Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Clin Endocrinol Metab ; 99(10): E1957-66, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25029423

ABSTRACT

CONTEXT: Despite the absence of progesterone receptor protein in human endometrial endothelial cells (HEECs), endometria of women receiving long-acting progestin-only contraceptives (LAPCs) display reduced uterine blood flow, elevated reactive oxygen species generation, increased angiogenesis, and irregularly distributed, enlarged, fragile microvessels resulting in abnormal uterine bleeding. OBJECTIVE: We propose that paracrine factors from LAPC-treated human endometrial stromal cells (HESCs) impair HEEC functions by shifting the balance between HEEC viability and death in favor of the latter. DESIGN AND SETTING: Proliferation, apoptosis, and transcriptome analyses were performed in HEECs treated with conditioned medium supernatant (CMS) derived from HESCs treated with estradiol (E2) ± medroxyprogesterone acetate or etonogestrel under normoxia or hypoxia. Mass spectrometry interrogated the CMS secretome while immunostaining for neuronal pentraxin-1 (NPTX1), cleaved caspase-3, and cytochrome c was performed in cultured HEECs and paired endometria from women using LAPCs. MAIN OUTCOME: HEEC apoptosis and its underlying mechanism. RESULTS: HESC CMS from E2 + medroxyprogesterone acetate or E2 + etonogestrel incubations under hypoxia induced HEEC apoptosis (P < .05), whereas mass spectrometry of the CMS revealed increased NPTX1 secretion (P < .05). Endothelial cleaved caspase-3 and stromal NPTX1 immunoreactivity were significantly higher in LAPC-treated endometria (P < .001). Transcriptomics revealed AKT signaling inhibition and mitochondrial dysfunction in HEECs incubated with HESC CMS. In vitro analyses proved that CMS decreased HEEC AKT phosphorylation (P < .05) and that recombinant NPTX1 (P < .05) or NPTX1 + H2O2 (P < .001) increase HEEC apoptosis and cytosolic cytochrome c levels. CONCLUSIONS: LAPC-enhanced NPTX1 secretion and reactive oxygen species generation in HESCs impair HEEC survival resulting in a loss in vascular integrity, demonstrating a novel paracrine mechanism to explain LAPC-induced abnormal uterine bleeding.


Subject(s)
Apoptosis/drug effects , C-Reactive Protein/metabolism , Contraceptive Agents, Female/administration & dosage , Endometrium/drug effects , Nerve Tissue Proteins/metabolism , Progestins/administration & dosage , Stromal Cells/drug effects , Caspase 3/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Contraceptive Agents, Female/adverse effects , Culture Media, Conditioned/pharmacology , Cytochromes c/metabolism , Endometrium/cytology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Estradiol/adverse effects , Estradiol/pharmacology , Female , Humans , Medroxyprogesterone Acetate/adverse effects , Medroxyprogesterone Acetate/pharmacology , Microvessels/metabolism , Paracrine Communication/drug effects , Paracrine Communication/physiology , Progestins/adverse effects , Reactive Oxygen Species/metabolism , Stromal Cells/metabolism
2.
J Mol Histol ; 43(1): 107-13, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22143492

ABSTRACT

The epithelial cells of the mouse endometrium comprises the recurring physiologic changes such as proliferation and apoptosis induced by the reproductive hormones throughout the estrus cycle. The c-Abl is a protein tyrosine kinase, localized in the different cellular compartments such as nucleus, cytoplasm, mitochondria, and endoplasmic reticulum, interacts with different cellular proteins, including signaling adaptors, kinases, phosphatases, cell-cycle regulators, transcription factors and cytoskeletal proteins. Our hypothesis is that the c-Abl expression in the mouse uterine epithelium shows cyclic changes during the estrus cycle that is involved in regulation of the endometrial epithelial cells. The regulation of c-Abl gene and protein expression in the uterus of intact animals in the different cycle phases was investigated using immunohistochemistry, western blot and semi-quantitative RT-PCR. The immunohistochemistry results revealed that the luminal and the glandular epithelium (LE) and (GE), respectively, showed gradually increase in the expression of c-Abl from the proestrus (P) to the metestrus and followed by a decrease in the diestrus (D). c-Abl immunoreactivity was detected in the both LE and GE cells, especially the LE showed diminished the c-Abl protein expression on the D phase and the minimal value was detected on the P day. The c-Abl protein level in the LE was increased during the M, presenting a high correlation with the hormonal level of this cell type. The result of c-Abl RT-PCR analysis was compatible with pattern of c-Abl protein expression. In conclusion, the stage-specific protein pattern of the mammalian c-Abl tyrosine kinase presented a good correlation with the mouse estrus cycle and may have a regulative function during the uterine remodeling.


Subject(s)
Endometrium/metabolism , Estrous Cycle/physiology , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Animals , Endometrium/physiology , Female , Gene Expression , Mice , Mice, Inbred BALB C , Proteins , RNA, Messenger/metabolism , Uterus/metabolism
3.
J Clin Endocrinol Metab ; 95(5): 2271-5, 2010 May.
Article in English | MEDLINE | ID: mdl-20237167

ABSTRACT

CONTEXT: Unchanging plasma progesterone (P4) levels suggest that human labor is initiated by reduced P4 receptor (PR) expression, which elicits functional P4 withdrawal. The glucocorticoid receptor (GR) is also implicated in this process. OBJECTIVE: Our objective was to compare PR and GR staining in human decidual cells (DCs) and interstitial trophoblasts (ITs) of gestational age-matched pre- and postcontraction specimens and to evaluate steroid effects on PR and GR expression in human DC cultures. INTERVENTIONS AND MAIN OUTCOME MEASURES: Decidua basalis and parietalis sections were immunostained for PR or GR and then for the cytoplasmic DC and IT markers vimentin and cytokeratin. Western blotting measured PR and GR levels in nuclear extracts of cultured leukocyte-free term DCs after incubation with estradiol-17beta (E2) with or without medroxyprogesterone acetate (MPA). RESULTS: PR histological scores (HSCOREs) were significantly higher in DC nuclei from pre- vs. post-uterine-contraction decidua basalis and parietalis sections with PR immunostaining absent from ITs. In contrast, immunoreactive GR was localized in IT and DC nuclei. GR HSCORES were significantly higher in ITs than DCs but similar in pre- vs. post-uterine-contraction specimens. In term DC monolayers, PR-A and PR-B were enhanced by E2 and inhibited by MPA, whereas E2 plus MPA produced intermediate PR expression. The GR was constitutively expressed. CONCLUSIONS: In post- vs. pre-uterine-contraction specimens, significantly lower HSCOREs in DC nuclei, but not IT, and unchanging GR levels in DCs and ITs suggest that functional P4 withdrawal may occur in DCs and is unlikely to involve the GR. Nuclear extracts from DC monolayer cultures express steroid-regulated PR-A and PR-B and constitutive GR.


Subject(s)
Decidua/metabolism , Labor, Obstetric/metabolism , Placenta/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Progesterone/metabolism , Blotting, Western , Cytoplasm/metabolism , Delivery, Obstetric , Female , Humans , Keratins/metabolism , Pregnancy , Progesterone/blood , Trophoblasts/metabolism , Vimentin/metabolism
4.
Thromb Res ; 124(5): 516-20, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19720393

ABSTRACT

Vascular injury increases access and binding of plasma-derived factor VII to perivascular cell membrane-bound tissue factor (TF). The resulting TF/VIIa complex promotes hemostasis by cleaving pro-thrombin to thrombin leading to the fibrin clot. In human pregnancy, decidual cell-expressed TF prevents decidual hemorrhage (abruption). During placentation, trophoblasts remodel decidual spiral arteries into high conductance vessels. Shallow trophoblast invasion impedes decidual vascular conversion, producing an inadequate uteroplacental blood flow that elicits abruption-related placental ischemia. Thrombin induces several biological effects via cell surface protease activated receptors. In first trimester human DCs thrombin increases synthesis of sFlt-1, which elicits placental ischemia by impeding angiogenesis-related decidual vascular remodeling. During pregnacy, the fibrillar collagen-rich amnion and choriodecidua extracellular matrix (ECM) provides greater than additive tensile strength and structural integrity. Thrombin acts as an autocrine/paracrine mediator that degrades these ECMs by augmenting decidual cell expression of: 1) matrix metalloproteinases and 2) interleukin-8, a key mediator of abruption-associated decidual infiltration of neutrophils, which express several ECM degrading proteases. Among the cell types at the maternal fetal interface at term, TF expression is highest in decidual cells indicating that this TF meets the hemostatic demands of labor and delivery. TF expression in cultured term decidual cells is enhanced by progestin and thrombin suggesting that the maintenance of elevated circulating progesterone provides hemostatic protection and that abruption-generated thrombin acts in an autocrine/paracrine fashion on decidual cells to promote hemostasis via enhanced TF expression.


Subject(s)
Abruptio Placentae/metabolism , Decidua/metabolism , Pregnancy Complications, Hematologic/metabolism , Thromboplastin/metabolism , Uterine Hemorrhage/metabolism , Uterus/blood supply , Abruptio Placentae/blood , Abruptio Placentae/pathology , Animals , Decidua/pathology , Female , Hemostasis , Humans , Mice , Mice, Transgenic , Pregnancy , Pregnancy Complications, Hematologic/blood , Pregnancy Complications, Hematologic/pathology , Thrombin/metabolism , Uterine Hemorrhage/blood , Uterine Hemorrhage/pathology , Uterus/metabolism
5.
J Clin Endocrinol Metab ; 94(6): 2164-70, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19276228

ABSTRACT

CONTEXT: Perivascular cell membrane-bound tissue factor (TF) initiates hemostasis via thrombin generation. The identity and potential regulation of TF-expressing cells at the human maternal-fetal interface that confers hemostatic protection during normal and preterm delivery is unclear. OBJECTIVES: The objective of the study were to identify TF-expressing cells at the maternal-fetal interface in term and preterm decidual sections by immunohistochemistry and evaluate progestin, thrombin, TNF-alpha, and IL-1beta effects on TF expression by cultured human term decidual cells (DCs). INTERVENTIONS AND MAIN OUTCOME MEASURES: Serial placental sections were immunostained for TF. Leukocyte-free term DC monolayers were incubated with 10(-8) M estradiol (E2) or E2 plus 10(-7) M medroxyprogestrone acetate (MPA) +/- thrombin or TNF-alpha or IL-1beta. ELISA and Western blotting assessed TF in cell lysates. Quantitative real-time RT-PCR measured TF mRNA levels. RESULTS: Immunolocalized TF in DC membranes in preterm and term placental sections displayed higher Histologic Scores than villous mesenchymal cells (P < 0.05). TF was undetected in interstitial or extravillous trophoblasts. Compared with DCs incubated with E2, MPA and 2.5 U/ml thrombin each doubled TF levels (P < 0.05) and E2 + MPA + thrombin further doubled TF levels (P < 0.05), whereas TNF-alpha and IL-1beta were ineffective. Western blotting confirmed the ELISA results. Quantitative RT-PCR revealed corresponding changes in TF mRNA levels. CONCLUSIONS: In human term placental sections, DC-expressed TF exceeds that of other cell types at the maternal-fetal interface and is localized at the cell membranes in which it can bind to factor VII and meet the hemostatic demands of labor and delivery via thrombin formation. Unlike the general concept that TF is constitutive in cells that highly express it, MPA and thrombin significantly enhanced TF expression in term DC monolayers.


Subject(s)
Decidua/metabolism , Progestins/pharmacology , Term Birth/genetics , Thrombin/pharmacology , Thromboplastin/genetics , Cells, Cultured , Decidua/drug effects , Female , Gene Expression Regulation/drug effects , Humans , Interleukin-1beta/pharmacology , Medroxyprogesterone Acetate/pharmacology , Pregnancy , Term Birth/metabolism , Thromboplastin/metabolism , Tumor Necrosis Factor-alpha/pharmacology
6.
J Physiol Biochem ; 62(1): 27-34, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16909929

ABSTRACT

We investigated the effects of lipopolysaccharide (LPS) administration on plasma nitrite, nitrotyrosine and 6-keto prostaglandin F1alpha, (PGF1alpha) levels and the related resultant changes in function and histochemistry of aorta in rats. Plasma nitrite and PGF1alpha nitrotyrosine levels were analysed after 5 mg/kg intravenous LPS was administered to rats compared with those in non-treated rats. The distribution of nitrotyrosine in the aorta was studied immunohistochemically. The contractile responses of aortic rings to phenylephrine (PE) from both the LPS-treated and control rats were studied in the organ baths. There were increases in plasma nitrite, PGF1alpha, and nitrotyrosine concentrations of LPS-treated rats compared to non-treated rats. Immunoreactivity of nitrotyrosine residues were detected in the endothelial and smooth muscle cells in LPS-treated but not in control rat aorta. The contractile responses to PE of the LPS-treated rat aortic rings were significantly reduced as compared with those of control rat's. Incubation of the aortic rings from LPS-treated rats with cyclooxygenase inhibitor indomethacine or with a combination of indomethacine and nitric oxide synthase inhibitor N(omega)-nitro-L-arginine methyl ester (L-NAME) increased the contractile responses to the levels observed in control rats suggesting that both prostanoids and particularly nitric oxide (NO) are involved in the reduced contractile responses in LPS-treated rats. These results supported the view that LPS might cause an increment in both NO and PGI2 levels. This increase in the NO and PGI2 levels may be responsible from the reduction in responses of aorta to contractile agents in LPS-treated rats. Increased peroxynitrite formation in LPS-treated rats may lead to nitration of the tyrosil residues of the proteins in the aorta.


Subject(s)
Lipopolysaccharides/pharmacology , Nitrites/blood , Prostaglandins F/blood , Tyrosine/analogs & derivatives , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/physiology , Enzyme Inhibitors/pharmacology , Indomethacin/pharmacology , Male , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Muscle, Smooth/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Phenylephrine/pharmacology , Rats , Rats, Wistar , Tyrosine/blood , Tyrosine/metabolism , Vasoconstriction/drug effects
7.
Placenta ; 27(6-7): 648-59, 2006.
Article in English | MEDLINE | ID: mdl-16026828

ABSTRACT

Vasculogenesis in the human placenta comprises differentiation and growth of newly forming blood vessels derived from hemangiogenic stem cells within the mesenchymal core of villi. In a second stage, angiogenesis leads to the expansion and remodeling of the already existing vessels. At present, relatively little is known about the regulatory mechanisms of vasculogenesis and angiogenesis during very early placentation. Using placental villous tissues from days 22 to 48 of pregnancy, we analyzed the spatial and temporal expression of Tie-1 and Tie-2 in parallel to vascular maturation in the human placenta. In immunohistochemistry both receptors, Tie-1 and Tie-2 show a cell and villous type specific expression during this early phase of placental development. Especially, cytotrophoblast and hemangiogenic cell cords in mesenchymal villi and Hofbauer cells in immature intermediate villi have the strongest immunoreactivities. Western blot analysis showed that no significant changes were detected for Tie-1 and Tie-2 as pregnancy advanced. Moreover, phospho-Tie-2 levels did not change significantly in parallel to pregnancy ages. We conclude that both receptors are involved in angiogenesis as well as vascular modulation of early vessels. Due to their spatial distribution we speculate on an additional role in regulation of villous and extravillous trophoblastic behavior.


Subject(s)
Chorionic Villi/metabolism , Neovascularization, Physiologic/physiology , Placental Circulation/physiology , Pregnancy Trimester, First , Receptor, TIE-1/metabolism , Receptor, TIE-2/metabolism , Trophoblasts/metabolism , Adult , Chorionic Villi/blood supply , Chorionic Villi/embryology , Female , Gestational Age , Humans , Pregnancy
8.
Placenta ; 27(6-7): 535-9, 2006.
Article in English | MEDLINE | ID: mdl-16029887

ABSTRACT

Development of blood vessels takes place via two subsequent processes, vasculogenesis and angiogenesis. During vasculogenesis, formation of first blood vessels is achieved by differentiation of hemangiogenic stem cells from pluripotent mesenchymal cells, while during angiogenesis new blood vessels form from already existing vessels. The combination of our data with those from the literature leads us to depict the chronological steps of cell differentiation in the mesenchymal core of placental villi during vasculogenesis and angiogenesis. This current opinion will focus on the temporal and spatial expression of VEGF and its receptors VEGFR-1 and VEGFR-2, and the angiopoietin receptors Tie-1 and Tie-2 in parallel to vascular maturation in human placental villi during very early stages of placental development. There is evidence that the interplay of a variety of growth factors secreted from different cell types during development is needed to trigger as well as maintain placental vasculogenesis and angiogenesis.


Subject(s)
Neovascularization, Physiologic/physiology , Placenta/blood supply , Placenta/embryology , Adult , Biomarkers/metabolism , Cell Differentiation , Female , Humans , Placenta/cytology , Pregnancy , Vascular Endothelial Growth Factor A/metabolism
9.
Thorax ; 60(7): 576-81, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15994266

ABSTRACT

BACKGROUND: Vasodilator stimulated phosphoprotein (VASP) is associated with focal adhesions and is thought to have an important role in actin filament assembly and cell motility. We hypothesise that an increase in the expression of VASP is involved in the progression and invasion of lung adenocarcinomas in parallel to tumour progression. A study was undertaken to analyse VASP expression in normal lung tissue and lung adenocarcinomas. METHODS: Human lung tissues with adenocarcinomas (n = 26) were used. Normal lung tissue specimens (n = 14) were taken from areas a standard distance (3 cm) from resected adenocarcinomas of patients who underwent surgical lung resection. Adenocarcinomas were classified according to pathological staging and histopathological grades. Tissues were stained for VASP using immunohistochemistry. RESULTS: Normal lung pneumocytes showed no VASP expression while alveolar macrophages had the strongest immunoreactivity for VASP. Bronchial epithelium (surface epithelium, goblet cells) and bronchial gland cells had a very weak immunoreactivity for VASP. Adenocarcinomas had significantly greater VASP expression than normal epithelium (p < 0.001). Moreover, VASP expression in adenocarcinomas increased significantly with more advanced tumour stage (p < 0.001). CONCLUSIONS: The spatial and differential expression of VASP in normal lung tissue and lung adenocarcinomas suggests that it is likely to be involved in the differentiation of normal lung cells to adenocarcinomas. The significant increase in the expression of VASP in adenocarcinomas in parallel to pathological staging suggests that it may regulate the invasive behaviour of lung adenocarcinomas as adenocarcinoma invasion is increased in more advanced tumours.


Subject(s)
Adenocarcinoma/metabolism , Cell Adhesion Molecules/metabolism , Lung Neoplasms/metabolism , Lung/metabolism , Phosphoproteins/metabolism , Aged , Disease Progression , Female , Humans , Immunohistochemistry , Male , Microfilament Proteins , Middle Aged
10.
Placenta ; 25(6): 560-72, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15135240

ABSTRACT

Vascularization within the human placenta is the result of the de novo formation of vessels derived from pluripotent precursor cells in the mesenchymal core of the villi. Vascularization of placental villi starts at around day 21 post conception (p.c.) with a four somite embryo. At this stage progenitors of haemangiogenic cells differentiate to form first vessels. These progenitor cells are thought to be directly derived from mesenchymal cells rather than originating from fetal blood cells. We investigated the relation between differentiation of stromal cells towards endothelial cells and vascular structures and the expression pattern of the respective growth factors. Using transmission electron microscopy and immunohistochemistry (for VEGF, Flt-1, Flk-1, CD14, CD34, and CD68) the development of placental vasculogenesis during very early stages of pregnancy (days 22-48 p.c.) was studied. We found that VEGF is strongly expressed in villous cytotrophoblast cells and subsequently in Hofbauer cells while its receptors Flt-1 and Flk-1 are found on vasculogenic and angiogenic precursor cells. The developmental expression and secretion of VEGF suggests its involvement in recruitment, maintenance and formation of first angiogenic cells and vessels. Interactions between VEGF and Flk-1 and Flt-1 may regulate placental vasculogenesis and angiogenesis in a paracrine and autocrine manner. The sequential expression of growth factors in different cell types may point to the fact that placental vasculogenesis and angiogenesis are clearly distinct events.


Subject(s)
Neovascularization, Physiologic/physiology , Placenta/blood supply , Placenta/chemistry , Receptors, Vascular Endothelial Growth Factor/analysis , Vascular Endothelial Growth Factor A/analysis , Antigens, CD/analysis , Antigens, CD34/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Blotting, Western , Cell Differentiation , Chorionic Villi/blood supply , Chorionic Villi/chemistry , Chorionic Villi/ultrastructure , Endothelial Cells/cytology , Female , Humans , Immunohistochemistry , Lipopolysaccharide Receptors/analysis , Microscopy, Electron , Placenta/cytology , Pregnancy , Stromal Cells/cytology , Trophoblasts/chemistry , Vascular Endothelial Growth Factor Receptor-1/analysis , Vascular Endothelial Growth Factor Receptor-2/analysis
11.
Mol Hum Reprod ; 10(6): 393-8, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15073283

ABSTRACT

When Fas ligand (FasL) interacts with the Fas receptor, it induces apoptosis through autocrine and/or paracrine signalling. Vascular endothelial growth factor (VEGF) is a potent mitogenic cytokine. VEGF plays a role during remodelling of the endometrium following menstruation. We hypothesized that, by regulating FasL expression, VEGF may play a role in endometrial stromal cell survival by decreasing autocrine apoptotic signalling. We aimed to determine the expression of FasL in cultured endometrial stromal cells and its modulation by VEGF. VEGF induced a decrease in both FasL-positive cell number and FasL intensity as determined by immunocytochemistry and western blot respectively (P < 0.05). These effects of VEGF were observed in a concentration-dependent manner (10-42%; P < 0.05). Anti-VEGF neutralizing antibody alone resulted in an increase in the FasL expression. When combined with VEGF, anti-VEGF reversed the VEGF-induced decrease in FasL level up to 100% (P < 0.05). In addition, western blot analysis showed that FasL expression in endometrial stromal cells demonstrated a cyclic change every 12 h during 48 h of incubation. These results suggest that down-regulation of FasL by VEGF may affect endometrial stromal cell survival in an autocrine or paracrine manner. The decrease in FasL level may be due to a stimulation of its degradation. Our results show that FasL in endometrial stromal cells in culture has a cyclic expression model, suggesting that there may be a regulation at the translation level.


Subject(s)
Endometrium/cytology , Gene Expression Regulation, Developmental , Membrane Glycoproteins/metabolism , Stromal Cells/physiology , Vascular Endothelial Growth Factor A/metabolism , Adult , Animals , Antibodies/metabolism , Apoptosis/physiology , Cell Proliferation , Cell Survival , Cells, Cultured , Endometrium/metabolism , Fas Ligand Protein , Female , Humans , Membrane Glycoproteins/genetics , Middle Aged , RNA, Messenger/metabolism , Stromal Cells/cytology
12.
Ann N Y Acad Sci ; 1034: 50-63, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15731299

ABSTRACT

The immune system is a complex entity designed to eliminate foreign intruding antigens and is influenced by and, in turn, influences the function of the reproductive system. Despite the widespread associations between immunology and reproductive medicine, the study of system interactions remains in its infancy. Many diverse facts are accumulating, and pieces of the puzzle are becoming available to provide a clearer picture. In this review article, we focus on the interactions between endocrine and immune systems in the human endometrium. Understanding the molecular pathways in endocrine-immune interactions in the human endometrium is crucial to understand events such as menstrual bleeding, tissue repair and regeneration, inflammation, angiogenesis, blastocyst implantation, and progression of pregnancy. These events require a balanced regulation of endometrial differentiation, proliferation, cell survival, leukocyte recruitment, apoptosis, and angiogenesis by sex steroids. In this review, we first outline the role of survival factors such as phosphoinositol 3-kinase/protein kinase B, PTEN, NFkappaB, and apoptotic molecules (Fas-FasL, Bcl-2). We then discuss their regulation by estrogen and progesterone in the endometrium. We present evidence for direct and/or indirect roles of steroid hormones on the expression of chemotactic cytokines (interleukin-8 and monocyte chemotactic protein-1) and on the survival versus apoptosis of resident endometrial cells (stromal, epithelial, and endothelial cells) and nonresident cells (leukocytes).


Subject(s)
Endocrine System/physiology , Endometrium/immunology , Hormones/physiology , Immune System/physiology , Female , Humans
13.
Reproduction ; 125(1): 75-84, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12622698

ABSTRACT

The significance of insulin, insulin-like growth factor I (IGF-I) and glucocorticoids to the early mammalian embryo is clear in that they are key regulators of both mitogenic and metabolic effects during development. In the present study, the temporal sequence of expression of the respective receptor proteins was investigated for the first time in the developing rat utero-embryonic unit between conception and day 12 of gestation using immunocytochemistry. Insulin, IGF-I and glucocorticoid receptor were expressed in embryonic tissues after the start of implantation, and were co-localized in the primary ectoderm, extraembryonic ectoderm as well as in the ectoplacental cone. The parietal endoderm was devoid of glucocorticoid receptor staining, whereas IGF-I receptor was absent in visceral endoderm. After completion of basic organogenesis, the neural tube, notochord, otic placode, Wolffian duct, mesonephros and intestinal tube expressed insulin, IGF-I and glucocorticoid receptor. The glucocorticoid receptor was not expressed in heart tube and dorsal aortae. Considerable amounts of insulin receptor were detected in trophoblast-derived giant cells. In the uterus, luminal epithelium, endometrial stromal and myometrial smooth muscle cells immunoreacted with antisera against insulin, IGF-I and glucocorticoid receptor. Endometrial glands remained negative for the glucocorticoid receptor throughout the gestational period investigated. Uterine hormone receptor expression reached a peak at days 4 and 5 of gestation in endometrial stromal cells and decidua, respectively. In conclusion, the demonstrated ontogenetic pattern of insulin, IGF-I and glucocorticoid receptor expression indicates the potential sites of biological action of the respective ligands, providing supportive evidence for their critical importance during the course of embryogenesis in rats.


Subject(s)
Embryo, Mammalian/chemistry , Insulin-Like Growth Factor I/analysis , Insulin/analysis , Receptors, Glucocorticoid/analysis , Uterus/chemistry , Animals , Decidua/chemistry , Embryo Implantation/physiology , Endometrium/chemistry , Female , Gestational Age , Giant Cells/chemistry , Immunohistochemistry/methods , Organogenesis/physiology , Pregnancy , Rats , Rats, Wistar , Trophoblasts/cytology
14.
Placenta ; 23(8-9): 672-84, 2002.
Article in English | MEDLINE | ID: mdl-12361686

ABSTRACT

The differentiation of human endometrial epithelium is a dynamic event that occurs throughout the menstrual cycle and early pregnancy. The structural transformation and differentiation of human uterine luminal and glandular epithelium of early human pregnancy (n=14) was investigated ultrastructurally and immunohistochemically using antibodies against cytokeratin (CT), endothelial marker CD31, Fas, and proliferating cell nuclear antigen (PCNA). Ultrastructurally, luminal epithelial cells showed distinctive euchromatic nuclei with prominent nucleoli and relatively loose cell membranes in all poles (apical to basal). Subcellular components were easily recognized in luminal epithelium except in degenerating cells. Mainly two cell types, dark and clear cells, formed the glandular epithelium. In the early gestation period, microvilli were abundant on the apical and apico-lateral poles of these cells. Only a few cytoplasmic projections were observed in dark cells. Numerous cilia were observed on the apical pole of some clear cells, located at the adluminal segment. In contrast, dark cells lacked cilia, nuclear channels, or giant mitochondrial profiles. Glycogen synthesis and apocrine secretion were recognizable for several days during early gestation. The apocrine secretory activity differed among dark cells of the glandular epithelium. The immunoreactivity of PCNA and Fas, and ultrastructural observations in the glandular epithelium suggest that, even in different segments of the same gland, epithelial cells do not regress during early gestation, but proliferate, perhaps representing a resistance against trophoblastic invasion. These morphological and molecular changes suggest that both luminal and glandular epithelium may play an important role in cellular defense and limitation for trophoblastic invasion during early pregnancy since plasma membrane alterations of the surface epithelium take place at the apical, basal and lateral poles compared to early secretory phase endometrial cells. Besides glandular epithelium may be consequently responsible for uterine secretions, which may be critical for early embryo development.


Subject(s)
Cell Differentiation , Decidua/cytology , Decidua/metabolism , Embryo Implantation/physiology , Adult , Apoptosis/physiology , Biomarkers , Cell Division/physiology , Epithelium/metabolism , Epithelium/ultrastructure , Female , Fluorescent Antibody Technique, Indirect , Gestational Age , Humans , Immunoenzyme Techniques , Keratins/metabolism , Microscopy, Electron , Microvilli/ultrastructure , Pregnancy , Proliferating Cell Nuclear Antigen/metabolism , fas Receptor/metabolism
15.
Biol Reprod ; 65(4): 979-85, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11566716

ABSTRACT

Implantation involves a complex set of events, including apoptosis in endometrial cells. Apoptosis in human endometrium coincides with the implantation window, suggesting a potential role for steroid hormones in its regulation. Fas ligand (FasL) is one of the mediators of apoptosis in differentiated cells and in embryonic development. Interaction of FasL with its receptor, Fas, induces apoptosis through autocrine and paracrine signaling. We hypothesized that FasL expression in human endometrium is cycle-dependent and that sex steroid hormones regulate FasL expression. We first studied menstrual cycle-dependent expression of FasL in human endometrium by immunohistochemistry in 24 samples. We then investigated the in vitro regulation of FasL expression by ovarian steroid hormones. Throughout the menstrual cycle immunohistochemical staining intensity was stronger in the functional layer of endometrium than it was in the basal layer. FasL immunoreactivity increased gradually through the mid- and late-proliferative phases in both endometrial stromal and glandular cells. Strong FasL expression was observed throughout the late-proliferative and secretory phases. Semiquantitative reverse transcription-polymerase chain reaction analysis in cultured endometrial glandular cells demonstrated that estradiol and progesterone stimulate FasL mRNA expression. Western blot analysis in endometrial glandular and stromal cells in culture revealed that estradiol alone and in combination with progesterone up-regulated FasL protein expression. These results suggest that estradiol and progesterone may have a role in the regulation of maternal immunotolerance for the implantation of a semiallograft embryo by inducing FasL expression. We speculate that increased FasL expression may mediate the apoptosis of endometrial cells and thus may play a role in trophoblast invasion.


Subject(s)
Endometrium/metabolism , Estradiol/pharmacology , Gene Expression Regulation/drug effects , Membrane Glycoproteins/genetics , Progesterone/pharmacology , Adult , Apoptosis , Blotting, Western , Cells, Cultured , Endometrium/chemistry , Fas Ligand Protein , Female , Humans , Immunohistochemistry , Membrane Glycoproteins/analysis , Menstrual Cycle/physiology , Middle Aged , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/chemistry , Stromal Cells/metabolism
16.
Menopause ; 8(4): 296-301, 2001.
Article in English | MEDLINE | ID: mdl-11449089

ABSTRACT

OBJECTIVE: The protective effect of estrogen against early atherosclerosis in animal models is well documented, but the mechanisms responsible for this effect are not well understood. The earliest recognizable event in the pathogenesis of atherosclerosis is an increased recruitment of macrophages into the arterial subendothelium. Macrophages first play a protective role by removing low-density lipoproteins, but when the cholesterol is in excess, macrophages are converted into foam cells and form atheromas. Recent human and animal data indicate that the recruitment of macrophages to the arterial wall is mediated by monocyte chemotactic protein-1 (MCP-1). We hypothesized that one of the mechanisms of estrogen's protective effect against atherosclerosis may be the down-regulation of MCP-1 expression in the arterial wall. DESIGN: Human coronary artery smooth muscle cells were replicated to confluence in smooth muscle cell basal medium supplemented with growth factors and 5% fetal bovine serum. Before each experiment, cells were incubated for 24 h with phenol red-free medium containing 5% charcoal-stripped calf serum, and then they were treated with various concentrations of 17beta-estradiol as well as selective estrogen receptor (ER) modulators, raloxifene and tamoxifen. MCP-1 messenger ribonucleic acid (mRNA) levels were quantified by Northern blots. MCP-1 protein was quantified using an enzyme-linked immunosorbent assay. ER expression was evaluated by reverse transcriptase-polymerase chain reaction. RESULTS: Human coronary artery smooth muscle cells expressed MCP-1 mRNA and produced MCP-1 protein. Estradiol induced up to 40% inhibition in mRNA expression at concentrations 10-9 M and higher. Raloxifene and tamoxifen also resulted in an inhibition, but the inhibition was less than when induced by estradiol. Estradiol also inhibited the MCP-1 protein production in a concentration-dependent manner (p < 0.05). Coronary smooth muscle cells expressed both ERalpha and ERbeta. CONCLUSION: Our findings suggest that one of the mechanisms by which estrogen prevents atherosclerosis is by down-regulating MCP-1 expression, thus decreasing macrophage recruitment to the arterial wall.


Subject(s)
Arteriosclerosis/drug therapy , Arteriosclerosis/etiology , Chemokine CCL2/immunology , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Estradiol/immunology , Estradiol/therapeutic use , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/immunology , Animals , Arteriosclerosis/immunology , Blotting, Northern , Cells, Cultured/drug effects , Chemokine CCL2/analysis , Disease Models, Animal , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Endothelium, Vascular/cytology , Estradiol/pharmacology , Female , Humans , Muscle, Smooth, Vascular/cytology , Raloxifene Hydrochloride/immunology , Raloxifene Hydrochloride/pharmacology , Raloxifene Hydrochloride/therapeutic use , Selective Estrogen Receptor Modulators/immunology , Selective Estrogen Receptor Modulators/pharmacology , Selective Estrogen Receptor Modulators/therapeutic use , Tamoxifen/immunology , Tamoxifen/pharmacology , Tamoxifen/therapeutic use
17.
Fertil Steril ; 75(6): 1172-6, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11384645

ABSTRACT

OBJECTIVES: To determine whether aminopeptidase N (APN) regulates the cycle-dependent bioavailability of interleukin-8 (IL-8) in the endometrium. DESIGN: Prospective study. SETTING: University medical center. PATIENT(S): Women without endometrial pathology from the proliferative (n = 25) or secretory (n = 18) phase of the menstrual cycle. INTERVENTION(S): We first immunolocalized APN in the endometrium using an anti-APN antibody. We then determined the regulation of APN kinetic activity by sex steroids in endometrial stromal cell cultures. MAIN OUTCOME MEASURE(S): Expression of APN in human endometrium throughout the menstrual cycle. Regulation of APN activity by estradiol and progesterone in cultured endometrial stromal cells. RESULT(S): Immunohistochemistry of endometrial sections revealed staining of endometrial stroma throughout the menstrual cycle. There was no detectable staining in glandular cells. The expression of APN as detected by immunohistochemistry was significantly lower in the early proliferative phase. In cultured cells, estradiol inhibited APN activity in a concentration-dependent manner. Progesterone did not have a significant effect. CONCLUSION(S): Stromal localization of APN in endometrium may explain the epithelial rather than stromal presence of IL-8 in vivo. Decreased expression of APN may increase IL-8 bioavailability thus contributing to angiogenesis and polymorphonuclear leukocyte chemotaxis in early proliferative phase.


Subject(s)
CD13 Antigens/metabolism , Endometrium/metabolism , Estrogens/physiology , Cells, Cultured , Endometrium/cytology , Endometrium/drug effects , Estradiol/pharmacology , Female , Humans , Immunohistochemistry , Interleukin-8/pharmacology , Menstrual Cycle/physiology , Progesterone/pharmacology , Prospective Studies , Tissue Distribution
18.
Biol Trace Elem Res ; 77(1): 65-81, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11097472

ABSTRACT

The aim of this study was to determine the changes in the function and fine structure of the gastric mucosa following exposure to high cadmium (Cd) for 30 d in rats. In the present study, control animals were fed with normal food and tap water and the remaining animals received Cd (15 ppm CdCl2) in drinking water for the same period. Receiving Cd for 30 d increased the mean blood (p < 0.01) and mucosa (p < 0.001) Cd levels, while decreased mucus thickness, mucin content (p < 0.01) significantly. Basal acid output fell significantly (p < 0.01). Light and electron microscopic examination revealed the following: (1) Cd decreases the mean number of surface mucous, isthmic-neck, parietal cells (p < 0.05) and chief cells (p < 0.001) per unit from the control value and (2) in some cells of zymogenic unit, the Cd-induced alterations were characterized with dilated Golgi cisternae, focal enlarged endoplasmic reticulum, broken tubulovesicles, degenerated mitochondria, dense nuclei, as well as lysosomal structures. We concluded that Cd augments the elimination rate of zymogenic unit's cells by increasing the alteration rate, and the reduced basal acid output, mucin content, and mucus thickness can be explained easily with the loss of zymogenic unit's cell population.


Subject(s)
Cadmium/toxicity , Gastric Mucosa/drug effects , Animals , Cadmium/metabolism , Free Radicals , Gastric Acid/metabolism , Gastric Mucosa/pathology , Gastric Mucosa/ultrastructure , Male , Microscopy, Electron , Mucins/analysis , Rats
19.
Biol Trace Elem Res ; 74(2): 153-70, 2000 May.
Article in English | MEDLINE | ID: mdl-11051589

ABSTRACT

The aim of this study was to determine the cadmium (Cd)-induced functional and structural changes in gastric parietal cells of male rats exposed to high Cd for 30 d. In the present study, control animals were fed with normal food and tap water; the remaining animals received Cd (15 ppm CdCl2) in drinking water for the same period. Receiving Cd for 30 d increased the mean blood Cd level, the mean tissue Cd content, and the mean blood pressure (p < 0.01, p < 0.001, p < 0.01, respectively). The basal acid output fell; however, the increases in stimulated acid output were not statistically significant. Light and electron microscopic examination revealed respectively that (1) Cd decreases the mean parietal cell number per unit from the control value of 23.46 +/- 3.84 to 19.46 +/- 2.12 (p < 0.05) and it affected preferentially the cells located at the distal half of the zymogenic unit and (2) in parietal cells, the Cd-induced alterations were characterized with swollen canalicular profiles, broken-down tubulovesicles, or degenerated mitochondria. We concluded that Cd augments the elimination rate of parietal cells by increasing the alteration rate and reduced basal acid output can be explained easily with the loss of parietal cell population.


Subject(s)
Cadmium/toxicity , Parietal Cells, Gastric/drug effects , Parietal Cells, Gastric/ultrastructure , Animals , Appetite/drug effects , Blood Pressure/drug effects , Cadmium/pharmacokinetics , Cell Count , Gastric Acid/metabolism , Gastric Mucosa/cytology , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Male , Microscopy, Electron , Parietal Cells, Gastric/physiology , Rats , Tissue Distribution , Vagus Nerve/physiology , Weight Gain/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...