Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

País/Región como asunto
País de afiliación
Intervalo de año de publicación
1.
Nucleic Acids Res ; 51(7): 3150-3165, 2023 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-36869674

RESUMEN

DNA double-strand breaks (DSBs) are functionally linked to genomic instability in spermatocytes and to male infertility. The heavy metal cadmium (Cd) is known to induce DNA damage in spermatocytes by unknown mechanisms. Here, we showed that Cd ions impaired the canonical non-homologous end-joining (NHEJ) repair pathway, but not the homologous recombination (HR) repair pathway, through stimulation of Ser2056 and Thr2609 phosphorylation of DNA-PKcs at DSB sites. Hyper-phosphorylation of DNA-PKcs led to its premature dissociation from DNA ends and the Ku complex, preventing recruitment of processing enzymes and further ligation of DNA ends. Specifically, this cascade was initiated by the loss of PP5 phosphatase activity, which results from the dissociation of PP5 from its activating ions (Mn), that is antagonized by Cd ions through a competitive mechanism. In accordance, in a mouse model Cd-induced genomic instability and consequential male reproductive dysfunction were effectively reversed by a high dosage of Mn ions. Together, our findings corroborate a protein phosphorylation-mediated genomic instability pathway in spermatocytes that is triggered by exchange of heavy metal ions.


Asunto(s)
Cadmio , Inestabilidad Genómica , Infertilidad Masculina , Espermatocitos , Animales , Humanos , Masculino , Ratones , Cadmio/toxicidad , ADN/metabolismo , Reparación del ADN por Unión de Extremidades , Reparación del ADN , Inestabilidad Genómica/efectos de los fármacos , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Iones/metabolismo , Fosforilación , Reparación del ADN por Recombinación , Espermatocitos/efectos de los fármacos
2.
Biol Reprod ; 109(1): 83-96, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37115805

RESUMEN

The aim of this study was to determine the impact of glycyrrhizin, an inhibitor of high mobility group box 1, on glucose metabolic disorders and ovarian dysfunction in mice with polycystic ovary syndrome. We generated a polycystic ovary syndrome mouse model by using dehydroepiandrosterone plus high-fat diet. Glycyrrhizin (100 mg/kg) was intraperitoneally injected into the polycystic ovary syndrome mice and the effects on body weight, glucose tolerance, insulin sensitivity, estrous cycle, hormone profiles, ovarian pathology, glucolipid metabolism, and some molecular mechanisms were investigated. Increased number of cystic follicles, hormonal disorders, impaired glucose tolerance, and decreased insulin sensitivity in the polycystic ovary syndrome mice were reverted by glycyrrhizin. The increased high mobility group box 1 levels in the serum and ovarian tissues of the polycystic ovary syndrome mice were also reduced by glycyrrhizin. Furthermore, increased expressions of toll-like receptor 9, myeloid differentiation factor 88, and nuclear factor kappa B as well as reduced expressions of insulin receptor, phosphorylated protein kinase B, and glucose transporter type 4 were restored by glycyrrhizin in the polycystic ovary syndrome mice. Glycyrrhizin could suppress the polycystic ovary syndrome-induced upregulation of high mobility group box 1, several inflammatory marker genes, and the toll-like receptor 9/myeloid differentiation factor 88/nuclear factor kappa B pathways, while inhibiting the insulin receptor/phosphorylated protein kinase B/glucose transporter type 4 pathways. Hence, glycyrrhizin is a promising therapeutic agent against polycystic ovary syndrome.


Asunto(s)
Resistencia a la Insulina , Síndrome del Ovario Poliquístico , Femenino , Humanos , Ratones , Animales , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Síndrome del Ovario Poliquístico/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Insulina/metabolismo , Ácido Glicirrínico/efectos adversos , Receptor Toll-Like 9/metabolismo , Receptor Toll-Like 9/uso terapéutico , FN-kappa B/metabolismo , Transportador de Glucosa de Tipo 4 , Factor 88 de Diferenciación Mieloide/metabolismo , Insulina/metabolismo , Glucosa/efectos adversos
3.
Reproduction ; 166(5): 323-336, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37651270

RESUMEN

In brief: Obese PCOS mice display metabolic and endocrine disorders that manifest as abnormal metabolism of glucose and dysfunctions in the reproductive system. This study demonstrates that emodin alleviates most of these conditions possibly via the HMGB1/TLR4/NF-kB pathway. Abstract: PCOS is a reproductive disorder with an unclear etiology. It affects 5-10% of women worldwide and is largely associated with impaired glucose metabolism and obesity. HMGB1 is a nuclear protein associated with impaired glucose metabolism and PCOS. We sought to investigate the potential therapeutic effects of emodin on glucose metabolism and ovarian functions in PCOS mice via the HMGB1 molecular pathway. A high-fat diet (HFD) and dehydroepiandrosterone (DHEA)- induced PCOS mouse model comprising four experimental groups was established: control, PCOS, PCOS plus emodin, and PCOS plus vehicle groups. Emodin administration attenuated obesity, elevated fasting glucose levels, impaired glucose tolerance, and insulin resistance, and improved the polycystic ovarian morphology of PCOS mice. Additionally, it lowered elevated serum HMGB1, LH, and testosterone levels in PCOS mice. Elevated ovarian protein and mRNA levels of HMGB1 and TLR4 in PCOS mice were also lowered following emodin treatment. Furthermore, emodin lowered high NF-ĸB/65 protein levels in the ovaries of PCOS mice. Immunohistochemical staining of the ovaries revealed strong HMGB1, TLR4, and AR expressions in PCOS mice, which were lowered by emodin treatment. Moreover, emodin significantly increased GLUT4, IRS2, and INSR levels that were lowered by PCOS. Overall, our study showed that emodin alleviated the impaired glucose metabolism and improved ovarian function in PCOS mice, possibly via the HMGB1/TLR4/NF-ĸB signaling pathway. Thus, emodin could be considered a potential therapeutic agent in the management of PCOS.


Asunto(s)
Emodina , Proteína HMGB1 , Síndrome del Ovario Poliquístico , Animales , Femenino , Humanos , Ratones , Emodina/farmacología , Emodina/uso terapéutico , Glucosa/metabolismo , Proteína HMGB1/genética , FN-kappa B , Obesidad/complicaciones , Síndrome del Ovario Poliquístico/metabolismo , Receptor Toll-Like 4/genética
4.
Biol Reprod ; 106(4): 756-765, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35098296

RESUMEN

PROBLEM: Natural killer (NK) cells from the peripheral blood and spleen represent the source from which various tissues replenish their immune cell populations. Hyperandrogenism and high interleukin-2 (IL-2) levels are factors present in polycystic ovary syndrome (PCOS). These factors and metformin, one of the commonest medications used in treating PCOS, may have an impact on NK cells. However, this is presently unknown. Here, we aimed to assess the distribution of peripheral blood and splenic NK cells and their CD2 and CD94 expression patterns in a PCOS mouse model and test whether metformin could reverse these effects. METHOD OF STUDY: Four mouse groups were designed as follows (n = 15/group): control, PCOS, PCOS plus vehicle, PCOS plus metformin. Dehydroepiandrosterone and a high-fat diet were administered to induce the PCOS mouse model. Flow cytometry was used to analyze the expressions of CD2 and CD94 on peripheral blood and splenic NK cells. RESULTS: PCOS mice had a low surface-density of CD2 on peripheral blood NK cells and a decreased percentage of CD2+ splenic NK cells. Metformin administration did not significantly influence these changes; however, it reduced the splenic NK cell counts. CONCLUSIONS: Our findings proved the association of PCOS with an altered expression of CD2 on peripheral blood and splenic NK cells and that of metformin with a lowered splenic NK cell reserve in PCOS conditions. These findings could further unlock key mechanisms in PCOS pathophysiology and in the mechanism of action of metformin, towards improving PCOS management.


Asunto(s)
Resistencia a la Insulina , Metformina , Síndrome del Ovario Poliquístico , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Células Asesinas Naturales , Metformina/farmacología , Metformina/uso terapéutico , Ratones
5.
Mol Hum Reprod ; 28(5)2022 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-35536241

RESUMEN

Human cytotrophoblast (CTB) differentiation into syncytiotrophoblast (STB) is essential for placental formation and function. Understanding the molecular mechanisms involved in trophoblast differentiation is necessary as it would help in the development of novel therapeutic agents to treat placentation-mediated pregnancy complications. In this study, we found a common upregulated gene, ADAM-like Decysin-1 (ADAMDEC1), from five published microarray and RNA-sequencing datasets. Interference to ADAMDEC1 impaired forskolin-induced BeWo cells differentiation, while ADAMDEC1 overexpression promoted BeWo cells and 3D JEG-3 spheroids differentiation. Interestingly, ADAMDEC1 may inhibit Thrombospondin 1 rather than E-cadherin to trigger the activation of the cAMP signal pathway during CTB differentiation into STB. More importantly, a decreasing in ADAMDEC1 might be involved in the development of preeclampsia. Therefore, ADAMDEC1 is expected to become a new target for prediction of and intervention in placenta-derived pregnancy diseases.


Asunto(s)
Preeclampsia , Trofoblastos , Diferenciación Celular/genética , Línea Celular Tumoral , Femenino , Humanos , Placenta , Placentación/genética , Preeclampsia/genética , Preeclampsia/metabolismo , Embarazo , Trofoblastos/metabolismo
6.
Reproduction ; 163(5): 309-321, 2022 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-35275842

RESUMEN

Decidualization of uterine stromal cells plays an important role in the establishment of normal pregnancy. Previous studies have demonstrated that Acyl-CoA binding protein (Acbp) is critical to cellular proliferation, differentiation, mitochondrial functions, and autophagy. The characterization and physiological function of Acbp during decidualization remain largely unknown. In the present study, we conducted the expression profile of Acbp in the endometrium of early pregnant mice. With the occurrence of decidualization, the expression of Acbp gradually increased. Similarly, Acbp expression was also strongly expressed in decidualized cells following artificial decidualization, both in vivo and in vitro. We applied the mice pseudopregnancy model to reveal that the expression of Acbp in the endometrium of early pregnant mice was not induced by embryonic signaling. Moreover, P4 significantly upregulated the expression of Acbp, whereas E2 appeared to have no regulating effect on Acbp expression in uterine stromal cells. Concurrently, we found that interfering with Acbp attenuated decidualization, and that might due to mitochondrial dysfunctions and the inhibition of fatty acid oxidation. The level of autophagy was increased after knocking down Acbp. During induced decidualization, the expression of ACBP was decreased with the treatment of rapamycin (an autophagy inducer), while increased with the addition of Chloroquine (an autophagy inhibitor). Our work suggests that Acbp plays an essential role in the proliferation and differentiation of stromal cells during decidualization through regulating mitochondrial functions, fatty acid oxidation, and autophagy.


Asunto(s)
Decidua , Inhibidor de la Unión a Diazepam , Animales , Decidua/metabolismo , Inhibidor de la Unión a Diazepam/metabolismo , Endometrio/metabolismo , Femenino , Ratones , Embarazo , Seudoembarazo , Células del Estroma/metabolismo
7.
Reprod Biomed Online ; 44(5): 791-802, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35370096

RESUMEN

RESEARCH QUESTION: What is the expression pattern of inflammatory mRNA profiles of a dehydroepiandrosterone (DHEA) plus high-fat diet (HFD)-induced polycystic ovary syndrome (PCOS) mouse model? DESIGN: RNA sequencing was performed to investigate the mRNA expression profiles in the ovarian tissues of a DHEA plus HFD-induced PCOS mouse model. Six samples were divided into two groups (control and PCOS), with three biological replicates in each group. This was followed by hierarchical clustering, gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses. The relative expression levels of nine inflammatory genes were validated via quantitative reverse-transcription polymerase chain reaction. RESULTS: A total of 436 genes were differentially expressed between the control and PCOS mice. Out of these, 137 genes were up-regulated while 299 genes were down-regulated. Gene ontology analysis indicated that differentially expressed mRNA were associated with T cell-mediated cytotoxicity and homocysteine metabolic processes. Pathway analysis further showed that these abnormally expressed mRNA were associated with signalling pathways, such as NF-kB signalling, tyrosine metabolism and phenylalanine metabolism. All these pathways are involved in chronic inflammation and PCOS. CONCLUSION: The differentially expressed genes are potentially involved in the inflammation that is evident in PCOS, and so could serve as therapeutic options against the disease. Nevertheless, prospective studies are needed to test this hypothesis.


Asunto(s)
Síndrome del Ovario Poliquístico , Animales , Deshidroepiandrosterona , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamación , Ratones , Síndrome del Ovario Poliquístico/complicaciones , ARN Mensajero/genética
8.
Exp Cell Res ; 409(1): 112885, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34662557

RESUMEN

The cytoskeleton, with its actin bundling proteins, plays crucial roles in a host of cellular function, such as cancer metastasis, antigen presentation and trophoblast migration and invasion, as a result of cytoskeletal remodeling. A key player in cytoskeletal remodeling is fascin. Upregulation of fascin induces the transition of epithelial phenotypes to mesenchymal phenotypes through complex interaction with transcription factors. Fascin expression also regulates mitochondrial F-actin to promote oxidative phosphorylation (OXPHOS) in some cancer cells. Trophoblast cells, on the other hand, exhibit similar physiological functions, involving the upregulation of genes crucial for its migration and invasion. Owing to the similar tumor-like characteristics among cancer and trophoblats, we review recent studies on fascin in relation to cancer and trophoblast cell biology; and based on existing evidence, link fascin to the establishment of the maternal-fetal interface.


Asunto(s)
Carcinogénesis/genética , Proteínas Portadoras/genética , Implantación del Embrión/genética , Proteínas de Microfilamentos/genética , Animales , Movimiento Celular/genética , Humanos , Fosforilación Oxidativa
9.
Exp Cell Res ; 403(1): 112597, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33862100

RESUMEN

Inadequate trophoblast proliferation, shallow invasion and exaggerated rate of trophoblast apoptosis are implicated in early recurrent miscarriage (ERM). However, the mechanistic bases of this association have not been fully established. We aimed at investigating the involvement of fascin, an actin-bundling protein, in trophoblast activities and ERM. We found that fascin was downregulated in the cytotrophoblasts (CTBs) and distal cytotrophoblasts (DCTs) of ERM placentae. Knockdown of fascin altered cellular and nucleolar morphology, and inhibited the proliferation but increased apoptosis of trophoblastic HTR8/SVneo cells. Furthermore, fascin knockdown decreased the expression of transcription factors such as Snail1/2, Twist and Zeb1/2, mesenchymal molecules such as Vimentin and N-cadherin, and the protein expression of phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2) and phosphorylates signal transducer and activator of transcript 3 (STAT3). Exposure of HTR-8/SVneo cells to hypoxia reoxygenation (H/R) decreased fascin expression to affect the cells' invasion. Our results indicate for the first time that the downregulation of fascin is involved in the pathogenesis of early recurrent miscarriage; and hence a potential therapeutic target against the disease.


Asunto(s)
Proteínas Portadoras/metabolismo , Proliferación Celular/fisiología , Vellosidades Coriónicas/metabolismo , Proteínas de Microfilamentos/metabolismo , Placenta/metabolismo , Aborto Habitual/metabolismo , Movimiento Celular/fisiología , Regulación hacia Abajo , Femenino , Humanos , Fosforilación , Embarazo , Transducción de Señal/fisiología
10.
Biol Reprod ; 104(1): 71-82, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-32940657

RESUMEN

Ephrins are ligands of Eph receptors (Ephs); both of which are sorted into two classes, A and B. There are five types of ephrin-As (ephrin-A1-5) and three types of ephrin-Bs (ephrin-B1-3). Also, there are 10 types of EphAs (EphA1-10) and six types of EphBs (EphB1-6). Binding of ephrins to the Eph receptors activates signaling cascades that regulate several biological processes such as cellular proliferation, differentiation, migration, angiogenesis, and vascular remodeling. Clarification of their roles in the female reproductive system is crucial to understanding the physiology and pathology of this system. Such knowledge will also create awareness regarding the importance of these molecules in diagnostic, prognostic, and therapeutic medicine. Hence, we have discussed the involvement of these molecules in the physiological and pathological events that occur within the female reproductive system. The evidence so far suggests that the ephrins and the Eph receptors modulate folliculogenesis, ovulation, embryo transport, implantation, and placentation. Abnormal expression of some of these molecules is associated with polycystic ovarian syndrome, ovarian cancer, tubal pregnancy, endometrial cancer, uterine leiomyoma (fibroids), cervical cancer, and preeclampsia, suggesting the need to utilize these molecules in the clinical setting. To enhance a quick development of this gradually emerging field in female reproductive medicine, we have highlighted some "gaps in knowledge" that need prospective investigation.


Asunto(s)
Efrinas/metabolismo , Receptores de la Familia Eph/metabolismo , Transducción de Señal/fisiología , Útero/metabolismo , Animales , Implantación del Embrión/fisiología , Femenino , Humanos
11.
Biol Reprod ; 102(1): 8-17, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-31494673

RESUMEN

Thyroid hormones (THs) regulate a number of metabolic processes during pregnancy. After implantation, the placenta forms and enhances embryonic growth and development. Dysregulated maternal THs signaling has been observed in malplacentation-mediated pregnancy complications such as preeclampsia, miscarriage, and intrauterine growth restriction (IUGR), but the molecular mechanisms involved in this association have not been fully characterized. In this review, we have discussed THs signaling and its roles in trophoblast proliferation, trophoblast differentiation, trophoblast invasion of the decidua, and decidual angiogenesis. We have also explored the relationship between specific pregnancy complications and placental THs transporters, deiodinases, and THs receptors. In addition, we have examined the effects of specific endocrine disruptors on placental THs signaling. The available evidence indicates that THs signaling is involved in the formation and functioning of the placenta and serves as the basis for understanding the pathogenesis and pathophysiology of dysthyroidism-associated pregnancy complications such as preeclampsia, miscarriage, and IUGR.


Asunto(s)
Placenta/metabolismo , Placentación/fisiología , Hormonas Tiroideas/metabolismo , Animales , Femenino , Humanos , Embarazo , Complicaciones del Embarazo/metabolismo , Trofoblastos/metabolismo
12.
Biol Reprod ; 102(1): 18-26, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-31566220

RESUMEN

During pregnancy, there is increased expression of some cytokines at the fetal-maternal interface; and the clarification of their roles in trophoblast-endometrium interactions is crucial to understanding the mechanism of placentation. This review addresses the up-to-date reported mechanisms by which the members of the transforming growth factor beta superfamily regulate trophoblast proliferation, differentiation, and invasion of the decidua, which are the main phases of placentation. The available information shows that these cytokines regulate placentation in somehow a synergistic and an antagonistic manner; and that dysregulation of their levels can lead to aberrant placentation. Nevertheless, prospective studies are needed to reconcile some conflicting reports; and identify some unknown mediators involved in the actions of these cytokines before their detailed mechanistic regulation of human placentation could be fully characterized. The TGF beta superfamily are expressed in the placenta, and regulate the process of placentation through the activation of several signaling pathways.


Asunto(s)
Placenta/metabolismo , Placentación/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Trofoblastos/metabolismo , Animales , Citocinas/metabolismo , Endometrio/metabolismo , Femenino , Humanos , Embarazo
13.
Biol Reprod ; 102(2): 276-291, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31621835

RESUMEN

Anti-androgenic endocrine-disrupting chemicals (EDCs) can cross the placenta to modify early offspring sexual dimorphic markers. These changes are linked to anogenital distance (AGD), which is an androgen-sensitive anthropometric parameter used as a biomarker of perineal growth and caudal migration of the genital tubercle. This review aimed to summarize strength of evidence for associations of in utero exposure to EDCs with AGD and to identify gaps and limitations in the literature so as to inform future research. We performed an electronic search of English literature in September 2019 in medical literature analysis and retrieval system online (MEDLINE), Web of Science and Toxline. We included epidemiological studies that examined in utero exposure to persistent and nonpersistent EDCs and considered AGD in offspring as an outcome. Our review contained 16 investigations examining exposure to persistent EDCs (nine studies) and nonpersistent EDCs (seven studies). Some individual studies reported an inverse association between exposure to bisphenol A (BPA), dioxins, perfluoroalkyl substances, and organochlorides and AGD in both male and female offspring. Meta-analysis of three studies found a small reduction of AGD in female offspring exposed to BPA. The number of studies per chemical is small, and number of subjects examined is limited; so, replication of these results is needed. To achieve more specificity and better replication of results, future studies should establish the association of nonpersistent EDCs using multiple urine samples, evaluate the cumulative impact of exposure to a mixture of anti-androgenic chemicals, and offer adequate consideration of more maternal- and children-related confounding factors.


Asunto(s)
Canal Anal/anatomía & histología , Disruptores Endocrinos/administración & dosificación , Genitales/anatomía & histología , Exposición Materna/efectos adversos , Efectos Tardíos de la Exposición Prenatal , Canal Anal/efectos de los fármacos , Antropometría , Femenino , Genitales/efectos de los fármacos , Humanos , Masculino , Embarazo
14.
Cytokine ; 133: 155105, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32438278

RESUMEN

Activins and inhibins - comprising activin A, B, AB, C and E, and inhibin A and B isoforms - belong to the transforming growth factor beta (TGFß) superfamily. They regulate several biological processes, including cellular proliferation, differentiation and invasiveness, to enhance the formation and functioning of many human tissues and organs. In this review, we have discussed the role of activin and inhibin signaling in the physiological and female-specific pathological events that occur in the female reproductive system. The up-to-date evidence indicates that these cytokines regulate germ cell development, follicular development, ovulation, uterine receptivity, decidualization and placentation through the activation of several signaling pathways; and that their dysregulated expression is involved in the pathogenesis and pathophysiology of the numerous diseases, including pregnancy complications, that disturb reproduction. Hence, some of the isoforms have been suggested as potential biomarkers and therapeutic targets for the management of some of these diseases. Tackling the research directions highlighted in this review will enhance a detailed comprehension and the clinical utility of these cytokines.


Asunto(s)
Activinas/metabolismo , Inhibinas/metabolismo , Reproducción/fisiología , Transducción de Señal/fisiología , Animales , Femenino , Humanos , Embarazo
15.
Mol Reprod Dev ; 87(8): 837-842, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32691498

RESUMEN

Decidualization is a crucial precedent to embryo implantation, as its impairment is a major contributor to female infertility and pregnancy complications. Unraveling the molecular mechanisms involved in the impairment of decidualization has been a subject of interest in the field of reproductive medicine. Evidence from several experimental settings show that exposure to bisphenol A (BPA), an endocrine-disrupting chemical, affects the expression of several molecules that are involved in decidualization. Both low and high doses of BPA impair decidualization through the dysregulation of estrogen (ER) and progesterone (PR) receptors. Exposure to low doses of BPA leads to decreased levels and activities of several antioxidant enzymes, increased activity of endothelial nitric oxide synthase (eNOS), and increased production of nitric oxide (NO) via the upregulation of ER and PR. Consequently, oxidative stress is induced and decidualization becomes impaired. On the other hand, exposure to high doses of BPA downregulates ER and PR and impairs decidualization through two distinct pathways. One is through the upregulation of early growth response-1 (EGR1) via increased phosphorylation of extracellular signal-regulated protein kinases 1 and 2; and the other is through a reduced serum glucocorticoid-induced kinase-1 (SGK1)-mediated downregulation of epithelial sodium channel-α and the induction of oxidative stress. Thus, regardless of the dose, BPA can impair decidualization to trigger infertility and pregnancy complications. This warrants the need to adopt lifestyles that will decrease the tendency of getting exposed to BPA.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Decidua/efectos de los fármacos , Implantación del Embrión/efectos de los fármacos , Fenoles/toxicidad , Animales , Decidua/fisiología , Implantación del Embrión/fisiología , Implantación Tardía del Embrión/efectos de los fármacos , Disruptores Endocrinos/toxicidad , Femenino , Humanos , Enfermedades Placentarias/inducido químicamente , Enfermedades Placentarias/patología , Embarazo , Transducción de Señal/efectos de los fármacos
16.
Rev Environ Contam Toxicol ; 251: 131-184, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31129734

RESUMEN

Maternal exposure to endocrine-disrupting chemicals (EDCs) is associated with long-term hormone-dependent effects that are sometimes not revealed until maturity, middle age, or adulthood. The aim of this study was to conduct descriptive reviews on animal experimental and human epidemiological evidence of the adverse health effects of in utero and lactational exposure to selected EDCs on the first generation and subsequent generation of the exposed offspring. PubMed, Web of Science, and Toxline databases were searched for relevant human and experimental animal studies on 29 October 29 2018. Search results were screened for relevance, and studies that met the inclusion criteria were evaluated and qualitative data extracted for analysis. The search yielded 73 relevant human and 113 animal studies. Results from studies show that in utero and lactational exposure to EDCs is associated with impairment of reproductive, immunologic, metabolic, neurobehavioral, and growth physiology of the exposed offspring up to the fourth generation without additional exposure. Little convergence is seen between animal experiments and human studies in terms of the reported adverse health effects which might be associated with methodologic challenges across the studies. Based on the available animal and human evidence, in utero and lactational exposure to EDCs is detrimental to the offspring. However, more human studies are necessary to clarify the toxicological and pathophysiological mechanisms underlying these effects.


Asunto(s)
Disruptores Endocrinos , Exposición Materna/estadística & datos numéricos , Animales , Femenino , Humanos , Embarazo
17.
Cell Biochem Funct ; 38(1): 106-117, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31746004

RESUMEN

Preeclampsia is not fully understood; and few biomarkers, therapeutic targets, and therapeutic agents for its management have been identified. Original investigative findings suggest that abnormal placentation triggers preeclampsia and leads to hypertension, proteinuria, endothelial dysfunction, and inflammation, which are characteristics of the disease. Because of the regulatory roles that it plays in several metabolic processes, adiponectin has become a cytokine of interest in metabolic medicine. In this review, we have discussed the role of adiponectin in trophoblast proliferation, trophoblast differentiation, trophoblast invasion of the decidua, and decidual angiogenesis, which are the major phases of placentation. Also, we have highlighted the physiological profile of adiponectin in the course of normal pregnancy. Moreover, we have discussed the involvement of adiponectin in hypertension, endothelial dysfunction, inflammation, and proteinuria. Furthermore, we have summarized the reported relationship between the maternal serum adiponectin level and preeclampsia. The available evidence indicates that adiponectin level physiologically falls as pregnancy advances, regulates placentation, and exhibits protective effects against the symptoms of preeclampsia and that while hyperadiponectinemia is evident in normal-weight preeclamptic women, hypoadiponectinemia is evident in overweight and obese preeclamptic women. Therefore, the clinical use of adiponectin as a biomarker, therapeutic target, or therapeutic agent against the disease looks promising and should be considered.


Asunto(s)
Adiponectina/metabolismo , Placentación , Preeclampsia/metabolismo , Adiponectina/deficiencia , Femenino , Humanos , Errores Innatos del Metabolismo/metabolismo , Embarazo
18.
J Cell Physiol ; 234(4): 4154-4166, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30132880

RESUMEN

Embryo implantation is essential for normal pregnancy, and the process of decidualization is critical for embryo implantation. However, the mechanism of decidualization during early pregnancy is still unknown. Forkhead box O3a (FOXO3a) is the most important functional transcription factor of the forkhead box family and is a highly conserved transcription factor of apoptosis-related genes. In the mouse uterus, FOXO3a was found to be expressed regularly from Days 1-7 of early pregnancy. Upon further exploration, it was found that FOXO3a was expressed at significantly higher levels at the implantation site than at the interimplantation site on Days 5-7 of pregnancy. Under artificial decidualization, FOXO3a was highly expressed in the first and second decidual zones. After decidualization, the expression of FOXO3a was significantly increased both in vivo and vitro. In primary stromal cells, apoptosis was reduced by decreased expression of FOXO3a after inducing decidualization. Moreover, when FOXO3a-small interfering RNA was transfected into the uteri of mice, the expression of decidualization- and apoptosis-related factors was impaired. Thus, FOXO3a might play an important role in decidualization during early pregnancy, and cell apoptosis might be one of pathways for FOXO3a-regulated decidualization.


Asunto(s)
Apoptosis , Implantación del Embrión , Endometrio/metabolismo , Proteína Forkhead Box O3/metabolismo , Células del Estroma/metabolismo , Aborto Espontáneo/metabolismo , Aborto Espontáneo/patología , Animales , Femenino , Proteína Forkhead Box O3/genética , Humanos , Ratones , Embarazo , Transducción de Señal , Factores de Tiempo , Regulación hacia Arriba
19.
Reprod Biomed Online ; 34(2): 191-202, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27986413

RESUMEN

The molecular mechanisms underlying endometrial stromal cell proliferation and differentiation (decidualization) are still not fully understood. This study revealed that increased Slp-2 expression is a significant factor modulating endometrial stromal cell proliferation and decidualization in both mice and humans. Our results showed a significant difference in the mRNA and protein levels between the implantation site and inter-implantation site on day 5 and day 6 of pregnancy in mice (all P < 0.05). Strong Slp-2 immunostaining was mainly localized within the decidual zone of mice through the post-implantation period. Mice with artificially induced deciduoma showed significantly higher expression of Slp-2 compared with uninduced controls (P < 0.005). Human stromal cells in the middle and late-secretory phases demonstrated significantly (all P < 0.05) upregulated SLP-2, compared with cells in the proliferative phase and early secretory phases. Further analyses of the SLP-2 gene knocked down revealed a significant (P < 0.005) repression of both the decidualization marker gene's expression (decidual/trophoblast prolactin-related protein in mice, insulin-like growth factor binding protein and prolactin in human) and the cell proliferation in in vitro-induced decidualized primary endometrial stromal cells in mice and humans.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Endometrio/citología , Regulación del Desarrollo de la Expresión Génica , Proteínas de la Membrana/metabolismo , Células del Estroma/citología , Animales , Diferenciación Celular , Proliferación Celular , Decidua/metabolismo , Deciduoma/metabolismo , Implantación del Embrión , Femenino , Humanos , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Ciclo Menstrual , Ratones , Embarazo , Prolactina/análogos & derivados , Prolactina/metabolismo , ARN Mensajero/metabolismo , Células del Estroma/metabolismo
20.
Reprod Fertil Dev ; 29(8): 1509-1520, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27439778

RESUMEN

DNA cytosine-5 methylation plays a vital role in regulating the expression of E-cadherin, which is encoded by the CDH1 gene. In this study, we characterised the DNA methylation and expression pattern of CDH1 in an extravillous trophoblast cell line (HTR-8/SVneo) and two trophoblast cell lines -- JEG-3 and JAR. Promoter hypermethylation with reduced E-cadherin expression in HTR-8/SVneo cells and promoter hypomethylation with increased E-cadherin expression in JEG-3 and JAR cells were observed. Demethylation treatment significantly restored E-cadherin expression, contributing to decreases in the motility and invasiveness of HTR-8/SVneo cells. Sense-methylated oligonucleotides (MONs) labelled with Cy5 and complementary to a region of the human CDH1 promoter were designed, with the cytosines in 5'-cytosine-phosphate-guanine-3' (CpG) dinucleotides being replaced by methylated cytosines. Following MON transfection into JEG-3 cells, the level of CDH1 promoter DNA methylation as well as cell motility and invasiveness were increased and gene expression was significantly repressed. Our results indicate that MON-mediated DNA methylation of the CDH1 promoter and subsequent alterations in gene expression may contribute to trophoblast motility and invasion, suggesting a potential method for controlling the biological function of trophoblasts in vitro through epigenetic modification.


Asunto(s)
Cadherinas/genética , Movimiento Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Oligonucleótidos/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Trofoblastos/citología , Antígenos CD , Cadherinas/metabolismo , Línea Celular , Femenino , Humanos , Trofoblastos/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA