Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Genes (Basel) ; 15(5)2024 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-38790245

RESUMEN

Insulin receptor signaling promotes cell differentiation, proliferation, and growth which are essential for oocyte maturation, embryo implantation, endometrial decidualization, and placentation. The dysregulation of insulin signaling in women with metabolic syndromes including diabetes exhibits poor pregnancy outcomes that are poorly understood. We utilized the Cre/LoxP system to target the tissue-specific conditional ablation of insulin receptor (Insr) and insulin-like growth factor-1 receptor (Igf1r) using an anti-Mullerian hormone receptor 2 (Amhr2) Cre-driver which is active in ovarian granulosa and uterine stromal cells. Our long-term goal is to examine insulin-dependent molecular mechanisms that underlie diabetic pregnancy complications, and our conditional knockout models allow for such investigation without confounding effects of ligand identity, source and cross-reactivity, or global metabolic status within dams. Puberty occurred with normal timing in all conditional knockout models. Estrous cycles progressed normally in Insrd/d females but were briefly stalled in diestrus in Igf1rd/d and double receptor (DKO) mice. The expression of vital ovulatory genes (Lhcgr, Pgr, Ptgs2) was not significantly different in 12 h post-hCG superovulated ovaries in knockout mice. Antral follicles exhibited an elevated apoptosis of granulosa cells in Igf1rd/d and DKO mice. However, the distribution of ovarian follicle subtypes and subsequent ovulations was normal in all insulin receptor mutants compared to littermate controls. While ovulation was normal, all knockout lines were subfertile suggesting that the loss of insulin receptor signaling in the uterine stroma elicits implantation and decidualization defects responsible for subfertility in Amhr2-Cre-derived insulin receptor mutants.


Asunto(s)
Ratones Noqueados , Ovario , Receptor IGF Tipo 1 , Receptor de Insulina , Animales , Femenino , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Ratones , Ovario/metabolismo , Ovario/patología , Infertilidad Femenina/genética , Infertilidad Femenina/metabolismo , Infertilidad Femenina/patología , Ovulación/genética , Células de la Granulosa/metabolismo , Células de la Granulosa/patología , Embarazo , Transducción de Señal/genética
2.
Front Endocrinol (Lausanne) ; 15: 1336496, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38559689

RESUMEN

Immune dysfunction is one of the central components in the development and progression of endometriosis by establishing a chronic inflammatory environment. Western-style high-fat diets (HFD) have been linked to greater systemic inflammation to cause metabolic and chronic inflammatory diseases, and are also considered an environmental risk factor for gynecologic diseases. Here, we aimed to examine how HFD cause an inflammatory environment in endometriosis and discern their contribution to endometriotic-associated hyperalgesia. Our results showed that HFD-induced obesity enhanced abdominal hyperalgesia that was induced by endometriotic lesions. Peritoneal inflammatory macrophages and cytokine levels increased by lesion induction were elevated by chronic exposure to HFD. Increased expression of pain-related mediators in the dorsal root ganglia was observed after lesion induction under the HFD condition. Although HFD did not affect inflammatory macrophages in the peritoneal cavity without lesion induction, the diversity and composition of the gut microbiota were clearly altered by HFD as a sign of low-grade systemic inflammation. Thus, HFD alone might not establish a local inflammatory environment in the pelvic cavity, but it can contribute to further enhancing chronic inflammation, leading to the exacerbation of endometriosis-associated abdominal hyperalgesia following the establishment and progression of the disease.


Asunto(s)
Endometriosis , Femenino , Humanos , Endometriosis/complicaciones , Endometriosis/metabolismo , Hiperalgesia/etiología , Dieta Alta en Grasa/efectos adversos , Inflamación/metabolismo , Abdomen
3.
bioRxiv ; 2023 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-38014254

RESUMEN

Immune dysfunction is one of the central components in the development and progression of endometriosis by establishing a chronic inflammatory environment. Western-style high-fat diets (HFD) have been linked to greater systemic inflammation to cause metabolic and chronic inflammatory diseases, and are also considered an environmental risk factor for gynecologic diseases. Here, we aimed to examine how HFD alter an inflammatory environment in endometriosis and discern their contribution to endometriotic-associated hyperalgesia. Our results showed that HFD-induced obesity enhanced abdominal mechanical allodynia that was induced by endometriotic lesions. Peritoneal inflammatory macrophages and cytokine levels increased by lesion induction were elevated by chronic exposure to HFD. Pain-related mediators in the dorsal root ganglia were further stimulated after lesion induction under the HFD condition. Although HFD did not affect inflammatory macrophages in the peritoneal cavity without lesion induction, the diversity and composition of the gut microbiota were clearly altered by HFD as a sign of low-grade systemic inflammation. Thus, HFD alone might not establish a local inflammatory environment in the pelvic cavity, but it can contribute to further enhancing chronic inflammation, leading to the exacerbation of endometriosis-associated abdominal hyperalgesia following the establishment and progression of the disease.

4.
Commun Biol ; 5(1): 1225, 2022 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-36369244

RESUMEN

Due to the vital roles of macrophages in the pathogenesis of endometriosis, targeting macrophages could be a promising therapeutic direction. Here, we investigated the efficacy of niclosamide for the resolution of a perturbed microenvironment caused by dysregulated macrophages in a mouse model of endometriosis. Single-cell transcriptomic analysis revealed the heterogeneity of macrophages including three intermediate subtypes with sharing characteristics of traditional "small" or "large" peritoneal macrophages (SPMs and LPMs) in the peritoneal cavity. Endometriosis-like lesions (ELL) enhanced the differentiation of recruited macrophages, promoted the replenishment of resident LPMs, and increased the ablation of embryo-derived LPMs, which were stepwise suppressed by niclosamide. In addition, niclosamide restored intercellular communications between macrophages and B cells. Therefore, niclosamide rescued the perturbed microenvironment in endometriosis through its fine regulations on the dynamic progression of macrophages. Validation of similar macrophage pathogenesis in patients will further promote the clinical usage of niclosamide for endometriosis treatment.


Asunto(s)
Endometriosis , Ratones , Humanos , Animales , Femenino , Endometriosis/tratamiento farmacológico , Niclosamida/farmacología , Niclosamida/uso terapéutico , Macrófagos/patología , Macrófagos Peritoneales/patología , Modelos Animales de Enfermedad
5.
Cells ; 11(4)2022 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-35203298

RESUMEN

Estrogen and progesterone and their signaling mechanisms are tightly regulated to maintain a normal menstrual cycle and to support a successful pregnancy. The imbalance of estrogen and progesterone disrupts their complex regulatory mechanisms, leading to estrogen dominance and progesterone resistance. Gynecological diseases are heavily associated with dysregulated steroid hormones and can induce chronic pelvic pain, dysmenorrhea, dyspareunia, heavy bleeding, and infertility, which substantially impact the quality of women's lives. Because the menstrual cycle repeatably occurs during reproductive ages with dynamic changes and remodeling of reproductive-related tissues, these alterations can accumulate and induce chronic and recurrent conditions. This review focuses on faulty progesterone signaling mechanisms and cellular responses to progesterone in endometriosis, adenomyosis, leiomyoma (uterine fibroids), polycystic ovary syndrome (PCOS), and endometrial hyperplasia. We also summarize the association with gene mutations and steroid hormone regulation in disease progression as well as current hormonal therapies and the clinical consequences of progesterone resistance.


Asunto(s)
Leiomioma , Enfermedades Uterinas , Endometrio/anomalías , Estrógenos , Femenino , Humanos , Embarazo , Progesterona
6.
Toxicol Sci ; 185(2): 128-142, 2022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-34865136

RESUMEN

This study was performed to examine whether vapor exposure to cannabis plant matter negatively impacts male reproductive functions and testis development in mice. Adult CD-1 male mice (F0) were exposed to air (control) or 200 mg of vaporized cannabis plant matter 3×/day over a 10-day period. Subsequently, F0 males were bred with drug-naïve CD-1 females to generate F1 males, and F1 offspring were used to generate F2 males. Cannabis vapor exposure decreased sperm count and/or motility in F0 and F1 males and disrupted the progression of germ cell development, as morphometric analyses exhibited an abnormal distribution of the stages of spermatogenesis in F0 males. Although plasma levels of testosterone were not affected by cannabis exposure in any ages or generations of males, dysregulated steroidogenic enzymes, Cyp11a1 and Cyp19a1, were observed in F0 testis. In the neonatal testis from F1 males, although apoptosis was not altered, DNA damage and DNMT1, but not DNMT3A and DNMT3B, were increased in germ cells following cannabis exposure. In contrast, the alterations of DNA damage and DNMT1 expression were not observed in F2 neonatal males. These results suggest that cannabis vapor exposure generationally affects male reproductive functions, probably due to disruption of spermatogenesis in the developing testis.


Asunto(s)
Cannabis , Efectos Tardíos de la Exposición Prenatal , Animales , Cannabis/toxicidad , Femenino , Masculino , Ratones , Efectos Tardíos de la Exposición Prenatal/metabolismo , Reproducción , Espermatogénesis , Testículo/metabolismo , Testosterona
7.
J Med Food ; 25(2): 158-165, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34936814

RESUMEN

Dietary soy protein isolate (SPI) and the isoflavones daidzein and genistein have been shown to provide neuroprotection from stroke. However, the mechanisms remain uncertain. We sought to determine whether the addition of isoflavones to a diet containing caseinate (CAS) as the protein source would induce behavioral neuroprotection similar to that seen previously in rats fed SPI. Furthermore, we aimed to characterize the baseline and poststroke expression of mRNAs involved in pathways previously published as perhaps mediating soy-based neuroprotection from stroke and other markers of neuronal plasticity, oxidative stress, and inflammation. Adult male rats were fed a semipurified diet containing (1) sodium caseinate (CAS), (2) CAS plus daidzein and genistein (CAS+ISO), or (3) SPI for 2 weeks. A subset of rats was euthanized, and tissue was collected for quantitative real-time PCR (qPCR). Remaining rats underwent a middle cerebral artery occlusion to induce a stroke. Samples for qPCR were collected on day 3 poststroke. Rats fed SPI made fewer errors on the skilled ladder rung walking task after stroke compared to rats fed CAS (P < .05). Rats fed CAS+ISO were not different from rats fed CAS or SPI. Significant effects of diet were found at day 0 for Syp, Pparg, and Ywhae and at day 3 for Rtn4 expression. We concluded that the benefits of SPI are not solely attributable to daidzein and genistein.


Asunto(s)
Isoflavonas , Proteínas de Soja , Animales , Dieta , Isoflavonas/farmacología , Masculino , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Caminata
8.
FASEB J ; 35(5): e21584, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33860549

RESUMEN

Endometriosis, a common gynecological disease, causes chronic pelvic pain and infertility in women of reproductive age. Due to the limited efficacy of current therapies, a critical need exists to develop new treatments for endometriosis. Inflammatory dysfunction, instigated by abnormal macrophage (MΦ) function, contributes to disease development and progression. However, the fundamental role of the heterogeneous population of peritoneal MΦ and their potential druggable functions is uncertain. Here we report that GATA6-expressing large peritoneal MΦ (LPM) were increased in the peritoneal cavity following lesion induction. This was associated with increased cytokine and chemokine secretion in the peritoneal fluid (PF), as well as MΦ infiltration, vascularization and innervation in endometriosis-like lesions (ELL). Niclosamide, an FDA-approved anti-helminthic drug, was effective in reducing LPM number, but not small peritoneal MΦ (SPM), in the PF. Niclosamide also inhibits aberrant inflammation in the PF, ELL, pelvic organs (uterus and vagina) and dorsal root ganglion (DRG), as well as MΦ infiltration, vascularization and innervation in the ELL. PF from ELL mice stimulated DRG outgrowth in vitro, whereas the PF from niclosamide-treated ELL mice lacked the strong stimulatory nerve growth response. These results suggest LPM induce aberrant inflammation in endometriosis promoting lesion progression and establishment of the inflammatory environment that sensitizes peripheral nociceptors in the lesions and other pelvic organs, leading to increased hyperalgesia. Our findings provide the rationale for targeting LPM and their functions with niclosamide and its efficacy in endometriosis as a new non-hormonal therapy to reduce aberrant inflammation which may ultimately diminish associated pain.


Asunto(s)
Anticestodos/farmacología , Endometriosis/complicaciones , Factor de Transcripción GATA6/metabolismo , Ganglios Espinales/efectos de los fármacos , Inflamación/tratamiento farmacológico , Macrófagos Peritoneales/efectos de los fármacos , Niclosamida/farmacología , Animales , Femenino , Factor de Transcripción GATA6/genética , Ganglios Espinales/patología , Inflamación/etiología , Inflamación/patología , Macrófagos Peritoneales/patología , Ratones , Ratones Endogámicos C57BL
9.
Biol Reprod ; 105(2): 403-412, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-33855343

RESUMEN

Recent evidence indicates that niclosamide is an anti-cancer compound that is able to inhibit several signaling pathways. Although niclosamide has previously been identified by high-throughput screening platforms as a potential effective compound against several cancer types, no direct binding interactions with distinct biological molecule(s) has been established. The present study identifies key signal transduction mechanisms altered by niclosamide in ovarian cancer. Using affinity purification with a biotin-modified niclosamide derivative and mass spectrometry analysis, several RNA-binding proteins (RBPs) were identified. We chose the two RBPs, FXR1 and IGF2BP2, for further analysis. A significant correlation exists in which high-expression of FXR1 or IGF2BP2 is associated with reduced survival of ovarian cancer patients. Knockdown of FXR1 or IGF2BP2 in ovarian cancer cells resulted in significantly reduced cell viability, adhesion, and migration. Furthermore, FXR1 or IGF2BP2 deficient ovarian cancer cells exhibited reduced response to most doses of niclosamide showing greater cell viability than those with intact RBPs. These results suggest that FXR1 and IGF2BP2 are direct targets of niclosamide and could have critical activities that drive multiple oncogenic pathways in ovarian cancer.


Asunto(s)
Antineoplásicos/farmacología , Niclosamida/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Proteínas de Unión al ARN/genética , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones
10.
FASEB J ; 35(4): e21440, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33749878

RESUMEN

Insulin signaling is critical for the development of preovulatory follicles and progression through the antral stage. Using a conditional knockout model that escapes this blockage, we recently described the role of insulin signaling in granulosa cells during the periovulatory window in mice lacking Insr and Igf1r driven by Pgr-Cre. These mice were infertile, exhibiting defects in ovulation, luteinization, steroidogenesis, and early embryo development. Herein, we demonstrate that while these mice exhibit normal uterine receptivity, uterine cell proliferation and decidualization are compromised resulting in complete absence of embryo implantation in uteri lacking both receptors. While the histological organization of double knockout mice appeared normal, the thickness of their endometrium was significantly reduced. This was supported by the reduced proliferation of both epithelial and stromal cells during the preimplantation stages of pregnancy. Expression and localization of the main drivers of uterine proliferation, ESR1 and PGR, was normal in knockouts, suggesting that insulin signaling acts downstream of these two receptors. While AKT/PI3K signaling was unaffected by insulin receptor ablation, activation of p44/42 MAPK was significantly reduced in both single and double knockout uteri at 3.5 dpc. Overall, we conclude that both INSR and IGF1R are necessary for optimal endometrial proliferation and implantation.


Asunto(s)
Endometrio/fisiología , Insulina/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Animales , Péptidos de Penetración Celular , Implantación del Embrión , Femenino , Regulación de la Expresión Génica/fisiología , Silenciador del Gen , Ratones , Ratones Endogámicos C57BL , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Embarazo , Receptor IGF Tipo 1/genética , Receptor de Insulina/genética , Transducción de Señal
11.
Biol Reprod ; 102(5): 1055-1064, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-31930396

RESUMEN

Ovarian cancer (OvCa) remains the most common cause of death from gynecological malignancies. Genetically engineered mouse models have been used to study initiation, origin, progression, and/or mechanisms of OvCa. Based on the clinical features of OvCa, we examined a quadruple combination of pathway perturbations including PTEN, TRP53, RB1, and/or CDH1. To characterize the cancer-promoting events in the ovarian surface epithelium (OSE), Amhr2cre/+ mice were used to ablate floxed alleles of Pten, Trp53, and Cdh1, which were crossed with TgK19GT121 mice to inactivate RB1 in KRT19-expressing cells. Inactivation of PTEN, TRP53, and RB1 with or without CDH1 led to the development of type I low-grade OvCa with enlarged serous papillary carcinomas and some high-grade serous carcinomas (HGSCs) in older mice. Initiation of epithelial hyperplasia and micropapillary carcinoma started earlier at 1 month in the triple mutations of Trp53, Pten, and Rb1 mice as compared to 2 months in quadruple mutations of Trp53, Pten, Rb1, and Cdh1 mice, whereas both genotypes eventually developed enlarged proliferating tumors that invaded into the ovary at 3-4 months. Mice with triple and quadruple mutations developed HGSC and/or metastatic tumors, which disseminated into the peritoneal cavity at 4-6 months. In summary, inactivation of PTEN, TRP53, and RB1 initiates OvCa from the OSE. Additional ablation of CDH1 further increased persistence of tumor dissemination and ascites fluid accumulation enhancing peritoneal metastasis.


Asunto(s)
Cadherinas/metabolismo , Neoplasias Ováricas/patología , Ovario/metabolismo , Fosfohidrolasa PTEN/metabolismo , Proteínas de Unión a Retinoblastoma/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Cadherinas/genética , Transformación Celular Neoplásica , Epitelio/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Endogámicos , Ratones Noqueados , Mutación , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/secundario , Fosfohidrolasa PTEN/genética , Proteínas de Unión a Retinoblastoma/genética , Proteína p53 Supresora de Tumor/genética
12.
FASEB J ; 34(2): 2376-2391, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31908002

RESUMEN

Recent studies have demonstrated an essential role for insulin signaling in folliculogenesis as conditional ablation of Igf1r in primary follicles elicits defective follicle-stimulating hormone responsiveness blocking development at the preantral stage. Thus the potential role of insulin action in the periovulatory window and in the corpus luteum is unknown. To examine this, we generated conditional Insr,Igf1r, and double receptor knockout mice driven by Pgr-Cre. These models escape the preantral follicle block and in response to superovulatory gonadotropins exhibit normal distribution of ovarian follicles and corpora lutea. However, single ablation of Igf1r leads to subfertility and mice lacking both receptors are infertile. Double knockout mice have impaired oocyte development and ovulation. While some oocytes are released and fertilized, subsequent embryo development is retarded, and the embryos potentially fail to thrive due to lack of luteal support. In support of this, we found reduced expression of key enzymes in the steroid synthesis pathway and reduced serum progesterone. In addition to metabolic and steroidogenic pathways, RNA-sequencing analysis revealed transcription factor-3 as an important transcription factor downstream of insulin signaling. Collectively, these results highlight the importance of growth factors of the insulin family during two distinct windows of follicular development, ovulation, and luteinization.


Asunto(s)
Diferenciación Celular/fisiología , Fertilidad/fisiología , Células de la Granulosa/metabolismo , Insulina/metabolismo , Ovulación/fisiología , Animales , Femenino , Células de la Granulosa/citología , Insulina/genética , Masculino , Ratones , Ratones Noqueados
13.
Biol Reprod ; 102(5): 1011-1019, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-31950153

RESUMEN

Endometriosis is a common gynecological disease, which causes chronic pelvic pain and infertility in women of reproductive age. Due to limited efficacy of current treatment options, a critical need exists to develop new and effective treatments for endometriosis. Niclosamide is an efficacious and FDA-approved drug for the treatment of helminthosis in humans that has been used for decades. We have reported that niclosamide reduces growth and progression of endometriosis-like lesions via targeting STAT3 and NFĸB signaling in a mouse model of endometriosis. To examine the effects of niclosamide on macrophage-induced inflammation in endometriosis, a total of 29 stage III-IV endometrioma samples were used to isolate human endometriotic stromal cells (hESCs). M1 or M2 macrophages were isolated and differentiated from fresh human peripheral blood samples. Then, hESCs were cultured in conditioned media (CM) from macrophages with/without niclosamide. Niclosamide dose dependently reduced cell viability and the activity of STAT3 and NFκB signaling in hESCs. While macrophage CM stimulated cell viability in hESCs, niclosamide inhibited this stimulation. Macrophage CM stimulated the secretion of proinflammatory cytokines and chemokines from hESCs. Most of these secreted factors were inhibited by niclosamide. These results indicate that niclosamide is able to reduce macrophage-induced cell viability and cytokine/chemokine secretion in hESCs by inhibiting inflammatory mechanisms via STAT3 and/or NFκB signaling.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Endometriosis/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Niclosamida/farmacología , Animales , Anticestodos/farmacología , Células Cultivadas , Endometriosis/tratamiento farmacológico , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/tratamiento farmacológico , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Células del Estroma
14.
Toxicol Sci ; 172(2): 303-315, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31532523

RESUMEN

This study was performed to examine the transgenerational effects of bisphenol (BP) A analogs, BPE, and BPS on male reproductive functions using mice as a model. CD-1 mice (F0) were orally exposed to control treatment (corn oil), BPA, BPE, or BPS (0.5 or 50 µg/kg/day) from gestational day 7 (the presence of vaginal plug = 1) to birth. Mice from F1 and F2 offspring were used to generate F3 males. Prenatal exposure to BPA, BPE, and BPS decreased sperm counts and/or motility and disrupted the progression of germ cell development as morphometric analyses exhibited an abnormal distribution of the stages of spermatogenesis in F3 males. Dysregulated serum levels of estradiol-17ß and testosterone, as well as expression of steroidogenic enzymes in F3 adult testis were also observed. In the neonatal testis, although apoptosis and DNA damage were not affected, mRNA levels of DNA methyltransferases, histone methyltransferases, and their associated factors were increased by BP exposure. Furthermore, BP exposure induced immunoreactive expression of DNMT3A in Sertoli cells, strengthened DNMT3B, and weakened H3K9me2 and H3K9me3 in germ cells of the neonatal testis, whereas DNMT1, H3K4me3, and H3K27ac were not affected. In adult testis, stage-specific DNMT3B was altered by BP exposure, although DNMT3A, H3K9me2, and H3K9me3 expression remained stable. These results suggest that prenatal exposure to BPA, BPE, and BPS induces transgenerational effects on male reproductive functions probably due to altered epigenetic modification following disruption of DNMTs and histone marks in the neonatal and/or adult testis.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Disruptores Endocrinos/toxicidad , Fenoles/toxicidad , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Reproducción/efectos de los fármacos , Sulfonas/toxicidad , Testículo/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Estradiol/sangre , Femenino , Masculino , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/metabolismo , Recuento de Espermatozoides , Espermatogénesis/efectos de los fármacos , Testículo/embriología , Testículo/metabolismo , Testículo/patología , Testosterona/sangre
15.
Toxicol Sci ; 170(2): 320-329, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31132128

RESUMEN

This study was performed to examine the transgenerational effects of bisphenol (BP) A analogs, BPE, and BPS on female reproductive functions using mice as a model. CD-1 mice (F0) were orally exposed to control treatment (corn oil), BPA, BPE, or BPS (0.5 or 50 µg/kg/day) from gestational day 7 (the presence of vaginal plug = 1) to birth. Mice from F1 and F2 offspring were used to generate F3 females. Prenatal exposure to BPA, BPE, and BPS accelerated the onset of puberty and exhibited abnormal estrous cyclicity in F3 females, and those females exhibited mating difficulties starting at 6 months of age. Various fertility problems including reduced pregnancy rates, parturition, and nursing issues were also observed starting at 6 months, which worsened at 9 months. The levels of serum estradiol-17ß were elevated by BPA or BPS exposure at the age of 6 months, whereas testosterone levels were not affected. The dysregulated expression of steroidogenic enzymes was observed in the ovary at 3 or 6 months of age by BPE or BPS exposure. However, BPA, BPE, and BPS exposure did not affect neonatal follicular development such as germ cell nest breakdown or follicle numbers in the ovary on postnatal day 4. These results suggest that prenatal exposure to BPA analogs, BPE and BPS, have transgenerational effects on female reproductive functions in mice.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Disruptores Endocrinos/toxicidad , Fenoles/toxicidad , Efectos Tardíos de la Exposición Prenatal , Reproducción/efectos de los fármacos , Sulfonas/toxicidad , Animales , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Femenino , Hormonas Esteroides Gonadales/sangre , Masculino , Ratones , Folículo Ovárico/efectos de los fármacos , Embarazo
16.
Toxicol Sci ; 168(2): 561-571, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30629253

RESUMEN

This study was performed to examine whether prenatal exposure to bisphenol (BP) A analogues, BPE and BPS, negatively impacts female reproductive functions and follicular development using mice as a model. CD-1 mice were orally exposed to control treatment (corn oil), BPA, BPE, or BPS (0.5, 20, or 50 µg/kg/day) from gestational day 11 (the presence of vaginal plug = 1) to birth. Exposure to BPA, BPE, and BPS accelerated the onset of puberty and exhibited abnormal estrous cyclicity, especially with lower doses. Females exposed to BPA, BPE, and BPS exhibited mating difficulties starting at 6 months of age. By 9 months, mice exhibited various fertility problems including reduced pregnancy rate, parturition issues, and increased dead pups at birth. Furthermore, the levels of serum testosterone were elevated by BPE or BPS exposure at the age of 9 months, whereas estrogen levels were not affected. On the other hand, the dysregulated expression of steroidogenic enzymes was observed in the ovary at 3, 6, or 9 months of age by BPE or BPS exposure. When we examined neonatal ovary on postnatal day 4, BPA, BPE, and BPS exposure inhibited germ cell nest breakdown and reduced number of primary and secondary follicles. These results suggest that prenatal exposure to BPA analogues, BPE, and BPS, have effects on fertility in later reproductive life probably due to the disruption of early folliculogenesis.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Contaminantes Ambientales/toxicidad , Ciclo Estral/efectos de los fármacos , Fertilidad/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Fenoles/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Sulfonas/toxicidad , Animales , Relación Dosis-Respuesta a Droga , Estradiol/sangre , Femenino , Masculino , Ratones Endogámicos , Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Razón de Masculinidad , Testosterona/sangre
17.
Biol Reprod ; 100(2): 398-408, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30329025

RESUMEN

Endometriosis causes severe chronic pelvic pain and infertility. We have recently reported that niclosamide treatment reduces growth and progression of endometriosis-like lesions and inflammatory signaling (NF${\rm \small K}$B and STAT3) in a mouse model. In the present study, we examined further inhibitory mechanisms by which niclosamide affects endometriotic lesions using an endometriotic epithelial cell line, 12Z, and macrophages differentiated from a monocytic THP-1 cell line. Niclosamide dose dependently reduced 12Z viability, reduced STAT3 and NF${\rm \small K}$B activity, and increased both cleaved caspase-3 and cleaved PARP. To model the inflammatory microenvironment in endometriotic lesions, we exposed 12Z cells to macrophage conditioned media (CM). Macrophages were differentiated from THP-1 cells using 12-O-tetradecanoylphorbol-13-acetate as M0, and then M0 macrophages were polarized into M1 or M2 using LPS/IFNγ or IL4/IL13, respectively. Conditioned media from M0, M1, or M2 cultures increased 12Z viability. This effect was blocked by niclosamide, and cell viability returned to that of CM from cells treated with niclosamide alone. To assess proteins targeted by niclosamide in 12Z cells, CM from 12Z cells cultured with M0, M1, or M2 with/without niclosamide were analyzed by cytokine/chemokine protein array kits. Conditioned media from M0, M1, and/or M2 stimulated the secretion of cytokines/chemokines from 12Z cells. Production of most of these secreted cytokines/chemokines in 12Z cells was inhibited by niclosamide. Knockdown of each gene in 12Z cells using siRNA resulted in reduced cell viability. These results indicate that niclosamide can inhibit the inflammatory factors in endometriotic epithelial cells stimulated by macrophages by targeting STAT3 and/or NF${\rm \small K}$B signaling.


Asunto(s)
Endometritis/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/metabolismo , Macrófagos/fisiología , Monocitos/efectos de los fármacos , Niclosamida/farmacología , Anticestodos/farmacología , Comunicación Celular , Línea Celular , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Células Epiteliales , Femenino , Humanos , FN-kappa B
18.
Toxicol Sci ; 163(2): 620-631, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29741722

RESUMEN

This study was performed to examine whether prenatal exposure to bisphenol (BP) A analogues, BPE and BPS, negatively impacts male reproductive functions and testis development using mice as a model. CD-1 mice were orally exposed to control treatment (corn oil), BPA, BPE, and BPS (0.5, 20, or 50 µg/kg/day) from gestational day 11 (the presence of vaginal plug = 1) to birth. Although sperm counts were significantly reduced by BPA, BPE, or BPS on postnatal day (PND) 60, only BPE or BPS exposure remained low for sperm motility. Exposure to BPA, BPE, and BPS disrupted the progression of germ cell development, as morphometric analyses exhibited an abnormal distribution of the stages of spermatogenesis. Furthermore, BPS with a dose of 50 µg/kg/day increased a level of serum estradiol-17ß in adult males. On PND12, BPE and BPS significantly increased TUNEL positive cells in neonatal testis, following disruption of the expression of apoptosis, autophagy, and oxidative stress-related factors. In addition, the expression of methyltransferases for DNA methylation and histone modification was also affected by prenatal exposure to BPA, BPE, or BPS in neonatal testis. These results suggest that prenatal exposure to BPE and BPS with physiologically relevant doses affects male reproductive functions probably due to spermatogenic defect in the developing testis.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Disruptores Endocrinos/toxicidad , Fenoles/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Reproducción/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Testículo/efectos de los fármacos , Animales , Compuestos de Bencidrilo/química , Metilación de ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Disruptores Endocrinos/química , Femenino , Masculino , Ratones , Ratones Endogámicos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Fenoles/química , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/patología , Motilidad Espermática/efectos de los fármacos , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Testículo/patología
19.
Reprod Toxicol ; 73: 280-291, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28676390

RESUMEN

This study was performed to examine whether bisphenol (BP) A analogues, BPE and BPS, negatively impacts reproductive functions using mice as a model. CD-1 mice were exposed to control treatment (corn oil), BPA, BPE and BPS (50µg/kg or 10mg/kg) from birth to postnatal day (PND) 60 by s.c. injection every three days. Sperm counts or motility was significantly reduced by BPA, BPE or BPS exposure on PND 60 or PND 90. Exposure of BPA, BPE and BPS disrupted the progression of germ cell development, as morphometric analyses exhibited an abnormal distribution of the stages of spermatogenesis. In females, postnatal BPA and BPE exposure accelerated the onset of puberty, and increased body weight after parturition. Furthermore, postnatal exposure of BPA, BPE and/or BPS increased steroid hormone levels in serum. These results suggest that BPA analogues (BPS and BPE) affects male and female reproductive functions.


Asunto(s)
Disruptores Endocrinos/toxicidad , Fenoles/toxicidad , Reproducción/efectos de los fármacos , Sulfonas/toxicidad , Animales , Compuestos de Bencidrilo , Estradiol/sangre , Femenino , Masculino , Ratones , Maduración Sexual/efectos de los fármacos , Recuento de Espermatozoides , Motilidad Espermática/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Testosterona/sangre
20.
Biol Reprod ; 95(4): 76, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27535961

RESUMEN

Endometriosis causes severe chronic pelvic pain and infertility. Because the standard medication and surgical treatments of endometriosis show high recurrence of symptoms, it is necessary to improve current treatment options. In the initial study, we examined whether niclosamide can be a useful drug for endometriosis in a preclinical setting. Endometriotic implants were induced using an established mouse model involving transimplantation of mouse endometrial fragments to the peritoneal wall of recipient mice. When the recipient mice were treated with niclosamide for 3 weeks, niclosamide reduced the size of endometriotic implants with inhibition of cell proliferation, and inflammatory signaling including RELA (NFKB) and STAT3 activation, but did not alter expression of steroid hormone receptors. To identify genes whose expression is regulated by niclosamide in endometriotic implants, RNA-sequencing was performed, and several genes downregulated by niclosamide were related to inflammatory responses, WNT and MAPK signaling. In a second study designed to assess whether niclosamide affects reproductive function, the recipient mice started receiving niclosamide after the induction of endometriosis. Then, the recipient mice were mated with wild type males, and treatments continued until the pups were born. Niclosamide treated recipient mice became pregnant and produced normal size and number of pups. These results suggest that niclosamide could be an effective therapeutic drug, and acts as an inhibitor of inflammatory signaling without disrupting normal reproductive function.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA