Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 5: 13237, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26272398

ABSTRACT

The placenta forms the interface between the maternal and fetal circulation and is critical for the establishment of a healthy pregnancy. Specialized trophoblast cells derived from the embryonic trophectoderm play a pivotal role in the establishment of the placenta. Leukemia inhibitory factor (LIF) is one of the predominant cytokines present in the placenta during early pregnancy. LIF has been shown to regulate trophoblast adhesion and invasion in vitro, however its precise role in vivo is unknown. We hypothesized that LIF would be required for normal placental development in mice. LIF and LIFRα were immunolocalized to placental trophoblasts and fetal vessels in mouse implantation sites during mid-gestation. Temporally blocking LIF action during specific periods of placental development via intraperitoneal administration of our specific LIFRα antagonist, PEGLA, resulted in abnormal placental trophoblast and vascular morphology and reduced activated STAT3 but not ERK. Numerous genes regulating angiogenesis and oxidative stress were altered in the placenta in response to LIF inhibition. Pregnancy viability was also significantly compromised in PEGLA treated mice. Our data suggest that LIF plays an important role in placentation in vivo and the maintenance of healthy pregnancy.


Subject(s)
Abortion, Spontaneous/physiopathology , Leukemia Inhibitory Factor/metabolism , Placenta Diseases/physiopathology , Placentation , Animals , Female , Leukemia Inhibitory Factor/antagonists & inhibitors , Leukemia Inhibitory Factor/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Polyethylene Glycols/pharmacology , Pregnancy , Receptors, OSM-LIF/antagonists & inhibitors
2.
J Microbiol Biotechnol ; 25(8): 1307-14, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25876596

ABSTRACT

Leukemia inhibitory factor (LIF) is a member of the IL-6 cytokine family, having pleiotropic actions such as maintaining stem cell pluripotency and enabling blastocyst implantation. Because the action of LIF is mediated by a ligand-receptor interaction with the LIF receptor (LIF-R), an antagonist for LIF-R has been developed to inhibit LIF-induced signaling. In this study, we present a novel method for the production and purification of an antagonist to human LIF-R (hLA). His-tagged hLA was expressed in E. coli, and simple purification methods without any endopeptidase cleavage were designed. In addition, we determined the optimal temperature conditions for enhancing the production of soluble hLA. Finally, the bioactivity of His-tagged hLA was examined using STAT3 phosphorylation and receptivity of human endometrial ECC-1 cells. Our strategy provides a rapid and efficient method to produce biologically active recombinant hLA.


Subject(s)
Chromatography/methods , Escherichia coli/genetics , Gene Expression , Receptors, OSM-LIF/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Cell Line , Escherichia coli/metabolism , Humans , Recombinant Proteins/metabolism
3.
Reproduction ; 138(5): 827-36, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19635738

ABSTRACT

Uteri of Lif null mice do not support embryo implantation. Since deletion of some genes often prevents the survival of null mice to adulthood, we have used a proven inhibitor of leukaemia inhibitory factor (LIF) signalling to identify the precise window of time during which LIF is required in vivo, and assessed the cellular expression of several LIF-associated targets. On day 4 of pregnancy, mice were injected with hLIF-05 (inhibitor) into the uterine lumen, with corresponding volumes of PBS (vehicle) injected into the contralateral horn. On days 5 and 6, the number of implantation sites was recorded and the uteri processed for immunohistochemistry. Blockade of LIF on day 4 reduced embryo implantation by 50% (P

Subject(s)
Embryo Implantation/drug effects , Leukemia Inhibitory Factor/pharmacology , Receptors, OSM-LIF/antagonists & inhibitors , Administration, Intravaginal , Amphiregulin , Animals , Cyclooxygenase 2/metabolism , Desmin/metabolism , EGF Family of Proteins , Embryo Implantation/physiology , Female , Gestational Age , Glycoproteins/metabolism , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/metabolism , Leukemia Inhibitory Factor/administration & dosage , Leukemia Inhibitory Factor/antagonists & inhibitors , Leukemia Inhibitory Factor/metabolism , Leukemia Inhibitory Factor/physiology , Mice , Phosphorylation , Pregnancy , Receptors, OSM-LIF/metabolism , STAT3 Transcription Factor/metabolism , Time Factors , Uterus/anatomy & histology , Uterus/drug effects , Uterus/metabolism
4.
Cardiovasc Res ; 75(3): 536-45, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17559824

ABSTRACT

OBJECTIVES: Cardiomyocyte loss is involved in the transition from compensatory left ventricular hypertrophy (LVH) to heart failure (HF). Our aim was to investigate the status of the leukaemia inhibitory factor receptor (LIFR)/gp130 survival pathway and its cytoprotective activity in intact cardiac tissue and in cardiomyocytes obtained from adult spontaneously hypertensive rats (SHR) with LVH (non-failing SHR) and from aged SHR with overt HF (failing SHR). METHODS: Cardiac morphometry was assayed by planimetry in an image analysis system. mRNA and protein expression were quantified by real time RT-PCR and Western blotting. Receptors were localized by immunocytochemistry. Trypan blue staining, TUNEL, and MTT cell viability assays were employed to study the cytoprotective activity of cardiotrophin-1 (CT-1) in isolated caridomyocytes. RESULTS: Compared to non-failing SHR, failing SHR exhibited enhanced myocardial cell death (p<0.01) demonstrated by the increase in Bax/Bcl-2 ratio, caspase-3 activation and poly (ADP-ribose) polymerase (PARP) fragmentation. Failing SHR had a 7-fold diminished expression (p<0.01) of LIFR, no changes in gp130, and 1.6-fold increased myocardial expression (p<0.01) of CT-1. In cardiomyocytes isolated from non-failing SHR, recombinant CT-1 inhibited apoptotic and non-apoptotic cell death induced by angiotensin II or hydrogen peroxide. LIFR protein was entirely absent in cardiomyocytes isolated from failing SHR, which were resistant to the cytoprotective effects of CT-1. Finally, stimulation of non-failing SHR cardiomyocytes with angiotensin II, aldosterone, norepinephrine or endothelin-1 significantly decreased (p<0.01) LIFR expression. CONCLUSIONS: These data suggest that loss of CT-1-dependent survival mechanisms may contribute to the increase of cell death associated with HF in SHR. Neurohumoral activation may contribute to this alteration via suppression of LIFR.


Subject(s)
Cytokines/physiology , Heart Failure/metabolism , Myocardium/metabolism , Receptors, OSM-LIF/antagonists & inhibitors , Aldosterone/pharmacology , Angiotensin II/pharmacology , Animals , Apoptosis , Cells, Cultured , Cytokines/pharmacology , Cytoprotection , Endothelin-1/pharmacology , In Situ Nick-End Labeling , Leukemia Inhibitory Factor/metabolism , Myocytes, Cardiac/metabolism , Norepinephrine/pharmacology , RNA, Messenger/analysis , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, OSM-LIF/analysis , Receptors, OSM-LIF/metabolism
5.
J Interferon Cytokine Res ; 27(4): 281-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17477816

ABSTRACT

Leukemia inhibitory factor (LIF) and oncostatin M (OSM) are found in appreciable concentrations in synovial fluid from patients with rheumatoid arthritis (RA) but not osteoarthritis. Accordingly, both are potential therapeutic targets in inflammatory diseases of the joints. Several LIF antagonists have been developed. They have the capacity to inhibit the biologic activities of not only LIF but also other interleukin-6 (IL-6) subfamily cytokines, including OSM. Both LIF and OSM share the same receptor, which is part of a cytokine receptor super family in which the glycoprotein 130 (gp130) subunit is a common constituent. The aim of this study was to evaluate the antagonistic potentials of two LIF mutants, LIF05 and MH35-BD. Both are mutant forms of human LIF with reduced affinity for gp130 and greater LIF receptor (LIFR) binding affinity. The results, using Ba/F3 cell proliferation assay, acute-phase protein (haptoglobin) induction analysis in HepG2 human hepatoma cells, a porcine cartilage glycosaminoglycan release assessment for proteoglycan degradation, and a collagen release assay, show that these antagonists inhibit relevant LIF, OSM, and other IL-6 subfamily cytokines in vitro albeit with differential potencies and have, therefore, therapeutic potential for treatment of RA and perhaps other diseases.


Subject(s)
Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/therapy , Receptors, OSM-LIF/antagonists & inhibitors , Animals , Cell Line , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/immunology , Glycosaminoglycans/metabolism , Humans , Hydroxyproline/metabolism , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/immunology , Oncostatin M/immunology , Receptors, OSM-LIF/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Swine
6.
Cancer Res ; 66(22): 10891-901, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17108126

ABSTRACT

The interleukin-6 cytokine oncostatin M (OSM) induces potent growth-inhibitory and morphogenic responses in several different tumor cell types, highlighting the importance of OSM signaling mechanisms as targets for therapeutic intervention. The specific molecular pathways involved are not well understood, as OSM can signal through two separate heterodimeric receptor complexes, glycoprotein 130 (gp130)/leukemia inhibitory factor receptor (LIFR) alpha and gp130/OSM receptor beta (OSMRbeta). In this investigation, we used a LIFR antagonist to help resolve signaling responses and identify patterns of gene expression elicited by the different receptor complexes. OSM-induced biological effects on breast tumor-derived cell lines were specifically mediated through the gp130/OSMRbeta complex. Each cytokine tested exhibited differential signaling capability and manifested both shared and unique patterns of gene activation, emphasizing compositional differences in activator protein-1 transcription factor activity and expression. In particular, OSM strongly activated the c-Jun NH(2)-terminal kinase (JNK) serine/threonine kinase and downstream components, including activating transcription factor (ATF)/cyclic AMP-responsive element binding protein family member, ATF3. JNK/stress-activated protein kinase kinase inhibition abrogated cell morphogenesis induced by OSM, indicating an important role for this pathway in OSM specificity. These findings identify a core signaling/transcriptional mechanism specific to the OSMRbeta in breast tumor cells.


Subject(s)
Breast Neoplasms/pathology , Leukemia Inhibitory Factor Receptor alpha Subunit/antagonists & inhibitors , Oncostatin M Receptor beta Subunit/metabolism , Oncostatin M/pharmacology , Receptors, OSM-LIF/antagonists & inhibitors , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Drug Interactions , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-6/pharmacology , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Oncostatin M Receptor beta Subunit/antagonists & inhibitors , Receptors, OSM-LIF/metabolism , STAT Transcription Factors/metabolism , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...