3.1 to 5.8 ± 0.8 nmol/mL, p < 0.0001) to minimize the risk of NTDs in all women, regardless of MTHFR genotype. Regardless of MTHFR genotype, multivitamin supplements could control folate and homocysteine levels. Tests for folate and homocysteine levels and optimal multivitamin supplementation in women with risk of NTDs one month or more before pregnancy should be recommended to women who are planning a pregnancy.


Asunto(s)
Pueblo Asiatico/genética , Suplementos Dietéticos , Ácido Fólico/sangre , Variación Genética , Homocisteína/sangre , Infertilidad Femenina/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Vitaminas/farmacología , Adulto , Femenino , Humanos , Infertilidad Femenina/sangre , Infertilidad Femenina/enzimología , Embarazo , Resultado del Embarazo , Vitamina D/sangre
3.
J Assist Reprod Genet ; 37(10): 2377-2387, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32767206

RESUMEN

OBJECTIVE: Increased oxidative stress has been identified as a pathogenetic mechanism in female infertility. However, the effect of specific antioxidants, such as coenzyme Q10 (CoQ10), on the outcomes after assisted reproductive technologies (ART) has not been clarified. The aim of this study was to systematically review and meta-analyze the best available evidence regarding the effect of CoQ10 supplementation on clinical pregnancy (CPR), live birth (LBR), and miscarriage rates (MR) compared with placebo or no-treatment in women with infertility undergoing ART. METHODS: A comprehensive literature search was conducted in PubMed (MEDLINE), Cochrane, and Scopus, from inception to March 2020. Data were expressed as odds ratio (OR) with 95% confidence intervals (CI). The I2 index was employed for heterogeneity. RESULTS: Five randomized-controlled trials fulfilled eligibility criteria (449 infertile women; 215 in CoQ10 group and 234 in placebo/no treatment group). Oral supplementation of CoQ10 resulted in an increase of CPR when compared with placebo or no-treatment (28.8% vs. 14.1%, respectively; OR 2.44, 95% CI 1.30-4.59, p = 0.006; I2 32%). This effect remained significant when women with poor ovarian response and polycystic ovarian syndrome were analyzed separately. No difference between groups was observed regarding LBR (OR 1.67, 95% CI 0.66-4.25, p = 0.28; I2 34%) and MR (OR 0.61, 95% CI 0.13-2.81, p = 0.52; I2 0%). CONCLUSIONS: Oral supplementation of CoQ10 may increase CPR when compared with placebo or no-treatment, in women with infertility undergoing ART procedures, without an effect on LBR or MR.


Asunto(s)
Suplementos Dietéticos , Infertilidad Femenina , Síndrome del Ovario Poliquístico , Ubiquinona , Femenino , Humanos , Embarazo , Antioxidantes/uso terapéutico , Fertilidad/efectos de los fármacos , Fertilidad/genética , Infertilidad Femenina/tratamiento farmacológico , Infertilidad Femenina/genética , Infertilidad Femenina/patología , Estrés Oxidativo/efectos de los fármacos , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/patología , Ubiquinona/análogos & derivados , Ubiquinona/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto
4.
Redox Biol ; 30: 101431, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31972508

RESUMEN

Endometriosis an important cause of female infertility and seriously impact physical and psychological health of patients. Endometriosis is now considered to be a public health problem that deserves in-depth investigation, especially the etiopathogenesis of endometriosis-associated infertility. We aimed to illuminate the etiopathogenesis of endometriosis-associated infertility that involve excessive oxidative stress (OS) induced pathological changes of ovary cumulus granulosa cell (GCs). Senescence-associated ß-galactosidase (SA ß-gal) activity in GCs from endometriosis patients, soluble isoform of advanced glycation end products receptor (sRAGE) expression in follicular fluid from endometriosis patients and differentially expressed senescence-associated secretory phenotype factors (IL-1ß, MMP-9, KGF and FGF basic protein) are all useful indexes to evaluate oocyte retrieval number and mature oocyte number. RNA-sequencing and bioinformatics analysis indicated senescent phenotype of endometriosis GCs and aggravated endoplasmic reticulum (ER) stress in endometriosis GCs. Targeting ER stress significantly alleviated OS-induced GCs senescence as well as mitochondrial membrane potential (MMP) and adenosine triphosphate (ATP) reduction in GCs. Moreover, melatonin administration rescued OS-enhanced ER stress, cellular senescence, and MMP and ATP abnormities of endometriosis GCs in vitro and in vivo. In conclusion, our results indicated excessive reactive oxygen species induces senescence of endometriosis GCs via arouse ER stress, which finally contributes to endometriosis-associated infertility, and melatonin may represent a novel adjuvant therapy strategy for endometriosis-associated infertility.


Asunto(s)
Antígenos de Neoplasias/genética , Células del Cúmulo/citología , Endometriosis/tratamiento farmacológico , Infertilidad Femenina/tratamiento farmacológico , Melatonina/administración & dosificación , Proteínas Quinasas Activadas por Mitógenos/genética , Estrés Oxidativo/efectos de los fármacos , Animales , Línea Celular , Senescencia Celular/efectos de los fármacos , Células del Cúmulo/metabolismo , Modelos Animales de Enfermedad , Endometriosis/complicaciones , Endometriosis/genética , Femenino , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Infertilidad Femenina/etiología , Infertilidad Femenina/genética , Melatonina/farmacología , Ratones , Inducción de la Ovulación , Análisis de Secuencia de ARN
5.
Biol Trace Elem Res ; 193(2): 319-325, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30963410

RESUMEN

This study was performed to determine the effects of selenium supplementation on clinical symptoms and gene expression related to inflammatory markers in infertile women with polycystic ovary syndrome (PCOS) who were candidate for in vitro fertilization (IVF). Thirty-six women candidate for IVF were recruited in this randomized double-blinded, placebo-controlled trial. They (n = 18/group) were randomly assigned into intervention groups to take either 200 µg/day of selenium or placebo for 8 weeks. RT-PCR findings indicated that selenium supplementation downregulated gene expression of interleukin-1 (IL-1) (P < 0.004) and tumor necrosis factor alpha (TNF-α) (P = 0.02) in lymphocytes of patients with PCOS compared with the placebo. In addition, selenium supplementation upregulated gene expression of vascular endothelial growth factor (VEGF) (P = 0.001) in lymphocytes of patients with PCOS compared with the placebo. Selenium supplementation had no significant effect on clinical symptoms and gene expression of IL-8 (P = 0.10) and transforming growth factor beta (TGF-ß) (P = 0.63). Overall, our findings documented that selenium supplementation for 8 weeks to infertile women candidate for IVF improved IL-1, TNF-α, and VEGF gene expression, though selenium had no effect on clinical symptoms and, IL-8 and TGF-ß gene expression. Clinical trial registration number: http://www.irct.ir: IRCT20170513033941N23.


Asunto(s)
Fertilización In Vitro , Expresión Génica/efectos de los fármacos , Infertilidad Femenina/genética , Inflamación/genética , Selenio/farmacología , Factor A de Crecimiento Endotelial Vascular/genética , Adulto , Suplementos Dietéticos , Método Doble Ciego , Femenino , Humanos , Infertilidad Femenina/diagnóstico , Interleucina-1/genética , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Síndrome del Ovario Poliquístico/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Selenio/administración & dosificación , Factor de Necrosis Tumoral alfa/genética
6.
Biol Reprod ; 102(1): 102-115, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-31373359

RESUMEN

Superoxide dismutase 1 suppresses oxidative stress within cells by decreasing the levels of superoxide anions. A dysfunction of the ovary and/or an aberrant production of sex hormones are suspected causes for infertility in superoxide dismutase 1-knockout mice. We report on attempts to rescue the infertility in female knockout mice by providing two antioxidants, ascorbic acid and/or coenzyme Q10, as supplements in the drinking water of the knockout mice after weaning and on an investigation of their reproductive ability. On the first parturition, 80% of the untreated knockout mice produced smaller litter sizes compared with wild-type mice (average 2.8 vs 7.3 pups/mouse), and supplementing with these antioxidants failed to improve these litter sizes. However, in the second parturition of the knockout mice, the parturition rate was increased from 18% to 44-75% as the result of the administration of antioxidants. While plasma levels of progesterone at 7.5 days of pregnancy were essentially the same between the wild-type and knockout mice and were not changed by the supplementation of these antioxidants, sizes of corpus luteum cells, which were smaller in the knockout mouse ovaries after the first parturition, were significantly ameliorated in the knockout mouse with the administration of the antioxidants. Moreover, the impaired vasculogenesis in uterus/placenta was also improved by ascorbic acid supplementation. We thus conclude that ascorbic acid and/or coenzyme Q10 are involved in maintaining ovarian and uterus/placenta homeostasis against insults that are augmented during pregnancy and that their use might have positive effects in terms of improving female fertility.


Asunto(s)
Ácido Ascórbico/farmacología , Infertilidad Femenina/tratamiento farmacológico , Reproducción/efectos de los fármacos , Superóxido Dismutasa-1/metabolismo , Ubiquinona/análogos & derivados , Animales , Ácido Ascórbico/uso terapéutico , Femenino , Infertilidad Femenina/genética , Infertilidad Femenina/metabolismo , Ratones , Ratones Noqueados , Progesterona/sangre , Reproducción/genética , Superóxido Dismutasa-1/genética , Ubiquinona/farmacología , Ubiquinona/uso terapéutico
7.
Anal Biochem ; 586: 113433, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31521670

RESUMEN

This study investigated the association of A419T (rs121909661) and T449I (rs28928870) with infertility among Iranian women and possible treatments by agonizing the mutated receptor. 151 women were genotyped at A419T and T449I sites. Homology modeling, pharmacophore modeling, virtual screening, docking and molecular dynamics (MD) were performed. A419T and T449I indicated a significant and a weak association with infertility among Iranian women (P = 0.005 and P = 0.03, respectively). Significant differences found among three genotypes of A419T with FSH (P = 0.01) and LH (P < 0.0001). G-allele carriers of A419T had susceptibility to display higher FSH and LH serum levels. In silico results revealed the most potent agonists among 3041 similar compounds and MD supported this finding. Altogether, genotyping of A419T and T449I as potential markers might be helpful in prognosis and treatment of infertility. Also, a new series of potent FSHR agonists were identified for future drug development and treatment of infertility related to FSHR dysfunction.


Asunto(s)
Infertilidad Femenina/tratamiento farmacológico , Simulación de Dinámica Molecular , Mutación Missense/efectos de los fármacos , Receptores de HFE/agonistas , Receptores de HFE/antagonistas & inhibidores , Adulto , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Infertilidad Femenina/genética , Irán , Ligandos , Masculino , Estructura Molecular , Embarazo , Receptores de HFE/genética , Adulto Joven
8.
Sci Rep ; 9(1): 12519, 2019 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-31467307

RESUMEN

In natural cycle or minimal stimulation cycle IVF, buserelin acetate (buserelin), a gonadotropin-releasing hormone agonist, is often used as a maturation trigger; however, its effect on pregnancy outcomes remains unclear. Therefore, in the present study, we compared uterine receptivity in buserelin-administered mice with that in human chorionic gonadotropin (hCG)-administered mice during the peri-implantation period. Implantation, decidualisation, and term-pregnancy were impaired following hCG, but not buserelin administration. hCG stimulated the synthesis and secretion of progesterone and oestradiol, whereas ovarian steroidogenesis in the buserelin-treated group was comparable with that in the control group. Furthermore, similar to the observation in controls, the buserelin-treated group exhibited activation of progesterone receptor signalling and inhibition of oestrogen receptor signalling in the endometrial epithelium on the day of implantation. However, epithelial progesterone signalling was not detected, and a high expression of genes downstream to oestrogen was observed on day 4 following hCG administration. These results suggest that buserelin administration does not impact uterine receptivity as it did not affect ovarian steroidogenesis and endometrial steroid signalling. Therefore, buserelin is preferred as an oocyte maturation trigger to optimise uterine receptivity during treatments involving timed intercourse, intrauterine insemination, or fresh embryo transfer following in vitro fertilisation.


Asunto(s)
Buserelina/administración & dosificación , Hormona Liberadora de Gonadotropina/agonistas , Infertilidad Femenina/tratamiento farmacológico , Oocitos/citología , Oogénesis/efectos de los fármacos , Útero/efectos de los fármacos , Animales , Buserelina/efectos adversos , Gonadotropina Coriónica/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Implantación del Embrión/efectos de los fármacos , Transferencia de Embrión , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Estradiol/metabolismo , Femenino , Fertilización In Vitro , Humanos , Infertilidad Femenina/genética , Infertilidad Femenina/metabolismo , Infertilidad Femenina/fisiopatología , Masculino , Ratones , Ratones Endogámicos ICR , Oocitos/efectos de los fármacos , Ovario/efectos de los fármacos , Ovario/metabolismo , Embarazo , Índice de Embarazo , Progesterona/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Útero/fisiopatología
9.
Reprod Biol Endocrinol ; 16(1): 94, 2018 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-30286768

RESUMEN

BACKGROUND: Vitamin D deficiency in women diagnosed with polycystic ovary syndrome (PCOS) remarkably decreases the chance of pregnancy, which might be related to its impact on metabolic abnormalities in these patients. It is hypothesized that vitamin D supplementation influences metabolic profile of these patients and indirectly might affect fertility and the outcomes. Therefore, this study was conducted to determine the effects of vitamin D supplementation on the levels of anti-Müllerian hormone (AMH), metabolic profiles, and gene expression of insulin and lipid metabolism in infertile women with PCOS who were candidate for in vitro fertilization (IVF). METHODS: This study was a randomized, double-blinded, placebo-controlled trial conducted among 40 infertile women, aged 18-40 years, diagnosed with PCOS and was candidate for IVF. Participants were randomly assigned into two intervention groups for receiving either 50,000 IU vitamin D or placebo (n = 20 each group) every other week for 8 weeks. Gene expression for insulin and lipid metabolism was conducted using peripheral blood mononuclear cells (PBMCs) of women with PCOS, via RT-PCR method. RESULTS: Vitamin D supplementation led to a significant reduction in serum AMH (- 0.7 ± 1.2 vs. - 0.1 ± 0.5 ng/mL, P = 0.02), insulin levels (- 1.4 ± 1.6 vs. -0.3 ± 0.9 µIU/mL, P = 0.007), homeostatic model of assessment for insulin resistance (- 0.3 ± 0.3 vs. -0.1 ± 0.2, P = 0.008), and a significant increase in quantitative insulin sensitivity check index (+ 0.009 ± 0.01 vs. + 0.001 ± 0.004, P = 0.04), compared with the placebo. Moreover, following vitamin D supplementation there was a significant decrease in serum total- (- 5.1 ± 12.6 vs. + 2.9 ± 10.9 mg/dL, P = 0.03) and LDL-cholesterol levels (- 4.5 ± 10.3 vs. + 2.5 ± 10.6 mg/dL, P = 0.04) compared with the placebo. CONCLUSION: Overall, the findings of this trial supported that 50,000 IU vitamin D supplementation every other week for 8 weeks had beneficial effects on insulin metabolism, and lipid profile of infertile women with PCOS who are candidate for IVF. These benefits might not be evident upon having sufficient vitamin D levels. TRIAL REGISTRATION: This study was retrospectively registered in the Iranian website ( www.irct.ir ) for clinical trials registration ( http://www.irct.ir : IRCT20170513033941N27).


Asunto(s)
Infertilidad Femenina/genética , Insulina/genética , Metabolismo de los Lípidos/genética , Síndrome del Ovario Poliquístico/genética , Transcriptoma/efectos de los fármacos , Vitamina D/administración & dosificación , Adolescente , Adulto , Suplementos Dietéticos , Método Doble Ciego , Femenino , Fertilización In Vitro , Humanos , Irán , Embarazo , Estudios Retrospectivos , Deficiencia de Vitamina D/tratamiento farmacológico , Deficiencia de Vitamina D/genética , Deficiencia de Vitamina D/metabolismo , Vitaminas/administración & dosificación , Adulto Joven
10.
J Neurosci ; 35(37): 12903-16, 2015 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-26377475

RESUMEN

The gonadotropin-releasing hormone (GnRH) is the master regulator of fertility and kisspeptin (KP) is a potent trigger of GnRH secretion from GnRH neurons. KP signals via KISS1R, a Gαq/11-coupled receptor, and mice bearing a global deletion of Kiss1r (Kiss1r(-/-)) or a GnRH neuron-specific deletion of Kiss1r (Kiss1r(d/d)) display hypogonadotropic hypogonadism and infertility. KISS1R also signals via ß-arrestin, and in mice lacking ß-arrestin-1 or -2, KP-triggered GnRH secretion is significantly diminished. Based on these findings, we hypothesized that ablation of Gαq/11 in GnRH neurons would diminish but not completely block KP-triggered GnRH secretion and that Gαq/11-independent GnRH secretion would be sufficient to maintain fertility. To test this, Gnaq (encodes Gαq) was selectively inactivated in the GnRH neurons of global Gna11 (encodes Gα11)-null mice by crossing Gnrh-Cre and Gnaq(fl/fl);Gna11(-/-) mice. Experimental Gnaq(fl/fl);Gna11(-/-);Gnrh-Cre (Gnaq(d/d)) and control Gnaq(fl/fl);Gna11(-/-) (Gnaq(fl/fl)) littermate mice were generated and subjected to reproductive profiling. This process revealed that testicular development and spermatogenesis, preputial separation, and anogenital distance in males and day of vaginal opening and of first estrus in females were significantly less affected in Gnaq(d/d) mice than in previously characterized Kiss1r(-/-) or Kiss1r(d/d) mice. Additionally, Gnaq(d/d) males were subfertile, and although Gnaq(d/d) females did not ovulate spontaneously, they responded efficiently to a single dose of gonadotropins. Finally, KP stimulation triggered a significant increase in gonadotropins and testosterone levels in Gnaq(d/d) mice. We therefore conclude that the milder reproductive phenotypes and maintained responsiveness to KP and gonadotropins reflect Gαq/11-independent GnRH secretion and activation of the neuroendocrine-reproductive axis in Gnaq(d/d) mice. SIGNIFICANCE STATEMENT: The gonadotropin-releasing hormone (GnRH) is the master regulator of fertility. Over the last decade, several studies have established that the KISS1 receptor, KISS1R, is a potent trigger of GnRH secretion and inactivation of KISS1R on the GnRH neuron results in infertility. While KISS1R is best understood as a Gαq/11-coupled receptor, we previously demonstrated that it could couple to and signal via non-Gαq/11-coupled pathways. The present study confirms these findings and, more importantly, while it establishes Gαq/11-coupled signaling as a major conduit of GnRH secretion, it also uncovers a significant role for non-Gαq/11-coupled signaling in potentiating reproductive development and function. This study further suggests that by augmenting signaling via these pathways, GnRH secretion can be enhanced to treat some forms of infertility.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP/deficiencia , Hormona Liberadora de Gonadotropina/fisiología , Hipogonadismo/fisiopatología , Infertilidad Femenina/fisiopatología , Infertilidad Masculina/fisiopatología , Animales , Blastocisto/patología , Desarrollo Embrionario , Femenino , Subunidades alfa de la Proteína de Unión al GTP/fisiología , Perfilación de la Expresión Génica , Genitales Femeninos/patología , Genitales Femeninos/fisiopatología , Genitales Masculinos/patología , Genitales Masculinos/fisiopatología , Hormonas Esteroides Gonadales/metabolismo , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Gonadotropinas Hipofisarias/metabolismo , Gonadotropinas Hipofisarias/farmacología , Hipogonadismo/genética , Hipogonadismo/patología , Sistema Hipotálamo-Hipofisario/fisiopatología , Hipotálamo/patología , Infertilidad Femenina/embriología , Infertilidad Femenina/genética , Infertilidad Masculina/embriología , Infertilidad Masculina/genética , Kisspeptinas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Oligopéptidos/farmacología , Ovariectomía , Ovulación/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Péptidos/farmacología , Fenotipo , Receptores Acoplados a Proteínas G , Receptores de Kisspeptina-1 , Espermatogénesis
11.
Nat Rev Endocrinol ; 11(7): 429-41, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25942654

RESUMEN

Hypoandrogenism in women with low functional ovarian reserve (LFOR, defined as an abnormally low number of small growing follicles) adversely affects fertility. The androgen precursor dehydroepiandrosterone (DHEA) is increasingly used to supplement treatment protocols in women with LFOR undergoing in vitro fertilization. Due to differences in androgen metabolism, however, responses to DHEA supplementation vary between patients. In addition to overall declines in steroidogenic capacity with advancing age, genetic factors, which result in altered expression or enzymatic function of key steroidogenic proteins or their upstream regulators, might further exacerbate variations in the conversion of DHEA to testosterone. In this Review, we discuss in vitro studies and animal models of polymorphisms and gene mutations that affect the conversion of DHEA to testosterone and attempt to elucidate how these variations affect female hormone profiles. We also discuss treatment options that modulate levels of testosterone by targeting the expression of steroidogenic genes. Common variants in genes encoding DHEA sulphotransferase, aromatase, steroid 5α-reductase, androgen receptor, sex-hormone binding globulin, fragile X mental retardation protein and breast cancer type 1 susceptibility protein have been implicated in androgen metabolism and, therefore, can affect levels of androgens in women. Short of screening for all potential genetic variants, hormonal assessments of patients with low testosterone levels after DHEA supplementation facilitate identification of underlying genetic defects. The genetic predisposition of patients can then be used to design individualized fertility treatments.


Asunto(s)
Andrógenos/deficiencia , Deshidroepiandrosterona/metabolismo , Infertilidad Femenina/genética , Testosterona/metabolismo , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/genética , Animales , Aromatasa/genética , Deshidroepiandrosterona/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Genes BRCA1 , Humanos , Reserva Ovárica , Polimorfismo Genético , Receptores Androgénicos/genética , Globulina de Unión a Hormona Sexual/genética , Sulfotransferasas/genética
12.
Elife ; 42015 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-25581095

RESUMEN

Whereas it is well established that chronic stress induces female reproductive dysfunction, whether stress negatively impacts fertility and fecundity when applied prior to mating and pregnancy has not been explored. In this study, we show that stress that concludes 4 days prior to mating results in persistent and marked reproductive dysfunction, with fewer successful copulation events, fewer pregnancies in those that successfully mated, and increased embryo resorption. Chronic stress exposure led to elevated expression of the hypothalamic inhibitory peptide, RFamide-related peptide-3 (RFRP3), in regularly cycling females. Remarkably, genetic silencing of RFRP3 during stress using an inducible-targeted shRNA completely alleviates stress-induced infertility in female rats, resulting in mating and pregnancy success rates indistinguishable from non-stress controls. We show that chronic stress has long-term effects on pregnancy success, even post-stressor, that are mediated by RFRP3. This points to RFRP3 as a potential clinically relevant single target for stress-induced infertility.


Asunto(s)
Pérdida del Embrión/etiología , Pérdida del Embrión/prevención & control , Técnicas de Silenciamiento del Gen , Hormonas Hipotalámicas/genética , Hipotálamo/metabolismo , Infertilidad Femenina/etiología , Estrés Psicológico/complicaciones , Animales , Doxiciclina/farmacología , Pérdida del Embrión/genética , Pérdida del Embrión/patología , Ciclo Estral/genética , Femenino , Hormonas Hipotalámicas/metabolismo , Infertilidad Femenina/genética , Infertilidad Femenina/patología , Infertilidad Femenina/prevención & control , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas Sprague-Dawley , Reproducción , Estrés Psicológico/patología , Regulación hacia Arriba/genética
13.
Acta Obstet Gynecol Scand ; 94(1): 65-71, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25283235

RESUMEN

OBJECTIVE: To study folic acid intake, folate status and pregnancy outcome after infertility treatment in women with different infertility diagnoses in relation to methylenetetrahydrofolate reductase (MTHFR) 677C>T, 1298A>C and 1793G>A polymorphisms. Also the use of folic acid supplements, folate status and the frequency of different gene variations were studied in women undergoing infertility treatment and fertile women. DESIGN: Observational study. SETTING: University hospital. POPULATION: Women undergoing infertility treatment and healthy, fertile, non-pregnant women. METHODS: A questionnaire was used to assess general background data and use of dietary supplements. Blood samples were taken to determine plasma folate and homocysteine levels, and for genomic DNA extraction. A comparison of four studies was performed to assess pregnancy outcome in relation to MTHFR 677 TT vs. CC, and 1298 CC vs. AA polymorphisms. MAIN OUTCOME MEASURES: Folic acid supplement intake, and plasma folate, homocysteine and genomic assays. RESULTS: Women in the infertility group used significantly more folic acid supplements and had better folate status than fertile women, but pregnancy outcome after fertility treatment was not dependent on folic acid intake, folate status or MTHFR gene variations. CONCLUSION: High folic acid intakes and MTHFR gene variations seem not to be associated with helping women to achieve pregnancy during or after fertility treatment.


Asunto(s)
Suplementos Dietéticos , Fertilización In Vitro/métodos , Ácido Fólico/administración & dosificación , Infertilidad Femenina/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Resultado del Embarazo , Adulto , Estudios de Casos y Controles , Distribución de Chi-Cuadrado , Femenino , Fertilización In Vitro/efectos adversos , Variación Genética , Hospitales Universitarios , Humanos , Infertilidad Femenina/terapia , Polimorfismo de Nucleótido Simple , Embarazo , Estudios Prospectivos , Valores de Referencia , Estadísticas no Paramétricas , Suecia , Adulto Joven
14.
Taiwan J Obstet Gynecol ; 53(4): 559-65, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25510701

RESUMEN

OBJECTIVE: Our previous study showed the potential benefits of dehydroepiandrosterone (DHEA) supplementation in women with a poor ovarian response (POR). Because the connection between cumulus cells (CCs) and oocytes is a key step for oocyte maturation, we supposed that altered gene expression of CCs in women with POR after DHEA supplementation might favor oocyte maturation. MATERIALS AND METHODS: Women with POR treated with flexible daily gonadotropin-releasing hormone antagonist in vitro fertilization (IVF) cycles at The Reproductive Center in Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan between January 2013 and October 2013 were enrolled for this prospective study. CCs were isolated during IVF before and after DHEA (CPH-Formulation, Oakdale, CA, USA) supplementation. Nine genes of isolated CCs, including hyaluronan synthase (HAS2), versican (VCAN), thrombospondin 1 (THBS1), runt-related transcription factor 2 (RUNX2), chromobox homolog 3 (CBX3), tripartite motif-containing 28 (TRIM28), B-cell lymphoma 2 (BCL2), BCL2-associated X protein (BAX), and ankyrin repeat domain 57 (ANKRD57), were compared. RESULTS: There was a significant difference in the expression of genes in women with POR before and after DHEA supplementation (all p < 0.05). All genes related to extracellular matrix (ECM) formation, including HAS2, VCAN, and THBS1, were upregulated. By contrast, all genes involving cell development, differentiation, and apoptosis regulation were downregulated. Unknown function gene ANKRD57 was also downregulated after DHEA supplementation. Although expressions of both BCL2 and BAX were decreased in women with POR after DHEA supplementation compared to those before treatment, the ratio of BCL2 and BAX was significantly increased in women with POR after DHEA supplementation, suggesting that DHEA supplementation might activate the antiapoptosis process of CCs, which might be beneficial to the improvement of ovarian function in women with POR. CONCLUSION: The study showed that DHEA therapy positively affected the gene expression of CCs in women with POR, and provided evidence to support the positive effect of DHEA supplementation on women with POR.


Asunto(s)
Células del Cúmulo/efectos de los fármacos , Deshidroepiandrosterona/uso terapéutico , Fármacos para la Fertilidad Femenina/uso terapéutico , Infertilidad Femenina/terapia , Inducción de la Ovulación/métodos , Deshidroepiandrosterona/farmacología , Suplementos Dietéticos , Regulación hacia Abajo , Femenino , Fármacos para la Fertilidad Femenina/farmacología , Fertilización In Vitro , Marcadores Genéticos , Humanos , Infertilidad Femenina/genética , Estudios Prospectivos , Reacción en Cadena en Tiempo Real de la Polimerasa , Regulación hacia Arriba
15.
Biol Reprod ; 90(2): 24, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24337314

RESUMEN

Female ESR2-null mice (betaERKO) display defects in ovarian function and are subfertile. Follicular maturation is impaired and explains smaller litters, but betaERKO also produce fewer litters, which may be partially due to inadequate ovulatory signals. To test this, the amplitude and timing of the naturally occurring luteinizing hormone (LH) surge was measured in individual intact betaERKO and wild-type (WT) mice. Vaginal cytology was evaluated daily, and blood samples were taken from mice in proestrus. The amplitude of the LH surge was severely blunted in betaERKO mice compared to WT, but pituitary LH levels revealed no differences. The betaERKO mice did not produce a preovulatory estradiol surge. To determine if the smaller LH surges and the reduced number of litters in betaERKO were due to the lack of ESR2 in the hypothalamic-pituitary axis or due to the absence of ESR2 in the ovary, ovaries were transplanted from WT into betaERKO mice and vice versa. The size of the LH surge was reduced only in mice lacking ESR2 within the ovary, and these mice had fewer litters. Fertility and size of the LH surge were rescued in betaERKO mice receiving a WT ovary. These data provide the first experimental evidence that the LH surge is impaired in betaERKO females and may contribute to their reduced fertility. ESR2 is not necessary within the pituitary and hypothalamus for the generation of a normal LH surge and for normal fertility, but ESR2 is essential within the ovary to provide proper signals.


Asunto(s)
Receptor beta de Estrógeno/genética , Hormona Luteinizante/sangre , Ovario/metabolismo , Animales , Regulación hacia Abajo , Receptor beta de Estrógeno/metabolismo , Ciclo Estral/sangre , Ciclo Estral/genética , Femenino , Hipotálamo/metabolismo , Infertilidad Femenina/sangre , Infertilidad Femenina/genética , Hormona Luteinizante/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovario/trasplante , Hipófisis/metabolismo
16.
Proc Natl Acad Sci U S A ; 110(43): 17474-9, 2013 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-24082083

RESUMEN

Primary ovarian insufficiency (POI) and polycystic ovarian syndrome are ovarian diseases causing infertility. Although there is no effective treatment for POI, therapies for polycystic ovarian syndrome include ovarian wedge resection or laser drilling to induce follicle growth. Underlying mechanisms for these disruptive procedures are unclear. Here, we explored the role of the conserved Hippo signaling pathway that serves to maintain optimal size across organs and species. We found that fragmentation of murine ovaries promoted actin polymerization and disrupted ovarian Hippo signaling, leading to increased expression of downstream growth factors, promotion of follicle growth, and the generation of mature oocytes. In addition to elucidating mechanisms underlying follicle growth elicited by ovarian damage, we further demonstrated additive follicle growth when ovarian fragmentation was combined with Akt stimulator treatments. We then extended results to treatment of infertility in POI patients via disruption of Hippo signaling by fragmenting ovaries followed by Akt stimulator treatment and autografting. We successfully promoted follicle growth, retrieved mature oocytes, and performed in vitro fertilization. Following embryo transfer, a healthy baby was delivered. The ovarian fragmentation-in vitro activation approach is not only valuable for treating infertility of POI patients but could also be useful for middle-aged infertile women, cancer patients undergoing sterilizing treatments, and other conditions of diminished ovarian reserve.


Asunto(s)
Infertilidad Femenina/metabolismo , Folículo Ovárico/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Adulto , Animales , Transferencia de Embrión , Femenino , Fertilización In Vitro , Vía de Señalización Hippo , Humanos , Immunoblotting , Recién Nacido , Infertilidad Femenina/genética , Infertilidad Femenina/terapia , Masculino , Ratones , Ratones SCID , Recuperación del Oocito , Folículo Ovárico/trasplante , Embarazo , Resultado del Embarazo , Insuficiencia Ovárica Primaria/genética , Insuficiencia Ovárica Primaria/metabolismo , Insuficiencia Ovárica Primaria/terapia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Resultado del Tratamiento
17.
Am J Physiol Endocrinol Metab ; 305(6): E717-26, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23880317

RESUMEN

Homozygous androgen receptor (AR)-knockout (ARKO) female mice are subfertile due to both intra- and extraovarian (neuroendocrine) defects as defined by ovary transplantation. Using ARKO mice, this study set out to reveal the precise AR-regulated pathways required for optimal androgen-regulated ovulation and fertility. ARKO females exhibit deficient neuroendocrine negative feedback, with a reduced serum luteinizing hormone (LH) response to ovariectomy (OVX) (P < 0.01). Positive feedback is also altered as intact ARKO females, at late proestrus, exhibit an often mistimed endogenous ovulatory LH surge. Furthermore, at late proestrus, intact ARKO females display diminished preovulatory serum estradiol (E2; P < 0.01) and LH (P < 0.05) surge levels and reduced Kiss1 mRNA expression in the anteroventral periventricular nucleus (P < 0.01) compared with controls. However, this reduced ovulatory LH response in intact ARKO females can be rescued by OVX and E2 priming or treatment with endogenous GnRH. These findings reveal that AR regulates the negative feedback response to E2, E2-positive feedback is compromised in ARKO mice, and AR-regulated negative and positive steroidal feedback pathways impact on intrahypothalamic control of the kisspeptin/GnRH/LH cascade. In addition, intraovarian AR-regulated pathways controlling antral to preovulatory follicle dynamics are disrupted because adult ARKO ovaries collected at proestrus have small antral follicles with reduced oocyte/follicle diameter ratios (P < 0.01) and increased proportions of unhealthy large antral follicles (P < 0.05) compared with controls. As a consequence of aberrant follicular growth patterns, proestrus ARKO ovaries also exhibit fewer preovulatory follicle (P < 0.05) and corpora lutea numbers (P < 0.01). However, embryo development to the blastocyst stage is unchanged in ARKO females, and hence, the subfertility is a consequence of reduced ovulations and not altered embryo quality. These findings reveal that the AR has a functional role in neuroendocrine regulation and timing of the ovulatory LH surge as well as antral/preovulatory follicle development.


Asunto(s)
Hipotálamo/metabolismo , Infertilidad Femenina/metabolismo , Ovario/metabolismo , Ovulación/metabolismo , Receptores Androgénicos/metabolismo , Animales , Cuerpo Lúteo/metabolismo , Estradiol/sangre , Ciclo Estral/sangre , Ciclo Estral/genética , Ciclo Estral/metabolismo , Femenino , Hipotálamo/fisiopatología , Infertilidad Femenina/genética , Infertilidad Femenina/fisiopatología , Kisspeptinas/genética , Kisspeptinas/metabolismo , Hormona Luteinizante/sangre , Hormona Luteinizante/metabolismo , Ratones , Ratones Noqueados , Folículo Ovárico/metabolismo , Ovario/fisiopatología , Ovulación/sangre , Ovulación/genética , Receptores Androgénicos/genética
18.
J Tradit Chin Med ; 33(5): 597-602, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24660581

RESUMEN

OBJECTIVE: To investigate the effect of Soothing liver therapy on infertile women undergoing in vitro fertilization and embryo transfer (IVF-ET) and to explore its mechanism. METHODS: Fifty-eight women with tubal infertility were randomized into two groups: 30 in an experimental group treated with Xiaoyao powder (Shugan) plus gonadotropin-releasing hormone analog (GnRHa)/follicle-stimulating hormone (FSH)/ human chorionic gonadotropin (hCG) and 28 in the control group who were treated with GnRHa/FSH/ hCG only. The total gonadotropin (Gn) doses required, endometrial thickness, oocyte numbers, high quality embryo production rate and pregnancy rate of the two groups were compared. The concentration of growth differentiation factor-9 (GDF-9) in follicular fluid was detected by western blotting and the expression of GDF-9 mRNA in granulosa cells was measured using reverse transcription-polymerase chain reaction amplification. RESULTS: In the experimental group, the Gn dose was significantly lower than that in the control group; the endometrial thickness, high quality embryo production and pregnancy rates were significantly higher and the expression of GDF-9 mRNA was also significantly higher than in the control group (all P < 0.05). CONCLUSION: Shugan treatment can improve the pregnancy rate of women with tubal infertility; its mechanism is possibly related to the increased expression of GDF-9 in granulosa cells.


Asunto(s)
Medicamentos Herbarios Chinos/uso terapéutico , Factor 9 de Diferenciación de Crecimiento/genética , Infertilidad Femenina/tratamiento farmacológico , Hígado/efectos de los fármacos , Oocitos/efectos de los fármacos , Adulto , Transferencia de Embrión , Femenino , Fertilización In Vitro , Hormona Folículo Estimulante/uso terapéutico , Hormona Liberadora de Gonadotropina/uso terapéutico , Factor 9 de Diferenciación de Crecimiento/metabolismo , Humanos , Infertilidad Femenina/genética , Infertilidad Femenina/metabolismo , Infertilidad Femenina/terapia , Hígado/fisiopatología , Oocitos/citología , Oocitos/metabolismo , Embarazo
19.
Biol Reprod ; 87(1): 10, 1-10, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22553218

RESUMEN

Treatment of neonatal mice with the phytoestrogen genistein (50 mg/kg/day) results in complete female infertility caused in part by preimplantation embryo loss in the oviduct between Days 2 and 3 of pregnancy. We previously demonstrated that oviducts of genistein-treated mice are "posteriorized" as compared to control mouse oviducts because they express numerous genes normally restricted to posterior regions of the female reproductive tract (FRT), the cervix and vagina. We report here that neonatal genistein treatment resulted in substantial changes in oviduct expression of genes important for the FRT mucosal immune response, including immunoglobulins, antimicrobials, and chemokines. Some of the altered immune response genes were chronically altered beginning at the time of neonatal genistein treatment, indicating that these alterations were a result of the posteriorization phenotype. Other alterations in oviduct gene expression were observed only in early pregnancy, immediately after the FRT was exposed to inflammatory or antigenic stimuli from ovulation and mating. The oviduct changes affected development of the surviving embryos by increasing the rate of cleavage and decreasing the trophectoderm-to-inner cell mass cell ratio at the blastocyst stage. We conclude that both altered immune responses to pregnancy and deficits in oviduct support for preimplantation embryo development in the neonatal genistein model are likely to contribute to infertility phenotype.


Asunto(s)
Desarrollo Embrionario/efectos de los fármacos , Genisteína/toxicidad , Inmunidad Mucosa/efectos de los fármacos , Oviductos/efectos de los fármacos , Oviductos/inmunología , Fitoestrógenos/toxicidad , Animales , Animales Recién Nacidos , Desarrollo Embrionario/genética , Desarrollo Embrionario/inmunología , Desarrollo Embrionario/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genes MHC Clase II/efectos de los fármacos , Genisteína/administración & dosificación , Inmunidad Mucosa/genética , Infertilidad Femenina/inducido químicamente , Infertilidad Femenina/genética , Infertilidad Femenina/inmunología , Infertilidad Femenina/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Oviductos/metabolismo , Oviductos/patología , Fitoestrógenos/administración & dosificación , Embarazo
20.
Mol Endocrinol ; 25(5): 833-46, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21436260

RESUMEN

GnRH is the central regulator of reproductive function responding to central nervous system cues to control gonadotropin synthesis and secretion. GnRH neurons originate in the olfactory placode and migrate to the forebrain, in which they are found in a scattered distribution. Congenital idiopathic hypogonadotropic hypogonadism (CIHH) has been associated with mutations or deletions in a number of genes that participate in the development of GnRH neurons and expression of GnRH. Despite the critical role of GnRH in mammalian reproduction, a comprehensive understanding of the developmental factors that are responsible for regulating the establishment of mature GnRH neurons and the expression of GnRH is lacking. orthodenticle homeobox 2 (OTX2), a homeodomain protein required for the formation of the forebrain, has been shown to be expressed in GnRH neurons, up-regulated during GnRH neuronal development, and responsible for increased GnRH promoter activity in GnRH neuronal cell lines. Interestingly, mutations in Otx2 have been associated with human hypogonadotropic hypogonadism, but the mechanism by which Otx2 mutations cause CIHH is unknown. Here we show that deletion of Otx2 in GnRH neurons results in a significant decrease in GnRH neurons in the hypothalamus, a delay in pubertal onset, abnormal estrous cyclicity, and infertility. Taken together, these data provide in vivo evidence that Otx2 is critical for GnRH expression and reproductive competence.


Asunto(s)
Eliminación de Gen , Hormona Liberadora de Gonadotropina/metabolismo , Hipogonadismo/genética , Neuronas/metabolismo , Factores de Transcripción Otx/genética , Animales , Apoptosis/genética , Caspasa 3/metabolismo , Cuerpo Lúteo/anomalías , Estro/genética , Femenino , Ingeniería Genética , Hormona Liberadora de Gonadotropina/genética , Hipogonadismo/metabolismo , Hipogonadismo/patología , Hipotálamo/metabolismo , Hipotálamo/patología , Infertilidad Femenina/genética , Hormona Luteinizante de Subunidad beta/genética , Hormona Luteinizante de Subunidad beta/metabolismo , Masculino , Ratones , Ratones Noqueados , Factores de Transcripción Otx/deficiencia , Prosencéfalo/metabolismo , Prosencéfalo/patología , Maduración Sexual/genética , Testículo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA