Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 323
Filtrar
1.
Biol Reprod ; 107(4): 945-955, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-35765194

RESUMEN

One mechanism by which the maternal environment regulates the early embryo is by secretion of cell-signaling molecules. One of these is dickkopf WNT signaling pathway inhibitor 1. Objectives were to (A) resolve discrepancies in the literature regarding effects of dickkopf WNT signaling pathway inhibitor 1 in the bovine embryo on development of trophectoderm and competence to establish pregnancy after embryo transfer and (B) determine whether there are long-term consequences of dickkopf WNT signaling pathway inhibitor 1 on placental function and postnatal phenotype. Embryos produced in vitro were cultured with vehicle or 100 ng/mL recombinant human dickkopf WNT signaling pathway inhibitor 1 from Days 5 to 7.5 of development (i.e., the morula and blastocyst stages of development). dickkopf WNT signaling pathway inhibitor 1 increased the number of cells positive for the trophectoderm marker CDX2 at Day 7.5 of development while having no effect on number of cells positive for the inner cell mass marker SOX2. There was no effect of dickkopf WNT signaling pathway inhibitor 1 on pregnancy or calving rate after transfer of blastocysts produced with Y-sorted semen to either lactating dairy cows or suckling beef cows. Treatment with dickkopf WNT signaling pathway inhibitor 1 at the morula-to-blastocyst stages programmed placental function, as measured by an effect of dickkopf WNT signaling pathway inhibitor 1 on plasma concentrations of pregnancy associated glycoproteins and placental lactogen at Day 160 of gestation (although not on other days examined). dickkopf WNT signaling pathway inhibitor 1 treatment also resulted in calves that were heavier at birth as compared to calves derived from control embryos. After birth, dickkopf WNT signaling pathway inhibitor 1 calves grew slower than controls. Results confirm that dickkopf WNT signaling pathway inhibitor 1 alters the developmental program of the bovine embryo to affect both prenatal and postnatal phenotypes.


Asunto(s)
Desarrollo Embrionario , Lactancia , Animales , Blastocisto/metabolismo , Bovinos , Desarrollo Embrionario/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Fenotipo , Placenta/metabolismo , Lactógeno Placentario/genética , Lactógeno Placentario/metabolismo , Lactógeno Placentario/farmacología , Embarazo
2.
Biomolecules ; 12(2)2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-35204716

RESUMEN

As patients recently diagnosed with T1D and patients with T2D have residual beta cell mass, there is considerable effort in beta cell biology to understand the mechanisms that drive beta cell regeneration as a potential cellular therapy for expanding patients' residual beta cell population. Both mouse and human studies have established that beta cell mass expansion occurs rapidly during pregnancy. To investigate the mechanisms of beta cell mass expansion during pregnancy, we developed a novel in vivo and in vitro models of pseudopregnancy. Our models demonstrate that pseudopregnancy promotes beta cell mass expansion in parous mice, and this expansion is driven by beta cell proliferation rather than hypertrophy. Importantly, estrogen, progesterone, and placental lactogen induce STAT5A signaling in the pseudopregnancy model, demonstrating that this model successfully recapitulates pregnancy-induced beta cell replication. We then created an in vitro model of pseudopregnancy and found that the combination of estrogen and placental lactogen induced beta cell replication in human islets and rat insulinoma cells. Therefore, beta cells both in vitro and in vivo increase proliferation when subjected to the pseudopregnancy cocktail compared to groups treated with estradiol or placental lactogen alone. The pseudopregnancy models described here may help inform novel methods of inducing beta cell replication in patients with diabetes.


Asunto(s)
Células Secretoras de Insulina , Islotes Pancreáticos , Animales , División Celular , Femenino , Humanos , Ratones , Placenta , Lactógeno Placentario/farmacología , Embarazo , Ratas
3.
Diabetes ; 69(7): 1463-1475, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32332156

RESUMEN

Diabetes occurs due to a loss of functional ß-cells, resulting from ß-cell death and dysfunction. Lactogens protect rodent and human ß-cells in vitro and in vivo against triggers of ß-cell cytotoxicity relevant to diabetes, many of which converge onto a common pathway of endoplasmic reticulum (ER) stress. However, whether lactogens modulate the ER stress pathway is unknown. This study examines whether lactogens can protect ß-cells against ER stress and mitigate diabetes incidence in Akita (Ak) mice, a rodent model of ER stress-induced diabetes, akin to neonatal diabetes in humans. We show that lactogens protect INS-1 cells, primary rodent and human ß-cells in vitro against two distinct ER stressors, tunicamycin and thapsigargin, through activation of the JAK2/STAT5 pathway. Lactogens mitigate expression of proapoptotic molecules in the ER stress pathway that are induced by chronic ER stress in INS-1 cells and rodent islets. Transgenic expression of placental lactogen in ß-cells of Ak mice drastically reduces the severe hyperglycemia, diabetes incidence, hypoinsulinemia, ß-cell death, and loss of ß-cell mass observed in Ak littermates. These are the first studies in any cell type demonstrating that lactogens modulate the ER stress pathway, causing enhanced ß-cell survival and reduced diabetes incidence in the face of chronic ER stress.


Asunto(s)
Diabetes Mellitus/prevención & control , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Lactógeno Placentario/farmacología , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Estrés del Retículo Endoplásmico/fisiología , Femenino , Glucosa/metabolismo , Humanos , Insulina/sangre , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/fisiología , Janus Quinasa 2/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Prolactina/farmacología , Factor de Transcripción STAT5/fisiología , Transducción de Señal/efectos de los fármacos
4.
Birth Defects Res B Dev Reprod Toxicol ; 107(4-5): 206-210, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27669115

RESUMEN

BACKGROUND: High fructose intake induces disruption of lipid metabolism via AMP-activated protein kinase (AMPK) signaling in the liver and peripheral tissues. Maternal lipid metabolism is physiologically altered by the activity of pregnancy hormones such as human placental lactogen (PL). To elucidate the influence of high fructose intake on hepatic lipid metabolism during pregnancy, we examined the effects of fructose on lipid metabolism via the AMPK pathway in hepatocytes stimulated with PL. METHODS: Human hepatoma cells (HepG2) were treated with D(-)-fructose in the presence or absence of PL. Intracellular lipid contents were measured. The total and phosphorylated protein content of AMPK and acetyl-CoA carboxylase (ACC) was quantified by Western blotting. RESULTS: The intracellular triacylglycerol level in fructose-treated HepG2 cells decreased significantly compared with that in untreated cells in the presence, but not absence, of PL. AMPK and ACC phosphorylation increased significantly and concentration-dependently in fructose-treated HepG2 cells in the presence of PL. CONCLUSION: Our results suggest that fructose treatment reduces triacylglycerol levels via AMPK/ACC signaling in PL-stimulated hepatocytes. These findings suggest that high fructose intake during pregnancy might impair lipid metabolism in the maternal liver.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Fructosa/toxicidad , Hepatocitos/efectos de los fármacos , Lactógeno Placentario/farmacología , Proteínas Quinasas Activadas por AMP/genética , Acetil-CoA Carboxilasa/genética , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Fosforilación , Transducción de Señal , Triglicéridos/metabolismo
5.
Horm Res Paediatr ; 84(5): 305-10, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26352728

RESUMEN

BACKGROUND/AIM: To evaluate the accuracy of the human chorionic gonadotropin (hCG) stimulation test in children with micropenis in predicting later Leydig cell function. METHODS: We conducted a retrospective investigation of testosterone response to a 3-day hCG test (3,000 IU/m2/day) in prepuberty to indicate the need for hormone replacement therapy (HRT) in adolescence. RESULTS: Fifty Japanese boys (range, 0.8-15.4 years of age; median, 8.9) with micropenis were enrolled. Thirty-four spontaneously developed puberty and preserved the ability of testosterone production (group 1), while 16 did not develop any pubertal signs without HRT (group 2). Serum testosterone levels after the hCG test (post-hCG T) in group 2 (range, <0.05-1.1 ng/ml; median, 0.24) were significantly lower than in group 1 (range, 0.5-8.7 ng/ml; median, 2.4; p < 0.0001). Based on true positives who required continuous HRT, the area under the receiver-operating characteristics curve for post-hCG T was 0.983 [95% confidence interval (CI), 0.90-1.00]. The post-hCG T cut-off level corresponding to the Youden index was 1.1 ng/ml (95% CI, 1.0-1.1), with a sensitivity of 100.0% (95% CI, 79.4-100.0) and a specificity of 94.1% (95% CI, 80.3-99.3). CONCLUSIONS: The hCG test in prepubertal children with micropenis can be useful for predicting Leydig cell function in pubertal or postpubertal adolescents. The post-hCG T cut-off level of 1.1 ng/ml is recommended to screen for those who will likely require HRT for pubertal development.


Asunto(s)
Enfermedades de los Genitales Masculinos/diagnóstico , Células Intersticiales del Testículo/efectos de los fármacos , Pene/anomalías , Lactógeno Placentario/farmacología , Adolescente , Pueblo Asiatico , Niño , Preescolar , Hormona Folículo Estimulante/sangre , Hormona Liberadora de Gonadotropina/sangre , Hormona Liberadora de Gonadotropina/farmacología , Terapia de Reemplazo de Hormonas , Humanos , Lactante , Masculino , Pene/anatomía & histología , Pene/crecimiento & desarrollo , Pubertad , Estudios Retrospectivos , Estimulación Química , Testosterona/sangre
6.
Acta Pharmacol Sin ; 36(5): 572-86, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25891087

RESUMEN

AIM: Prolactin family hormones include growth hormone, placental lactogen and prolactin, which are able to regulate angiogenesis via NO and prostaglandins. However, their effects on vascular tone are not fully understood. The aim of this study was to evaluate the effects of prolactin family hormones on rat vascular tone in vitro. METHODS: Aortic rings were prepared from adult male rats and precontracted with phenylephrine, then treated with the hormones and drugs. The tension was measured with isometric force displacement transducer connected to a polygraph. NO production and prostacyclin release in physiological solution was determined. Cultured rat aortic endothelial cells (RAECs) were treated with the hormones and drugs, and the phosphorylation of eNOS at serine 1177 was assessed using Western bolt analysis. RESULTS: Administration of growth hormone or placental lactogen (0.01-100 nmol/L) induced endothelium-dependent vasodilation. Both the hormones significantly increased the phosphorylation of eNOS in RAECs and NO level in physiological solution. Preincubation with L-NAME blocked growth hormone- or placental lactogen-induced vasodilation and NO production. Preincubation with an antibody against growth hormone receptors blocked growth hormone- and placental lactogen-induced vasodilation. Addition of a single dose of prolactin (0.01 nmol/L) induced sustained vessel relaxation, whereas multiple doses of prolactin induced a biphasic contraction-relaxation effect. The vascular effects of prolactin depended on endothelium. Prolactin significantly increased the level of prostacyclin I2 in physiological solution. Preincubation with indomethacin or an antibody against prolactin receptors blocked prolactin-induced vasodilation. CONCLUSION: The prolactin family hormones regulate rat vascular tone, selectively promoting either relaxation or contraction of vascular smooth muscle via activation of either growth hormone receptors or prolactin receptors within the endothelium.


Asunto(s)
Aorta/efectos de los fármacos , Epoprostenol/metabolismo , Hormona de Crecimiento Humana/farmacología , Óxido Nítrico/metabolismo , Lactógeno Placentario/farmacología , Prolactina/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Aorta/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Técnicas In Vitro , Masculino , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Ratas Wistar , Receptores de Somatotropina/efectos de los fármacos , Receptores de Somatotropina/metabolismo , Serina , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología
7.
PLoS One ; 10(3): e0121868, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25816302

RESUMEN

Pregnancy requires a higher functional beta cell mass and this is associated with profound changes in the gene expression profile of pancreatic islets. Taking Tph1 as a sensitive marker for pregnancy-related islet mRNA expression in female mice, we previously identified prolactin receptors and placental lactogen as key signalling molecules. Since beta cells from male mice also express prolactin receptors, the question arose whether male and female islets have the same phenotypic resilience at the mRNA level during pregnancy. We addressed this question in vitro, by stimulating cultured islets with placental lactogen and in vivo, by transplanting male or female islets into female acceptor mice. Additionally, the islet mRNA expression pattern of pregnant prolactin receptor deficient mice was compared with that of their pregnant wild-type littermates. When cultured with placental lactogen, or when transplanted in female recipients that became pregnant (day 12.5), male islets induced the 'islet pregnancy gene signature', which we defined as the 12 highest induced genes in non-transplanted female islets at day 12.5 of pregnancy. In addition, serotonin immunoreactivity and beta cell proliferation was also induced in these male transplanted islets at day 12.5 of pregnancy. In order to further investigate the importance of prolactin receptors in these mRNA changes we used a prolactin receptor deficient mouse model. For the 12 genes of the signature, which are highly induced in control pregnant mice, no significant induction of mRNA transcripts was found at day 9.5 of pregnancy. Together, our results support the key role of placental lactogen as a circulating factor that can trigger the pregnancy mRNA profile in both male and female beta cells.


Asunto(s)
Células Secretoras de Insulina/trasplante , Lactógeno Placentario/sangre , Embarazo/genética , ARN Mensajero/genética , Receptores de Prolactina/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Femenino , Perfilación de la Expresión Génica , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Ratones , Lactógeno Placentario/farmacología , Embarazo/sangre , Receptores de Prolactina/genética
8.
Drug Metab Dispos ; 42(4): 492-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24408518

RESUMEN

The state of pregnancy is known to alter hepatic drug metabolism. Hormones that rise during pregnancy are potentially responsible for the changes. Here we report the effects of prolactin (PRL), placental lactogen (PL), and growth hormone variant (GH-v) on expression of major hepatic cytochromes P450 expression and a potential molecular mechanism underlying CYP2E1 induction by PL. In female human hepatocytes, PRL and GH-v showed either no effect or small and variable effects on mRNA expression of CYP1A2, 2A6, 2B6, 2C9, 2C19, 2D6, 2E1, 3A4, and 3A5. On the other hand, PL increased expression level of CYP2E1 mRNA with corresponding increases in CYP2E1 protein and activity levels. Results from hepatocytes and HepaRG cells indicate that PL does not affect the expression or activity of HNF1α, the known transcriptional activator of basal CYP2E1 expression. Furthermore, transient transfection studies and Western blot results showed that STAT signaling, the previously known mediator of PL actions in certain tissues, does not play a role in CYP2E1 induction by PL. A chemical inhibitor of PI3-kinase signaling significantly repressed the CYP2E1 induction by PL in human hepatocytes, suggesting involvement of PI3-kinase pathway in CYP2E1 regulation by PL. CYP2E1-humanized mice did not exhibit enhanced CYP2E1 expression during pregnancy, potentially because of interspecies differences in PL physiology. Taken together, these results indicate that PL induces CYP2E1 expression via PI3-kinase pathway in human hepatocytes.


Asunto(s)
Citocromo P-450 CYP2E1/biosíntesis , Hepatocitos/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Lactógeno Placentario/fisiología , Adolescente , Adulto , Animales , Western Blotting , Citocromo P-450 CYP2E1/metabolismo , Inducción Enzimática , Femenino , Células Hep G2 , Hepatocitos/efectos de los fármacos , Hormona de Crecimiento Humana/farmacología , Hormona de Crecimiento Humana/fisiología , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Preparaciones Farmacéuticas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Lactógeno Placentario/farmacología , Embarazo/metabolismo , Cultivo Primario de Células , Prolactina/farmacología , Prolactina/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Adulto Joven
9.
Diabetologia ; 55(6): 1721-32, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22382519

RESUMEN

AIMS/HYPOTHESIS: A leading cause of type 2 diabetes is a reduction in functional beta cell mass partly due to increased beta cell death, triggered by stressors such as glucolipotoxicity (GLT). This study evaluates the hypothesis that lactogens can protect beta cells against GLT and examines the mechanism behind the pro-survival effect. METHODS: The effect of exogenous treatment or endogenous expression of lactogens on GLT-induced beta cell death was examined in INS-1 cells, and in rodent and human islets. The mechanism behind the pro-survival effect of lactogens was determined using an inhibitor, siRNAs, a dominant negative (DN) mutant, and Cre-lox-mediated gene deletion analysis. RESULTS: Lactogens significantly protect INS-1 and primary rodent beta cells against GLT-induced cell death. The pro-survival effect of lactogens in rodent beta cells is mediated through activation of the Janus kinase-2 (JAK2)/signal transducer and activator of transcription-5 (STAT5) signalling pathway. Lactogen-induced increase in the anti-apoptotic B cell lymphoma-extra large (BCLXL) protein is required to mediate its pro-survival effects in both INS-1 cells and primary rodent beta cells. Most importantly, lactogens significantly protect human beta cells against GLT-induced cell death, and their pro-survival effect is also mediated through the JAK2/STAT5 pathway. CONCLUSIONS/INTERPRETATION: These studies, together with previous work, clearly demonstrate the pro-survival nature of lactogens and identify the JAK2/STAT5 pathway as an important mediator of this effect in both rodent and human beta cells. Future studies will determine the effectiveness of this peptide in vivo in the pathophysiology of type 2 diabetes.


Asunto(s)
Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Janus Quinasa 2/metabolismo , Lactógeno Placentario/farmacología , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular Tumoral , Células Cultivadas , Femenino , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Janus Quinasa 2/genética , Masculino , Ratones , Ratones Transgénicos , ARN Interferente Pequeño , Ratas , Factor de Transcripción STAT5/genética , Transducción de Señal/genética
10.
Islets ; 3(5): 250-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21765243

RESUMEN

The search for factors either promoting islets proliferation or survival during adult life is a major issue for both type 1 and 2 diabetes mellitus. Among factors with mitogenic activity on pancreatic ß-cells, human placental lactogen (hPL) showed stronger activity when compared to the other lactogen hormones: growth hormone (GH) and prolactin (PRL). The aim of the present work is to elucidate the biological and molecular events of hPL isoform A (hPL-A) activity on human cultured islets. We used pure human pancreatic islets and insulinoma cell lines (ßTC-1 and RIN, murine and rat respectively) stimulated with hPL-A recombinant protein and we compared hPL-A activity with that of hGH. We showed that hPL-A inhibits apoptosis, both in insulinoma and human islets, by the phosphorylation of AKT protein. Indeed, the antiapoptotic role of hPL-A was mediated by PI3K, p38 and it was independent by PKA, Erk1/2. Compared with hGH, hPL-A modulated at different intervals and/or intensity by the phosphorylation of JAKs/STATs and MAPKinases. Moreover, hPL-A induced PDX-1 intracellular expression, improving beta cell activity and ameliorating insulin secretion in response to high glucose stimulation. Our data support the idea that hPL-A is involved in the regulation of beta cells activity. Importantly, we found that hPL-A can preserve and improve the ability of purified human pancreatic islets cultured to secrete insulin in vitro.


Asunto(s)
Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Lactógeno Placentario/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hormona de Crecimiento Humana/metabolismo , Hormona de Crecimiento Humana/farmacología , Humanos , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Insulinoma , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Neoplasias Pancreáticas , Lactógeno Placentario/farmacología , Prolactina/metabolismo , Prolactina/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Transgenic Res ; 20(4): 721-33, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20960057

RESUMEN

Human placental lactogen (hPL) is a peptidic hormone that belongs to the short list of growth factors that could treat type-1 diabetes through pancreatic islet transplantation. Placental lactogen has the capacity to improve islet survival and function before or after transplantation. In this study, transgenic tobacco plants were used as a novel expression system for the production of recombinant hPL protein (rhPL). The expression vector pNEKhPL2 containing hPL cDNA was introduced into tobacco plants; the transcriptional activity was confirmed by real-time PCR, and the rhPL levels reached 1% of the total soluble protein (TSP) content in plants cultivated in the greenhouse. In vitro bioassays using the rat insulinoma (INS-1) cell line showed that recombinant protein was able to induce cell proliferation and activate the JAK-2/STAT-5 signal transduction pathway, demonstrating that plant cells can produce the biologically active hPL protein. To further characterize the plant expression system for hPL production, we analyzed the stability of the protein during the life cycle of tobacco plants as well as the transmission of the transgenic trait to the progeny. The recombinant protein was stably accumulated in young leaves, reaching the maximum level in the first month (6.51 µg/g of fresh weight), but showing a decreasing trend of 26% from the initial sampling time until the end of plant's life cycle. The progeny of the selected pNEKhPL2 plant showed in vitro expression levels of up to 1.1% of TSP. Our results therefore indicate that transgenic plants are a suitable expression system for hPL production.


Asunto(s)
Células Secretoras de Insulina/efectos de los fármacos , Nicotiana/genética , Lactógeno Placentario/biosíntesis , Lactógeno Placentario/farmacología , Plantas Modificadas Genéticamente , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/cirugía , Retículo Endoplásmico/metabolismo , Expresión Génica , Vectores Genéticos , Humanos , Trasplante de Islotes Pancreáticos , Lactógeno Placentario/química , Estabilidad Proteica , Ratas , Proteínas Recombinantes/genética , Transducción de Señal
12.
Diabetologia ; 53(12): 2589-99, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20938637

RESUMEN

AIMS/HYPOTHESIS: Upregulation of the functional beta cell mass is required to match the physiological demands of mother and fetus during pregnancy. This increase is dependent on placental lactogens (PLs) and prolactin receptors, but the mechanisms underlying these events are only partially understood. We studied the mRNA expression profile of mouse islets during pregnancy to gain a better insight into these changes. METHODS: RNA expression was measured ex vivo via microarrays and quantitative RT-PCR. In vivo observations were extended by in vitro models in which ovine PL was added to cultured mouse islets and MIN6 cells. RESULTS: mRNA encoding both isoforms of the rate-limiting enzyme of serotonin biosynthesis, tryptophan hydroxylase (TPH), i.e. Tph1 and Tph2, were strongly induced (fold change 25- to 200-fold) during pregnancy. This induction was mimicked by exposing islets or MIN6 cells to ovine PLs for 24 h and was dependent on janus kinase 2 and signal transducer and activator of transcription 5. Parallel to Tph1 mRNA and protein induction, islet serotonin content increased to a peak level that was 200-fold higher than basal. Interestingly, only a subpopulation of the beta cells was serotonin-positive in vitro and in vivo. The stored serotonin pool in pregnant islets and PL-treated MIN6 cells was rapidly released (turnover once every 2 h). CONCLUSIONS/INTERPRETATION: A very strong lactogen-dependent upregulation of serotonin biosynthesis occurs in a subpopulation of mouse islet beta cells during pregnancy. Since the newly formed serotonin is rapidly released, this lactogen-induced beta cell function may serve local or endocrine tasks, the nature of which remains to be identified.


Asunto(s)
Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Lactógeno Placentario/farmacología , Embarazo/metabolismo , Serotonina/biosíntesis , Animales , Células Cultivadas , Embrión de Mamíferos , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Edad Gestacional , Células Secretoras de Insulina/clasificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Lactógeno Placentario/fisiología , Triptófano Hidroxilasa/genética , Triptófano Hidroxilasa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
13.
Biol Reprod ; 79(2): 376-86, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18448839

RESUMEN

Gastrin-releasing peptide (GRP) is abundantly expressed by endometrial glands of the ovine uterus and processed into different bioactive peptides, including GRP1-27, GRP18-27, and a C-terminus, that affect cell proliferation and migration. However, little information is available concerning the hormonal regulation of endometrial GRP and expression of GRP receptors in the ovine endometrium and conceptus. These studies determined the effects of pregnancy, progesterone (P4), interferon tau (IFNT), placental lactogen (CSH1), and growth hormone (GH) on expression of GRP in the endometrium and GRP receptors (GRPR, NMBR, BRS3) in the endometrium, conceptus, and placenta. In pregnant ewes, GRP mRNA and protein were first detected predominantly in endometrial glands after Day 10 and were abundant from Days 18 through 120 of gestation. Treatment with IFNT and progesterone but not CSH1 or GH stimulated GRP expression in the endometrial glands. Western blot analyses identified proGRP in uterine luminal fluid and allantoic fluid from Day 80 unilateral pregnant ewes but not in uterine luminal fluid of either cyclic or early pregnant ewes. GRPR mRNA was very low in the Day 18 conceptus and undetectable in the endometrium and placenta; NMBR and BRS3 mRNAs were undetectable in ovine uteroplacental tissues. Collectively, the present studies validate GRP as a novel IFNT-stimulated gene in the glands of the ovine uterus, revealed that IFNT induction of GRP is dependent on P4, and found that exposure of the ovine uterus to P4 for 20 days induces GRP expression in endometrial glands.


Asunto(s)
Péptido Liberador de Gastrina/genética , Regulación de la Expresión Génica/efectos de los fármacos , Interferón Tipo I/farmacología , Proteínas Gestacionales/farmacología , Progesterona/farmacología , Ovinos/genética , Útero/efectos de los fármacos , Líquido Amniótico/metabolismo , Animales , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Femenino , Feto/metabolismo , Péptido Liberador de Gastrina/metabolismo , Hormona del Crecimiento/farmacología , Masculino , Placenta/metabolismo , Lactógeno Placentario/farmacología , Embarazo , ARN Mensajero/metabolismo , Receptores de Bombesina/genética , Receptores de Bombesina/metabolismo , Ovinos/metabolismo , Ovinos/fisiología , Útero/metabolismo
14.
Pharm Res ; 25(2): 444-52, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17823853

RESUMEN

PURPOSE: We investigated whether the pregnancy-related hormones, estriol (E3), testosterone, human placental lactogen (hPL), human prolactin (hPRL), and human chorionic gonadotropin (hCG) affect BCRP expression in human placental BeWo cells. MATERIALS AND METHODS: The effects of these hormones on BCRP protein and mRNA expression in BeWo cells were determined by immunoblotting and quantitative real-time RT-PCR, respectively. The effects of these hormones on membrane localization of BCRP in BeWo cells were examined by immunofluorescent confocal microscopy. RESULTS: E3, hPL, and hPRL significantly increased BCRP protein and mRNA approximately two to threefold at physiological concentrations. Induction of BCRP by E3 was abrogated by the estrogen receptor (ER) antagonist ICI-182,780. However, knock-down of ER alpha by RNA interference did not abolish the inductive effect of E3. Testosterone by itself did not affect BCRP expression at physiological concentrations. However, testosterone together with 17beta-estradiol (E2) increased BCRP protein and mRNA approximately twofold, and this induction was abolished by ICI-182,780 or the testosterone receptor (TR) antagonist flutamide or knock-down of ER alpha expression. Further analysis revealed that E2 increased TR mRNA approximately 5.9-fold, suggesting that testosterone in combination with E2 increases BCRP expression, possibly through E2-mediated up-regulation of TR. hCG at physiological concentrations had no effect on BCRP expression. CONCLUSIONS: E3, hPL, hPRL, and testosterone in combination with E2 may up-regulate BCRP expression in the placenta during pregnancy.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hormonas/farmacología , Proteínas de Neoplasias/genética , Placenta/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Células Cultivadas , Gonadotropina Coriónica/farmacología , Estradiol/farmacología , Estriol/farmacología , Receptor alfa de Estrógeno/fisiología , Humanos , Lactógeno Placentario/farmacología , Prolactina/farmacología , ARN Mensajero/análisis , Testosterona/farmacología
15.
J Endocrinol ; 188(3): 559-68, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16522735

RESUMEN

Ovine placental lactogen (oPL) is produced by the conceptus trophectoderm and is secreted into both the maternal and fetal circulations. The present study was designed to examine in vivo the luteotropic effect of recombinant oPL (roPL), as determined by monitoring progesterone concentration and cycle length (experiment 1), and the antioxidative and antiapoptotic effects of roPL, as determined respectively by monitoring antioxidant enzymatic activity and apoptosis in the corpus luteum (CL) of cyclic ewes (experiment 2). We also studied whether roPL is capable of stimulating progesterone secretion in vitro by cultured luteal tissue of functionally active CL obtained from day-10 cyclic ewes (experiment 3) and day-60 pregnant ewes (experiment 4). Circulating concentrations of progesterone and cycle length were not affected by treatment of ewes with 80 microg/kg body weight per day of roPL (n = 4 ewes) for 10 days beginning on day 11 post-estrus, as compared with saline-treated ewes (n = 4 ewes). Luteolysis occurred between days 15 and 16 post-estrus in the four saline-treated ewes and in 3/4 roPL-treated ewes. The activities of the key antioxidant enzymes copper-zinc superoxide dismutase (Cu,Zn-SOD), manganese SOD (Mn-SOD), glutathione peroxidase (GPX), glutathione reductase (GSR) and glutathione S-transferase (GST) were unaffected by treatment of ewes with 80 microg/kg per day of roPL (n = 4 ewes) for 3 days, between days 11 and 14 post-estrus, as compared with saline-treated ewes (n = 4 ewes). In situ TUNEL method revealed that the number of apoptotic cells was not different between the two groups of ewes. There was no significant change in progesterone secretion by explants from day-10 estrous cycle (n = 3 ewes) or day-60 pregnancy (n = 3 ewes) CL cultured with different concentrations (10, 100 and 1000 ng/ml) of roPL, whereas treatment with oLH at the concentration of 100 or 1000 ng/ml caused a significant increase in progesterone secretion by explants from day-10 estrous cycle CL (P < 0.05) and by explants from day-60 pregnancy CL (P < 0.01). In conclusion, our results demonstrate that oPL has no luteotropic and/or luteoprotective actions in sheep, either in vivo or in vitro.


Asunto(s)
Cuerpo Lúteo/efectos de los fármacos , Infertilidad/metabolismo , Lactógeno Placentario/farmacología , Ovinos/fisiología , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Cuerpo Lúteo/metabolismo , Ciclo Estral , Femenino , Etiquetado Corte-Fin in Situ , Embarazo , Progesterona/sangre , Progesterona/metabolismo , Proteínas Recombinantes/farmacología , Técnicas de Cultivo de Tejidos
16.
J Vasc Res ; 43(2): 205-13, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16410683

RESUMEN

In 22 pigs anesthetized with sodium pentobarbitone, changes in blood flow caused by infusion of human placental lactogen into the left renal, external iliac, and anterior descending coronary arteries were assessed using electromagnetic flowmeters. In 17 pigs, infusion of human placental lactogen whilst keeping the heart rate and arterial pressure constant decreased coronary, renal and iliac flow. In 5 additional pigs, increasing the dose of human placental lactogen produced a dose-related decrease in regional blood flow. The mechanisms of the above response were studied in 15 of the 17 pigs by repeating the experiment of infusion. The human placental lactogen-induced decrease in regional blood flow was not affected by blockade of cholinergic receptors (5 pigs) or of alpha-adrenergic receptors (5 pigs), but it was abolished by blockade of beta2-adrenergic receptors (5 pigs). The present study showed that intra-arterial infusion of human placental lactogen primarily decreased coronary, renal and iliac blood flow. The mechanism of this response was shown to be due to the inhibition of a vasodilatory beta2-adrenergic receptor-mediated effect.


Asunto(s)
Anestesia/veterinaria , Lactógeno Placentario/farmacología , Flujo Sanguíneo Regional/efectos de los fármacos , Antagonistas Adrenérgicos alfa/farmacología , Antagonistas Adrenérgicos beta/farmacología , Animales , Atropina/farmacología , Butoxamina/farmacología , Antagonistas Colinérgicos/farmacología , Circulación Coronaria/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Humanos , Arteria Ilíaca/efectos de los fármacos , Riñón/irrigación sanguínea , Pentobarbital , Fentolamina/farmacología , Porcinos
17.
J Dairy Res ; 72 Spec No: 14-9, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16180716

RESUMEN

Fluorescence resonance energy transfer (FRET) microscopy was used to study interactions between proteins in intact cells. We showed that growth hormone (GH) causes transient homodimerization of GH receptors tagged with yellow or cyan fluorescent proteins. The peak of FRET signaling occurred 2 to 4 min after hormonal stimulation and was followed by a decrease in FRET signal. Repeating those experiments in cells pretreated with the inhibitor of internalization methyl-beta-cyclodextrin, or in potassium-depleted cells showed no difference in the kinetics of FRET signaling as compared with the non-treated cells, indicating that the decrease in FRET signal does not result from receptor internalization by the pathways inhibited by methyl-beta-cyclodextrin or potassium depleted but might occur by other pathways of internalization. Using a similar methodology, we also demonstrated that ovine placental lactogen (oPL) causes transient heterodimerization of GH and prolactin (PRL) receptors 2.5 to 3 min after oPL application. On the other hand, oGH or oPRL had no effect at all, further substantiating the finding the oPL, which lacks a specific receptor, acts in homologous systems by heterodimerization of GH and PRL receptors. We also demonstrated that both PRL and leptin (LEP) are capable of transactivation of the oncogenic receptors erbB2 and erbB3. Upon PRL or LEP stimulation of HEK-293T cells transfected with LEP or PRL receptors and erbB2 or erbB3, erbB proteins are first phosphorylated and then activate MAPK (erk1/erk2). However, the FRET experiments failed to document any evidence of a direct interaction between erbB2 and the PRL or LEP receptors, suggesting that erbB activation probably occurs via activated JAK2, translocated from the respective receptors to erbB2.


Asunto(s)
Células/metabolismo , Citocinas/farmacología , Transferencia Resonante de Energía de Fluorescencia , Microscopía Fluorescente/métodos , Animales , Células/efectos de los fármacos , Dimerización , Hormona del Crecimiento/farmacología , Humanos , Leptina/farmacología , Lactógeno Placentario/farmacología , Prolactina/farmacología , Receptor ErbB-2/metabolismo , Receptores de Prolactina/química , Receptores de Somatotropina/química , Receptores de Somatotropina/fisiología , Ovinos , Activación Transcripcional
18.
Gynecol Endocrinol ; 21(1): 27-32, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16048798

RESUMEN

The placenta is an important source of leptin production that contributes to the state of hyperleptinemia observed in pregnant women. Moreover, the synthesis of leptin and its receptors by syncytiotrophoblast cells suggests a potential paracrine or autocrine action of leptin in the placenta. In the present study we examined the effect of gestational hormones, human chorionic gonadotropin (hCG), human placental lactogen (hPL), progesterone and estradiol, on in vitro leptin release by human term trophoblast cells in culture. Placentas at term were obtained immediately after delivery from mothers with uncomplicated pregnancies. Leptin levels were measured by enzyme-linked immunosorbent assay in culture media of trophoblasts maintained in monolayer culture for 24, 48 and 72 h with different hormonal treatments or placebo. Treatment with hPL and progesterone led to a time- and dose-dependent decrease in leptin release that was statistically significant after 24 h, with a maximal effect after 72 h of incubation. In contrast, incubation with estradiol and hCG did not have exhibit any effect on leptin secretion at any of the doses and times assayed in this work. The results obtained in this study support that leptin can be considered a gestational hormone implied in the endocrine function of the placenta and that its secretion is at least partially regulated by steroid and peptidic reproductive hormones in trophoblast cells in vitro.


Asunto(s)
Leptina/metabolismo , Placenta/efectos de los fármacos , Lactógeno Placentario/farmacología , Progesterona/farmacología , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Células Cultivadas , Gonadotropina Coriónica/farmacología , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Estradiol/farmacología , Femenino , Humanos , Leptina/análisis , Embarazo , Factores de Tiempo
19.
Gene ; 313: 83-9, 2003 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-12957379

RESUMEN

A macroarray approach used to list genes differentially expressed in goat mammary gland (gestation vs. lactation), other than milk protein genes, allowed us to detect the Glycosylation-dependent Cell Adhesion Molecule 1 (GLYCAM1) gene. GLYCAM1, a member of the glycoprotein mucin family, is a component of the milk fat globule membrane (MFGM). Its complete cDNA and gene sequences were determined and it was mapped by fluorescent in situ hybridization (FISH) on goat and cattle chromosome 5 (CHI5q21 and BTA5q21), and on sheep chromosome 3 (OAR3q21). Northern blot analyses confirmed its differential expression during the development and differentiation of the mammary gland of ruminants with a significantly higher mRNA amount during lactation than during pregnancy. An experimental in vivo induction model for lactation, developed by Kann et al., showed that the expression of GLYCAM1 is hormonally regulated in the mammary gland of ewes. Interspecies comparison of the gene promoter revealed the evolutionary conservation of a short proximal nucleotide sequence encompassing several transcription factor binding sites that could mediate the above-mentioned hormonal regulation.


Asunto(s)
Cabras/genética , Mucinas/genética , Animales , Secuencia de Bases , Sitios de Unión/genética , Mapeo Cromosómico , Secuencia Conservada/genética , ADN/química , ADN/genética , ADN Complementario/química , ADN Complementario/genética , ADN Complementario/aislamiento & purificación , Evolución Molecular , Exones , Femenino , Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genes/genética , Hibridación Fluorescente in Situ , Intrones , Lactancia/genética , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Datos de Secuencia Molecular , Lactógeno Placentario/farmacología , Embarazo , Regiones Promotoras Genéticas/genética , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Ácido Nucleico , Especificidad de la Especie , Factores de Transcripción/metabolismo
20.
Arch Biochem Biophys ; 416(2): 249-56, 2003 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12893303

RESUMEN

Nitric oxide (NO) is an important modulator involved in immune regulation. Here, we describe conditions under which NO-donors induce apoptosis on Nb2 lymphoma cells, as evidenced by decreased cell viability and increased hypodiploid DNA content determined by flow cytometry. In addition, DNA fragmentation typical of apoptosis was shown by agarose gel electrophoresis. This apoptosis was accompanied by a significant increase of caspase-3-like enzymatic activity. Both ovine prolactin (oPRL) and ovine placental lactogen (oPL) exerted a protective effect on the NO-donor-induced apoptosis. Furthermore, dexamethasone (Dex)-induced cell death was also associated with caspase-3-like activity and oPL had the same potency as oPRL in its protective effect on Dex-induced apoptosis of Nb2 cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Linfoma/fisiopatología , Óxido Nítrico/metabolismo , Lactógeno Placentario/farmacología , Prolactina/farmacología , Animales , Caspasa 3 , Caspasas/biosíntesis , Bovinos , Supervivencia Celular/efectos de los fármacos , Fragmentación del ADN/efectos de los fármacos , Dexametasona/farmacología , Diploidia , Linfoma/enzimología , Linfoma/genética , Linfoma/metabolismo , Sustancias Protectoras/farmacología , Ratas , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...