Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Placenta ; 134: 39-47, 2023 03 24.
Article in English | MEDLINE | ID: mdl-36870301

ABSTRACT

The placenta and the extraembryonic tissues represent a valuable source of cells for regenerative medicine. In particular, the amniotic membrane possesses cells with stem cells characteristics that have attracted research attention. Human amniotic epithelial cells (hAECs) have unique and desirable features that position them over other stem cells, not only because of the unlimited potential supplied of, the easy access to placental tissues, and the minimal ethical and legal barriers associated, but also due to the embryonic stem cells markers expression and their ability to differentiate into the three germ layers. In addition, they are non-tumorigenic and have immunomodulatory and anti-inflammatory properties. Hepatic failure is one of the major causes of morbidity and mortality worldwide. Organ transplantation is the best way to treat acute and chronic liver failure, but there are several associated obstacles. Stem cells have been highlighted as alternative hepatocytes source because of their potential for hepatogenic differentiation. HAECs, in particular, have some properties that make them suitable for hepatocyte differentiation. In this work, we review the general characteristics of the epithelial stem cells isolated from human amniotic membrane as well as their ability to differentiate to hepatic cells. We also revise their regenerative properties, with the focus on their potential application in the liver disease treatment.


Subject(s)
Epithelial Cells , Liver Diseases , Humans , Female , Pregnancy , Placenta , Liver Diseases/therapy , Cell Differentiation , Embryonic Stem Cells
2.
Stem Cell Res Ther ; 12(1): 155, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33648582

ABSTRACT

A new coronavirus respiratory disease (COVID-19) caused by the SARS-CoV-2 virus, surprised the entire world, producing social, economic, and health problems. The COVID-19 triggers a lung infection with a multiple proinflammatory cytokine storm in severe patients. Without effective and safe treatments, COVID-19 has killed thousands of people, becoming a pandemic. Stem cells have been suggested as a therapy for lung-related diseases. In particular, mesenchymal stem cells (MSCs) have been successfully tested in some clinical trials in patients with COVID-19. The encouraging results positioned MSCs as a possible cell therapy for COVID-19. The amniotic membrane from the human placenta at term is a valuable stem cell source, including human amniotic epithelial cells (hAECs) and human mesenchymal stromal cells (hAMSCs). Interestingly, amnion cells have immunoregulatory, regenerative, and anti-inflammatory properties. Moreover, hAECs and hAMSCs have been used both in preclinical studies and in clinical trials against respiratory diseases. They have reduced the inflammatory response and restored the pulmonary tissue architecture in lung injury in vivo models. Here, we review the existing data about the stem cells use for COVID-19 treatment, including the ongoing clinical trials. We also consider the non-cellular therapies that are being applied. Finally, we discuss the human amniotic membrane cells use in patients who suffer from immune/inflammatory lung diseases and hypothesize their possible use as a successful treatment against COVID-19.


Subject(s)
Amnion/cytology , COVID-19/therapy , Mesenchymal Stem Cell Transplantation , Stem Cells/cytology , Clinical Trials as Topic , Female , Humans , Inflammation , Mesenchymal Stem Cells/cytology , Placenta/cytology , Pregnancy , Risk
3.
Reproduction ; 160(4): 591-602, 2020 10.
Article in English | MEDLINE | ID: mdl-32698160

ABSTRACT

Pregnancy success requires a proper fetal maternal interaction at the establishment of implantation. Leptin has been described as a multitasking cytokine in pregnancy, particularly in the placenta, where it acts as an autocrine hormone. The expression of leptin in normal trophoblastic cells is regulated by different endogenous signals. We have previously reported that 17ß-estradiol upregulates placental leptin expression through genomic and non-genomic mechanisms. To improve the knowledge of estrogen receptor mechanisms in regulating leptin gene expression, we examined transcription nuclear factor kappa B (NFκB) effect on estradiol leptin induction in human BeWo cell line and human term placental explants. We demonstrated that estradiol induction effect on leptin expression is blocked by the inhibition of NFκB signaling. We also found that the overexpression of p65 subunit, the active form of NFκB, induces leptin expression. Moreover, downregulation of estrogen receptor alpha (ERα), through a specific siRNA, abolished NFκB effect on leptin expression. We also demonstrated that ERα enhanced NFκB signaling pathway activation in trophoblastic cells. Estradiol treatment significantly increased p65 expression and phosphorylation of the inhibitory protein κB alpha (IκBα). A reporter plasmid containing NFκB elements was also induced in response to estradiol stimulation. Localization experiments revealed that estradiol treatment induced nuclear localization of overexpressed p65. Moreover, the overexpression of ERα produced a complete displacement of p65 protein to the nucleus. Finally, immunoprecipitation experiments showed the presence of a complex containing ERα and NFκB. All these evidences suggest a cooperative behavior between ERα and NFκB transcription factors to induce leptin transcription.


Subject(s)
Choriocarcinoma/pathology , Estrogens/pharmacology , Leptin/metabolism , NF-kappa B/metabolism , Placenta/metabolism , Uterine Neoplasms/pathology , Cell Nucleus , Choriocarcinoma/genetics , Choriocarcinoma/metabolism , Female , Humans , Leptin/genetics , NF-kappa B/genetics , Phosphorylation , Placenta/drug effects , Pregnancy , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism
4.
Vitam Horm ; 112: 311-326, 2020.
Article in English | MEDLINE | ID: mdl-32061347

ABSTRACT

Water is the major component of cells and tissues. The fetal body consists of about 70-90% water and its fluid balance is dependent on the mother. In fact, abortion, premature birth, amniotic fluid volume abnormality, malformation and fetal growth restrictions might result when the homeostasis of the maternal-fetal fluid exchange is disrupted. Thus, maternal-fetal fluid balance is critical during pregnancy. In this sense, several mechanisms, including aquaporins (AQPs) have been reported to play important roles in maternal-fetal fluid balance. AQPs are small membrane proteins (about 30kDa), present in different organs, that increase the permeability of water, as well as other small uncharged molecules to be transported across the bilayer cell membranes. Several aquaporins are expressed in placenta, and aquaporins play key roles in the placental function. Even though aquaporins have a proven crucial role in water homeostasis, the physiological and pathological importance of aquaporins as glycerol channels is not fully understood. This review focuses on advances in our knowledge of the roles of aquaporins in placental cells, particularly the roles of AQP3 and AQP9 in placental metabolism and points to the pathophysiological importance of glycerol channels in placenta, as well as the signal transduction pathways activated by them. Moreover, the regulation of aquaporins expression by different placental hormones, such as leptin and the mechanisms involved will be discussed.


Subject(s)
Aquaporins , Placenta , Water-Electrolyte Balance , Animals , Aquaporins/metabolism , Female , Humans , Placenta/physiology , Pregnancy , Water-Electrolyte Balance/physiology
5.
Sci Rep ; 9(1): 14193, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31578445

ABSTRACT

The placental stem cells have called the focus of attention for their therapeutic potential to treat different diseases, including cancer. There is plenty evidence about the antiproliferative, antiangiogenic and proapoptotic properties of the amniotic membrane. Liver cancer is the fifth cause of cancer in the world, with a poor prognosis and survival. Alternative treatments to radio- or chemotherapy have been searched. In this work we aimed to study the antiproliferative properties of the human amniotic membrane conditioned medium (AM-CM) in hepatocarcinoma cells. In addition, we have analyzed the regulation of pro and antiOncomiRs expression involved in hepatocarcinoma physiology. We have determined by 3H-thymidine incorporation assay that AM-CM inhibits DNA synthesis in HepG2 cells after 72 h of treatment. AM-CM pure or diluted at 50% and 25% also diminished HepG2 and HuH-7 cells viability and cell number. Furthermore, AM-CM induced cell cycle arrest in G2/M. When proliferation mechanisms were analyzed we found that AM-CM reduced the expression of both Cyclin D1 mRNA and protein. Nuclear expression of Ki-67 was also reduced. We observed that this CM was able to promote the expression of p53 and p21 mRNA and proteins, leading to cell growth arrest. Moreover, AM-CM induced an increase in nuclear p21 localization, observed by immunofluorescence. As p53 levels were increased, Mdm-2 expression was downregulated. Interestingly, HepG2 and HuH-7 cells treatment with AM-CM during 24 and 72 h produced an upregulation of antiOncomiRs 15a and 210, and a downregulation of proOncomiRs 206 and 145. We provide new evidence about the promising novel applications of human amniotic membrane in liver cancer.


Subject(s)
Amnion/metabolism , Carcinoma, Hepatocellular/drug therapy , Culture Media, Conditioned/pharmacology , Liver Neoplasms/drug therapy , Amnion/growth & development , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Culture Media, Conditioned/metabolism , Cyclin D1/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , MicroRNAs/genetics , Placenta/metabolism , Pregnancy , Proto-Oncogene Proteins c-mdm2/genetics , Stem Cells/metabolism , Tumor Suppressor Protein p53/genetics
6.
Cell Tissue Res ; 375(3): 733-742, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30338379

ABSTRACT

Development of the human placenta is critical for a successful pregnancy. The placenta allows the exchange of oxygen and carbon dioxide and is crucial to manage acid-base balance within a narrow pH. It is known that low pH levels are a risk of apoptosis in several tissues. However, there has been little discussion about the effect of acidic stress in the placenta. Leptin is produced by the placenta with a trophic autocrine effect. Previous results of our group have demonstrated that leptin prevents apoptosis of trophoblast cells under different stress conditions such as serum deprivation and hyperthermia. The purpose of the present work is to evaluate acidic stress consequences in trophoblast explant survival and to determine leptin action in these conditions. For this objective, term human trophoblast explants were cultured at physiological pH (pH 7.4) and at acidic pH (pH 6.8) in the presence or absence of leptin. Western blot assays were performed to study the abundance of active caspase-3 and the p89 fragment of PARP-1. Pro-apoptotic and pro-survival members of Bcl-2 family, as Bax, t-Bid, and Bcl-2, were studied. Moreover, p53 pathway was also evaluated including Mdm-2, the main p53 regulator. Active caspase-3 and cleaved PARP-1 abundances were increased at low extracellular pH. Moreover, t-Bid levels were also augmented as well as p53 expression and phosphorylation on S46. Leptin treatment prevents the consequences of acidosis, decreasing p53 expression and increasing Mdm-2 expression. In summary, this work demonstrated for first time that low pH induces apoptosis of human trophoblast explants involving apoptotic intrinsic pathway, and leptin impairs this effect.


Subject(s)
Acids/toxicity , Apoptosis/drug effects , Cytoprotection/drug effects , Leptin/pharmacology , Placenta/cytology , Stress, Physiological/drug effects , Adult , BH3 Interacting Domain Death Agonist Protein/metabolism , Female , Humans , Hydrogen-Ion Concentration , Models, Biological , Phosphorylation/drug effects , Pregnancy , Proto-Oncogene Proteins c-mdm2/metabolism , Trophoblasts/cytology , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
7.
PLoS One ; 13(1): e0191489, 2018.
Article in English | MEDLINE | ID: mdl-29346426

ABSTRACT

Stem cells derived from placental tissues are an attractive source of cells for regenerative medicine. Amniotic epithelial cells isolated from human amnion (hAECs) have desirable and competitive characteristics that make them stand out between other stem cells. They have the ability to differentiate toward all three germ layers, they are not tumorigenic and they have immunosuppressive properties. Although liver transplantation is the best way to treat acute and chronic hepatic failure patients, there are several obstacles. Recently, stem cells have been spotlighted as alternative source of hepatocytes because of their potential for hepatogenic differentiation. In this work, we aimed to study the proliferation and survival of the hAECs during their hepatic differentiation. We have also analyzed the changes in pluripotency and hepatic markers. We differentiated amniotic cells applying a specific hepatic differentiation (HD) protocol. We determined by qRT-PCR that hAECs express significant levels of SOX-2, OCT-4 and NANOG during at least 15 days in culture and these pluripotent markers diminish during HD. SSEA-4 expression was reduced during HD, measured by immunofluorescence. Morphological characteristics became more similar to hepatic ones in differentiated cells and representative hepatic markers significantly augmented their expression, measured by qRT-PCR and Western blot. Cells achieved a differentiation efficiency of 75%. We observed that HD induced proliferation and promoted survival of hAECs, during 30 days in culture, evaluated by 3H-thymidine incorporation and MTT assay. HD also promoted changes in hAECs cell cycle. Cyclin D1 expression increased, while p21 and p53 levels were reduced. Immunofluorescence analysis showed that Ki-67 expression was upregulated during HD. Finally, ERK 1/2 phosphorylation, which is intimately linked to proliferation and cell survival, augmented during all HD process and the inhibition of this signaling pathway affected not only proliferation but also differentiation. Our results suggest that HD promotes proliferation and survival of hAECs, providing important evidence about the mechanisms governing their hepatic differentiation. We bring new knowledge concerning some of the optimal transplantation conditions for these hepatic like cells.


Subject(s)
Amnion/cytology , Cell Proliferation , Cell Survival , Liver/cytology , Biomarkers/metabolism , Cells, Cultured , Epithelial Cells/cytology , Female , Humans , Liver/metabolism , MAP Kinase Signaling System , Phosphorylation , Pregnancy , Real-Time Polymerase Chain Reaction
8.
J Cell Mol Med ; 22(2): 716-727, 2018 02.
Article in English | MEDLINE | ID: mdl-29160594

ABSTRACT

Leptin is now considered an important signalling molecule of the reproductive system, as it regulates the production of gonadotrophins, the blastocyst formation and implantation, the normal placentation, as well as the foeto-placental communication. Leptin is a peptide hormone secreted mainly by adipose tissue, and the placenta is the second leptin-producing tissue in humans. Placental leptin is an important cytokine which regulates placental functions in an autocrine or paracrine manner. Leptin seems to play a crucial role during the first stages of pregnancy as it modulates critical processes such as proliferation, protein synthesis, invasion and apoptosis in placental cells. Furthermore, deregulation of leptin levels has been correlated with the pathogenesis of various disorders associated with reproduction and gestation, including polycystic ovary syndrome, recurrent miscarriage, gestational diabetes mellitus, pre-eclampsia and intrauterine growth restriction. Due to the relevant incidence of the mentioned diseases and the importance of leptin, we decided to review the latest information available about leptin action in normal and pathological pregnancies to support the idea of leptin as an important factor and/or predictor of diverse disorders associated with reproduction and pregnancy.


Subject(s)
Leptin/metabolism , Pregnancy Complications/metabolism , Adipose Tissue/metabolism , Female , Humans , Immunologic Factors/metabolism , Placenta/embryology , Placenta/metabolism , Pregnancy , Reproduction
9.
Gynecol Endocrinol ; 34(2): 175-177, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28942694

ABSTRACT

Aquaporins are integral membrane proteins that have permeability functions in many tissues. Aquaporin 9 may transport not only water but also small molecules, such as glycerol, monocarboxylates, purines and pyrimidines. Aquaporin 9 is expressed in syncytiotrophoblast of human term placenta, and it may contribute to the embryonic/fetal growth and survival. We have previously found that Aquaporin 9 expression levels seem to be increased in placenta from gestational diabetes. Since leptin plasma levels and leptin expression are increased in placenta from gestational diabetes, we aimed to study the possible role of leptin on Aquaporin 9 expression in human placenta in vitro. The present work shows that leptin produces a dose-dependent increase of Aquaporin 9 expression, resulting in an increase in Aquaporin-9 protein in human trophoblast explants.


Subject(s)
Aquaporins/metabolism , Gene Expression Regulation, Developmental , Leptin/metabolism , Placenta/metabolism , Up-Regulation , Adult , Aquaporins/genetics , Cesarean Section , Female , Glycosylation , Humans , Immunoblotting , Osmolar Concentration , Placenta/cytology , Pregnancy , Protein Processing, Post-Translational , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Term Birth/metabolism , Tissue Culture Techniques , Trophoblasts/cytology , Trophoblasts/metabolism
10.
Reproduction ; 155(1): R1-R12, 2018 01.
Article in English | MEDLINE | ID: mdl-29018059

ABSTRACT

Leptin is a homeostatic regulator in the placenta where it promotes proliferation, protein synthesis and the expression of tolerogenic maternal response molecules such as HLA-G. Leptin also exerts an anti-apoptotic action in placenta controlling the expression of p53 master cell cycle regulator under different stress conditions. On the other hand, leptin is an integrative target of different placental stimuli. The expression of leptin in placenta is regulated by hCG, insulin, steroids, hypoxia and many other growth hormones, suggesting that it might have an important endocrine function in the trophoblastic cells. The leptin expression is induced involving the cAMP/PKA or cAMP/Epac pathways which have profound actions upon human trophoblast function. The activation of PI3K and MAPK pathways also participates in the leptin expression. Estrogens play a central role during pregnancy, particularly 17ß-estradiol upregulates the leptin expression in placental cells through genomic and non-genomic actions. The leptin promoter analysis reveals specific elements that are active in placental cells. The transcription factors CREB, AP1, Sp1, NFκB and the coactivator CBP are involved in the placental leptin expression. Moreover, placental leptin promoter is a target of epigenetic marks such as DNA methylation and histone acetylation that regulates not only the leptin expression in placenta during pregnancy but also determines the predisposition of acquiring adult metabolism diseases. Taken together, all these results allow a better understanding of leptin function and regulatory mechanisms of leptin expression in human placental trophoblasts, and support the importance of leptin during pregnancy and in programming adult health.


Subject(s)
Leptin/metabolism , Placenta/physiology , Animals , Female , Humans , Placenta/cytology , Pregnancy , Signal Transduction
11.
Placenta ; 57: 152-162, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28864005

ABSTRACT

INTRODUCTION: Pleiotropic effects of leptin have been identified in reproduction and pregnancy, particularly in the placenta, where it functions as an autocrine hormone. The synthesis of leptin in normal trophoblastic cells is regulated by different endogenous biochemical agents, but the regulation of placental leptin expression is still poorly understood. We have previously reported that 17ß-estradiol up-regulates placental leptin expression through genomic and nongenomic mechanisms. METHODS: To improve the understanding of estrogen receptor mechanisms in regulating leptin gene expression, we examined Sp1 transcription factor effect on estradiol leptin induction in human BeWo cell line. RESULTS: We demonstrated that Sp1 induces leptin expression determined by qRT-PCR, Western blot and transient transfection experiments. We also found that estradiol induction effect on leptin expression is enhanced by the over expression of Sp1 factor. Moreover, estradiol effect was not evidenced when Sp1 binding site on leptin promoter is mutated, suggesting that estradiol action is dependent on Sp1. On the other hand we showed data that demonstrate that Sp1 induction of leptin expression is insensitive to the antiestrogen ICI 182 780. By over expression experiments, we have also found that Sp1 effect on leptin expression could be mediated by estrogen receptor alpha. Supporting this idea, the downregulation of estrogen receptor alpha level through a specific siRNA, abolished Sp1 effect on leptin expression. DISCUSSION: Taken together all these evidences suggest a cooperative behavior between estrogen receptor alpha and Sp1 transcription factors to induce leptin transcription.


Subject(s)
Estradiol/physiology , Estrogen Receptor alpha/metabolism , Leptin/metabolism , Placenta/metabolism , Sp1 Transcription Factor/metabolism , Cell Line, Tumor , Estradiol/analogs & derivatives , Female , Fulvestrant , Humans , Pregnancy
12.
Placenta ; 42: 106-13, 2016 06.
Article in English | MEDLINE | ID: mdl-27238720

ABSTRACT

Maternal fever is common during pregnancy and has for many years been suspected to harm the developing fetus. Whether increased maternal temperature produces exaggerated apoptosis in trophoblast cells remains unclear. Since p53 is a critical regulator of apoptosis we hypothesized that increased temperature in placenta produces abnormal expression of proteins in the p53 pathway and finally caspase-3 activation. Moreover, leptin, produced by placenta, is known to promote the proliferation and survival of trophoblastic cells. Thus, we aimed to study the possible role of leptin preventing apoptosis triggered by high temperature, as well as the molecular mechanisms underlying this effect. Fresh placental tissue was collected from normal pregnancies. Explants of placental villi were exposed to 37 °C, 40 °C and 42 °C during 3 h in the presence or absence of 10 nM leptin in DMEM-F12 medium. Western blotting and qRT-PCR was performed to analyze the expression of p53 and downstream effector, P53AIP1, Mdm2, p21, BAX and BCL-2 as well as the activated cleaved form of caspase-3 and the fragment of cytokeratin-18 (CK-18) cleaved at Asp396 (neoepitope M30). Phosphorylation of the Ser 46 residue on p53, the expression of P53AIP1, Mdm2, p21, as well as caspase-3 and CK-18 were significantly increased in explants at 40 °C and 42 °C. Conversely, these effects were significantly attenuated by leptin 10 nM at both 40 °C and 42 °C. The BCL2/BAX ratio was also significantly decreased in explants at 40 °C and 42 °C compared with explants incubated at 37 °C, which was prevented by leptin stimulation. These data illustrate the potential role of leptin for reducing apoptosis in trophoblast explants, including trophoblastic cells, triggered by high temperature, by preventing the activation of p53 signaling.


Subject(s)
Apoptosis/drug effects , Hot Temperature , Leptin/pharmacology , Placenta/drug effects , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Apoptosis/physiology , Caspase 3/metabolism , Chorionic Villi/drug effects , Chorionic Villi/metabolism , Female , Humans , Keratin-18/metabolism , Phosphorylation/drug effects , Placenta/metabolism , Pregnancy
13.
Sci Rep ; 6: 26364, 2016 05 23.
Article in English | MEDLINE | ID: mdl-27212399

ABSTRACT

Trophoblast cells migrate and invade the decidual stroma in a tightly regulated process to maintain immune homeostasis at the maternal-placental interface during the first weeks of pregnancy. Locally synthesized factors modulate trophoblast cell function and their interaction with maternal leukocytes to promote the silent clearance of apoptotic cells. The vasoactive intestinal peptide (VIP) is a pleiotropic polypeptide with trophic and anti-inflammatory effects in murine pregnancy models. We explored the effect of VIP on two human first trimester trophoblast cell lines, particularly on their migration, invasiveness and interaction with phagocytic cells, and the signalling and regulatory pathways involved. We found that VIP enhanced trophoblast cell migration and invasion through the activation of high affinity VPAC receptors and PKA-CRE signalling pathways. VIP knocked-down trophoblast cells showed reduced migration in basal and leukemic inhibitor factor (LIF)-elicited conditions. In parallel, VIP-silenced trophoblast cells failed to induce the phagocytosis of apoptotic bodies and the expression of immunosuppressant markers by human monocytes. Our results suggest that VIP-mediated autocrine pathways regulate trophoblast cell function and contribute to immune homeostasis maintenance at placentation and may provide new clues for therapeutic intervention in pregnancies complicated by defective deep placentation.


Subject(s)
Autocrine Communication , Phagocytes/cytology , Trophoblasts/cytology , Vasoactive Intestinal Peptide/metabolism , Cell Line , Cell Movement , Female , Gene Knockdown Techniques , Humans , Pregnancy , Pregnancy Trimester, First , Protein Kinases/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Signal Transduction , Vasoactive Intestinal Peptide/genetics
14.
Placenta ; 36(11): 1266-75, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26386653

ABSTRACT

Leptin, a 16-kDa polypeptide hormone, is produced by the adipocyte and can also be synthesized by placenta. We previously demonstrated that leptin promotes proliferation and survival in placenta, in part mediated by the p53 pathway. In this work, we investigated the mechanisms involved in leptin down-regulation of p53 level. The human first trimester cytotrophoblastic Swan-71 cell line and human placental explants at term were used. In order to study the late phase of apoptosis, triggered by serum deprivation, experiments of DNA fragmentation were carried out. Exogenous leptin added to human placental explants, showed a decrease on DNA ladder formation and MAPK pathway is involved in this leptin effect. We also found that under serum deprivation condition, leptin decreases p53 levels and the inhibitory leptin effect is lost when cells were pretreated with 50 µM PD98059 or 10 µM LY29004; or were transfected with dominant negative mutants of intermediates of these pathways, suggesting that MAPK and PI3K signaling pathways are necessaries for leptin action. Additionally, leptin diminished Ser-46 p53 phosphorylation and this effect in placental explants was mediated by the activation of MAPK and PI3K pathways. Finally, in order to assess leptin effect on p53 half-life experiments with cycloheximide were performed and MDM-2 expression was analyzed. Leptin diminished p53 half-life and up-regulated MDM-2 expression. In summary, we provided evidence suggesting that leptin anti-apoptotic effect is mediated by MAPK and PI3K pathways.


Subject(s)
Leptin/metabolism , Trophoblasts/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis , Cell Line , Cell Proliferation , Humans , MAP Kinase Signaling System , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism
15.
Reproduction ; 149(4): 357-66, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25602035

ABSTRACT

Leptin exerts both stimulatory and inhibitory effects on the ovulatory process. In this study, we investigated whether these opposite effects involve changes in the oxidative status in response to different levels of leptin. To this end, we performed both in vivo and in vitro assays using ovaries of immature rats primed with gonadotropins to induce ovulation. Superoxide dismutase (SOD) and catalase (CAT) activity, lipid peroxidation, glutathione (GSH) content, and reactive oxygen species (ROS) were studied as oxidative damage-related parameters. The expression of BCL2, BAX, and caspase 3 were measured by western blot as apoptosis-related biomarkers. The acute treatment with leptin, which inhibits ovulation, decreased SOD activity and increased active caspase 3 expression. No differences were found in CAT activity, lipid peroxidation, or total GSH. In contrast, the daily administration of leptin, which induces ovulation, decreased GSH content, ROS levels, and Bax and active caspase 3 expression, but caused no changes in other parameters. In addition, the daily administration of leptin induced follicular growth, measured by the number of antral follicles in ovarian sections. Using ovarian explant cultures, we found increased BCL2 expression and decreased SOD activity at low and high concentrations of leptin respectively. Thus, leptin can modulate the oxidative status of the ovarian tissue, during the ovulatory process, by acting on different targets according to its circulating levels. At low concentration, leptin seems to play a protective role against the oxidative stress, whereas at high concentrations, this protein seems to be involved in cell death.


Subject(s)
Leptin/pharmacology , Ovary/metabolism , Ovulation/drug effects , Oxidative Stress/drug effects , Animals , Blotting, Western , Caspase 3/metabolism , Catalase/metabolism , Female , Glutathione/metabolism , Lipid Peroxidation/drug effects , Ovulation/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism
16.
Clin Chem Lab Med ; 53(1): 15-28, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25014521

ABSTRACT

Reproductive function is dependent on energy resources. The role of weight, body composition, fat distribution and the effect of diet have been largely investigated in experimental female animals as well as in women. Any alteration in diet and/or weight may induce abnormalities in timing of sexual maturation and fertility. However, the cellular mechanisms involved in the fine coordination of energy balance and reproduction are largely unknown. The brain and hypothalamic structures receive endocrine and/or metabolic signals providing information on the nutritional status and the degree of fat stores. Adipose tissue acts both as a store of energy and as an active endocrine organ, secreting a large number of biologically important molecules termed adipokines. Adipokines have been shown to be involved in regulation of the reproductive functions. The first adipokine described was leptin. Extensive research over the last 10 years has shown that leptin is not only an adipose tissue-derived messenger of the amount of energy stores to the brain, but also a crucial hormone/cytokine for a number of diverse physiological processes, such as inflammation, angiogenesis, hematopoiesis, immune function, and most importantly, reproduction. Leptin plays an integral role in the normal physiology of the reproductive system with complex interactions at all levels of the hypothalamic-pituitary gonadal (HPG) axis. In addition, leptin is also produced by placenta, where it plays an important autocrine function. Observational studies have demonstrated that states of leptin excess, deficiency, or resistance can be associated with abnormal reproductive function. This review focuses on the leptin action in female reproduction.


Subject(s)
Leptin/metabolism , Reproduction , Animals , Embryo Implantation , Female , Fetal Development , Gonadotrophs/metabolism , Humans , Ovary/physiology , Pregnancy
17.
PLoS One ; 9(6): e99187, 2014.
Article in English | MEDLINE | ID: mdl-24922063

ABSTRACT

Leptin, a peripheral signal synthetized by the adipocyte to regulate energy metabolism, can also be produced by placenta, where it may work as an autocrine hormone. We have previously demonstrated that leptin promotes proliferation and survival of trophoblastic cells. In the present work, we aimed to study the molecular mechanisms that mediate the survival effect of leptin in placenta. We used the human placenta choriocarcinoma BeWo and first trimester Swan-71 cell lines, as well as human placental explants. We tested the late phase of apoptosis, triggered by serum deprivation, by studying the activation of Caspase-3 and DNA fragmentation. Recombinant human leptin added to BeWo cell line and human placental explants, showed a decrease on Caspase-3 activation. These effects were dose dependent. Maximal effect was achieved at 250 ng leptin/ml. Moreover, inhibition of endogenous leptin expression with 2 µM of an antisense oligonucleotide, reversed Caspase-3 diminution. We also found that the cleavage of Poly [ADP-ribose] polymerase-1 (PARP-1) was diminished in the presence of leptin. We analyzed the presence of low DNA fragments, products from apoptotic DNA cleavage. Placental explants cultivated in the absence of serum in the culture media increased the apoptotic cleavage of DNA and this effect was prevented by the addition of 100 ng leptin/ml. Taken together these results reinforce the survival effect exerted by leptin on placental cells. To improve the understanding of leptin mechanism in regulating the process of apoptosis we determined the expression of different intermediaries in the apoptosis cascade. We found that under serum deprivation conditions, leptin increased the anti-apoptotic BCL-2 protein expression, while downregulated the pro-apoptotic BAX and BID proteins expression in Swan-71 cells and placental explants. In both models leptin augmented BCL-2/BAX ratio. Moreover we have demonstrated that p53, one of the key cell cycle-signaling proteins, is downregulated in the presence of leptin under serum deprivation. On the other hand, we determined that leptin reduced the phosphorylation of Ser-46 p53 that plays a pivotal role for apoptotic signaling by p53. Our data suggest that the observed anti-apoptotic effect of leptin in placenta is in part mediated by the p53 pathway. In conclusion, we provide evidence that demonstrates that leptin is a trophic factor for trophoblastic cells.


Subject(s)
Apoptosis , Down-Regulation , Leptin/metabolism , Placenta/cytology , Placenta/metabolism , Tumor Suppressor Protein p53/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Cell Line, Tumor , Female , Humans , Phosphorylation , Phosphoserine/metabolism , Pregnancy , Trophoblasts/cytology , Trophoblasts/metabolism , bcl-2-Associated X Protein/metabolism
18.
Biol Reprod ; 89(1): 20, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23718986

ABSTRACT

Leptin, one of the adipokines that controls energy metabolism via the central nervous system, also has pleiotropic peripheral effects, acting as a proinflammatory cytokine. Leptin is also produced by trophoblastic cells in the placenta, where leptin seems to function as a trophic autocrine hormone. Leptin expression is regulated by various tissue-specific factors, such as insulin, in the adipocyte. However, the complete regulation of leptin production in the placenta is still poorly understood. That is why we investigated the regulation of leptin expression by insulin in JEG-3 trophoblastic cells and human placental explants from normal pregnancies. Western blot analysis and quantitative real time RT-PCR was performed to determine the leptin expression level after treatment of cells or trophoblast explants with different concentrations of insulin (0.1-100 nM). Leptin promoter activity was evaluated by transient transfection with a plasmid construct containing different promoter regions and the reporter luciferase gene. We found a stimulatory, dose-dependent effect of insulin on endogenous leptin expression in human placental explants. Maximal effect was achieved at 10 nM insulin, and this effect can be totally prevented both by blocking phosphatidylinositol 3 kinase (PI3K) pathways and mitogen-activated protein kinase (MAPK). Moreover, insulin treatment significantly enhanced leptin promoter activity up to 40% in JEG-3 trophoblastic cells. Deletion analysis demonstrated that a minimal promoter region between -1951 and -1546 bp is necessary to achieve insulin effects. In conclusion, we provide evidence suggesting that insulin induces leptin expression in trophoblastic cells, enhancing the activity of leptin promoter region between -1951 and -1546 bp, via both PI3K- and MAPK-signaling pathways.


Subject(s)
Insulin/physiology , Leptin/biosynthesis , Leptin/genetics , Trophoblasts/metabolism , Female , Humans , Leptin/physiology , MAP Kinase Signaling System , Pregnancy , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation
19.
PLoS One ; 7(10): e46216, 2012.
Article in English | MEDLINE | ID: mdl-23056265

ABSTRACT

Pleiotropic effects of leptin have been identified in reproduction and pregnancy, particularly in the placenta, where it works as an autocrine hormone. In this work, we demonstrated that human chorionic gonadotropin (hCG) added to JEG-3 cell line or to placental explants induces endogenous leptin expression. We also found that hCG increased cAMP intracellular levels in BeWo cells in a dose-dependent manner, stimulated cAMP response element (CRE) activity and the cotransfection with an expression plasmid of a dominant negative mutant of CREB caused a significant inhibition of hCG stimulation of leptin promoter activity. These results demonstrate that hCG indeed activates cAMP/PKA pathway, and that this pathway is involved in leptin expression. Nevertheless, we found leptin induction by hCG is dependent on cAMP levels. Treatment with (Bu)(2)cAMP in combination with low and non stimulatory hCG concentrations led to an increase in leptin expression, whereas stimulatory concentrations showed the opposite effect. We found that specific PKA inhibition by H89 caused a significant increase of hCG leptin induction, suggesting that probably high cAMP levels might inhibit hCG effect. It was found that hCG enhancement of leptin mRNA expression involved the MAPK pathway. In this work, we demonstrated that hCG leptin induction through the MAPK signaling pathway is inhibited by PKA. We observed that ERK1/2 phosphorylation increased when hCG treatment was combined with H89. In view of these results, the involvement of the alternative cAMP/Epac signaling pathway was studied. We observed that a cAMP analogue that specifically activates Epac (CPT-OMe) stimulated leptin expression by hCG. In addition, the overexpression of Epac and Rap1 proteins increased leptin promoter activity and enhanced hCG. In conclusion, we provide evidence suggesting that hCG induction of leptin gene expression in placenta is mediated not only by activation of the MAPK signaling pathway but also by the alternative cAMP/Epac signaling pathway.


Subject(s)
Chorionic Gonadotropin/physiology , Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Leptin/biosynthesis , MAP Kinase Signaling System , Placenta/metabolism , Base Sequence , Blotting, Western , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA Primers , Enzyme Activation , Female , Humans , In Vitro Techniques , Pregnancy , Real-Time Polymerase Chain Reaction
20.
Biochim Biophys Acta ; 1823(4): 900-10, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22310000

ABSTRACT

The placenta produces a wide number of molecules that play essential roles in the establishment and maintenance of pregnancy. In this context, leptin has emerged as an important player in reproduction. The synthesis of leptin in normal trophoblastic cells is regulated by different endogenous biochemical agents, but the regulation of placental leptin expression is still poorly understood. We have previously reported that 17ß-estradiol (E(2)) up-regulates placental leptin expression. To improve the understanding of estrogen receptor mechanisms in regulating leptin gene expression, in the current study we examined the effect of membrane-constrained E(2) conjugate, E-BSA, on leptin expression in human placental cells. We have found that leptin expression was induced by E-BSA both in BeWo cells and human placental explants, suggesting that E(2) also exerts its effects through membrane receptors. Moreover E-BSA rapidly activated different MAPKs and AKT pathways, and these pathways were involved in E(2) induced placental leptin expression. On the other hand we demonstrated the presence of ERα associated to the plasma membrane of BeWo cells. We showed that E(2) genomic and nongenomic actions could be mediated by ERα. Supporting this idea, the downregulation of ERα level through a specific siRNA, decreased E-BSA effects on leptin expression. Taken together, these results provide new evidence of the mechanisms whereby E(2) regulates leptin expression in placenta and support the importance of leptin in placental physiology.


Subject(s)
Cell Membrane/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Gene Expression Regulation/drug effects , Leptin/genetics , Placenta/cytology , Placenta/metabolism , Cell Membrane/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Estradiol/analogs & derivatives , Female , Fulvestrant , Gene Silencing/drug effects , Humans , Leptin/metabolism , Models, Biological , Placenta/drug effects , Pregnancy , Protein Binding/drug effects , Protein Transport/drug effects , Serum Albumin, Bovine , Signal Transduction/drug effects , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...