Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Cell Mol Med ; 24(24): 14571-14582, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33174363

RESUMEN

Cranial radiotherapy induces endocrine disorders and reproductive abnormalities, particularly in long-term female cancer survivors, and this might in part be caused by injury to the pituitary gland, but the underlying mechanisms are unknown. The aim of this study was to investigate the influence of cranial irradiation on the pituitary gland and related endocrine function. Female Wistar rat pups on postnatal day 11 were subjected to a single dose of 6 Gy whole-head irradiation, and hormone levels and organ structure in the reproductive system were examined at 20 weeks after irradiation. We found that brain irradiation reduced cell proliferation and induced persistent inflammation in the pituitary gland. The whole transcriptome analysis of the pituitary gland revealed that apoptosis and inflammation-related pathways were up-regulated after irradiation. In addition, irradiation led to significantly decreased levels of the pituitary hormones, growth hormone, adrenocorticotropic hormone, thyroid-stimulating hormone and the reproductive hormones testosterone and progesterone. To conclude, brain radiation induces reduction of pituitary and reproduction-related hormone secretion, this may due to reduced cell proliferation and increased pituitary inflammation after irradiation. Our results thus provide additional insight into the molecular mechanisms underlying complications after head irradiation and contribute to the discovery of preventive and therapeutic strategies related to brain injury following irradiation.


Asunto(s)
Irradiación Craneana , Hipopituitarismo/etiología , Hipopituitarismo/metabolismo , Hipófisis/metabolismo , Hipófisis/efectos de la radiación , Hormonas Hipofisarias/biosíntesis , Hormona Adrenocorticotrópica/biosíntesis , Animales , Biomarcadores , Proliferación Celular/efectos de la radiación , Biología Computacional/métodos , Irradiación Craneana/efectos adversos , Modelos Animales de Enfermedad , Ciclo Estral/efectos de la radiación , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de la radiación , Hipopituitarismo/patología , Inmunohistoquímica , Hipófisis/patología , Hormonas Hipofisarias/deficiencia , Traumatismos por Radiación/complicaciones , Ratas , Transducción de Señal/efectos de la radiación , Transcriptoma , Proteína p53 Supresora de Tumor/metabolismo
2.
EBioMedicine ; 18: 157-170, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28336389

RESUMEN

Adult rats treated concomitantly with insulin and human chorionic gonadotropin exhibit endocrine, metabolic, and reproductive abnormalities that are very similar to those observed in polycystic ovary syndrome (PCOS) patients. In this study, we used this rat model to assess the effects of metformin on PCOS-related uterine dysfunction. In addition to reducing androgen levels, improving insulin sensitivity, and correcting the reproductive cycle, metformin treatment induced morphological changes in the PCOS-like uterus. At the molecular and cellular levels, metformin normalized the androgen receptor-mediated transcriptional program and restored epithelial-stromal interactions. In contrast to glucose transport, uterine inflammatory gene expression was suppressed through the PI3K-Akt-NFκB network, but without affecting apoptosis. These effects appeared to be independent of AMPK subunit and autophagy-related protein regulation. We found that when metformin treatment partially restored implantation, several implantation-related genes were normalized in the PCOS-like rat uterus. These results improve our understanding of how metformin rescues the disruption of the implantation process due to the uterine defects that result from hyperandrogenism and insulin resistance. Our data provide insights into the molecular and functional clues that might help explain, at least in part, the potential therapeutic options of metformin in PCOS patients with uterine dysfunction.


Asunto(s)
Metformina/farmacología , Síndrome del Ovario Poliquístico/patología , Útero/efectos de los fármacos , Animales , Gonadotropina Coriónica/farmacología , Modelos Animales de Enfermedad , Implantación del Embrión/efectos de los fármacos , Estradiol/sangre , Ciclo Estral/efectos de los fármacos , Femenino , Hormona Folículo Estimulante/sangre , Hiperandrogenismo/inducido químicamente , Hiperandrogenismo/patología , Insulina/farmacología , Hormona Luteinizante/sangre , Metformina/uso terapéutico , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Androgénicos/metabolismo , Transducción de Señal/efectos de los fármacos , Testosterona/sangre , Útero/metabolismo , Útero/patología , Proteína Wnt4/genética , Proteína Wnt4/metabolismo
3.
J Ovarian Res ; 10(1): 10, 2017 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-28193283

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) regulate the expression of genes involved in various cellular functions related to metabolism, inflammation, and reproduction. This study evaluated the effects of sex hormones and obesity on the expression of circulating miR-23a and miR-23b in women with polycystic ovary syndrome (PCOS) and healthy women. METHODS: Serum sex hormones concentrations and body mass index (BMI) were measured in 18 women with PCOS and in 30 healthy women from the East China area and these measurements were correlated with serum miR-23a/b levels. The effect of miR-23a and miR-23b risk factors on occurrence of PCOS and predisposing factors of PCOS on these miRNA expressions were evaluated. RESULTS: The expressions of miR-23a/b were significantly lower in the women with PCOS than the normal women, and the expression levels of miR-23a/b were positively correlated with each other in the normal women (p = 0.001) but not in the women with PCOS (p > 0.05). In the women with PCOS, miR-23a was positively correlated with BMI (p = 0.03). However, no correlations were found between the levels of miR-23a/b and the sex hormones in the normal and PCOS women. On the other hand, without considering the presence or absence of PCOS, increase in BMI had a positive effect on the levels of circulating miR-23b; while testosterone had negative effects on the levels of circulating miR-23a. Furthermore, the likelihood of women with PCOS decreased by 0.01-fold for every 1 fold increase of miR-23a expression. CONCLUSIONS: Both reduced levels and discordance between the expressions of miR-23a/b were observed in the women with PCOS and miR-23a/b were affected from testosterone and BMI, reversely. Therefore, miR-23a alteration in contrast with miR-23b is a better indicator for evaluation of PCOS than the miR-23b.


Asunto(s)
Expresión Génica , MicroARNs/genética , Síndrome del Ovario Poliquístico/genética , Adulto , Biomarcadores , Índice de Masa Corporal , Estudios de Casos y Controles , China , Hormonas del Cuerpo Lúteo/sangre , Femenino , Hormona Folículo Estimulante , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Hormonas Esteroides Gonadales/sangre , Humanos , MicroARNs/sangre , Obesidad/sangre , Obesidad/complicaciones , Obesidad/metabolismo , Oportunidad Relativa , Síndrome del Ovario Poliquístico/sangre , Síndrome del Ovario Poliquístico/etiología , Interferencia de ARN , Adulto Joven
4.
Sci Rep ; 6: 30679, 2016 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-27461373

RESUMEN

Peripheral insulin resistance and hyperandrogenism are the primary features of polycystic ovary syndrome (PCOS). However, how insulin resistance and hyperandrogenism affect uterine function and contribute to the pathogenesis of PCOS are open questions. We treated rats with insulin alone or in combination with human chorionic gonadotropin (hCG) and showed that peripheral insulin resistance and hyperandrogenism alter uterine morphology, cell phenotype, and cell function, especially in glandular epithelial cells. These defects are associated with an aberration in the PI3K/Akt signaling pathway that is used as an indicator for the onset of insulin resistance in classical metabolic tissues. Concomitantly, increased GSK3ß (Ser-9) phosphorylation and decreased ERK1/2 phosphorylation in rats treated with insulin and hCG were also observed. We also profiled the expression of glucose transporter (Glut) isoform genes in the uterus under conditions of insulin resistance and/or hyperandrogenism. Finally, we determined the expression pattern of glycolytic enzymes and intermediates during insulin resistance and hyperandrogenism in the uterus. These findings suggest that the PI3K/Akt and MAPK/ERK signaling pathways play a role in the onset of uterine insulin resistance, and they also suggest that changes in specific Glut isoform expression and alterations to glycolytic metabolism contribute to the endometrial dysfunction observed in PCOS patients.


Asunto(s)
Glucosa/metabolismo , Hiperandrogenismo/patología , Resistencia a la Insulina/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Síndrome del Ovario Poliquístico/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Gonadotropina Coriónica/farmacología , Células Epiteliales/patología , Femenino , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucólisis/genética , Humanos , Insulina/farmacología , Resistencia a la Insulina/fisiología , Fosforilación , Ratas , Ratas Sprague-Dawley , Útero/patología
5.
Data Brief ; 6: 135-42, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26858978

RESUMEN

In this data, non-pregnant women during the menstrual cycle, women with normal intrauterine pregnancy (IUP), and women with tubal ectopic pregnancy (EP) after informed consent were included. The serum levels of 17ß-estradiol, progesterone, testosterone, beta-human chorionic gonadotropin, interleukin (IL)-1ß, IL-4, IL-6, IL-7, IL-8, IL-10, tumor necrosis factor α (TNFα), and interferon-γ (IFN-γ), epidermal growth factor, the Chlamydia (C.) trachomatis IgG and HSP60 were analyzed. Receiver operating characteristic analysis was used to assess the diagnostic discrimination of tubal EP and gestational age-matched IUP. Our data show that C. trachomatis infection is associated with IL-8 levels, which had excellent discriminative validity in positively identifying tubal EP (concomitant with C. trachomatis infection) from IUP and non-pregnant conditions regardless of C. trachomatis infection.

6.
N Engl J Med ; 374(3): 223-32, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26789871

RESUMEN

Background Human reproduction depends on the fusion of a mature oocyte with a sperm cell to form a fertilized egg. The genetic events that lead to the arrest of human oocyte maturation are unknown. Methods We sequenced the exomes of five members of a four-generation family, three of whom had infertility due to oocyte meiosis I arrest. We performed Sanger sequencing of a candidate gene, TUBB8, in DNA samples from these members, additional family members, and members of 23 other affected families. The expression of TUBB8 and all other ß-tubulin isotypes was assessed in human oocytes, early embryos, sperm cells, and several somatic tissues by means of a quantitative reverse-transcriptase-polymerase-chain-reaction assay. We evaluated the effect of the TUBB8 mutations on the assembly of the heterodimer consisting of one α-tubulin polypeptide and one ß-tubulin polypeptide (α/ß-tubulin heterodimer) in vitro, on microtubule architecture in HeLa cells, on microtubule dynamics in yeast cells, and on spindle assembly in mouse and human oocytes. Results We identified seven mutations in the primate-specific gene TUBB8 that were responsible for oocyte meiosis I arrest in 7 of the 24 families. TUBB8 expression is unique to oocytes and the early embryo, in which this gene accounts for almost all the expressed ß-tubulin. The mutations affect chaperone-dependent folding and assembly of the α/ß-tubulin heterodimer, disrupt microtubule behavior on expression in cultured cells, alter microtubule dynamics in vivo, and cause catastrophic spindle-assembly defects and maturation arrest on expression in mouse and human oocytes. Conclusions TUBB8 mutations have dominant-negative effects that disrupt microtubule behavior and oocyte meiotic spindle assembly and maturation, causing female infertility. (Funded by the National Basic Research Program of China and others.).


Asunto(s)
Infertilidad Femenina/genética , Meiosis/genética , Microtúbulos/patología , Mutación , Oocitos/fisiología , Huso Acromático/fisiología , Tubulina (Proteína)/genética , Adulto , Animales , Femenino , Humanos , Meiosis/fisiología , Ratones , Microtúbulos/fisiología , ARN
7.
BBA Clin ; 4: 85-91, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26675316

RESUMEN

Background Determination of the role of steroid hormones in expression and regulation of endometrial glucose transport 4 (GLUT4) in humans is important for understanding endometrial disorders such as polycystic ovary syndrome (PCOS), a common hormone-imbalance disease. Methods Endometrial biopsy samples were collected from non-PCOS patients with regular menstrual cycles or with hyperplasia and from PCOS patients with or without hyperplasia. In addition, endometrial tissues from postmenopausal women were incubated with human chorionic gonadotropin (hCG, 10 IU/ml), 17ß-estradiol (E2, 10 nM), progesterone (P4, 100 nM), or a combination of E2 and P4 for 24 h. The expression of GLUT4 was measured at the mRNA level using quantitative real-time polymerase chain reaction (qRT-PCR) and at the protein level using Western blot analysis and immunohistochemistry. Results A cyclical change in GLUT4 expression pattern was observed in non-PCOS patients, and a high level of GLUT4 expression was seen in the proliferative phase compared to the secretory phase. Low levels of GLUT4 expression were found in PCOS patients compared to menstrual cycle phase-matched non-PCOS patients, and there was no significant change in GLUT4 expression in PCOS patients during the menstrual cycle. GLUT4 was localized in both epithelial and stromal cells, with notable changes in epithelial cells. We postulate that decreased GLUT4 expression might be regulated by steroid hormones. In support of this, we showed that in cultured endometrial tissues hCG and E2 alone had no effect on GLUT4 expression. However, P4 alone and P4 in combination with E2 decreased GLUT4 expression. Compared with non-PCOS controls, PCOS patients with endometrial hyperplasia exhibited decreased GLUT4 expression in particular in the epithelial cells. Conclusion We conclude that P4 can induce changes in endometrial GLUT4 expression during the menstrual cycle and that abnormal hormonal conditions such as PCOS disrupt normal patterns of GLUT4 expression in endometrial cells.

8.
Int J Biol Sci ; 11(12): 1376-89, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26681917

RESUMEN

The failure of reproductive success in polycystic ovary syndrome (PCOS) patients could be in part due to endometrial dysfunction. However, no studies have investigated any causality between androgen, androgen receptor (AR) expression, and adenosine monophosphate activated protein kinase (AMPK) activation in the endometrium under physiological and pathological conditions. In the present study, we show that 1) endometrial AR expression levels fluctuate in non-PCOS and PCOS patients during the menstrual cycle; 2) the menstrual phase-dependent alteration of p-AMPKα expression occurs in non-PCOS patients but not in PCOS patients; 3) AR expression is higher in PCOS patients than non-PCOS patients during hyperplasia while AMPKα activation (indicated by the ratio of p-AMPKα to AMPKα); and 4) co-localization of AR and Ki-67 in epithelial cell nuclei is observed in endometrial hyperplasia. Importantly, using in vitro human tissue culture and an in vivo 5α-dihydrotestosterone-treated rat model, we show that the action of androgen on AMPKα activation is likely mediated through nuclear AR, especially in epithelial cells. Collectively, we present evidence that AR expression and AMPKα activation depend on menstrual cycle phase and the presence of PCOS, and the data suggest that AR-mediated regulation of AMPKα activation might play a role in the development of endometrial hyperplasia.


Asunto(s)
Adenilato Quinasa/metabolismo , Endometrio/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Receptores Androgénicos/metabolismo , Animales , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Dihidrotestosterona/farmacología , Endometrio/enzimología , Femenino , Flutamida/farmacología , Humanos , Técnicas In Vitro , Antígeno Ki-67/metabolismo , Fosforilación , Síndrome del Ovario Poliquístico/enzimología , Ratas , Ratas Wistar
9.
Am J Transl Res ; 7(3): 574-86, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26045896

RESUMEN

Conflicting results have been reported regarding whether or not insulin-regulated glucose transporter 4 (GLUT4) is expressed in human and rodent endometria. There is an inverse relationship between androgen levels and insulin-dependent glucose metabolism in women. Hyperandrogenemia, hyperinsulinemia, and insulin resistance are believed to contribute to endometrial abnormalities in women with polycystic ovary syndrome (PCOS). However, it has been unclear in previous studies if endometrial GLUT4 expression is regulated by androgen-dependent androgen receptors (ARs) and/or the insulin receptor/Akt/mTOR signaling network. In this study, we demonstrate that GLUT4 is expressed in normal endometrial cells (mainly in the epithelial cells) and is down-regulated under conditions of hyperandrogenemia in tissues from PCOS patients and in a 5α-dihydrotestosterone-induced PCOS-like rat model. Western blot analysis revealed reduced endometrial GLUT4 expression and increased AR expression in PCOS patients. However, the reduced GLUT4 level was not always associated with an increase in AR in PCOS patients when comparing non-hyperplasia with hyperplasia. Using a human tissue culture system, we investigated the molecular basis by which GLUT4 regulation in endometrial hyperplasia tissues is affected by metformin in PCOS patients. We show that specific endogenous organic cation transporter isoforms are regulated by metformin, and this suggests a direct effect of metformin on endometrial hyperplasia. Moreover, we demonstrate that metformin induces GLUT4 expression and inhibits AR expression and blocks insulin receptor/PI3K/Akt/mTOR signaling in the same hyperplasia human tissues. These findings indicate that changes in endometrial GLUT4 expression in PCOS patients involve the androgen-dependent alteration of AR expression and changes in the insulin receptor/PI3K/Akt/mTOR signaling network.

10.
Reprod Biol Endocrinol ; 13: 40, 2015 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-25967158

RESUMEN

BACKGROUND: The fallopian tube transports the gametes to the fertilization site and delivers the embryo to the uterus at the optimal time for implantation. Progesterone and the classical progesterone receptor are involved in regulating both tubal ciliary beating and muscular contractions, likely via both genomic and non-genomic actions. METHODS: To provide more details of the underlying mechanisms, we investigated the effect of progesterone on gene expression in mice fallopian tubes in vitro at 20 min, 2 h and 8 h post progesterone treatment using microarray and/or quantitative PCR. In parallel, oocyte cumulus complex transport was investigated in ovulating mice that were injected with one of the progesterone receptor antagonists, Org 31710 or CDB2194. RESULTS: Microarray analyses did not reveal any apparently regulated genes 20 min after progesterone treatment, consistent with the proposed non-genomic action of progesterone controlling ciliary beating. After 2 h, 11 genes were identified as up-regulated. Analyses using quantitative PCR at 2 h and 8 h showed a consistent up-regulation of endothelin1 and a down-regulation of its receptor Endothelin receptor A by progesterone. We also confirmed that treatment with progesterone receptor antagonists before ovulation accelerates the transport of the oocyte cumulus complex. CONCLUSIONS: This is the first study showing that progesterone regulates the expression of endothelin1 and endothelin receptor A in the fallopian tube. Together with previous studies of the effects of endothelin on muscular contractions in the fallopian tube, the results from this study suggest that endothelin is a mediator of the progesterone-controlled effects on muscular contraction and eventually gamete transport in the fallopian tube.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Células del Cúmulo/metabolismo , Trompas Uterinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Oocitos/metabolismo , Progesterona/farmacología , Animales , Movimiento Celular/fisiología , Trompas Uterinas/fisiología , Femenino , Ratones , Ratones Endogámicos C57BL , Progesterona/fisiología
11.
Sci Rep ; 5: 8689, 2015 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-25732513

RESUMEN

Previous work from our laboratory demonstrated the existence of miRNAs in human follicular fluid. In the current study, we have sought to identify miRNAs that might affect oocyte/embryo quality in patients undergoing intracytoplasmic sperm injection and to investigate their roles in in vitro fertilization outcomes in mouse oocytes. 53 samples were classified as Group 1 (high quality) if the day-3 embryos had seven and more cells or as Group 2 (low quality) if the embryos had six and fewer cells. TaqMan Human microRNAs cards and qRT-PCR were performed to verify differently expressed miRNAs. The function of the corresponding miRNA was investigated in mouse oocytes by injecting them with miRNA-inhibitor oligonucleotides. We found that hsa-miR-320a and hsa-miR-197 had significantly higher expression levels in the Group 1 follicular fluids than in Group 2 (p = 0.0073 and p = 0.008, respectively). Knockdown of mmu-miR-320 in mouse oocytes strongly decreased the proportions of MII oocytes that developed into two-cell and blastocyst stage embryos (p = 0.0048 and p = 0.0069, respectively). Wnt signaling pathway components had abnormal expression level in miR-320 inhibitor-injected oocytes. This study provides the first evidence that miRNAs in human follicular fluid are indicative of and can influence embryo quality.


Asunto(s)
Desarrollo Embrionario/genética , Fertilización In Vitro , Líquido Folicular , MicroARNs/genética , Animales , Blastocisto/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Oocitos/metabolismo , Interferencia de ARN , Vía de Señalización Wnt
12.
Mol Neurobiol ; 51(1): 252-67, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24781382

RESUMEN

One of the most unfortunate side effects of aminoglycoside (AG) antibiotics such as neomycin is that they target sensory hair cells (HCs) and can cause permanent hearing impairment. We have observed HC loss and microglia-like cell (MLC) activation in the inner ear (cochlea) following neomycin administration. We focused on CX3CL1, a membrane-bound glycoprotein expressed on neurons and endothelial cells, as a way to understand how the MLCs are activated and the role these cells play in HC loss. CX3CL1 is the exclusive ligand for CX3CR1, which is a chemokine receptor expressed on the surface of macrophages and MLCs. In vitro experiments showed that the expression levels of CX3CL1 and CX3CR1 increased in the cochlea upon neomycin treatment, and CX3CL1 was expressed on HCs, while CX3CR1 was expressed on MLCs. When cultured with 1 µg/mL exogenous CX3CL1, MLCs were activated by CX3CL1, and the cytokine level was increased in the cochleae leading to apoptosis in the HCs. In CX3CR1 knockout mice, a significantly greater number of cochlear HCs survived than in wild-type mice when the cochlear explants were cultured with neomycin in vitro. Furthermore, inhibiting the activation of MLCs with minocycline reduced the neomycin-induced HC loss and improved the hearing function in neomycin-treated mice in vivo. Our results demonstrate that CX3CL1-induced MLC activation plays an important role in the induction of HC death and provide evidence for CX3CL1 and CX3CR1 as promising new therapeutic targets for the prevention of hearing loss.


Asunto(s)
Enfermedades del Oído/inducido químicamente , Enfermedades del Oído/patología , Microglía/patología , Neomicina/efectos adversos , Animales , Anticuerpos Neutralizantes/farmacología , Receptor 1 de Quimiocinas CX3C , Células Cultivadas , Quimiocina CX3CL1/metabolismo , Quimiocina CX3CL1/farmacología , Cóclea/efectos de los fármacos , Cóclea/patología , Enfermedades del Oído/metabolismo , Células Ciliadas Auditivas/efectos de los fármacos , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patología , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/complicaciones , Pérdida Auditiva/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/efectos de los fármacos , Minociclina/farmacología , Cadenas Ligeras de Miosina/metabolismo , Neomicina/administración & dosificación , Biosíntesis de Proteínas/efectos de los fármacos , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/metabolismo , Regulación hacia Arriba/efectos de los fármacos
13.
Fertil Steril ; 102(6): 1545-52.e1, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25439799

RESUMEN

OBJECTIVE: To investigate whether allogeneic uterine grafts in a rat model, with tacrolimus immunosuppression, can harbor pregnancies that result in offspring with normal postnatal growth. DESIGN: Experimental animal study. SETTING: University hospital. ANIMAL(S): Lewis rats as uterus donors and Piebald-Virol-Glaxo rats as recipients. INTERVENTION(S): Animals were allocated to one of the following three groups: allogeneic uterus transplantation with end-to-side anastomosis to the external iliac vessels and immunosuppression with tacrolimus (UT+Tac; n = 10); sham surgery and immunosuppression with tacrolimus (Sham+Tac; n = 10); or sham surgery (Sham; n = 10). The rats were subsequently introduced to male rats with proven fertility and in the event of resulting pregnancy cesarean sections were performed on day 22 of pregnancy. MAIN OUTCOME MEASURE(S): Graft viability, fertility rate, perinatal death, birth weight, postnatal birth trajectory. RESULT(S): Pregnancy rate was higher in the control groups (70% Sham and 80% Sham+Tac) than in the transplanted group (50% UT+Tac), although these differences did not reach the significance threshold. There were no differences between groups regarding number of living pups or neonatal deaths. Pups born from UT+Tac mothers had birth weights similar to external control animals from our breeding colony (BC): UT+Tac males 6.2 ± 0.2 g, UT+Tac females 5.5 ± 0.6 g, BC males 5.8 ± 0.2 g, BC females 5.2 ± 0.3 g; n.s. Evaluation of uteri and placentas of pregnant animals revealed a somewhat reduced vascular density in both tissues in the UT+Tac group, and that was not seen in the Sham+Tac group. CONCLUSION(S): Allogeneic uterus transplantation and immunosuppression with tacrolimus is compatible with normal pregnancy and perinatal outcome in a rat model.


Asunto(s)
Inmunosupresores/uso terapéutico , Preñez/inmunología , Tacrolimus/uso terapéutico , Útero/trasplante , Animales , Femenino , Supervivencia de Injerto , Tamaño de la Camada , Masculino , Embarazo , Preñez/fisiología , Ratas , Ratas Endogámicas Lew , Donantes de Tejidos , Trasplante Homólogo
14.
Int J Mol Sci ; 16(1): 49-67, 2014 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-25546387

RESUMEN

Nitric oxide (NO) is highly unstable and has a half-life of seconds in buffer solutions. It is synthesized by NO-synthase (NOS), which has been found to exist in the following three isoforms: neuro nitric oxide synthase (nNOS), inducible nitric oxide synthase (iNOS), and endothelial nitric oxide synthase (eNOS). NOS activity is localized in the reproductive tracts of many species, although direct evidence for NOS isoforms in the Fallopian tubes of mice is still lacking. In the present study, we investigated the expression and regulation of NOS isoforms in the mouse and human Fallopian tubes during the estrous and menstrual cycles, respectively. We also measured isoform expression in humans with ectopic pregnancy and in mice treated with lipopolysaccharide (LPS). Our results confirmed the presence of different NOS isoforms in the mouse and human Fallopian tubes during different stages of the estrous and menstrual cycles and showed that iNOS expression increased in the Fallopian tubes of women with ectopic pregnancy and in LPS-treated mice. Elevated iNOS activity might influence ovulation, cilia beats, contractility, and embryo transportation in such a manner as to increase the risk of ectopic pregnancy. This study has provided morphological and molecular evidence that NOS isoforms are present and active in the human and mouse Fallopian tubes and suggests that iNOS might play an important role in both the reproductive cycle and infection-induced ectopic pregnancies.


Asunto(s)
Trompas Uterinas/enzimología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Embarazo Ectópico/enzimología , Adulto , Animales , Ciclo Estral , Femenino , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Ciclo Menstrual , Ratones , Ratones Endogámicos , Óxido Nítrico Sintasa de Tipo II/genética , Embarazo
15.
J Exp Clin Cancer Res ; 33: 41, 2014 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-24887156

RESUMEN

Although a number of in vitro studies have demonstrated the antiproliferative, anti-invasive, and antimetastatic effects of metformin in multiple cancer cell types, its cellular and molecular mechanisms of anti-cancer action in the endometrium of women with polycystic ovary syndrome (PCOS) have not yet been fully elucidated. Organic cation transporters (OCTs) and multidrug and toxin extrusion proteins (MATEs) are known to be involved in metformin uptake and excretion in cells. In this article, we discuss the novel therapeutic possibilities for early-stage endometrial carcinoma (EC) in women with PCOS focusing on metformin, which might have a direct effect in the endometrium through the OCTs and MATEs. We then review the molecular mechanism(s) of the action of metformin in the endometrium and highlight possible mechanistic insights into the inhibition of cell proliferation and tumor growth and, ultimately, the reversal of early-stage EC into normal endometria in women with PCOS.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma/tratamiento farmacológico , Neoplasias Endometriales/tratamiento farmacológico , Endometrio/efectos de los fármacos , Metformina/farmacología , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Endometrio/metabolismo , Femenino , Humanos , Metformina/uso terapéutico , Proteínas de Transporte de Catión Orgánico/metabolismo , Comunicación Paracrina , Células del Estroma/metabolismo
16.
J Cancer ; 5(3): 173-81, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24563672

RESUMEN

BACKGROUND: Young women with polycystic ovary syndrome (PCOS) have a high risk of developing endometrial carcinoma. There is a need for the development of new medical therapies that can reduce the need for surgical intervention so as to preserve the fertility of these patients. The aim of the study was to describe and discuss cases of PCOS and insulin resistance (IR) women with early endometrial carcinoma while being co-treated with Diane-35 and metformin. METHODS: Five PCOS-IR women who were scheduled for diagnosis and therapy for early endometrial carcinoma were recruited. The hospital records and endometrial pathology reports were reviewed. All patients were co-treated with Diane-35 and metformin for 6 months to reverse the endometrial carcinoma and preserve their fertility. Before, during, and after treatment, endometrial biopsies and blood samples were obtained and oral glucose tolerance tests were performed. Endometrial pathology was evaluated. Body weight (BW), body mass index (BMI), follicle-stimulating hormone (FSH), luteinizing hormone (LH), total testosterone (TT), sex hormone-binding globulin (SHBG), free androgen index (FAI), insulin area under curve (IAUC), and homeostasis model assessment of insulin resistance (HOMA-IR) were determined. RESULTS: Clinical stage 1a, low grade endometrial carcinoma was confirmed before treatment. After 6 months of co-treatment, all patients showed normal epithelia. No evidence of atypical hyperplasia or endometrial carcinoma was found. Co-treatment resulted in significant decreases in BW, BMI, TT, FAI, IAUC, and HOMA-IR in parallel with a significant increase in SHBG. There were no differences in the FSH and LH levels after co-treatment. CONCLUSIONS: Combined treatment with Diane-35 and metformin has the potential to revert the endometrial carcinoma into normal endometrial cells in PCOS-IR women. The cellular and molecular mechanisms behind this effect merit further investigation.

17.
Am J Cancer Res ; 4(1): 73-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24482740

RESUMEN

Endometrial cancer (EC) is the most common gynecological malignancy in women and is the leading cause of cancer-related deaths worldwide. Estrogenic stimulation significantly increases endometrial cell proliferation, and both insulin resistance and hyperinsulinemia are associated with the development of EC in women. It has long been known that insulin resistance occurs in women with polycystic ovary syndrome (PCOS) and/or obesity, but one important unanswered question is whether the insulin resistance associated with PCOS and obesity is part of the etiology of the initiation and development of EC. Therefore, research efforts to understand the common and specific underlying endometrial responses to insulin resistance in women with PCOS and obesity could provide further therapeutic options for early endometrial carcinoma.

18.
Am J Transl Res ; 6(2): 104-13, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24489989

RESUMEN

Endometriosis is a complex and challenging disease that involves aberrant adhesion, growth, and progression of endometrial tissues outside of the uterine cavity, and there is evidence to suggest that estrogen plays a key role in its development and progression. Numerous in vivo clinical studies have described the ectopic expression and regulation of estrogen receptor (ER) and progesterone receptor (PR) in the different types of endometriosis compared to normal or eutopic endometrium. However, we have noticed that conflicting and contradictory results have been presented in terms of ER subtype (ERα and ERß) and PR isoform (PRA and PRB) expression. Both ER and PR are transcription factors and ER/PR-mediated responses depend on the coordinated, opposing, and compensatory functions of ER subtypes and PR isoforms. Moreover, analysis of the uterine phenotypes of ERα/ERß and PRA/PRB knockout mice indicates that different ER subtypes and PR isoforms mediate distinct responses to steroid hormones and play different roles in uterine function. In this review, we outline studies that have elucidated the molecules and signaling pathways that are linked to ER and/or PR signaling pathways in the development and progression of endometriosis.

19.
Int J Clin Exp Pathol ; 7(1): 64-79, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24427327

RESUMEN

Human ectopic pregnancy (EP) is a leading cause of pregnancy-related death, but the molecular basis underlying the onset of tubal EP is largely unknown. Female Dicer1 conditional knockout mice are infertile with dysfunctional Fallopian tube and have a different miRNA expression profile compared to wild-type mice, and we speculated that Dicer-mediated regulation of miRNA expression and specific miRNA-controlled targets might contribute to the onset of tubal EP. In the present study, we used microarray analysis and quantitative RT-PCR to examine the expression of miRNAs and core miRNA regulatory components in Fallopian tube tissues from women with EP. We found that the levels of DICER1, four miRNAs (let-7i, miR-149, miR-182, and miR-424), and estrogen receptor α distinguished the tubal implantation site from the non-implantation site. Computational algorithms and screening for interactions with the estrogen and progesterone receptor signaling pathways showed that the four miRNAs were predicted to target ten genes, including NEDD4, TAF15, and SPEN. Subsequent experiments showed differences in NEDD4 mRNA and protein levels between the implantation and non-implantation sites. Finally, we revealed that increases in smooth muscle cell NEDD4 and stromal cell TAF15, in parallel with a decrease in epithelial cell SPEN, were associated with tubal implantation. Our study suggests that changes in miRNA levels by the DICER-mediated miRNA-processing machinery result in aberrant expression of cell type-specific proteins that are potentially involved in the onset of tubal EP.


Asunto(s)
Trompas Uterinas/metabolismo , MicroARNs/genética , Embarazo Tubario/genética , Adulto , Western Blotting , ARN Helicasas DEAD-box , Trompas Uterinas/patología , Femenino , Humanos , Inmunohistoquímica , Análisis de Secuencia por Matrices de Oligonucleótidos , Embarazo , Embarazo Tubario/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Ribonucleasa III , Transcriptoma
20.
J Biomed Sci ; 21: 2, 2014 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-24405633

RESUMEN

Polycystic ovary syndrome (PCOS) is a state of altered steroid hormone production and activity. Chronic estrogen exposure or lack of progesterone due to ovarian dysfunction can result in endometrial hyperplasia and carcinoma. A key contributor to our understanding of progesterone as a critical regulator for normal uterine function has been the elucidation of progesterone receptor (PR) expression, regulation, and signaling pathways. Several human studies indicate that PR-mediated signaling pathways in the nucleus are associated with progesterone resistance in women with PCOS. The aim of this review is to provide an overview of endometrial progesterone resistance in women with PCOS; to present the PR structure, its different isoforms, and their expression in the endometrium; to illustrate the possible regulation of PR and PR-mediated signaling in progesterone resistance in women with PCOS; and to discuss current clinical treatments for atypical endometrial hyperplasia and endometrial carcinoma in women with PCOS and accompanying progesterone resistance.


Asunto(s)
Endometrio/anomalías , Síndrome del Ovario Poliquístico/genética , Progesterona/biosíntesis , Progesterona/genética , Enfermedades Uterinas/genética , Hiperplasia Endometrial/genética , Hiperplasia Endometrial/patología , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Femenino , Regulación de la Expresión Génica , Humanos , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Progesterona/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA