Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(15)2022 Jul 28.
Article in English | MEDLINE | ID: mdl-35955469

ABSTRACT

Ciliary neurotrophic factor (CNTF) was identified as a survival factor in various types of peripheral and central neurons, glia and non-neural cells. At present, there is no available data on the expression and localization of CNTF-receptors in cementoblasts as well as on the role of exogenous CNTF on this cell line. The purpose of this study was to determine if cementoblasts express CNTF-receptors and analyze the mechanism of its apoptotic regulation effects on cementoblasts. OCCM-30 cementoblasts were cultivated and stimulated kinetically using CNTF protein (NBP2-35168, Novus Biologicals). Quantified transcriptional (RT-qPCR) and translational (WB) products of CNTFRα, IL-6Rα (CD126), LIFR, p-GP130, GP130, p-ERK1/2, ERK1/2, Caspase-8, -9, -3 and cleaved-caspase-3 were evaluated. Immunofluorescence (IF) staining was applied to visualize the localization of the CNTF-receptors within cells. The apoptosis ratio was measured with an Annexin-V FITC/PI kit. The ERK1/2 antagonist (FR180204, Calbiochem) was added for further investigation by flow cytometry analysis. The CNTF-receptor complex (CNTFRα, LIFR, GP130) was functionally up-regulated in cementoblasts while cultivated with exogenous CNTF. CNTF significantly attenuated cell viability and proliferation for long-term stimulation. Flow cytometry analysis shows that CNTF enhanced the apoptosis after prolonged duration. However, after only a short-term period, CNTF halts the apoptosis of cementoblasts. Further studies revealed that CNTF activated phosphorylated GP130 and the anti-apoptotic molecule ERK1/2 signaling to participate in the regulation of the apoptosis ratio of cementoblasts. In conclusion, CNTF elicited the cellular functions through a notable induction of its receptor complex in cementoblasts. CNTF has an inhibitory effect on the cementoblast homeostasis. These data also elucidate a cellular mechanism for an exogenous CNTF-triggered apoptosis regulation in a mechanism of ERK1/2 and caspase signaling and provides insight into the complex cellular responses induced by CNTF in cementoblasts.


Subject(s)
Ciliary Neurotrophic Factor Receptor alpha Subunit , Ciliary Neurotrophic Factor , Apoptosis , Caspases/metabolism , Ciliary Neurotrophic Factor/metabolism , Ciliary Neurotrophic Factor Receptor alpha Subunit/metabolism , Cytokine Receptor gp130/metabolism , Dental Cementum/metabolism , MAP Kinase Signaling System , Receptor, Ciliary Neurotrophic Factor/metabolism
2.
Cell Tissue Res ; 390(1): 113-129, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35794391

ABSTRACT

Ciliary neurotrophic factor (CNTF) is a pleiotropic cytokine that signals through a receptor complex containing a specific subunit, CNTF receptor α (CNTFRα). The two molecules are constitutively expressed in key structures for human placental growth and differentiation. The possible role of CNTF in enhancing cell proliferation and/or invasion during placental development and remodelling was investigated using HTR-8/SVneo and BeWo cells, taken respectively as cytotrophoblast and syncytiotrophoblast models. In both cell lines, treatment with human recombinant (hr) CNTF activated JAK2/STAT3 signalling and inhibited the ERK pathway. Interestingly, in HTR-8/SVneo cells, 50 ng hrCNTF induced significant downregulation of matrix metalloprotease (MMP)-1 and significant upregulation of MMP-9. Moreover, pharmacological inhibition of JAK2/STAT3 signalling by AG490 and curcumin resulted in MMP-9 downregulation; it activated the ERK signalling pathway and upregulated MMP-1 expression. Collectively, these data suggest a role for CNTF signalling in extravillous cytotrophoblast invasion through the modulation of specific MMPs.


Subject(s)
Ciliary Neurotrophic Factor , Curcumin , Ciliary Neurotrophic Factor/metabolism , Ciliary Neurotrophic Factor/pharmacology , Ciliary Neurotrophic Factor Receptor alpha Subunit/metabolism , Cytokines/metabolism , Female , Humans , Matrix Metalloproteinase 1 , Matrix Metalloproteinase 9 , Placenta/metabolism , Placentation , Pregnancy , Receptor, Ciliary Neurotrophic Factor/metabolism
3.
PLoS One ; 17(3): e0265749, 2022.
Article in English | MEDLINE | ID: mdl-35316287

ABSTRACT

Ciliary neurotrophic factor (CNTF) is a neurotrophic cytokine able to induce appetite reduction, weight loss and antidiabetic effects. However, its susceptibility to neutralizing anti-CNTF antibodies in patients hampered its use for treatment of human obesity and diabetes. In addition, CNTF has a very short plasma half-life, which limits its use as a therapeutic agent. Solutions, directed to prolong its in vivo effects, vary from the implantation of encapsulated secreting cells to identification of more active variants or chemical modification of the protein itself. PEGylation is a widely used modification for shielding proteins from circulating antibodies and for increasing their plasma half-life. Here, we have selected DH-CNTF, a CNTF variant which has a 40-fold higher affinity for the CNTF receptor α accompanied by an increased activity in cellular assays. The PEGylated DH-CNTF retained the biological activity of native protein in vitro and showed a significant improvement of pharmacokinetic parameters. In an acute model of glucose tolerance, the PEG-DH-CNTF was able to reduce the glycemia in diet-induced obese animals, with a performance equaled by a 10-fold higher dose of DH-CNTF. In addition, the PEGylated DH-CNTF analog demonstrated a more potent weight loss effect than the unmodified protein, opening to the use of CNTF as weight reducing agent with treatment regimens that can better meet patient compliance thanks to reduced dosing schedules.


Subject(s)
Ciliary Neurotrophic Factor , Obesity , Animals , Ciliary Neurotrophic Factor/pharmacology , Diet , Humans , Mice , Mice, Obese , Obesity/drug therapy , Obesity/etiology , Polyethylene Glycols/pharmacology , Proteins , Receptor, Ciliary Neurotrophic Factor/metabolism , Weight Loss
4.
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
5.
Int J Mol Sci ; 19(11)2018 Nov 10.
Article in English | MEDLINE | ID: mdl-30423807

ABSTRACT

We demonstrated that capsaicin (CAP), an agonist of transient receptor potential vanilloid subtype 1 (TRPV1), inhibits microglia activation and microglia-derived oxidative stress in the substantia nigra (SN) of MPP⁺-lesioned rat. However, the detailed mechanisms how microglia-derived oxidative stress is regulated by CAP remain to be determined. Here we report that ciliary neurotrophic factor (CNTF) endogenously produced by CAP-activated astrocytes through TRPV1, but not microglia, inhibits microglial activation and microglia-derived oxidative stress, as assessed by OX-6 and OX-42 immunostaining and hydroethidine staining, respectively, resulting in neuroprotection. The significant increase in levels of CNTF receptor alpha (CNTFRα) expression was evident on microglia in the MPP⁺-lesioned rat SN and the observed beneficial effects of CNTF was abolished by treatment with CNTF receptor neutralizing antibody. It is therefore likely that CNTF can exert its effect via CNTFRα on microglia, which rescues dopamine neurons in the SN of MPP⁺-lesioned rats and ameliorates amphetamine-induced rotations. Immunohistochemical analysis revealed also a significantly increased expression of CNTFRα on microglia in the SN from human Parkinson's disease patients compared with age-matched controls, indicating that these findings may have relevance to the disease. These data suggest that CNTF originated from TRPV1 activated astrocytes may be beneficial to treat neurodegenerative disease associated with neuro-inflammation such as Parkinson's disease.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Ciliary Neurotrophic Factor/pharmacology , Dopaminergic Neurons/pathology , Microglia/pathology , Neuroprotection/drug effects , Neurotoxicity Syndromes/pathology , Oxidative Stress , Aged , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Capsaicin/pharmacology , Cell Survival/drug effects , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Female , Gene Knockdown Techniques , Humans , Male , Microglia/drug effects , Microglia/metabolism , Models, Biological , Nerve Degeneration/pathology , Oxidative Stress/drug effects , RNA, Small Interfering/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptor, Ciliary Neurotrophic Factor/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , TRPV Cation Channels/metabolism
6.
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
7.
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
8.
Bone ; 64: 47-56, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24721701

ABSTRACT

Muscle and bone are intimately linked by bi-directional signals regulating both muscle and bone cell gene expression and proliferation. It is generally accepted that muscle cells secrete factors (myokines) that influence adjacent bone cells, but these myokines are yet to be identified. We have previously shown that osteocyte-specific deletion of the co-receptor subunit utilized by IL-6 family cytokines, glycoprotein 130 (gp130), resulted in impaired bone formation in the trabecular bone, but enhanced periosteal expansion, suggesting a gp130-dependent periosteum-specific inhibition of osteoblast function, potentially induced by the local muscle fibres. We report here that differentiated primary calvarial osteoblasts cultured in myotube-conditioned media (CM) from myogenic C2C12 cells show reduced mRNA levels of genes associated with osteoblast differentiation. Alkaline phosphatase protein activity and all mRNA markers of osteoblast differentiation in the tested panel (runx2, osterix, alkaline phosphatase, parathyroid hormone (PTH) receptor, osteoprotegerin, osteocalcin, sclerostin) were reduced following culture with myotube CM. The exception was RANKL, which was significantly elevated in differentiated primary osteoblast cultures expressing osteocytic genes. A cytokine array of the C2C12 myotube-conditioned media identified TIMP-1 and MCP-1 as the most abundant myokines, but treatment with recombinant TIMP-1 or MCP-1 did not inhibit osteoblast gene expression. Rather, the IL-6 family cytokine ciliary neurotrophic factor (CNTF), which we found abundantly expressed by mouse muscle at the transcript and protein level, reduced osteoblast gene expression, although not to the same extent as the myotube-conditioned media. These data indicate that muscle cells secrete abundant TIMP-1, MCP-1, and CNTF, and that of these, only CNTF has the ability to suppress osteoblast function and gene expression in a similar manner to myotube-conditioned medium. This suggests that CNTF is an inhibitory myokine for osteoblasts.


Subject(s)
Cell Differentiation/physiology , Chemokines/physiology , Ciliary Neurotrophic Factor/physiology , Cytokines/physiology , Osteoblasts/cytology , Animals , Cell Line , Ciliary Neurotrophic Factor/metabolism , Culture Media, Conditioned , Gene Expression/physiology , Mice , Mice, Inbred C57BL , Osteoblasts/metabolism , RANK Ligand/metabolism , Receptor, Ciliary Neurotrophic Factor/metabolism
9.
PLoS One ; 8(4): e61616, 2013.
Article in English | MEDLINE | ID: mdl-23626705

ABSTRACT

Ciliary neurotrophic factor (CNTF) induces neurogenesis, reduces feeding, and induces weight loss. However, the central mechanisms by which CNTF acts are vague. We employed the mHypoE-20/2 line that endogenously expresses the CNTF receptor to examine the direct effects of CNTF on mRNA levels of urocortin-1, urocortin-2, agouti-related peptide, brain-derived neurotrophic factor, and neurotensin. We found that treatment of 10 ng/ml CNTF significantly increased only urocortin-1 mRNA by 1.84-fold at 48 h. We then performed intracerebroventricular injections of 0.5 mg/mL CNTF into mice, and examined its effects on urocortin-1 neurons post-exposure. Through double-label immunohistochemistry using specific antibodies against c-Fos and urocortin-1, we showed that central CNTF administration significantly activated urocortin-1 neurons in specific areas of the hypothalamus. Taken together, our studies point to a potential role for CNTF in regulating hypothalamic urocortin-1-expressing neurons to mediate its recognized effects on energy homeostasis, neuronal proliferaton/survival, and/or neurogenesis.


Subject(s)
Appetite Depressants/pharmacology , Ciliary Neurotrophic Factor/pharmacology , Hypothalamus/drug effects , Neurons/drug effects , Urocortins/genetics , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Appetite/drug effects , Appetite/physiology , Appetite Depressants/metabolism , Appetite Regulation/drug effects , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cell Line , Ciliary Neurotrophic Factor/metabolism , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Gene Expression Regulation/drug effects , Hypothalamus/cytology , Hypothalamus/metabolism , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism , Neurotensin/genetics , Neurotensin/metabolism , Receptor, Ciliary Neurotrophic Factor/genetics , Receptor, Ciliary Neurotrophic Factor/metabolism , Urocortins/agonists , Urocortins/metabolism
10.
J Interferon Cytokine Res ; 33(2): 65-71, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23398366

ABSTRACT

A balanced proinflammatory cytokine response to Plasmodium ssp. infection is crucial to control the disease outcome. To elucidate the effect of cytokines and Plasmodium falciparum-infected erythrocytes on the regulation of interleukin (IL)-6 receptor (IL-6R), ciliary neurotrophic factor alpha (CNTFR-α) and glycoprotein (gp)130 in natural killer (NK) cells in the context of malaria, we assessed their gene expression and surface expression in NK92 cells. P. falciparum alone did not alter gene expression of the investigated receptors in NK92 cells. Analysis revealed a low effect of IL-6 on IL-6R surface expression in NK92 cells. However, at transcriptional level, a downregulation of IL-6R was observed following IL-6 stimulation. Thus, IL-6 might act within a negative feedback loop to terminate signal transduction by downregulating IL-6R expression. Additionally, we observed that IL-6R and CNTFR-α surface expression were regulated by a combination of IL-2, 12, and 18, and gp130 was influenced by interferon-α. Our results show that the IL-6 family receptors in NK92 cells are not directly influenced by P. falciparum. However, cytokines usually derived from accessory cells during malaria episodes may regulate IL-6 receptor signaling pathways. This finding encourages future studies in a more physiological context and with primary cells isolated from humans with and without malaria.


Subject(s)
Cytokine Receptor gp130/metabolism , Cytokines/pharmacology , Erythrocytes/parasitology , Killer Cells, Natural/metabolism , Plasmodium falciparum/physiology , Receptor, Ciliary Neurotrophic Factor/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cytokine Receptor gp130/genetics , Erythrocytes/drug effects , Erythrocytes/metabolism , Humans , Interferon-alpha/pharmacology , Killer Cells, Natural/drug effects , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Plasmodium falciparum/drug effects , Protein Binding/drug effects , Receptor, Ciliary Neurotrophic Factor/genetics , Transcription, Genetic/drug effects , Up-Regulation/drug effects
11.
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
12.
Neurochem Int ; 61(6): 874-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22306348

ABSTRACT

Leukemia inhibitory factor (LIF) and ciliary neurotrophic factor (CNTF) are neurally active cytokines, or neurokines. LIF signals through a receptor consisting of gp130 and the low affinity LIF receptor (LIFR), while the CNTF receptor consists of gp130, LIFR, and the low affinity CNTF receptor (CNTFR). Ser1044 of the LIFR is phosphorylated by Erk1/2 MAP kinase. Stimulation of neural cells with growth factors which strongly activate Erk1/2 decreases LIF-mediated signal transduction due to increased degradation of the LIFR as a consequence of Erk1/2-dependent phosphorylation of the receptor at Ser1044. The gp130 receptor subunit is phosphorylated, at least in part by calmodulin-dependent protein kinase II, at Ser782, which is adjacent to a dileucine internalization motif. Ser782 appears to negatively regulate cytokine receptor expression, as mutagenesis of Ser782 results in increased gp130 expression and cytokine-induced neuropeptide gene transcription. The LIFR and gp130 are transmembrane proteins, while CNTFR is a peripheral membrane protein attached to the cell surface via a glycosylphosphatidylinositol tail. In unstimulated cells, CNTFR but not LIFR and gp130 is localized to detergent-resistant lipid rafts. Stimulation of cells with CNTFR causes translocation of LIFR and gp130 into the lipid rafts, while stimulation with LIF does not induce receptor translocation, raising the possibility that CNTF could induce different patterns of signaling and/or receptor trafficking than caused by LIF. We used a compartmentalized culture system to examine the mechanisms for retrograde signaling by LIF and CNTF from distal neurites to the cell bodies of mouse sympathetic neurons. Stimulation with neurokines of the distal neurites of sympathetic neurons grown in a compartmentalized culture system resulted in the activation and nuclear translocation of the transcription factor Stat3. Retrograde signaling required Jak kinase activity in the cell body but not the distal neurites, and could be blocked by inhibitors of microtubule but not microfilament function. The results are consistent with a signaling endosomes model in which the ctyokine/receptor complex is transported back to the cell body where Stat3 is activated. While both LIF and CNTF mediate retrograde activation of Stat3, the kinetics for retrograde signaling differ for the two neurokines.


Subject(s)
Protein Transport , Receptor, Ciliary Neurotrophic Factor/metabolism , Receptors, OSM-LIF/metabolism , Signal Transduction , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Phosphorylation
13.
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
14.
Exp Neurol ; 233(1): 243-52, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22037350

ABSTRACT

We demonstrated previously that the hypothalamic supraoptic nucleus (SON) undergoes a robust axonal sprouting response following unilateral transection of the hypothalamo-neurohypophysial tract. Concomitant with this response is an increase in ciliary neurotrophic factor (CNTF) and CNTF receptor alpha (CNTFRα) expression in the contralateral non-uninjured SON from which the axonal outgrowth occurs. While these findings suggest that CNTF may act as a growth factor in support of neuronal plasticity in the SON, it remained to be determined if the observed increase in neurotrophin expression was related to the sprouting response per se or more generally to the increased neurosecretory activity associated with the post-lesion response. Therefore we used immunocytochemistry and Western blot analysis to examine the expression of CNTF and the components of the CNTF receptor complex in sprouting versus osmotically-stimulated SON. Western blot analysis revealed a significant increase in CNTF, CNTFRα, and gp130, but not LIFRß, protein levels in the sprouting SON at 10days post lesion in the absence of neuronal loss. In contrast, osmotic stimulation of neurosecretory activity in the absence of injury resulted in a significant decrease in CNTF protein levels with no change in CNTFRα, gp130, or LIFRß protein levels. Immunocytochemical analysis further demonstrated gp130 localization on magnocellular neurons and astrocytes while the LIFRß receptor was found only on astrocytes in the SON. These results are consistent with the hypothesis that increased CNTF and CNTFR complex in the sprouting, metabolically active SON are related directly to the sprouting response and not the increase in neurosecretory activity.


Subject(s)
Axons/physiology , Ciliary Neurotrophic Factor/metabolism , Neurons/pathology , Receptor, Ciliary Neurotrophic Factor/metabolism , Regeneration/physiology , Supraoptic Nucleus/pathology , Animals , Axons/drug effects , Cytokine Receptor gp130/metabolism , Gene Expression Regulation , Glial Fibrillary Acidic Protein/metabolism , Male , Neurons/drug effects , Neurons/metabolism , Oxytocin/metabolism , Rats , Rats, Sprague-Dawley , Salts/administration & dosage , Supraoptic Nucleus/injuries , Time Factors , Vasopressins/metabolism
15.
PLoS One ; 6(9): e23148, 2011.
Article in English | MEDLINE | ID: mdl-21912637

ABSTRACT

Ciliary neurotrophic factor (CNTF) protects photoreceptors and regulates their phototransduction machinery, but little is known about CNTF's effects on retinal pigment epithelial (RPE) physiology. Therefore, we determined the expression and localization of CNTF receptors and the physiological consequence of their activation in primary cultures of human fetal RPE (hfRPE). Cultured hfRPE express CNTF, CT1, and OsM and their receptors, including CNTFRα, LIFRß, gp130, and OsMRß, all localized mainly at the apical membrane. Exogenous CNTF, CT1, or OsM induces STAT3 phosphorylation, and OsM also induces the phosphorylation of ERK1/2 (p44/42 MAP kinase). CNTF increases RPE survivability, but not rates of phagocytosis. CNTF increases secretion of NT3 to the apical bath and decreases that of VEGF, IL8, and TGFß2. It also significantly increases fluid absorption (J(V)) across intact monolayers of hfRPE by activating CFTR chloride channels at the basolateral membrane. CNTF induces profound changes in RPE cell biology, biochemistry, and physiology, including the increase in cell survival, polarized secretion of cytokines/neurotrophic factors, and the increase in steady-state fluid absorption mediated by JAK/STAT3 signaling. In vivo, these changes, taken together, could serve to regulate the microenvironment around the distal retinal/RPE/Bruch's membrane complex and provide protection against neurodegenerative disease.


Subject(s)
Body Fluids/metabolism , Ciliary Neurotrophic Factor/metabolism , Retinal Pigment Epithelium/metabolism , Absorption , Cell Membrane/metabolism , Cell Polarity , Cell Proliferation , Cell Survival , Cells, Cultured , Fetus/cytology , Gene Expression Regulation , Humans , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oncostatin M/genetics , Oncostatin M/metabolism , Organic Cation Transporter 1/genetics , Phagocytosis , Phosphorylation , Protein Subunits/metabolism , Protein Transport , Receptor, Ciliary Neurotrophic Factor/metabolism , Retinal Pigment Epithelium/cytology , STAT3 Transcription Factor/metabolism
16.
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
17.
Proteomics ; 10(11): 2138-50, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20340160

ABSTRACT

Accumulating evidence suggests that extracellular alpha-synuclein (eSNCA) plays an important role in the pathogenesis of Parkinson's disease or related synucleinopathies by inducing neurotoxicity directly or indirectly via microglial or astroglial activation. However, the mechanisms by which this occurs remain to be characterized. To explore these mechanisms, we combined three biochemical techniques - stable isotope labeling of amino acid in cell cultures (SILAC), biotin labeling of plasma membrane proteins followed by affinity purification, and analysis of unique proteins binding to SNCA peptides on membrane arrays. The SILAC proteomic analysis identified 457 proteins, of which, 245 or 172 proteins belonged to membrane or membrane associated proteins, depending on the various bioinformatics tools used for interpretation. In dopamine neuronal cells treated with eSNCA, the levels of 86 membrane proteins were increased and 35 were decreased compared with untreated cells. In peptide array analysis, 127 proteins were identified as possibly interacting with eSNCA. Of those, seven proteins were overlapped with the membrane proteins that displayed alterations in relative abundance after eSNCA treatment. One was ciliary neurotrophic factor receptor, which appeared to modulate eSNCA-mediated neurotoxicity via mechanisms related to JAK1/STAT3 signaling but independent of eSNCA endocytosis.


Subject(s)
Neurons/drug effects , Receptor, Ciliary Neurotrophic Factor/metabolism , alpha-Synuclein/pharmacology , Blotting, Western , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Ciliary Neurotrophic Factor Receptor alpha Subunit/genetics , Ciliary Neurotrophic Factor Receptor alpha Subunit/metabolism , Humans , Immunohistochemistry , Immunoprecipitation , Neurons/cytology , Neurons/metabolism , Protein Binding/drug effects , Substantia Nigra/cytology , Tandem Mass Spectrometry
18.
Neuroreport ; 20(16): 1481-5, 2009 Oct 28.
Article in English | MEDLINE | ID: mdl-19786922

ABSTRACT

To evaluate the effect of angiotensin II type1 receptor blocker on nerve regeneration delay in diabetic rats, nerve regeneration was monitored by a pinch test on the crushed sciatic nerves of streptozotocin-induced diabetic rats. Nerve regeneration was significantly delayed in diabetic rats and was partly ameliorated by treatment with olmesartan medoxomil (3 mg/kg/day, orally). In the ipsilateral dorsal root ganglia, the mRNA level of insulin-like growth factor-1 and ciliary neurotrophic factor (CNTF) was downregulated, whereas the mRNA level of neurotrophin-3 and CNTF receptor was upregulated. Olmesartan medoxomil significantly enhanced the CNTF expression. These results showed that angiotensin II type1 receptor blocker treatment is effective on nerve regeneration delay in diabetic animals and may provide an effective therapy for clinical diabetic neuropathy.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Diabetes Mellitus, Experimental/physiopathology , Imidazoles/pharmacology , Nerve Regeneration/drug effects , Sciatic Neuropathy/physiopathology , Tetrazoles/pharmacology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Animals , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/metabolism , Diabetes Mellitus, Experimental/chemically induced , Disease Models, Animal , Dose-Response Relationship, Drug , Functional Laterality/drug effects , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Gene Expression Regulation/drug effects , Imidazoles/therapeutic use , Insulin-Like Growth Factor I/analogs & derivatives , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Male , Neurotrophin 3/genetics , Neurotrophin 3/metabolism , Olmesartan Medoxomil , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Ciliary Neurotrophic Factor/genetics , Receptor, Ciliary Neurotrophic Factor/metabolism , Sciatic Neuropathy/drug therapy , Streptozocin , Tetrazoles/therapeutic use
19.
Biochim Biophys Acta ; 1788(9): 1890-900, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19482006

ABSTRACT

Ciliary neurotrophic factor (CNTF) signals via a receptor complex consisting of the specific CNTF receptor (CNTFR) and two promiscuous signal transducers, gp130 and leukemia inhibitory factor receptor (LIFR). Whereas earlier studies suggested that the signaling complex is a hexamer, more recent analyses strongly support a tetrameric structure. However, all studies so far analyzed the stoichiometry of the CNTF receptor complex in vitro and not in the context of living cells. We generated and expressed in mammalian cells acyl carrier protein-tagged versions of both CNTF and CNTFR. After labeling CNTF and CNTFR with different dyes we analyzed their diffusion behavior at the cell surface. Fluorescence (cross) correlation spectroscopy (FCS/FCCS) measurements reveal that CNTFR diffuses with a diffusion constant of about 2 x 10(-9) cm(2) s(-1) independent of whether CNTF is bound or not. FCS and FCCS measurements detect the formation of receptor complexes containing at least two CNTFs and CNTFRs. In addition, we measured Förster-type fluorescence resonance energy transfer between two differently labeled CNTFs within a receptor complex indicating a distance of 5-7 nm between the two. These findings are not consistent with a tetrameric structure of the CNTFR complex suggesting that either hexamers and or even higher-order structures (e.g. an octamer containing two tetramers) are formed.


Subject(s)
Receptor, Ciliary Neurotrophic Factor/chemistry , Animals , Cells, Cultured , Ciliary Neurotrophic Factor/metabolism , Cytokine Receptor gp130/metabolism , Dimerization , Fluorescence Resonance Energy Transfer , Humans , Microscopy, Confocal , Models, Chemical , Receptor, Ciliary Neurotrophic Factor/metabolism , Receptors, OSM-LIF/metabolism , Spectrometry, Fluorescence
20.
Int J Dev Biol ; 53(7): 947-53, 2009.
Article in English | MEDLINE | ID: mdl-19378247

ABSTRACT

Neural stem cells (NSC) self-renew and generate specialized cell types. There are reports indicating that Notch and Leukemia Inhibitory Factor (LIF) signaling are involved in cell determination of NSC, either preventing differentiation or promoting astrocytic fate. In this work, we aimed to compare the astrocytogenic effect of activated Notch with that induced by LIF. To this end, rat cerebral cortex neural progenitors/NSC were transduced with retroviral vectors in order to express green fluorescent protein (GFP), or a fusion protein of GFP with the active Notch1 intracellular domain (NICD). In parallel, other cultures were treated with increasing concentrations of LIF. We confirmed, in proliferating NSC, that LIF activated intracellular effectors by measuring STAT3 phosphorylation and Socs3 transcription. In NICD-expressing cells, Hes5 mRNA was induced, an effect not present in GFP-transduced NSC. We quantified the proportion of cells expressing Nestin in the presence of Fibroblast Growth Factor-2 (FGF-2) with LIF or NICD treatments. LIF significantly increased the proportion of cells co-expresssing Nestin and Glial Fibrillary Acidic Protein (GFAP), an effect absent in cells with activated Notch. After FGF2 withdrawal to promote differentiation, Nestin was markedly down-regulated, and neuronal and glial markers appeared in control cultures. LIF treatment caused a significant increase in the proportion of GFAP-positive cells, but cells expressing NICD showed a significantly higher percentage of astrocytes than control and LIF-treated cultures. These experiments show that cells stimulated with NICD differentiate more readily to astrocytes than LIF-treated NSC.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Leukemia Inhibitory Factor/pharmacology , Neurons/cytology , Neurons/metabolism , Receptor, Notch1/metabolism , Animals , Astrocytes/drug effects , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/drug effects , Cell Proliferation , Cells, Cultured , Ciliary Neurotrophic Factor/pharmacology , DNA Primers/genetics , Embryonic Stem Cells/drug effects , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Intermediate Filament Proteins/metabolism , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Nerve Tissue Proteins/metabolism , Nestin , Neurons/drug effects , Rats , Receptor, Ciliary Neurotrophic Factor/metabolism , Receptor, Notch1/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...