Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 230
Filter
1.
J Endocrinol ; 237(3): 301-310, 2018 06.
Article in English | MEDLINE | ID: mdl-29661800

ABSTRACT

Several developmental windows, including placentation, must be negotiated to establish and maintain pregnancy. Impaired placental function can lead to preeclampsia and/or intrauterine growth restriction (IUGR), resulting in increased infant mortality and morbidity. It has been hypothesized that chorionic somatomammotropin (CSH) plays a significant role in fetal development, potentially by modifying maternal and fetal metabolism. Recently, using lentiviral-mediated in vivo RNA interference in sheep, we demonstrated significant reductions in near-term (135 days of gestation; dGA) fetal and placental size, and altered fetal liver gene expression, resulting from CSH deficiency. We sought to examine the impact of CSH deficiency on fetal and placental size earlier in gestation (50 dGA), and to examine placental gene expression at 50 and 135 dGA. At 50 dGA, CSH-deficient pregnancies exhibited a 41% reduction (P ≤ 0.05) in uterine vein concentrations of CSH, and significant (P ≤ 0.05) reductions (≈21%) in both fetal body and liver weights. Placentae harvested at 50 and 135 dGA exhibited reductions in IGF1 and IGF2 mRNA concentrations, along with reductions in SLC2A1 and SLC2A3 mRNA. By contrast, mRNA concentrations for various members of the System A, System L and System y+ amino acid transporter families were not significantly impacted. The IUGR observed at the end of the first-third of gestation indicates that the near-term IUGR reported previously, began early in gestation, and may have in part resulted from deficits in the paracrine action of CSH within the placenta. These results provide further compelling evidence for the importance of CSH in the progression and outcome of pregnancy.


Subject(s)
Fetal Development , Placenta/metabolism , Placental Lactogen/physiology , Animals , Animals, Genetically Modified , Female , Fetal Development/genetics , Fetal Growth Retardation/genetics , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Gene Expression , Gene Expression Regulation, Developmental , Gestational Age , Male , Placental Lactogen/blood , Placentation/genetics , Pregnancy , Sheep/genetics , Sheep/physiology
2.
Int J Mol Sci ; 18(11)2017 Oct 25.
Article in English | MEDLINE | ID: mdl-29068419

ABSTRACT

Background: Diabetes mellitus (DM) is a multifactorial disease orphan of a cure. Regenerative medicine has been proposed as novel strategy for DM therapy. Human fibroblast growth factor (FGF)-2b controls ß-cell clusters via autocrine action, and human placental lactogen (hPL)-A increases functional ß-cells. We hypothesized whether FGF-2b/hPL-A treatment induces ß-cell differentiation from ductal/non-endocrine precursor(s) by modulating specific genes expression. Methods: Human pancreatic ductal-cells (PANC-1) and non-endocrine pancreatic cells were treated with FGF-2b plus hPL-A at 500 ng/mL. Cytofluorimetry and Immunofluorescence have been performed to detect expression of endocrine, ductal and acinar markers. Bromodeoxyuridine incorporation and annexin-V quantified cells proliferation and apoptosis. Insulin secretion was assessed by RIA kit, and electron microscopy analyzed islet-like clusters. Results: Increase in PANC-1 duct cells de-differentiation into islet-like aggregates was observed after FGF-2b/hPL-A treatment showing ultrastructure typical of islets-aggregates. These clusters, after stimulation with FGF-2b/hPL-A, had significant (p < 0.05) increase in insulin, C-peptide, pancreatic and duodenal homeobox 1 (PDX-1), Nkx2.2, Nkx6.1, somatostatin, glucagon, and glucose transporter 2 (Glut-2), compared with control cells. Markers of PANC-1 (Cytokeratin-19, MUC-1, CA19-9) were decreased (p < 0.05). These aggregates after treatment with FGF-2b/hPL-A significantly reduced levels of apoptosis. Conclusions: FGF-2b and hPL-A are promising candidates for regenerative therapy in DM by inducing de-differentiation of stem cells modulating pivotal endocrine genes.


Subject(s)
Cell Differentiation , Fibroblast Growth Factor 2/physiology , Insulin-Secreting Cells , Pancreatic Ducts/physiology , Placental Lactogen/physiology , Diabetes Mellitus/therapy , Female , Fibroblast Growth Factor 2/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , Male , Middle Aged , Nuclear Proteins , Pancreatic Ducts/cytology , Pancreatic Ducts/metabolism , Placental Lactogen/metabolism , Regenerative Medicine/methods , Transcription Factors
3.
Adv Exp Med Biol ; 846: 1-35, 2015.
Article in English | MEDLINE | ID: mdl-25472532

ABSTRACT

New information concerning the effects of prolactin (PRL) on metabolic processes warrants reevaluation of its overall metabolic actions. PRL affects metabolic homeostasis by regulating key enzymes and transporters associated with glucose and lipid metabolism in several target organs. In the lactating mammary gland, PRL increases the production of milk proteins, lactose, and lipids. In adipose tissue, PRL generally suppresses lipid storage and adipokine release and affect adipogenesis. A specific case is made for PRL in the human breast and adipose tissues, where it acts as a circulating hormone and an autocrine/paracrine factor. Although its overall effects on body composition are both modest and species-specific, PRL may be involved in the manifestation of insulin resistance.


Subject(s)
Adipose Tissue/metabolism , Prolactin/genetics , Prolactin/physiology , Adipogenesis/physiology , Animals , Gene Expression Regulation , Growth Hormone/physiology , Humans , Lipid Metabolism , Placental Lactogen/physiology , Receptors, Prolactin/chemistry , Receptors, Prolactin/physiology
4.
Drug Metab Dispos ; 42(4): 492-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24408518

ABSTRACT

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.


Subject(s)
Cytochrome P-450 CYP2E1/biosynthesis , Hepatocytes/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Placental Lactogen/physiology , Adolescent , Adult , Animals , Blotting, Western , Cytochrome P-450 CYP2E1/metabolism , Enzyme Induction , Female , Hep G2 Cells , Hepatocytes/drug effects , Human Growth Hormone/pharmacology , Human Growth Hormone/physiology , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Pharmaceutical Preparations/metabolism , Phosphoinositide-3 Kinase Inhibitors , Placental Lactogen/pharmacology , Pregnancy/metabolism , Primary Cell Culture , Prolactin/pharmacology , Prolactin/physiology , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Young Adult
5.
Placenta ; 34(5): 407-15, 2013 May.
Article in English | MEDLINE | ID: mdl-23465880

ABSTRACT

UNLABELLED: Betamethasone (BET) is a widely used treatment for women who are at high risk of preterm delivery. In sheep, BET-induced growth restriction was found to be associated with reduced placenta lactogen (PL), a key regulator of fetal growth. We therefore hypothesized that also in humans a single course of BET administration is associated with a reduction of PL, associated with a deceleration in fetal growth. OBJECTIVE: To investigate effects of a single course of antenatal BET in humans on birth weight and PL. METHODS: Women exposed to BET (2 × 12 mg; n = 44) with normally grown fetuses between 23 + 5 and 34 + 0 wks (weeks + days of gestation) who delivered between 23 + 5 to 42 + 0 wks were compared to gestational age-matched controls (n = 49). Maternal gestational blood samples were obtained before, during and after BET treatment and at the time of birth. MAIN OUTCOME MEASURES: BET effects on fetal anthropometrics, placental morphometry and placental PL-protein and maternal plasma levels. RESULTS: The mean duration of days between BET administration and birth was 52 days. BET treatment was associated with decreased birth weight (-18.2%), head circumference (-8.6%), body length (-6.0%), and placental width (-5.5%), as compared to controls. These changes were irrespective of possible maternal confounders (gestational age at birth, maternal age, maternal BMI gain during pregnancy, smoking etc.). However, neither PL-plasma levels within 48 h after BET treatment nor placental PL-protein levels and maternal plasma levels at birth were changed after BET treatment. In central regions of the placenta, BET treatment increased the circumference of syncytiotrophoblast nuclei by +4.7% and nucleus surface area by +9.4% compared to controls, but these changes were not related to placental PL-protein or maternal PL-plasma levels at birth. CONCLUSION: A single course of BET treatment was accompanied with reduced fetal growth, but this growth restricting effect was not associated with altered placental or maternal plasma PL levels. Altered expression of PL appears not to be causal for BET-induced fetal growth restriction in the human.


Subject(s)
Betamethasone/adverse effects , Fetal Development/drug effects , Fetal Growth Retardation/chemically induced , Glucocorticoids/adverse effects , Placental Lactogen/physiology , Adult , Betamethasone/administration & dosage , Birth Weight/drug effects , Body Height , Cephalometry , Female , Gestational Age , Humans , Infant, Newborn , Placenta/chemistry , Placenta/pathology , Placental Lactogen/analysis , Placental Lactogen/blood , Pregnancy , Premature Birth/prevention & control
6.
Diabetologia ; 53(12): 2589-99, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20938637

ABSTRACT

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.


Subject(s)
Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Placental Lactogen/pharmacology , Pregnancy/metabolism , Serotonin/biosynthesis , Animals , Cells, Cultured , Embryo, Mammalian , Female , Gene Expression Regulation, Enzymologic/drug effects , Gestational Age , Insulin-Secreting Cells/classification , Mice , Mice, Inbred C57BL , Mice, Knockout , Placental Lactogen/physiology , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism , Up-Regulation/drug effects
7.
Diabetes ; 59(1): 143-52, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19833884

ABSTRACT

OBJECTIVE: The objectives of the study were to determine whether the cell cycle transcription factor, FoxM1, is required for glucose homeostasis and beta-cell mass expansion in maternal islets during pregnancy and whether FoxM1 is essential for placental lactogen (PL)-induced beta-cell proliferation. RESEARCH DESIGN AND METHODS: beta-Cell mass, beta-cell proliferation, and glucose homeostasis were assessed in virgin, pregnant, and postpartum mice with a pancreas-wide Foxm1 deletion (FoxM1(Deltapanc)). Wild-type islets were cultured with or without PL and examined for Foxm1 induction. Transgenic mice overexpressing PL in beta-cells were bred with FoxM1(Deltapanc) mice, and beta-cell proliferation was examined. RESULTS: Foxm1 was upregulated in maternal islets during pregnancy. In contrast to controls, beta-cell proliferation did not increase in pregnant FoxM1(Deltapanc) females. Mutant islets showed increased Menin and nuclear p27. FoxM1(Deltapanc) females developed gestational diabetes mellitus as pregnancy progressed. After parturition, euglycemia was restored in FoxM1(Deltapanc) females, but islet size was significantly reduced. Strikingly, beta-cell mass was normal in postpartum FoxM1(Deltapanc) pancreata due to a combination of increased beta-cell size and islet neogenesis. Evidence for neogenesis included increased number of endocrine clusters, increased proportion of smaller islets, and increased neurogenin 3 or insulin expression in cells adjacent to ducts. PL induced Foxm1 expression in cultured islets, and FoxM1 was essential for PL-mediated increases in beta-cell proliferation in vivo. CONCLUSIONS: FoxM1 is essential for beta-cell compensation during pregnancy. In the absence of increased beta-cell proliferation, neogenesis is induced in postpartum FoxM1(Deltapanc) pancreata. Our results suggest that FoxM1 functions downstream of PL to mediate its effects on beta-cell proliferation.


Subject(s)
Diabetes, Gestational/genetics , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Placental Lactogen/physiology , Animals , Blotting, Western , Cell Division , DNA Primers , Female , Forkhead Box Protein M1 , Gene Deletion , Gene Expression Regulation, Developmental , Glucose/metabolism , Glucose Tolerance Test , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/physiology , Islets of Langerhans/physiology , Mice , Mice, Transgenic , Pregnancy , RNA/genetics , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction
8.
Int. j. morphol ; 26(4): 1029-1033, Dec. 2008. ilus, tab
Article in English | LILACS | ID: lil-532940

ABSTRACT

The aim of this study was observe differences in the immunological distribution of the placental lactogene and IGF-1 receptor on free-chorionic villi, between studied groups and to relate the neonatal diagnosis of PEG with morphometric and immunohystochemical characteristics of the placenta. A total of, twelve placentas from AEG newborn and twelve from PEG newborn were obtained from the Maternity Ward of Temuco, Chile. H&E, Alcian blue and Masson's trichromic stains, as well as Hematoxilyn-PAS. In the immunoperoxidase technique, were used: 1) placental lactogen (polyclonal, dilution 1:200, NCL-PLP, Novocastra) 2) Insuline-1 like growth factor (monoclonal, dilution 1:200, NCL-GHR, Novocastra). Differences between PEG and AEG placentae in the immnunostaing for placental lactogen and IGF-1 receptor in the sincitial throphoblast were not observed.


El objetivo de este estudio fue observar diferencias en la distribución del lactogeno placentario y receptor del factor de crecimiento similar a la insulina, entre placentas de recién nacidos normales para la edad gestacional AEG y pequeños para la edad gestacional PEG. Un total de 12 placentas de recién nacidos AEG y 12 PEG obtenidas de la maternidad del Hospital de Temuco, Chile fueron procesadas con técnicas histológicas H&E, azul de Alcián y un método de tinción tricrómico. La técnica de inmunoperoxidasa utilizada fue: 1) lactogeno placentario (pohclonal, dilución 1:200, NCL-PLP, Novocastra) 2) Factor de crecimiento similar a la insulina (monoclonal, dilución 1:200, NCL-GHR, Novocastra). No se observaron diferencias en la distribución de lactogeno placentario ni factor de crecimiento similar a la insulina entre las placentas provenientes de recién nacidos pequeños para la edad gestacional y adecuados para la edad gestacional.


Subject(s)
Humans , Infant, Newborn , Placental Lactogen/physiology , Receptor, IGF Type 1/physiology , Chorionic Villi/anatomy & histology , Chorionic Villi/physiology , Infant, Small for Gestational Age
9.
Rev Endocr Metab Disord ; 9(4): 329-43, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18777097

ABSTRACT

Type 2 diabetes mellitus is a complex disease characterized by beta-cell failure in the setting of insulin resistance. In early stages of the disease, pancreatic beta-cells adapt to insulin resistance by increasing mass and function. As nutrient excess persists, hyperglycemia and elevated free fatty acids negatively impact beta-cell function. This happens by numerous mechanisms, including the generation of reactive oxygen species, alterations in metabolic pathways, increases in intracellular calcium and the activation of endoplasmic reticulum stress. These processes adversely affect beta-cells by impairing insulin secretion, decreasing insulin gene expression and ultimately causing apoptosis. In this review, we will first discuss the regulation of beta-cell mass during normal conditions. Then, we will discuss the mechanisms of beta-cell failure, including glucotoxicity, lipotoxicity and endoplasmic reticulum stress. Further research into mechanisms will reveal the key modulators of beta-cell failure and thus identify possible novel therapeutic targets. Type 2 diabetes mellitus is a multifactorial disease that has greatly risen in prevalence in part due to the obesity and inactivity that characterize the modern Western lifestyle. Pancreatic beta-cells possess the potential to greatly expand their function and mass in both physiologic and pathologic states of nutrient excess and increased insulin demand. beta-cell response to nutrient excess occurs by several mechanisms, including hypertrophy and proliferation of existing beta-cells, increased insulin production and secretion, and formation of new beta-cells from progenitor cells [1, 2]. Failure of pancreatic beta-cells to adequately expand in settings of increased insulin demand results in hyperglycemia and diabetes. In this review, we will first discuss the factors involved in beta-cell growth and then discuss the mechanisms by which beta-cell expansion fails and leads to beta-cell failure and diabetes (Fig. 1).


Subject(s)
Diabetes Mellitus, Type 2/complications , Insulin-Secreting Cells/pathology , Animals , Apoptosis , Cell Proliferation , Endoplasmic Reticulum/physiology , Fatty Acids, Nonesterified/adverse effects , Glucose/metabolism , Glucose/toxicity , Growth Hormone/physiology , Hepatocyte Growth Factor/physiology , Humans , Incretins/physiology , Insulin/physiology , Insulin Resistance/physiology , Insulin-Secreting Cells/drug effects , Parathyroid Hormone-Related Protein/physiology , Placental Lactogen/physiology , Prolactin/physiology , Somatomedins/physiology
10.
Reprod Biol ; 8(2): 85-106, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18677398

ABSTRACT

The bovine placenta produces a wide variety of proteins that are structurally and functionally similar to the pituitary proteins from the GH/PRL gene family. Bovine placental lactogen (bPL) is a 200-amino acid long glycoprotein hormone that exhibits both lactogenic and somatogenic properties. The apparent molecular masses of purified native (n) bPL molecules (31-33 kDa) exceed 23 041 Da, which is the theoretical molecular mass of the protein core. At least six isoelectric variants (pI: 4.85-6.3) of bPL were described in cotyledonary extracts and three different bPL isoforms (pI: 4.85-5.25) were found in fetal sera. The bPL molecules that are detected in higher concentrations in peripheral circulation exhibit a more acidic pI than those present in placental homogenates. This may reflect an important glycosylation process occurring just prior to the bPL secretion. The bPL mRNA is transcribed in trophectoderm binucleate cells starting from Day 30 of pregnancy until the end of gestation. In mothers, bPL is involved in the regulation of ovarian function, mammogenesis, lactogenesis, and pregnancy stage-dependent adaptation of nutrient supplies to the fetus. Due to the higher fetal, compared to maternal concentrations of circulating hormone, it has been suggested that bPL primarily targets fetal tissues.


Subject(s)
Placental Lactogen , Animals , Cattle , Female , Fetus/blood supply , Glycosylation , Humans , Models, Molecular , Placenta/chemistry , Placenta/metabolism , Placental Lactogen/blood , Placental Lactogen/chemistry , Placental Lactogen/physiology , Pregnancy , Recombinant Proteins , Trophoblasts/metabolism
12.
Trends Endocrinol Metab ; 17(8): 301-7, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16934485

ABSTRACT

Vasoinhibins are a family of peptides derived from prolactin, growth hormone and placental lactogen that act on endothelial cells to suppress vasodilation and angiogenesis and to promote apoptosis-mediated vascular regression. Some of the pathways by which vasoinhibins act have now been defined, and recent developments indicate that endogenous vasoinhibins exert tonic and essential actions on blood vessel growth, dilation and regression in vivo. By studying the pathways that can generate vasoinhibins, and the nature of their receptors and key biological mediators, it should be possible to clarify the role of vasoinhibins in controlling vascular function in health and disease.


Subject(s)
Endothelium, Vascular/physiology , Growth Hormone/physiology , Neovascularization, Physiologic , Placental Lactogen/physiology , Prolactin/physiology , Angiogenesis Modulating Agents/chemistry , Angiogenesis Modulating Agents/metabolism , Animals , Growth Hormone/chemistry , Humans , Models, Biological , Models, Molecular , Placental Lactogen/chemistry , Prolactin/chemistry , Protein Binding , Signal Transduction , Vasoconstrictor Agents/chemistry , Vasoconstrictor Agents/metabolism
13.
Horm Res ; 65 Suppl 3: 41-9, 2006.
Article in English | MEDLINE | ID: mdl-16612113

ABSTRACT

This review outlines the regulation of maternal metabolism by hormones, cytokines and growth factors, highlighting recent studies that implicate disordered somatolactogen signalling in the pathogenesis of perinatal growth failure and the development of the metabolic syndrome.


Subject(s)
Fetal Development/physiology , Pituitary Hormones/physiology , Placental Hormones/physiology , Pregnancy/metabolism , Animals , Female , Growth Hormone/physiology , Humans , Placental Lactogen/physiology , Prolactin/physiology
14.
Biol Reprod ; 74(6): 1041-50, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16481593

ABSTRACT

Defects in placental development lead to pregnancies at risk for miscarriage and intrauterine growth retardation and are associated with preeclampsia, a leading cause of maternal death and premature birth. In preeclampsia, impaired placental formation has been associated with alterations in a specific trophoblast lineage, the invasive trophoblast cells. In this study, an RT-PCR Trophoblast Gene Expression Profile previously developed by our laboratory was utilized to examine the lineage-specific gene expression of the rat Rcho-1 trophoblast cell line. Our results demonstrated that Rcho-1 cells represent an isolated, trophoblast population committed to the giant cell lineage. RT-PCR analysis revealed that undifferentiated Rcho-1 cells expressed trophoblast stem cell marker, Id2, and trophoblast giant cell markers. On differentiation, Rcho-1 cells downregulated Id2 and upregulated Csh1, a marker of the trophoblast giant cell lineage. Neither undifferentiated nor differentiated Rcho-1 cells expressed spongiotrophoblast marker Tpbpa or labyrinthine markers Esx1 and Tec. Differentiating Rcho-1 cells in hypoxia did not alter the expression of lineage-specific markers; however, hypoxia did inhibit the downregulation of the trophoblast stem cell marker Id2. Differentiation in hypoxia also blocked the induction of CSH1 protein. In addition, hypoxia inhibited stress fiber formation and abolished the induction of palladin, a protein associated with stress fiber formation and focal adhesions. Thus, Rcho-1 cells can be maintained as a proliferative, lineage-specific cell line that is committed to the trophoblast giant cell lineage on differentiation in both normoxic and hypoxic conditions; however, hypoxia does inhibit aspects of trophoblast giant cell differentiation at the molecular, morphological, and functional levels.


Subject(s)
Cell Differentiation/physiology , Cell Hypoxia/physiology , Trophoblasts/cytology , Trophoblasts/physiology , Animals , Cell Differentiation/genetics , Cell Line , Cell Lineage/genetics , Cell Lineage/physiology , Cell Proliferation , Cytoskeletal Proteins/analysis , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/physiology , Down-Regulation , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Inhibitor of Differentiation Protein 2/analysis , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/physiology , Phosphoproteins/analysis , Phosphoproteins/genetics , Phosphoproteins/physiology , Placenta/pathology , Placental Lactogen/analysis , Placental Lactogen/genetics , Placental Lactogen/physiology , Placentation , Pregnancy Proteins/analysis , Pregnancy Proteins/genetics , Pregnancy Proteins/physiology , Protein-Tyrosine Kinases/analysis , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/physiology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/analysis , Transcription Factors/genetics , Transcription Factors/physiology , Trophoblasts/chemistry , Up-Regulation
15.
Placenta ; 27(4-5): 357-66, 2006.
Article in English | MEDLINE | ID: mdl-16005513

ABSTRACT

Here we report the selective expression of two POU transcription factor genes, PLA-1 and OCT-1, in human placenta and choriocarcinoma cell lines JAR, JEG-3 and BeWo. Pla-1 protein binds to a POU-consensus DNA sequence in the human placental lactogen-3 (PL-3) promoter and it is capable of trans-activating its transcription up to 18-fold. Other tissue-specific or ubiquitous POU transcription factors such as Pit-1/GHF-1 or Oct-1 showed none or low levels of trans-activation of the PL-3 promoter. In addition, we identified an unique and highly charged region in the N-terminal portion of Pla-1 protein required for full trans-activation of the PL-3 promoter.


Subject(s)
POU Domain Factors/metabolism , Placenta/metabolism , Placental Lactogen/metabolism , Placental Lactogen/physiology , Binding Sites , Cell Line, Tumor , Female , Gene Expression Regulation , Humans , Placental Lactogen/genetics , Promoter Regions, Genetic , Trans-Activators
16.
Int J Biochem Cell Biol ; 38(5-6): 931-50, 2006.
Article in English | MEDLINE | ID: mdl-16168703

ABSTRACT

Recent studies have demonstrated that human islet allograft transplantation can be a successful therapeutic option in the treatment of patients with Type I diabetes. However, this impressive recent advance is accompanied by a very important constraint. There is a critical paucity of pancreatic islets or pancreatic beta cells for islet transplantation to become a large-scale therapeutic option in patients with diabetes. This has prompted many laboratories around the world to invigorate their efforts in finding ways for increasing the availability of beta cells or beta cell surrogates that potentially could be transplanted into patients with diabetes. The number of studies analyzing the mechanisms that govern beta cell proliferation and growth in physiological and pathological conditions has increased exponentially during the last decade. These studies exploring the role of growth factors, intracellular signaling molecules and cell cycle regulators constitute the substrate for future strategies aimed at expanding human beta cells in vitro and/or in vivo after transplantation. In this review, we describe the current knowledge on the effects of several beta cell growth factors that have been shown to increase beta cell proliferation and expand beta cell mass in vitro and/or in vivo and that they could be potentially deployed in an effort to increase the number of patients transplanted with islets. Furthermore, we also analyze in this review recent studies deciphering the relevance of these specific islet growth factors as physiological and pathophysiological regulators of beta cell proliferation and islet growth.


Subject(s)
Cell Proliferation/drug effects , Growth Substances/physiology , Insulin-Secreting Cells/cytology , Animals , Glucagon-Like Peptide 1/physiology , Growth Hormone/physiology , Hepatocyte Growth Factor/physiology , Humans , Insulin/physiology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/transplantation , Parathyroid Hormone-Related Protein/physiology , Placental Lactogen/physiology , Prolactin/physiology , Signal Transduction/physiology , Somatomedins/physiology
17.
Endocrinology ; 146(1): 103-12, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15388648

ABSTRACT

To delineate the roles of the lactogens and GH in the control of perinatal and postnatal growth, fat deposition, insulin production, and insulin action, we generated a novel mouse model that combines resistance to all lactogenic hormones with a severe deficiency of pituitary GH. The model was created by breeding PRL receptor (PRLR)-deficient (knockout) males with GH-deficient (little) females. In contrast to mice with isolated GH or PRLR deficiencies, double-mutant (lactogen-resistant and GH-deficient) mice on d 7 of life had growth failure and hypoglycemia. These findings suggest that lactogens and GH act in concert to facilitate weight gain and glucose homeostasis during the perinatal period. Plasma insulin and IGF-I and IGF-II concentrations were decreased in both GH-deficient and double-mutant neonates but were normal in PRLR-deficient mice. Body weights of the double mutants were reduced markedly during the first 3-4 months of age, and adults had striking reductions in femur length, plasma IGF-I and IGF binding protein-3 concentrations, and femoral bone mineral density. By age 6-12 months, however, the double-mutant mice developed obesity, hyperleptinemia, fasting hyperglycemia, relative hypoinsulinemia, insulin resistance, and glucose intolerance; males were affected to a greater degree than females. The combination of perinatal growth failure and late-onset obesity and insulin resistance suggests that the lactogen-resistant/GH-deficient mouse may serve as a model for the development of the metabolic syndrome.


Subject(s)
Animals, Newborn/growth & development , Animals, Newborn/metabolism , Growth Hormone/physiology , Placental Lactogen/physiology , Prolactin/physiology , Adipose Tissue/growth & development , Aging , Animals , Animals, Newborn/blood , Blood Glucose/metabolism , Bone Density , Drug Resistance , Female , Femur/growth & development , Glucose/physiology , Growth Hormone/deficiency , Insulin/blood , Insulin/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osmolar Concentration , Receptors, Prolactin/deficiency , Somatomedins/metabolism , Weight Gain
18.
Obstet Gynecol Clin North Am ; 31(4): 963-79, xii, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15550345

ABSTRACT

The endocrine control of lactation is one of the most complex physiologic mechanisms of human parturition. Mammogenesis, lactogenesis, galactopoiesis, and galactokinesis are all essential to assure proper lactation. Prolactin is the key hormone of lactation and seems to be the single most important galactopoietic hormone. Oxytocin, serotonin, opioids, histamine, substance P, and arginine-leucine modulate prolactin release by means of an autocrine/paracrine mechanism, whereas estrogen and progesterone hormones can act at the hypothalamic and adenohypophysial levels. Human placental lactogen and growth factors play an essential role to assure successful lactation during pregnancy. Oxytocin is the most powerful galactokinetic hormone.


Subject(s)
Lactation/physiology , Mammary Glands, Human/metabolism , Female , Humans , Mammary Glands, Human/anatomy & histology , Mammary Glands, Human/embryology , Milk, Human/metabolism , Oxytocin/physiology , Placental Lactogen/physiology , Pregnancy , Prolactin/physiology
19.
Anim Reprod Sci ; 82-83: 551-66, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15271479

ABSTRACT

Production of growth promoting substances by the placenta is regulated differently from the way production of similar compounds is regulated by maternal organs in various cases. Gene duplication is one of the mechanisms that facilitated the evolution of placental specific endocrine activity. Cattle, sheep and goats, although evolutionarily related, differ significantly from each other in the way their placental growth hormone (GH) and prolactin (PRL)-like hormones have evolved. Cattle carry one copy of the GH gene and there is no evidence yet for expression of that single GH gene copy in the placenta. On the other hand, the ovine GH gene has been duplicated and both oGH copies are expressed in the placenta during early stages of gestation. Prolactin gene duplication in ruminants resulted in the formation of specific placental-expressed prolactin-related genes including the placental lactogen (PL) gene. In homologous state, ovine PL manifests PRL activity, but antagonizes GH activity. Ovine PL activity which can be mediated by PRL receptors or by hetero-dimerization of GH and PRL receptors, provide a novel regulatory mechanism for somatogenic activity dependent on the coexistence of both GH and PRL receptors in the same cells. Another mechanism for specific placental endocrine activity is silencing of the alleles through genetic imprinting. Disruption of genetic imprinting of placental genes has been proposed as one of the explanations for the loss of cloned fetuses generated by somatic cell nuclear transfer.


Subject(s)
Embryonic and Fetal Development , Placental Hormones/physiology , Placentation , Animals , Cattle , Evolution, Molecular , Female , Gene Expression Regulation , Genomic Imprinting , Goats , Growth Hormone/genetics , Growth Hormone/physiology , Humans , Placental Lactogen/genetics , Placental Lactogen/physiology , Pregnancy , Prolactin/physiology , Receptors, Prolactin/physiology , Receptors, Somatotropin/physiology , Sheep
20.
J Endocrinol ; 173(2): 219-38, 2002 May.
Article in English | MEDLINE | ID: mdl-12010630

ABSTRACT

Prolactin, growth hormone and placental lactogen are members of a family of polypeptide hormones which share structural similarities and biological activities. Numerous functions have been attributed to these hormones, among which stand out their recently discovered effects on angiogenesis, the process by which new blood vessels are formed from the pre-existing microvasculature. Prolactin, growth hormone and placental lactogen, along with two non-classical members of the family, proliferin and proliferin-related protein, can act both as circulating hormones and as paracrine/autocrine factors to either stimulate or inhibit various stages of the formation and remodeling of new blood vessels, including endothelial cell proliferation, migration, protease production and apoptosis. Such opposing actions can reside in similar but independent molecules, as is the case of proliferin and proliferin-related protein, which stimulate and inhibit angiogenesis respectively. The potential to exert opposing effects on angiogenesis can also reside within the same molecule as the parent protein can promote angiogenesis (i.e. prolactin, growth hormone and placental lactogen), but after proteolytic processing the resulting peptide fragment acquires anti-angiogenic properties (i.e. 16 kDa prolactin, 16 kDa growth hormone and 16 kDa placental lactogen). The unique properties of the peptide fragments versus the full-length molecules, the regulation of the protease responsible for specific protein cleavage, the selective expression of specific receptors and their associated signal transduction pathways are issues that are being investigated to further establish the precise contribution of these hormones to angiogenesis under both physiological and pathological situations. In this review article, we summarize the known and speculative issues underlying the effects of the prolactin, growth hormone and placental lactogen family of proteins on angiogenesis, and address important remaining enigmas in this field of research.


Subject(s)
Neovascularization, Physiologic/physiology , Prolactin/physiology , Signal Transduction/physiology , Animals , Endothelium, Vascular/metabolism , Female , Glycoproteins/metabolism , Growth Hormone/physiology , Humans , Intercellular Signaling Peptides and Proteins , Pituitary Gland, Posterior/metabolism , Placental Lactogen/physiology , Pregnancy Proteins/metabolism , Protein Isoforms/physiology
SELECTION OF CITATIONS
SEARCH DETAIL