Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 31(5): 737-48, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18775332

ABSTRACT

gp130 is a shared receptor for at least nine cytokines and can signal either as a homodimer or as a heterodimer with Leukemia Inhibitory Factor Receptor (LIF-R). Here, we biophysically and structurally characterize the full-length, transmembrane form of a quaternary cytokine receptor complex consisting of gp130, LIF-R, the cytokine Ciliary Neurotrophic Factor (CNTF), and its alpha receptor (CNTF-Ralpha). Thermodynamic analysis indicates that, unlike the cooperative assembly of the symmetric gp130/Interleukin-6/IL-6Ralpha hexameric complex, CNTF/CNTF-Ralpha heterodimerizes gp130 and LIF-R via noncooperative energetics to form an asymmetric 1:1:1:1 complex. Single particle electron microscopic analysis of the full-length gp130/LIF-R/CNTF-Ralpha/CNTF quaternary complex elucidates an asymmetric structural arrangement, in which the receptor extracellular and transmembrane segments join as a continuous, rigid unit, poised to sensitively transduce ligand engagement to the membrane-proximal intracellular signaling regions. These studies also enumerate the organizing principles for assembly of the "tall" class of gp130 family cytokine receptor complexes including LIF, IL-27, IL-12, and others.


Subject(s)
Cytokine Receptor gp130/chemistry , Multiprotein Complexes/chemistry , Protein Structure, Quaternary , Receptors, OSM-LIF/chemistry , Signal Transduction/physiology , Animals , Ciliary Neurotrophic Factor/chemistry , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/metabolism , Crystallography, X-Ray , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes/ultrastructure , Receptor, Ciliary Neurotrophic Factor/chemistry , Receptor, Ciliary Neurotrophic Factor/genetics , Receptor, Ciliary Neurotrophic Factor/metabolism , Receptors, OSM-LIF/genetics , Receptors, OSM-LIF/metabolism , Thermodynamics
2.
J Biol Chem ; 289(26): 18442-50, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24802752

ABSTRACT

Ciliary neurotrophic factor (CNTF) is a neurotrophic factor with therapeutic potential for neurodegenerative diseases. Moreover, therapeutic application of CNTF reduced body weight in mice and humans. CNTF binds to high or low affinity receptor complexes consisting of CNTFR·gp130·LIFR or IL-6R·gp130·LIFR, respectively. Clinical studies of the CNTF derivative Axokine revealed intolerance at higher concentrations, which may rely on the low-affinity binding of CNTF to the IL-6R. Here, we aimed to generate a CNTFR-selective CNTF variant (CV). CV-1 contained the single amino acid exchange R28E. Arg(28) is in close proximity to the CNTFR binding site. Using molecular modeling, we hypothesized that Arg(28) might contribute to IL-6R/CNTFR plasticity of CNTF. CV-2 to CV-5 were generated by transferring parts of the CNTFR-binding site from cardiotrophin-like cytokine to CNTF. Cardiotrophin-like cytokine selectively signals via the CNTFR·gp130·LIFR complex, albeit with a much lower affinity compared with CNTF. As shown by immunoprecipitation, all CNTF variants retained the ability to bind to CNTFR. CV-1, CV-2, and CV-5, however, lost the ability to bind to IL-6R. Although all variants induced cytokine-dependent cellular proliferation and STAT3 phosphorylation via CNTFR·gp130·LIFR, only CV-3 induced STAT3 phosphorylation via IL-6R·gp130·LIFR. Quantification of CNTF-dependent proliferation of CNTFR·gp130·LIFR expressing cells indicated that only CV-1 was as biologically active as CNTF. Thus, the CNTFR-selective CV-1 will allow discriminating between CNTFR- and IL-6R-mediated effects in vivo.


Subject(s)
Amino Acid Substitution , Ciliary Neurotrophic Factor/genetics , Cytokine Receptor gp130/metabolism , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Receptor, Ciliary Neurotrophic Factor/metabolism , Receptors, Interleukin-6/metabolism , Ciliary Neurotrophic Factor/metabolism , Cytokine Receptor gp130/genetics , Humans , Interleukin-6/metabolism , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Mutation, Missense , Phosphorylation , Receptor, Ciliary Neurotrophic Factor/genetics , Receptors, Interleukin-6/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction
3.
J Neurosci ; 33(3): 1241-58, 2013 Jan 16.
Article in English | MEDLINE | ID: mdl-23325260

ABSTRACT

Appropriately targeted manipulation of endogenous neural stem progenitor (NSP) cells may contribute to therapies for trauma, stroke, and neurodegenerative disease. A prerequisite to such therapies is a better understanding of the mechanisms regulating adult NSP cells in vivo. Indirect data suggest that endogenous ciliary neurotrophic factor (CNTF) receptor signaling may inhibit neuronal differentiation of NSP cells. We challenged subventricular zone (SVZ) cells in vivo with low concentrations of CNTF to anatomically characterize cells containing functional CNTF receptors. We found that type B "stem" cells are highly responsive, whereas type C "transit-amplifying" cells and type A neuroblasts are remarkably unresponsive, as are GFAP(+) astrocytes found outside the SVZ. CNTF was identified in a subset of type B cells that label with acute BrdU administration. Disruption of in vivo CNTF receptor signaling in SVZ NSP cells, with a "floxed" CNTF receptor α (CNTFRα) mouse line and a gene construct driving Cre recombinase (Cre) expression in NSP cells, led to increases in SVZ-associated neuroblasts and new olfactory bulb neurons, as well as a neuron subtype-specific, adult-onset increase in olfactory bulb neuron populations. Adult-onset receptor disruption in SVZ NSP cells with a recombinant adeno-associated virus (AAV-Cre) also led to increased neurogenesis. However, the maintenance of type B cell populations was apparently unaffected by the receptor disruption. Together, the data suggest that endogenous CNTF receptor signaling in type B stem cells inhibits adult neurogenesis, and further suggest that the regulation may occur in a neuron subtype-specific manner.


Subject(s)
Lateral Ventricles/physiology , Neurogenesis/physiology , Neurons/physiology , Prosencephalon/physiology , Receptor, Ciliary Neurotrophic Factor/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Ciliary Neurotrophic Factor/metabolism , Lateral Ventricles/cytology , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Olfactory Bulb/cytology , Olfactory Bulb/physiology , Receptor, Ciliary Neurotrophic Factor/genetics , Signal Transduction/physiology
4.
Dev Neurosci ; 36(5): 422-31, 2014.
Article in English | MEDLINE | ID: mdl-25171250

ABSTRACT

Neuroblastoma is a childhood cancer caused by the transformation of sympathoadrenal progenitors. By following the formation of tumors in homozygous TH-MYCN mice, an established mouse model of neuroblastoma, we were able to capture transformed cells prior to the formation of large, vascularized tumors in order to determine the responsiveness of cells to neurotrophic factors. We discovered that the ciliary neurotrophic factor (CNTF) receptor is abundantly expressed in tumor cells from these mice. Furthermore, CNTF - but not nerve growth factor, brain-derived nerve growth factor, neurotrophin 3, or glial cell line-derived neurotrophic factor - promoted neuronal differentiation and withdrawal from the cell cycle. Thus, the transformation of sympathoadrenal progenitors by MYCN overexpression differentially affects responsiveness to neurotrophic molecules.


Subject(s)
Abdominal Neoplasms/drug therapy , Cell Differentiation/drug effects , Ciliary Neurotrophic Factor/pharmacology , Neuroblastoma/drug therapy , Receptor, Ciliary Neurotrophic Factor/metabolism , Abdominal Neoplasms/metabolism , Animals , Cell Proliferation/drug effects , Ciliary Neurotrophic Factor/therapeutic use , Disease Models, Animal , Mice , Neuroblastoma/metabolism , Neurons/drug effects , Neurons/metabolism , Receptor, Ciliary Neurotrophic Factor/genetics
5.
Mol Hum Reprod ; 18(2): 88-95, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21965347

ABSTRACT

Fetal ovarian development and primordial follicle formation are imperative for adult fertility in the female. Data suggest the interleukin (IL)6-type cytokines, leukaemia inhibitory factor (LIF), IL6, oncostatin M (OSM) and ciliary neurotrophic factor (CNTF), are able to regulate the survival, proliferation and differentiation of fetal murine germ cells (GCs) in vivo and in vitro. We postulated that these factors may play a similar role during early human GC development and primordial follicle formation. To test this hypothesis, we have investigated the expression and regulation of IL6-type cytokines, using quantitative reverse transcription polymerase chain reaction and immunohistochemistry. Expression of transcripts encoding OSM increased significantly across the gestational range examined (8-20 weeks), while expression of IL6 increased specifically between the first (8-11 weeks) and early second (12-16 weeks) trimesters, co-incident with the initiation of meiosis. LIF and CNTF expression remained unchanged. Expression of the genes encoding the LIF and IL6 receptors, and their common signalling subunit gp130, was also found to be developmentally regulated, with expression increasing significantly with increasing gestation. LIF receptor and gp130 proteins localized exclusively to GCs, including oocytes in primordial follicles, indicating this cell type to be the sole target of IL6-type cytokine signalling in the human fetal ovary. These data establish that IL6-type cytokines and their receptors are expressed in the human fetal ovary and may directly influence GC development at multiple stages of maturation.


Subject(s)
Gene Expression Regulation, Developmental , Oocytes/metabolism , Ovarian Follicle/metabolism , RNA, Messenger/biosynthesis , Signal Transduction/genetics , Adult , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/metabolism , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Female , Fetus , Gestational Age , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/metabolism , Oncostatin M/genetics , Oncostatin M/metabolism , Oocytes/growth & development , Ovarian Follicle/growth & development , Pregnancy , Pregnancy Trimesters , Real-Time Polymerase Chain Reaction , Receptor, Ciliary Neurotrophic Factor/genetics , Receptor, Ciliary Neurotrophic Factor/metabolism , Receptors, Oncostatin M/genetics , Receptors, Oncostatin M/metabolism
6.
Cytokine ; 55(3): 325-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21715184

ABSTRACT

Cytokine-like factor 1 (CLF1) is a secreted receptor belonging to the interleukin-6 family of cytokines. CLF1 and its physiologic partner, cardiotrophin-like cytokine (CLC) are secreted as a heterodimer and engage the tripartite signaling complex of ciliary neurotrophic factor receptor (CNTFR), leukemia inhibitory factor (LIFR) and gp130. Ligation of this receptor complex leads to activation of the STAT3 and MAPK pathways and mediates survival pathways in neurons. Mutations in CLF1, CLC, or CNTFR in mice lead to the birth of mice that die on post-natal day 1 because of an inability to nurse. These animals exhibit significant decreases in the number of motor neurons in the facial nucleus and the spinal cord. CLF1 or CLC deficiency is associated with the development of the human cold-induced sweating syndromes. A growing body of research suggests that CLF1 expression may be associated with several post-natal disease processes. In this review, we summarize the current understanding of CLF1 expression and suggest future studies to understand the potentially important role of CLF1 in postnatal life and disease.


Subject(s)
Cytokines , Receptor, Ciliary Neurotrophic Factor/metabolism , Receptors, Cytokine , Receptors, Interleukin-6/metabolism , Animals , Cytokine Receptor gp130/metabolism , Cytokines/deficiency , Cytokines/genetics , Cytokines/metabolism , Humans , Interleukin-6/metabolism , Leukemia Inhibitory Factor , MAP Kinase Signaling System , Mice , Mitogen-Activated Protein Kinases/metabolism , Motor Neurons/metabolism , Receptor, Ciliary Neurotrophic Factor/genetics , Receptors, Cytokine/deficiency , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , STAT3 Transcription Factor/metabolism
7.
Calcif Tissue Int ; 86(3): 261-70, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20157807

ABSTRACT

Ciliary neurotrophic factor (CNTF) receptor (CNTFR) expression has been described in osteoblast-like cells, suggesting a role for CNTF in bone metabolism. When bound to CNTF, neuropoietin (NP), or cardiotrophin-like-cytokine (CLC), CNTFR forms a signaling complex with gp130 and the leukemia inhibitory factor receptor, which both play critical roles in bone cell biology. This study aimed to determine the role of CNTFR-signaling cytokines in bone. Immunohistochemistry detected CNTF in osteoblasts, osteocytes, osteoclasts, and proliferating chondrocytes. CNTFR mRNA was detected in primary calvarial osteoblasts and was upregulated during osteoblast differentiation. Treatment of osteoblasts with CNTF or CLC, but not NP, significantly inhibited mineralization and osterix mRNA levels. Twelve-week-old male CNTF ( -/- ) mice demonstrated reduced femoral length, cortical thickness, and periosteal circumference; but femoral trabecular bone mineral density (Tb.BMD) and tibial trabecular bone volume (BV/TV) were not significantly different from wild-type, indicating a unique role for CNTF in bone growth in male mice. In contrast, female CNTF ( -/- ) femora were of normal width, but femoral Tb.BMD, tibial BV/TV, trabecular number, and trabecular thickness were all increased. Female CNTF ( -/- ) tibiae also demonstrated high osteoblast number and mineral apposition rate compared to wild-type littermates, and this was intrinsic to the osteoblast lineage. CNTF is expressed locally in bone and plays a unique role in female mice as an inhibitor of trabecular bone formation and in male mice as a stimulus of cortical growth.


Subject(s)
Bone Remodeling/drug effects , Ciliary Neurotrophic Factor/metabolism , Cytokines/metabolism , Osteogenesis/physiology , Receptor, Ciliary Neurotrophic Factor/genetics , Signal Transduction/physiology , Animals , Bone Regeneration/drug effects , Bone Regeneration/physiology , Bone Remodeling/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Lineage/drug effects , Cell Lineage/physiology , Cells, Cultured , Ciliary Neurotrophic Factor/pharmacology , Cytokines/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , RNA, Messenger/metabolism , Sex Characteristics , Sex Factors , Signal Transduction/drug effects , Up-Regulation/drug effects , Up-Regulation/physiology
8.
Yonsei Med J ; 60(10): 924-934, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31538427

ABSTRACT

PURPOSE: Acute leukemia (AL) is classified as acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML). This study aimed to investigate the effect of miR-146a on childhood AL and its underlying molecular mechanisms. MATERIALS AND METHODS: Bone marrow samples were obtained from 39 AL children and 10 non-cancer controls. The expressions of miR-146a and ciliary neurotrophic factor receptor (CNTFR) were detected by quantitative real-time polymerase chain reaction (qRT-PCR) in ALL and AML pediatric patients, as well as ALL (Jurkat) and AML (HL-60) cells. Correlations between miR-146a and clinical indicators were explored. A targeting relationship between miR-146a and CNTFR was detected by dual luciferase reporter gene assay. Cell proliferation, apoptosis, migration, and invasion of Jurkat and HL-60 cells were measured by MTT assay, flow cytometry, and transwell assay, respectively. LIF expression was detected by qRT-PCR in Jurkat and HL-60 cells. The expression of p-JAK2, JAK2, p-STAT3, and STAT3 in HL-60 cells was measured by Western blot. RESULTS: miR-146a was increased in ALL and AML pediatric patients, while CNTFR was decreased. miR-146a expression was associated with immunophenotype, karyotype, fusion gene, and SIL-TAL1. CNTFR was a target gene of miR-146a. miR-146a could promote cell proliferation, migration, and invasion, as well as inhibit cell apoptosis in Jurkat and HL-60 cells by downregulating CNTFR. Meanwhile, miR-146a inhibited the expression of LIF and activated JAK2/STAT3 pathway by downregulating CNTFR. CONCLUSION: miR-146a could promote the proliferation, migration, and invasion and inhibit the apoptosis of AL Jurkat and HL-60 cells by downregulating CNTFR and activating the JAK2/STAT3 pathway.


Subject(s)
Down-Regulation/genetics , Janus Kinase 2/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , MicroRNAs/metabolism , Receptor, Ciliary Neurotrophic Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Apoptosis/genetics , Base Sequence , Cell Movement/genetics , Cell Proliferation/genetics , Child , Child, Preschool , Gene Expression Regulation, Leukemic , HL-60 Cells , Humans , Infant , Jurkat Cells , Leukemia Inhibitory Factor/metabolism , MicroRNAs/genetics , Neoplasm Invasiveness , Receptor, Ciliary Neurotrophic Factor/metabolism , Treatment Outcome , Up-Regulation/genetics
9.
Eur J Neurosci ; 27(11): 2830-7, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18588528

ABSTRACT

Indirect evidence suggests that endogenous ciliary neurotrophic factor (CNTF) receptor signaling can promote motor neuron (MN) survival in the adult. If so, proper targeting of this signaling may selectively counteract the effects of adult MN diseases. However, direct evidence for CNTF receptor involvement in adult MN survival is lacking, presumably because the unconditional blockade of the mouse CNTF receptor in vivo [through genetic disruption of the essential CNTF receptor alpha (CNTFRalpha) gene] leads to uniform perinatal death of the mice. To overcome this limitation, we have developed a method to selectively disrupt CNTF receptor function in a targeted subset of adult MNs that are not required for survival. A 'floxed CNTFRalpha' mouse line was generated and characterized. In addition, an adeno-associated virus (AAV) vector that drives Cre recombinase (Cre) expression was constructed and shown, with reporter mouse lines, to selectively excise floxed genes in facial MNs following its stereotaxic injection into the facial motor nucleus. Adult floxed CNTFRalpha mice were then injected with the AAV-Cre vector to excise the CNTFRalpha gene in the targeted MNs. The resulting data indicate that adult CNTF receptor signaling, likely by the MNs themselves, can play an essential role in MN survival. The data further indicate that this role is independent of any developmental contributions CNTF receptor signaling makes to MN survival or function.


Subject(s)
Central Nervous System/metabolism , Ciliary Neurotrophic Factor/metabolism , Gene Targeting/methods , Motor Neurons/metabolism , Receptor, Ciliary Neurotrophic Factor/genetics , Age Factors , Animals , Cell Survival/genetics , Dependovirus/genetics , Down-Regulation/genetics , Facial Nerve/cytology , Facial Nerve/metabolism , Genetic Vectors/genetics , Integrases/genetics , Mice , Mice, Knockout , Signal Transduction/genetics , Transfection
10.
J Neurosci Res ; 86(12): 2615-23, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18478544

ABSTRACT

In this study, we evaluated the role of the GABA(A) receptor (GABA(A)R), expressed by undifferentiated neural progenitors isolated from fetal mouse neocortex, in the mechanisms relevant to self-replication and differentiation toward neuronal and astroglial lineages. Round spheres were formed with clusters of proliferating cells within 2-4 days during culture with epidermal growth factor (EGF), whereas the size of these clusters was drastically increased in proportion to increasing durations up to 10 days. Sustained exposure to the GABA(A)R agonist muscimol at 100 microM led to significant increases in the size of neurospheres cultured for 6-10 days, with increased proliferative activity and unchanged lactate dehydrogenase release in a manner sensitive to the GABA(A)R antagonist bicuculline. Muscimol also significantly increased the incorporation of 5-bromo-2'-deoxyuridine in neurospheres in a bicuculline-sensitive manner, whereas both high potassium and nifedipine significantly decreased the neurosphere area with increased numbers of apoptotic cells. Prior activation of GABA(A)R significantly promoted the subsequent expression of an astroglial marker protein in cells differentiated by ciliary neurotrophic factor (CNTF) toward an astroglial lineage after the removal of EGF, with a concomitant decrease in neuronal marker protein expression. In neurospheres with GABA(A)R activation, a significant and predominant increase was seen in mRNA expression of CNTF receptors. These results suggest that prior tonic activation of GABA(A)R may preferentially promote the differentiation by CNTF of neural progenitor cells toward an astroglial lineage through selective up-regulation of CNTF receptor expression in the developing mouse brain.


Subject(s)
Brain/growth & development , Cell Differentiation/physiology , Gene Expression Regulation, Developmental/physiology , Receptor, Ciliary Neurotrophic Factor/biosynthesis , Receptors, GABA-A/physiology , Stem Cells/physiology , Up-Regulation/physiology , Animals , Brain/cytology , Brain/embryology , Cells, Cultured , Mice , Neurons/cytology , Neurons/physiology , Receptor, Ciliary Neurotrophic Factor/genetics , Stem Cells/cytology
11.
Brain ; 130(Pt 10): 2566-76, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17898009

ABSTRACT

Neuronal and axonal degeneration results in irreversible neurological disability in multiple sclerosis (MS) patients. A number of adaptive or neuroprotective mechanisms are thought to repress neurodegeneration and neurological disability in MS patients. To investigate possible neuroprotective pathways in the cerebral cortex of MS patients, we compared gene transcripts in cortices of six control and six MS patients. Out of 67 transcripts increased in MS cortex nine were related to the signalling mediated by the neurotrophin ciliary neurotrophic factor (CNTF). Therefore, we quantified and localized transcriptional (RT-PCR, in situ hybridization) and translational (western, immunohistochemistry) products of CNTF-related genes. CNTF-receptor complex members, CNTFRalpha, LIFRbeta and GP130, were increased in MS cortical neurons. CNTF was increased and also expressed by neurons. Phosphorylated STAT3 and the anti-apoptotic molecule, Bcl2, known down stream products of CNTF signalling were also increased in MS cortical neurons. We hypothesize that in response to the chronic insults or stress of the pathogenesis of multiple sclerosis, cortical neurons up regulate a CNTF-mediated neuroprotective signalling pathway. Induction of CNTF signalling and the anti-apoptotic molecule, Bcl2, thus represents a compensatory response to disease pathogenesis and a potential therapeutic target in MS patients.


Subject(s)
Ciliary Neurotrophic Factor/metabolism , Motor Cortex/metabolism , Multiple Sclerosis/metabolism , Neurons/metabolism , Ciliary Neurotrophic Factor/genetics , Humans , Nerve Growth Factors/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , Receptor, Ciliary Neurotrophic Factor/genetics , Receptor, Ciliary Neurotrophic Factor/metabolism , STAT3 Transcription Factor/physiology , Signal Transduction , Transcription Factors/metabolism , Up-Regulation
12.
Mol Vis ; 13: 758-63, 2007 May 23.
Article in English | MEDLINE | ID: mdl-17563726

ABSTRACT

PURPOSE: Ciliary neurotrophic factor (CNTF) promotes gene expression, cell survival and differentiation in various types of peripheral and central neurons, glia and nonneural cells. The level of CNTF rises rapidly upon injury to neural tissue, suggesting that CNTF exerts its cytoprotective effects after release from cells via mechanisms induced by cell injury. The purpose of this study was to determine if cells in the optic nerve head express CNTF and its tripartite receptor complex. METHODS: Well-established optic nerve head astrocytes (ONHA) and lamina cribrosa (LC) cell cultures were derived from normal human donors. Total RNA was reverse transcribed and polymerase chain reaction (PCR) amplified for mRNA detection. Cytoplasmic protein expression was determined by immunocytochemistry and Western blot analysis of the cellular lysates. Serum free medium was concentrated and used for detecting extracellular proteins. CNTF complexed with CNTFR-alpha was assayed by immunoprecipitation. RESULTS: Cells isolated from the human optic nerve head express CNTF and its tripartite receptor complex members (CNTFR-alpha, gp130, LIFR-beta). CONCLUSIONS: Taken together, these data suggest a possible neuroprotective role of CNTF in the optic nerve head.


Subject(s)
Ciliary Neurotrophic Factor/metabolism , Optic Disk/metabolism , Receptor, Ciliary Neurotrophic Factor/metabolism , Aged , Aged, 80 and over , Astrocytes/metabolism , Blotting, Western , Cells, Cultured , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor Receptor alpha Subunit/metabolism , Cytokine Receptor gp130/metabolism , Cytoplasm/metabolism , Humans , Immunohistochemistry , Immunoprecipitation , Infant , Optic Disk/cytology , RNA, Messenger/metabolism , Receptor, Ciliary Neurotrophic Factor/genetics , Receptors, OSM-LIF/metabolism
13.
J Appl Physiol (1985) ; 102(5): 1824-31, 2007 May.
Article in English | MEDLINE | ID: mdl-17272411

ABSTRACT

Genotypic associations between polymorphisms in the ciliary neurotrophic factor (CNTF) and CNTF receptor (CNTFR) genes and muscular strength phenotypes in 154 middle-aged men (45-49 yr) and 138 women (38-44 yr) and 99 older men (60-78 yr) and 102 older women (60-80 yr) were tested to validate earlier association studies. Allelic interaction effects were hypothesized between alleles of CNTF and CNTFR. We performed analysis of covariance with age, height, and fat-free mass (FFM) as covariates. FFM was anthropometrically estimated by the equation of Durnin-Womersley. Isometric, concentric, and eccentric torques for the knee flexors (KF) and extensors (KE) were measured using Biodex dynamometry. In the older male group, T-allele carriers of the C-1703T polymorphism in CNTFR performed significantly better on all noncorrected KF torques, whereas only noncorrected KE isometric torque at 120 degrees and concentric torque at 240 degrees/s were higher than the C/C homozygotes (P < 0.05). When age, height, and FFM were used as covariates, T-allele carriers performed only better on KE and KF isometric torque at 120 degrees (P < 0.05). Concentric KF torque at 180 degrees/s was lower in middle-aged female A-allele carriers compared with the T/T subjects for the T1069A polymorphism in CNTFR. After correction for age, height, and FFM, middle-aged female A-allele carriers exhibited lower values on all concentric KF strength measures and isometric torque at 120 degrees . There was a lack of association with the CNTF G-6A polymorphism in men, with inconclusive results for a limited number of phenotypes in women. No significant CNTF/CNTFR allele interaction effects were found. Results indicate that CNTFR C-1703T and T1069A polymorphisms are significantly associated with muscle strength in humans.


Subject(s)
Aging/genetics , Ciliary Neurotrophic Factor/genetics , Muscle Strength/physiology , Muscle, Skeletal/physiology , Receptor, Ciliary Neurotrophic Factor/genetics , Adult , Age Factors , Aged , Cohort Studies , Female , Gene Frequency , Genotype , Humans , Knee , Longitudinal Studies , Male , Middle Aged , Muscle Strength/genetics , Phenotype , Sex Factors , Torque
14.
Cancer Res ; 62(12): 3544-8, 2002 Jun 15.
Article in English | MEDLINE | ID: mdl-12068002

ABSTRACT

The loss of gap junctional intercellular communication has been proposedas playing a major role in the process of carcinogenesis. Most neoplastic cells, including C6 gliomas, express less connexins and have fewer gap junctions, reduced gap junctional intercellular communication, and increased growth rates compared with their nonneoplastic counterparts. The purpose of this study was to determine whether ciliary neurotrophic factor (CNTF) can be used to increase endogenous connexin43 levels, increase intercellular coupling, and retard the growth rate of C6 glioma cells. C6 cells were grown in serum-reduced medium (1% serum) and exposed to the following agents: vehicle (PBS), CNTF (20 ng/ml), CNTF soluble receptor (CNTFRalpha; 200 ng/ml), or Complex (CNTF + CNTFRalpha). Reverse transcription-PCR analysis indicated that C6 cells express CNTF mRNA but not CNTFRalpha mRNA. When cells were exposed to the above agents, only Complex caused an up-regulation of connexin43 protein (based on immunocytochemical and immunoblot analysis). Furthermore, Complex increased gap junctional coupling in C6 cells as noted by the passage of the gap junction permeable dye calcein. Finally, it was demonstrated that Complex-treatment reduces the growth rate of C6 cells compared with all of the other agents tested. Taken together, this study has demonstrated that CNTF in combination with its soluble receptor can increase connexin43 expression, increase gap junctional coupling, and reduce the in vitro proliferation of C6 glioma cells.


Subject(s)
Cell Communication/physiology , Ciliary Neurotrophic Factor/pharmacology , Connexin 43/biosynthesis , Gap Junctions/physiology , Glioma/pathology , Receptor, Ciliary Neurotrophic Factor/physiology , Animals , Cell Communication/drug effects , Cell Division/physiology , Ciliary Neurotrophic Factor/biosynthesis , Ciliary Neurotrophic Factor/genetics , Gap Junctions/drug effects , Glioma/genetics , Glioma/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Receptor, Ciliary Neurotrophic Factor/biosynthesis , Receptor, Ciliary Neurotrophic Factor/genetics , Tumor Cells, Cultured , Up-Regulation
15.
J Neurosci ; 21(9): 3144-50, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11312299

ABSTRACT

Approximately half of the motoneurons produced during development die before birth or shortly after birth. Although it is believed that survival depends on a restricted supply of a trophic sustenance produced by the synaptic target tissue (i.e., muscle), it is unclear whether synapse formation per se is involved in motoneuron survival. To address this issue, we counted cranial motoneurons in a set of mutant mice in which formation of neuromuscular junctions is dramatically impaired (i.e., null mutants for agrin, nerve-derived agrin, rapsyn, and MuSK). We demonstrate that in the absence of synaptogenesis, there is an 18-34% increase in motoneuron survival in the facial, trochlear, trigeminal motor, and hypoglossal nuclei; the highest survival occurred in the MuSK-deficient animals in which synapse formation is most severely compromised. There was no change in the size of the mutant motoneurons as compared with control animals, and the morphology of the mutant motoneurons appeared normal. We postulate that the increased axonal branching observed in these mutants leads to a facilitated "access" of the motoneurons to muscle-derived trophic factors at sites other than synapses or that inactivity increases the production of such factors. Finally, we examined motoneurons in double mutants of CNTFRalpha(-/-) (in which there is a partial loss of motoneurons) and MuSK(-/-) (in which there is an increased survival of motoneurons). The motoneuron numbers in the double mutants parallel those of the single MuSK-deficient mice, indicating that synapse disruption can even overcome the deleterious effect of CNTFRalpha ablation.


Subject(s)
Motor Neurons/metabolism , Neuromuscular Junction Diseases/metabolism , Neuromuscular Junction Diseases/pathology , Neuromuscular Junction/pathology , Receptors, Cholinergic , Agrin/deficiency , Agrin/genetics , Animals , Animals, Newborn , Axons/pathology , Cell Count , Cell Survival/genetics , Cranial Nerves/cytology , Cranial Nerves/embryology , Mice , Mice, Mutant Strains , Motor Neurons/cytology , Muscle Proteins/deficiency , Muscle Proteins/genetics , Neuromuscular Junction/embryology , Neuromuscular Junction/genetics , Neuromuscular Junction Diseases/embryology , Neuromuscular Junction Diseases/genetics , Protein Isoforms/deficiency , Protein Isoforms/genetics , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Ciliary Neurotrophic Factor/deficiency , Receptor, Ciliary Neurotrophic Factor/genetics
16.
J Neurosci ; 21(19): 7642-53, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11567054

ABSTRACT

The cytokines that signal through the common receptor subunit gp130, including ciliary neurotrophic factor (CNTF), interleukin-6, leukemia inhibitory factor (LIF) and oncostatin M, have pleiotropic functions in CNS development. Given the restricted expression domain of the CNTF receptor alpha (CNTFR) in the developing forebrain germinal zone and adult forebrain periventricular area, we have examined the putative role of CNTFR/LIFR/gp130-mediated signaling in regulating forebrain neural stem cell fate in vivo and in vitro. Analysis of LIFR-deficient mice revealed that a decreased level of LIFR expression results in a reduction in the number of adult neural stem cells. In adult LIFR heterozygote (+/-) mice, the number of neural stem cells and their progeny in the forebrain subependyma and TH-immunoreactive neurons in the olfactory bulb were significantly reduced. Intraventricular infusion of CNTF into the adult mouse forebrain, in the absence or presence of epidermal growth factor (EGF), enhanced self-renewal of neural stem cells in vivo. Analyses of EGF-responsive neural stem cells proliferating in vitro found that CNTF inhibits lineage restriction of neural stem cells to glial progenitors, which in turn results in enhanced expansion of stem cell number. These results suggest that CNTFR/LIFR/gp130-mediated signaling supports the maintenance of forebrain neural stem cells, likely by suppressing restriction to a glial progenitor cell fate.


Subject(s)
Neurons/metabolism , Prosencephalon/metabolism , Receptor, Ciliary Neurotrophic Factor/metabolism , Stem Cells/metabolism , Animals , Antigens, CD/metabolism , Cell Count , Cell Differentiation/drug effects , Cell Lineage , Cells, Cultured , Ciliary Neurotrophic Factor/metabolism , Ciliary Neurotrophic Factor/pharmacology , Cytokine Receptor gp130 , Epidermal Growth Factor/pharmacology , Heterozygote , Injections, Intraventricular , Leukemia Inhibitory Factor Receptor alpha Subunit , Macromolecular Substances , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Prosencephalon/cytology , Receptor, Ciliary Neurotrophic Factor/genetics , Receptors, Cytokine/deficiency , Receptors, Cytokine/genetics , Receptors, OSM-LIF , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/drug effects , Tyrosine 3-Monooxygenase/biosynthesis
17.
J Neurosci ; 21(4): 1283-91, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11160399

ABSTRACT

Developing motoneurons require trophic support from their target, the skeletal muscle. Despite a large number of neurotrophic molecules with survival-promoting activity for isolated embryonic motoneurons, those factors that are required for motoneuron survival during development are still not known. Cytokines of the ciliary neurotrophic factor (CNTF)-leukemia inhibitory factor (LIF) family have been shown to play a role in motoneuron (MN) survival. Importantly, in mice lacking the LIFRbeta or the CNTFRalpha there is a significant loss of MNs during embryonic development. Because genetic deletion of either (or both) CNTF or LIF fails, by contrast, to perturb MN survival before birth, it was concluded that another ligand exists that is functionally inactivated in the receptor deleted mice, resulting in MN loss during development. One possible candidate for this ligand is the CNTF-LIF family member cardiotrophin-1 (CT-1). CT-1 is highly expressed in embryonic skeletal muscle, secreted by myotubes, and promotes the survival of cultured embryonic mouse and rat MNs. Here we show that ct-1 deficiency causes increased motoneuron cell death in spinal cord and brainstem nuclei of mice during a period between embryonic day 14 and the first postnatal week. Interestingly, no further loss was detectable during the subsequent postnatal period, and nerve lesion in young adult ct-1-deficient mice did not result in significant additional loss of motoneurons, as had been previously observed in mice lacking both CNTF and LIF. CT-1 is the first bona fide muscle-derived neurotrophic factor to be identified that is required for the survival of subgroups of developing motoneurons.


Subject(s)
Cytokines/metabolism , Interleukin-6 , Motor Neurons/metabolism , Muscle, Skeletal/metabolism , Neurodegenerative Diseases/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Axotomy , Brain Stem/embryology , Brain Stem/metabolism , Brain Stem/pathology , Cell Death , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Chick Embryo , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/metabolism , Cytokine Receptor gp130 , Cytokines/deficiency , Cytokines/genetics , Cytokines/pharmacology , Dose-Response Relationship, Drug , Facial Nerve , Growth Inhibitors/genetics , Growth Inhibitors/metabolism , Leukemia Inhibitory Factor , Lymphokines/genetics , Lymphokines/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Motor Neurons/drug effects , Motor Neurons/pathology , Muscle, Skeletal/embryology , Muscle, Skeletal/innervation , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , RNA, Messenger/biosynthesis , Receptor, Ciliary Neurotrophic Factor/genetics , Receptor, Ciliary Neurotrophic Factor/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Spinal Cord/pathology
18.
J Neurosci ; 24(48): 10806-15, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15574731

ABSTRACT

We have shown previously that intraocular elevation of cAMP using the cAMP analog 8-(4-chlorophenylthio)-cAMP (CPT-cAMP) failed to promote axonal regeneration of axotomized adult retinal ganglion cells (RGCs) into peripheral nerve (PN) grafts but significantly potentiated ciliary neurotrophic factor (CNTF)-induced axonal regeneration. Using the PN graft model, we now examine the mechanisms underlying spontaneous and CNTF/CPT-cAMP-induced neuronal survival and axonal regrowth. We found that blockade of the cAMP pathway executor protein kinase A (PKA) using the cell-permeable inhibitor KT5720 did not affect spontaneous survival and axonal regeneration but essentially abolished the CNTF/CPT-cAMP-induced RGC survival and axonal regeneration. Blockade of CNTF signaling pathways such as phosphotidylinositol 3-kinase (PI3K)/akt by 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (LY294002), mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) by 2-(2-diamino-3-methoxyphenyl-4H-1-benzopyran-4-one (PD98059), or Janus kinase (JAK)/signal transducer and activators of transcription (STAT3) by tyrphostin AG490 also blocked the CNTF/CPT-cAMP-dependent survival and regeneration effects. PKA activity assay and Western blots showed that KT5720, LY294002, and PD98059 almost completely inhibited PKA, PI3K/akt, and MAPK/ERK signal transduction, respectively, whereas AG490 substantially decreased JAK/STAT3 signal transduction. Intraocular injection of CPT-cAMP resulted in a small PKA-dependent increase in CNTF receptor alpha mRNA expression in the retinas, an effect that may facilitate CNTF action on survival and axonal regeneration. Surprisingly, in the absence of CNTF/CPT-cAMP, LY294002, PD98059, and AG490, but not KT5720, significantly enhanced spontaneous RGC survival, suggesting differential roles of these pathways in RGC survival under different conditions. Our data suggest that CNTF/CPT-cAMP-induced RGC survival and axonal regeneration are a result of multiple pathway actions, with PKA as an essential component, but that these pathways can function in an antagonistic manner under different conditions.


Subject(s)
Ciliary Neurotrophic Factor/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Cyclic AMP/analogs & derivatives , Nerve Regeneration , Retinal Ganglion Cells/physiology , Signal Transduction/physiology , Animals , Axons/drug effects , Axons/physiology , Carbazoles/pharmacology , Cell Survival/drug effects , Chromones/pharmacology , Ciliary Neurotrophic Factor/antagonists & inhibitors , Ciliary Neurotrophic Factor/pharmacology , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , DNA-Binding Proteins/antagonists & inhibitors , Flavonoids/pharmacology , Indoles/pharmacology , MAP Kinase Signaling System/drug effects , Morpholines/pharmacology , Nerve Regeneration/drug effects , Optic Nerve Injuries/enzymology , Optic Nerve Injuries/physiopathology , Peroneal Nerve/transplantation , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-akt , Pyrroles/pharmacology , Rats , Rats, Inbred F344 , Receptor, Ciliary Neurotrophic Factor/biosynthesis , Receptor, Ciliary Neurotrophic Factor/drug effects , Receptor, Ciliary Neurotrophic Factor/genetics , Retinal Ganglion Cells/drug effects , STAT3 Transcription Factor , Signal Transduction/drug effects , Thionucleotides/pharmacology , Trans-Activators/antagonists & inhibitors , Tyrphostins/pharmacology , Up-Regulation
19.
J Neuropathol Exp Neurol ; 60(11): 1087-98, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11706938

ABSTRACT

Oncostatin M (OSM) is a member of the interleukin (IL)-6 cytokine family and modulates inflammatory responses. Here we investigated the role of OSM as an immunoregulatory factor for human cerebral endothelial cells (HCEC). Using RT-PCR we detected transcripts of the receptor components involved in OSM signaling, gp130, OSM receptor (OSMR)-beta, and leukemia inhibitory factor receptor (LIFR), in HCEC. A parallel FACS analysis revealed surface expression of gp130 and OSMR-beta, but not of LIFR on these cells. Functionally, OSM upregulated intercellular adhesion molecule-1, but did not induce vascular cell adhesion molecule-1 in HCEC. Further, OSM upregulated IL-6 and monocyte chemoattractant protein (MCP)-1, whereas IL-8 was unaffected. Combined application of tumor necrosis factor (TNF)-alpha and OSM synergistically enhanced IL-6 and MCP-1 production, but downregulated TNF-alpha-induced IL-8. As OSM regulated molecules relevant in inflammatory brain diseases, we investigated its expression in normal and pathological human brains. OSM was detected by immunohistochemistry in brains from multiple sclerosis patients in microglia, reactive astrocytes, and infiltrating leukocytes, whereas in normal brains and noninflammatory neurological diseases. immunoreactivity was absent from the parenchyma. These data suggest that immunoregulatory functions in human cerebral endothelial cells may be a mechanism by which OSM participates in the pathophysiology of inflammatory brain disease.


Subject(s)
Blood-Brain Barrier/physiology , Endothelium, Vascular/chemistry , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Peptides/analysis , Adult , Aged , Antigens, CD/analysis , Antigens, CD/genetics , Cells, Cultured , Chemokine CCL2/analysis , Chemokine CCL2/genetics , Cytokine Receptor gp130 , Endothelium, Vascular/cytology , Female , Flow Cytometry , Gene Expression/immunology , Humans , Intercellular Adhesion Molecule-1/metabolism , Interleukin-6/analysis , Interleukin-6/genetics , Leukemia Inhibitory Factor Receptor alpha Subunit , Male , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , Middle Aged , Multiple Sclerosis/pathology , Oncostatin M , Peptides/genetics , RNA, Messenger/analysis , Receptor, Ciliary Neurotrophic Factor/genetics , Receptors, Cytokine/analysis , Receptors, Cytokine/genetics , Receptors, Interleukin-6/genetics , Receptors, OSM-LIF , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/immunology , Vascular Cell Adhesion Molecule-1/metabolism
20.
Endocrinology ; 141(5): 1746-53, 2000 May.
Article in English | MEDLINE | ID: mdl-10803585

ABSTRACT

Two of the most potent cytokines regulating anterior pituitary cell function are leukemia inhibitory factor and interleukin-6 (IL-6), which belong to the cytokine receptor family using the common gp130 signal transducer. We studied the actions of two other members of this family, IL-11 and ciliary neurotropic factor (CNTF), on folliculostellate (FS) cells (TtT/GF cell line) and lactosomatotropic cells (GH3 cell line). The messenger RNA (mRNA) for the alpha-chain specific for the IL-11 receptor (1.7 kb) and CNTF receptor (2 kb) are expressed on both cell types. In addition, we detected CNTF receptor mRNA in normal rat anterior pituitary cells. IL-11 (1.25-5 nM) dose dependently stimulated the proliferation of FS cells. CNTF, at doses from 0.4-2 nM, also significantly stimulated the growth of these cells. In addition, both cytokines significantly stimulated proliferation of lactosomatotropic GH3 cells, and CNTF stimulated hormone production (GH and PRL) at 24 h by these cells. At 16-72 h, IL-11 stimulates the secretion of the angiogenic factor vascular endothelial growth factor by FS cells. In addition, both GH3 and FS cells express CNTF mRNA. These data suggest that IL-11 and CNTF may act as growth and regulatory factors in anterior pituitary cells.


Subject(s)
Ciliary Neurotrophic Factor/physiology , Interleukin-11/physiology , Lactation/physiology , Pituitary Gland, Anterior/physiology , Receptor, Ciliary Neurotrophic Factor/biosynthesis , Receptors, Interleukin/biosynthesis , Animals , Cell Division , Cell Line , Endothelial Growth Factors/metabolism , Female , Interleukin-11 Receptor alpha Subunit , Lymphokines/metabolism , Male , Pituitary Gland, Anterior/cytology , Rats , Rats, Sprague-Dawley , Receptor, Ciliary Neurotrophic Factor/genetics , Receptors, Interleukin/genetics , Receptors, Interleukin-11 , Recombinant Proteins/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL