Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Front Mol Neurosci ; 13: 143, 2020.
Article in English | MEDLINE | ID: mdl-32982687

ABSTRACT

Patients suffering from temporal lobe epilepsy (TLE) show severe problems in hippocampus dependent memory consolidation. Memory consolidation strongly depends on an intact dialog between the hippocampus and neocortical structures. Deficits in hippocampal signal transmission are known to provoke disturbances in memory formation. In the present study, we investigate changes of synaptic plasticity at hippocampal output structures in an experimental animal model of TLE. In pilocarpine-treated rats, we found suppressed long-term potentiation (LTP) in hippocampal and parahippocampal regions such as the subiculum and the entorhinal cortex (EC). Subsequently we focused on the subiculum, serving as the major relay station between the hippocampus proper and downstream structures. In control animals, subicular pyramidal cells express different forms of LTP depending on their intrinsic firing pattern. In line with our extracellular recordings, we could show that LTP could only be induced in a minority of subicular pyramidal neurons. We demonstrate that a well-characterized cAMP-dependent signaling pathway involved in presynaptic forms of LTP is perturbed in pilocarpine-treated animals. Our findings suggest that in TLE, disturbances of synaptic plasticity may influence the information flow between the hippocampus and the neocortex.

2.
Article in English | MEDLINE | ID: mdl-31057390

ABSTRACT

The group I metabotropic glutamate (mGlu) receptor subtypes, mGlu1 and mGlu5, strongly regulate hippocampal synaptic plasticity. Both harbor PSD-95/discs-large/ZO-1 (PDZ) motifs at their extreme carboxyl terminals, which allow interaction with the PDZ domain of Tamalin, regulate the cell surface expression of group I mGlu receptors, and may modulate their coupling to signaling proteins. We investigated the functional role of this interaction in hippocampal long-term depression (LTD). Acute intracerebral treatment of adult rats with a cell-permeable PDZ-blocking peptide (pep-mGluR-STL), designed to competitively inhibit the interaction between Tamalin and group 1 mGlu receptors, prevented expression of LTD in the hippocampal CA1 region without affecting long-term potentiation (LTP) or basal synaptic transmission. Pep-mGluR-STL prevented facilitation by the group I mGlu receptor agonist, (S)-3,5-Dihydroxyphenylglycine (DHPG), and the mGlu5 agonist, (R,S)-2-chloro-5-Hydroxyphenylglycine (CHPG), of short-term depression (STD) into LTD, suggesting that Tamalin preferentially acts by mediating signaling through mGlu5. These data support that Tamalin is essential for the persistent expression of LTD and that it subserves the effective signaling of group 1 mGlu receptors.

3.
Gene Expr Patterns ; 12(1-2): 53-62, 2012.
Article in English | MEDLINE | ID: mdl-22138150

ABSTRACT

Visinin like 1 (Vsnl1) encodes a calcium binding protein which is well conserved between species. It was originally found in the brain and its biological functions in central nervous system have been addressed in several studies. Low expression levels have also been found in some peripheral organs, but very little information is available regarding its physiological roles in non-neuronal tissues. Except for the kidney, the expression pattern of Vsnl1 mRNA and protein has not yet been addressed during embryogenesis. By in situ hybridization and immunolabeling we have extensively analyzed the expression pattern of Vsnl1 during murine development. Vsnl1 specifies the cardiac primordia and its expression becomes restricted to the atrial myocardium after heart looping. However, in the adult heart, Vsnl1 is expressed by all four cardiac chambers. It also serves as a specific marker for the cardiomyocyte-derived structures in the systemic and pulmonary circulation. Vsnl1 is dynamically expressed also by many other organs during development e.g. taste buds, cochlea, thyroid, tooth, salivary and adrenal gland. The stage specific expression pattern of Vsnl1 makes it a potentially useful marker particularly in studies of cardiac and vascular morphogenesis.


Subject(s)
Embryonic Development , Gene Expression Regulation, Developmental , Neurocalcin/metabolism , Animals , Biomarkers , Brain/cytology , Brain/embryology , Brain/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Heart Atria/cytology , Heart Atria/embryology , Heart Atria/metabolism , In Situ Hybridization , Mice , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Neurocalcin/genetics , Pregnancy , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
PLoS One ; 6(11): e26793, 2011.
Article in English | MEDLINE | ID: mdl-22073194

ABSTRACT

The NCS protein Visinin-like Protein 1 (VILIP-1) transduces calcium signals in the brain and serves as an effector of the non-retinal receptor guanylyl cyclases (GCs) GC-A and GC-B, and nicotinic acetyl choline receptors (nAchR). Analysis of the quaternary structure of VILIP-1 in solution reveals the existence of monomeric and dimeric species, the relative contents of which are affected but not exclusively regulated by divalent metal ions and Redox conditions. Using small-angle X-ray scattering, we have investigated the low resolution structure of the calcium-bound VILIP-1 dimer under reducing conditions. Scattering profiles for samples with high monomeric and dimeric contents have been obtained. The dimerization interface involves residues from EF-hand regions EF3 and EF4.Using monolayer adsorption experiments, we show that myristoylated and unmyristoylated VILIP-1 can bind lipid membranes. The presence of calcium only marginally improves binding of the protein to the monolayer, suggesting that charged residues at the protein surface may play a role in the binding process.In the presence of calcium, VILIP-1 undergoes a conformational re-arrangement, exposing previously hidden surfaces for interaction with protein partners. We hypothesise a working model where dimeric VILIP-1 interacts with the membrane where it binds membrane-bound receptors in a calcium-dependent manner.


Subject(s)
Neurocalcin/chemistry , Neurocalcin/physiology , Amino Acid Sequence , Cations, Divalent , Cell Line , Chromatography, Gel , Models, Molecular , Molecular Sequence Data , Oxidation-Reduction , Protein Conformation , Scattering, Radiation , Sequence Homology, Amino Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
5.
Mol Carcinog ; 50(5): 319-33, 2011 May.
Article in English | MEDLINE | ID: mdl-21480386

ABSTRACT

VILIP-1 (visinin-like protein 1) is downregulated in various human squamous cell carcinoma (SCC). In a mouse skin SCC model VILIP-1 expression is reduced in aggressive tumor cells, accompanied by reduced cAMP levels. Overexpression of VILIP-1 in aggressive SCC cells led to enhanced cAMP production, in turn causing a reduction in invasive properties. Moreover, in primary neurons and neuronal tumor lines VILIP-1 enhanced cGMP signaling. Here, we set out to determine whether and how cAMP and cGMP signaling contribute to the VILIP-1 effect on enhanced SCC model cell migration, and thus most likely invasiveness in vivo. We found stronger increase in cGMP levels in aggressive, VILIP-1-negative SCC cells following stimulation of guanylyl cyclases NPR-A and -B with the natriuretic peptides ANP and CNP, respectively. Incubation with ANP or 8Br-cGMP to increase cGMP levels further enhanced the migration capacity of aggressive cells, whereas cell adhesion was unaffected. Increased cGMP was caused by elevated expression levels of NPR-A and -B. However, the expression level of VILIP-1 did not affect cGMP signaling and guanylyl cyclase expression in SCC. In contrast, VILIP-1 led to reduced migration of aggressive SCC cells depending on cAMP levels as shown by use of adenylyl cyclase (AC) inhibitor 2',3'-dideoxyadenosine. Involvement of cAMP-effectors PKA and EPAC play a role downstream of AC activation. VILIP-1-positive and -negative cells did not differ in mRNA expression of ACs, but an effect on enhanced protein expression and membrane localization of ACs was shown to underlie enhancement of cAMP production and, thus, reduction in cell migration by VILIP-1.


Subject(s)
Cell Movement/physiology , Cyclic AMP/metabolism , Guanosine Monophosphate/metabolism , Neurocalcin/metabolism , Signal Transduction/physiology , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Animals , Atrial Natriuretic Factor/pharmacology , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/drug effects , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Dideoxyadenosine/pharmacology , Humans , Mice , Microscopy, Fluorescence , Neurocalcin/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
6.
J Am Soc Nephrol ; 22(2): 274-84, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21289216

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) is indispensable for ureteric budding and branching. If applied exogenously, GDNF promotes ectopic ureteric buds from the Wolffian duct. Although several downstream effectors of GDNF are known, the identification of early response genes is incomplete. Here, microarray screening detected several GDNF-regulated genes in the Wolffian duct, including Visinin like 1 (Vsnl1), which encodes a neuronal calcium-sensor protein. We observed renal Vsnl1 expression exclusively in the ureteric epithelium, but not in Gdnf-null kidneys. In the tissue culture of Gdnf-deficient kidney primordium, exogenous GDNF and alternative bud inducers (FGF7 and follistatin) restored Vsnl1 expression. Hence, Vsnl1 characterizes the tip of the ureteric bud epithelium regardless of the inducer. In the tips, Vsnl1 showed a mosaic expression pattern that was mutually exclusive with ß-catenin transcriptional activation. Vsnl1 was downregulated in both ß-catenin-stabilized and ß-catenin-deficient kidneys. Moreover, in a mouse collecting duct cell line, Vsnl1 compromised ß-catenin stability, suggesting a counteracting relationship between Vsnl1 and ß-catenin. In summary, Vsnl1 marks ureteric bud tips in embryonic kidneys, and its mosaic pattern demonstrates a heterogeneity of cell types that may be critical for normal ureteric branching.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/physiology , Neurocalcin/physiology , Ureter/embryology , Animals , Biomarkers , Calcium/metabolism , Cell Cycle , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Phosphorylation , beta Catenin/physiology
7.
PLoS One ; 5(4): e10196, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20419170

ABSTRACT

VILIP-1, a member of the neuronal Ca(2+) sensor protein family, is able to act as a tumor suppressor in carcinoma cells by inhibiting cell proliferation and migration. In order to study the role of VILIP-1 in skin carcinogenesis we generated transgenic mice overexpressing VILIP-1 in epidermis under the control of the bovine keratin K5 promoter (K5-VILIP-1). We studied the susceptibility of FVB wild type and VILIP-1 transgenic mice to chemically mediated carcinogenesis. After 30 weeks of treatment with a two-stage carcinogenesis protocol, all animals showed numerous skin tumors. Nevertheless, K5-VILIP-1 mice showed decreased squamous cell carcinoma (SCC) multiplicity of approximately 49% (p<0.02) with respect to the corresponding SCC multiplicity observed in wild type (WT) mice. In addition, the relative percentage of low-grade cutaneous SCCs grade I (defined by the differentiation pattern according to the Broders grading scale) increased approximately 50% in the K5-VILIP1 mice when compared with SCCs in WT mice. Similar tendency was observed using a complete carcinogenesis protocol for skin carcinogenesis using benzo(a)pyrene (B(a)P). Further studies of tumors and primary epidermal keratinocyte cultures showed that matrix metalloproteinase 9 (MMP-9) levels and cell proliferation decreased in K5-VILIP-1 mice when compared with their wild counterparts. In addition tissue inhibitor of metalloproteinase 1 (TIMP-1) expression was higher in K5-VILIP-1 keratinocytes. These results show that VILIP-1 overexpression decreases the susceptibility to skin carcinogenesis in experimental mouse cancer models, thus supporting its role as a tumor suppressor gene.


Subject(s)
Keratinocytes/cytology , Neurocalcin/physiology , Skin Neoplasms/etiology , Animals , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Disease Susceptibility , Genes, Tumor Suppressor , Matrix Metalloproteinase 9 , Mice , Mice, Transgenic , Neurocalcin/genetics , Skin Neoplasms/pathology , Tissue Inhibitor of Metalloproteinase-1 , Tumor Suppressor Proteins/genetics
8.
Regul Pept ; 161(1-3): 51-7, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20079378

ABSTRACT

Accumulating evidence indicates that Visinin-like protein-1 (VILIP-1), a member of the family of neuronal calcium sensor proteins (NCS), modulates a variety of processes in extra-neuronal tissues. In this study, we describe VILIP-1 expression in the human heart, rat cardiomyocytes, and H9c2 cells, and demonstrate that VILIP-1 regulates the cell surface localization of natriuretic peptide receptor B (NPR-B). In preparations from failing hearts, we observed VILIP-1 downregulation and reduced NPR-B signalling. In conclusion, VILIP-1 deficiency may be responsible for the reduced efficiency of the natriuretic peptide system in cardiac hypertrophy and heart failure and may therefore serve as pharmacological target.


Subject(s)
Myocytes, Cardiac/metabolism , Neurocalcin/physiology , Receptors, Atrial Natriuretic Factor/metabolism , Animals , Blotting, Western , Cell Line , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Hemodynamics , Humans , Immunohistochemistry , Myocardial Infarction/metabolism , Myocardium/metabolism , Neurocalcin/deficiency , Neurocalcin/genetics , Neurocalcin/metabolism , Polymerase Chain Reaction , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology , Rats , Rats, Sprague-Dawley , Receptors, Atrial Natriuretic Factor/genetics
9.
J Biol Chem ; 284(40): 27577-86, 2009 Oct 02.
Article in English | MEDLINE | ID: mdl-19674972

ABSTRACT

VILIP-1 (gene name VSNL1), a member of the neuronal Ca(2+) sensor protein family, acts as a tumor suppressor gene by inhibiting cell proliferation, adhesion, and invasiveness. VILIP-1 expression is down-regulated in several types of human cancer. In human non-small cell lung cancer, we found that down-regulation was due to epigenetic changes. Consequently, in this study we analyzed the VSNL1 promoter and its regulation. Serial truncation of the proximal 2-kb VSNL1 promoter (VP-1998) from its 5' terminus disclosed that the last 3' terminal 100-bp promoter fragment maintained similar promoter activity as compared with VP-1998 and therefore was referred to as VSNL1 minimal promoter. When the 5' terminal 50 bp were deleted from the minimal promoter, the activity was dramatically decreased, suggesting that the deleted 50 bp contained a potential cis-acting element crucial for promoter activity. Deletion and site-directed mutagenesis combined with in silico transcription factor binding analysis of VSNL1 promoter identified nuclear respiratory factor (NRF)-1/alpha-PAL as a major player in regulating VSNL1 minimal promoter activity. The function of NRF-1 was further confirmed using dominant-negative NRF-1 overexpression and NRF-1 small interfering RNA knockdown. Electrophoretic mobility shift assay and chromatin immunoprecipitation provided evidence for direct NRF-1 binding to the VSNL1 promoter. Methylation of the NRF-1-binding site was found to be able to regulate VSNL1 promoter activity. Our results further indicated that NRF-1 could be a regulatory factor for gene expression of the other visinin-like subfamily members including HPCAL4, HPCAL1, HPCA, and NCALD.


Subject(s)
Gene Expression Regulation , Neurocalcin/genetics , Nuclear Respiratory Factor 1/metabolism , Promoter Regions, Genetic/genetics , Animals , Binding Sites , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA/metabolism , Gene Knockdown Techniques , Humans , Methylation , Neurocalcin/deficiency , Neurocalcin/metabolism , RNA, Small Interfering/genetics , Sequence Deletion , Transcription, Genetic , Transfection
11.
Cell Tissue Res ; 335(2): 301-16, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18989702

ABSTRACT

The visinin-like protein (VSNL) subfamily, including VILIP-1 (the founder protein), VILIP-2, VILIP-3, hippocalcin, and neurocalcin delta, constitute a highly homologous subfamily of neuronal calcium sensor (NCS) proteins. Comparative studies have shown that VSNLs are expressed predominantly in the brain with restricted expression patterns in various subsets of neurons but are also found in peripheral organs. In addition, the proteins display differences in their calcium affinities, in their membrane-binding kinetics, and in the intracellular targets to which they associate after calcium binding. Even though the proteins use a similar calcium-myristoyl switch mechanism to translocate to cellular membranes, they show calcium-dependent localization to various subcellular compartments when expressed in the same neuron. These distinct calcium-myristoyl switch properties might be explained by specificity for defined phospholipids and membrane-bound targets; this enables VSNLs to modulate various cellular signal transduction pathways, including cyclic nucleotide and MAPK signaling. An emerging theme is the direct or indirect effect of VSNLs on gene expression and their interaction with components of membrane trafficking complexes, with a possible role in membrane trafficking of different receptors and ion channels, such as glutamate receptors of the kainate and AMPA subtype, nicotinic acetylcholine receptors, and Ca(2+)-channels. One hypothesis is that the highly homologous VSNLs have evolved to fulfil specialized functions in membrane trafficking and thereby affect neuronal signaling and differentiation in defined subsets of neurons. VSNLs are involved in differentiation processes showing a tumor-invasion-suppressor function in peripheral organs. Finally, VSNLs play neuroprotective and neurotoxic roles and have been implicated in neurodegenerative diseases.


Subject(s)
Calcium/metabolism , Central Nervous System/metabolism , Neurocalcin/metabolism , Neuronal Calcium-Sensor Proteins/metabolism , Amino Acid Sequence , Animals , Gene Expression , Humans , Molecular Sequence Data , Protein Transport , Signal Transduction
12.
Cell Mol Neurobiol ; 29(2): 273-86, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18925431

ABSTRACT

Visinin-like protein (VILIP-1) belongs to the neuronal Ca2+ sensor family of EF-hand Ca2+-binding proteins that regulate a variety of Ca2+-dependent signal transduction processes in neurons. It is an interaction partner of alpha4beta2 nicotinic acetylcholine receptor (nAChR) and increases surface expression level and agonist sensitivity of the receptor in oocytes. Nicotine stimulation of nicotinic receptors has been reported to lead to an increase in intracellular Ca2+ concentration by Ca2+-permeable nAChRs, which in turn might lead to activation of VILIP-1, by a mechanism described as the Ca2+-myristoyl switch. It has been postulated that this will lead to co-localization of the proteins at cell membranes, where VILIP-1 can influence functional activity of alpha4-containing nAChRs. In order to test this hypothesis we have investigated whether a nicotine-induced and reversible Ca2+-myristoyl switch of VILIP-1 exists in primary hippocampal neurons and whether pharmacological agents, such as antagonist specific for distinct nAChRs, can interfere with the Ca2+-dependent membrane localization of VILIP-1. Here we report, that only alpha7- but not alpha4-containing nAChRs are able to elicit a Ca2+-dependent and reversible membrane-translocation of VILIP-1 in interneurons as revealed by employing the specific receptor antagonists dihydro-beta-erythroidine and methylallylaconitine. The nAChRs are associated with processes of synaptic plasticity in hippocampal neurons and they have been implicated in the pathology of CNS disorders, including Alzheimer's disease and schizophrenia. VILIP-1 might provide a novel functional crosstalk between alpha4- and alpha7-containing nAChRs.


Subject(s)
Calcium Signaling/physiology , Hippocampus/metabolism , Neurocalcin/metabolism , Neurons/metabolism , Receptors, Calcium-Sensing/metabolism , Receptors, Nicotinic/metabolism , Animals , Calcium Signaling/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Dihydro-beta-Erythroidine/pharmacology , Hippocampus/drug effects , Neurocalcin/drug effects , Neurons/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Protein Transport/drug effects , Protein Transport/physiology , Rats , Rats, Wistar , Receptor Cross-Talk/drug effects , Receptor Cross-Talk/physiology , Receptors, Calcium-Sensing/drug effects , Receptors, Nicotinic/drug effects , alpha7 Nicotinic Acetylcholine Receptor
13.
Biochem Biophys Res Commun ; 377(3): 820-5, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-18952057

ABSTRACT

The cGMP producing natriuretic peptide receptor B (NPR-B) and its ligand C-type natriuretic peptide (CNP) are widely distributed in the brain and are highly expressed in the hippocampal regions CA1-CA3. To date only limited functional data is available concerning the physiological effects of the peptide hormone in the hippocampus. Therefore, we were interested in how bath application of the peptide hormone might influence synaptic plasticity following high frequency stimulation (HFS). We found that CNP application decreased the population spike (PS) amplitude after HFS, thereby affecting long-term potentiation (LTP) in acute hippocampal slices. To investigate the molecular consequences of CNP application leading to a decrease in PS amplitude, we further analyzed the impact of the hormone on the number of presynaptic synapsin I clusters and number of postsynaptic AMPA receptor subunit GluR1 clusters as well as their co-localization in a primary hippocampal cell culture system. The observed pre-and postsynaptic effects after CNP stimulation of the cGMP pathway in hippocampal cell cultures may underlie the effect of the peptide hormone on LTP.


Subject(s)
Hippocampus/physiology , Long-Term Potentiation , Natriuretic Peptide, C-Type/physiology , Synapses/physiology , Animals , Cells, Cultured , Hippocampus/drug effects , Hippocampus/metabolism , Natriuretic Peptide, C-Type/pharmacology , Rats , Rats, Wistar , Receptors, AMPA/metabolism , Synapses/drug effects , Synapsins/metabolism , Synaptic Potentials/drug effects
14.
Neurobiol Dis ; 32(1): 162-75, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18691652

ABSTRACT

Post mortem studies in the hippocampus of schizophrenia patients revealed increased expression of neuronal Ca(2+)-sensor VILIP-1 (visinin-like protein) and enhanced co-localization with alpha4beta2 nAChR in interneurons. To study the pathological role of VILIP-1, particularly in interneurons, in the context of the glutamate hypothesis of schizophrenia, we have used ketamine-treated rats, a NMDA receptor hypofunction model, and hippocampal cultures as model systems for schizophrenia. Treatment with ketamine leads to enhanced VILIP-1 expression in interneurons in rat hippocampal CA1 region. In cultures glutamate treatment led to an increase in VILIP-1-positive interneurons, which is not dependent on NMDA receptor but metabotropic glutamate receptor activation. VILIP-1 mainly co-localizes with the interneuron marker calretinin, mGluR1alpha and the VILIP-1 interaction partner alpha4beta2 nAChR in hippocampal slices. Overexpression of VILIP-1 leads to enhanced nAChR-dependent inhibitory postsynaptic current (IPSC) generation by interneurons. This novel molecular link between the pathological role of mGluRs, VILIP-1 and its interaction partner alpha4beta2 nAChR by converging pathological glutamatergic and nicotinergic transmission may underlie cognitive impairments in schizophrenia.


Subject(s)
Calcium/metabolism , Glutamic Acid/physiology , Hippocampus/metabolism , Intracellular Calcium-Sensing Proteins/metabolism , Neurocalcin/metabolism , Neurons/metabolism , Schizophrenia/metabolism , Animals , Cells, Cultured , Dentate Gyrus/metabolism , Female , Glutamic Acid/metabolism , Hippocampus/pathology , Humans , Interneurons/metabolism , Interneurons/pathology , Ketamine/administration & dosage , Male , Neurons/pathology , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/metabolism , Receptors, Metabotropic Glutamate/physiology , Receptors, Nicotinic/metabolism , Schizophrenia/pathology
15.
PLoS One ; 3(2): e1698, 2008 Feb 27.
Article in English | MEDLINE | ID: mdl-18301774

ABSTRACT

VILIP-1, a member of the neuronal Ca++ sensor protein family, acts as a tumor suppressor gene in an experimental animal model by inhibiting cell proliferation, adhesion and invasiveness of squamous cell carcinoma cells. Western Blot analysis of human tumor cells showed that VILIP-1 expression was undetectable in several types of human tumor cells, including 11 out of 12 non-small cell lung carcinoma (NSCLC) cell lines. The down-regulation of VILIP-1 was due to loss of VILIP-1 mRNA transcripts. Rearrangements, large gene deletions or mutations were not found. Hypermethylation of the VILIP-1 promoter played an important role in gene silencing. In most VILIP-1-silent cells the VILIP-1 promoter was methylated. In vitro methylation of the VILIP-1 promoter reduced its activity in a promoter-reporter assay. Transcriptional activity of endogenous VILIP-1 promoter was recovered by treatment with 5'-aza-2'-deoxycytidine (5'-Aza-dC). Trichostatin A (TSA), a histone deacetylase inhibitor, potently induced VILIP-1 expression, indicating that histone deacetylation is an additional mechanism of VILIP-1 silencing. TSA increased histone H3 and H4 acetylation in the region of the VILIP-1 promoter. Furthermore, statistical analysis of expression and promoter methylation (n = 150 primary NSCLC samples) showed a significant relationship between promoter methylation and protein expression downregulation as well as between survival and decreased or absent VILIP-1 expression in lung cancer tissues (p<0.0001). VILIP-1 expression is silenced by promoter hypermethylation and histone deacetylation in aggressive NSCLC cell lines and primary tumors and its clinical evaluation could have a role as a predictor of short-term survival in lung cancer patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Down-Regulation/genetics , Gene Silencing , Neurocalcin/genetics , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/mortality , DNA Methylation , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Promoter Regions, Genetic , RNA, Messenger/genetics , Survival Rate
16.
J Biol Chem ; 281(31): 21942-21953, 2006 Aug 04.
Article in English | MEDLINE | ID: mdl-16731532

ABSTRACT

Visinin-like protein-1 (VILIP-1) is a member of the neuronal Ca2+ sensor protein family that modulates Ca2+-dependent cell signaling events. VILIP-1, which is expressed primarily in the brain, increases cAMP formation in neural cells by modulating adenylyl cyclase, but its functional role in other tissues remains largely unknown. In this study, we demonstrate that VILIP-1 is expressed in murine pancreatic islets and beta-cells. To gain insight into the functions of VILIP-1 in beta-cells, we used both overexpression and small interfering RNA knockdown strategies. Overexpression of VILIP-1 in the MIN6 beta-cell line or isolated mouse islets had no effect on basal insulin secretion but significantly increased glucose-stimulated insulin secretion. cAMP accumulation was elevated in VILIP-1-overexpressing cells, and the protein kinase A inhibitor H-89 attenuated increased glucose-stimulated insulin secretion. Overexpression of VILIP-1 in isolated mouse beta-cells increased cAMP content accompanied by increased cAMP-responsive element-binding protein gene expression and enhanced exocytosis as detected by cell capacitance measurements. Conversely, VILIP-1 knockdown by small interfering RNA caused a reduction in cAMP accumulation and produced a dramatic increase in preproinsulin mRNA, basal insulin secretion, and total cellular insulin content. The increase in preproinsulin mRNA in these cells was attributed to enhanced insulin gene transcription. Taken together, we have shown that VILIP-1 is expressed in pancreatic beta-cells and modulates insulin secretion. Increased VILIP-1 enhanced insulin secretion in a cAMP-associated manner. Down-regulation of VILIP-1 was accompanied by decreased cAMP accumulation but increased insulin gene transcription.


Subject(s)
Insulin/metabolism , Islets of Langerhans/metabolism , Neurocalcin/physiology , Animals , Cyclic AMP , Cyclic AMP-Dependent Protein Kinases , Exocytosis , Gene Expression Regulation , Insulin/genetics , Insulin Secretion , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred Strains , Neurocalcin/biosynthesis , Neurocalcin/genetics , RNA, Small Interfering/pharmacology , Transcription, Genetic , Transfection
17.
Mol Carcinog ; 45(8): 572-81, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16683251

ABSTRACT

EF-hand Ca(2+)-sensor proteins are key molecules for transducing Ca(2+) signals into physiological answers and changes in cytosolic Ca(2+) concentration control a variety of cellular responses, including proliferation, migration, and differentiation, which are relevant for tumor progression. The Ca(2+)-sensor visinin-like protein-1 (VILIP-1) has recently attracted major interest due to its putative tumor suppressor function. Whereas VILIP-1 is expressed in normal skin, it is downregulated in skin tumors in a murine tumor model. The aim of this study was to investigate the expression of the Ca(2+)-sensor VILIP-1 in squamous cell carcinoma of the esophagus and to correlate expression levels with clinicopathological features of the tumor. We examined VILIP-1 expression in 54 specimens of esophageal squamous cell carcinomas and 24 normal esophagus tissues, with immunohistochemical staining and immunofluorescence co-staining techniques. VILIP-1 expression was completely lost or significantly reduced in esophageal tumor tissue compared with normal squamous epithelium. Correlation with clinicopathological features indicated that there was significantly less VILIP-1 expression in lymph node positive (N = 1) versus lymph node negative (N = 0) tumors (P = 0.002). Although there was no significant difference between highly (G(1)), moderately (G(2)) and poorly differentiated (G(3)) tumors (P = 0.177), VILIP-1 expression in tumors is significantly correlated with the depth of tumor invasion (P = 0.028 between T1, T2, T3, and T4). In contrast, co-staining with the proliferation marker Ki-67 indicated no significant correlation with proliferation rates in tumors (Ki-67 index of the tumor). In summary, the expression of the Ca(2+)-sensor VILIP-1 was found to be lost during development of squamous cell carcinoma of the esophagus. The protein expression level significantly correlates with invasive features, such as depth of tumor invasion and local lymph node metastasis, but not with proliferation rate of tumor cells.


Subject(s)
Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , Neurocalcin/analysis , Aged , Aged, 80 and over , Calcium Signaling , Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Female , Humans , Ki-67 Antigen/analysis , Lymphatic Metastasis , Male , Middle Aged , Neurocalcin/metabolism
18.
J Cell Sci ; 118(Pt 11): 2495-505, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15923662

ABSTRACT

The family of neuronal Ca2+ sensor (NCS) proteins is known to influence a variety of physiological and pathological processes by affecting signalling of different receptors and ion channels. Recently, it has been shown that the NCS protein VILIP-1 influences the activity of the receptor guanylyl cyclase GC-B. In transfected cell lines, VILIP-1 performs a Ca2+-dependent membrane association, the reversible Ca2+-myristoyl switch of VILIP-1, which leads to an increase in natriuretic peptide-stimulated cGMP levels. In this study, we have investigated the effect of VILIP-1 on cGMP signalling in C6 cells and in primary hippocampal neurons, where VILIP-1 and GC-B are co-expressed in many but not all neurons and partially co-localize in the soma and in dendrites. Our data indicate that VILIP-1 modulates GC-B activity by influencing clathrin-dependent receptor recycling. These data support a general physiological role for VILIP-1 in membrane trafficking in the intact hippocampus, where the NCS protein may affect processes, such as neuronal differentiation and synaptic plasticity e.g. by influencing cGMP-signalling.


Subject(s)
Calcium Signaling/physiology , Dendrites/metabolism , Guanylate Cyclase/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Receptors, Calcium-Sensing/metabolism , Animals , Calcium Signaling/genetics , Cells, Cultured , Clathrin/metabolism , Cyclic GMP/metabolism , Hippocampus/cytology , Nerve Tissue Proteins/genetics , Neurocalcin , Protein Transport/genetics , Protein Transport/physiology , Rats , Rats, Wistar , Receptors, Calcium-Sensing/genetics , Transfection
19.
Trends Pharmacol Sci ; 26(7): 345-51, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15982480

ABSTRACT

Neuronal Ca2+-sensor (NCS) proteins constitute a subfamily of closely related EF-hand Ca2+-binding proteins that are expressed mainly in neurons or retinal photoreceptor cells. A variety of different neuronal functions have been attributed to these proteins. However, important new discoveries indicate that these proteins also have key roles in pathological processes of disease. Thus, a 'darker side' of NCS protein signaling has become evident in some CNS disorders, such as Alzheimer's disease and schizophrenia, and in cancer. In this article, I will provide an overview of the current, and rapidly expanding, knowledge of how this important family of proteins might be involved in various major CNS diseases, the proposed role of NCS proteins in pathophysiological signaling and the development of pharmacological tools and novel research strategies.


Subject(s)
Alzheimer Disease/physiopathology , Calcium Signaling/physiology , Calcium-Binding Proteins/physiology , Neoplasms/physiopathology , Nerve Tissue Proteins/physiology , Neurons/physiology , Humans , Neurodegenerative Diseases/physiopathology , Neuronal Calcium-Sensor Proteins , Neuropeptides , Schizophrenia/physiopathology
20.
Oncogene ; 24(14): 2307-16, 2005 Mar 31.
Article in English | MEDLINE | ID: mdl-15735716

ABSTRACT

Tumor cell invasion is a highly integrated and complex process comprising several biologically distinct functions such as cell adhesion, motility, proteolysis, etc. Visinin-like protein-1 (VILIP-1), a member of the neuronal EF-hand calcium-sensor protein family, plays a role in regulating tumor cell invasiveness of mouse squamous cell carcinoma (SCC). VILIP-1 enhances cyclic adenosine monophosphate levels through PKA induction. However, the mechanism by which VILIP-1 reduces cell invasiveness is not well understood. In this study, we show that VILIP-1 decreased cell adhesion and migration/invasiveness of highly invasive mouse SCC cells. Forced expression of VILIP-1 reduced cell adhesion to fibronectin in parallel to downregulating alphav and alpha5 integrin subunit levels. VILIP-1 overexpression also led to decreased migration ability. Conversely, short hairpin RNA-mediated VILIP-1 knock-down of SCC cells' characterized by little or no invasiveness, correlated with increased adhesion to fibronectin and enhanced expression of alphav and alpha5 integrin subunits together with increased cell migration. Function-blocking assays with inhibitory anti-alpha5 and anti-alphav integrin antibodies showed that both subunits contributed to cell adhesion, migration, and invasiveness of highly invasive SCC cell lines. These results point to a critical role of VILIP-1 in regulating cell adhesion and migration by downregulation of fibronectin receptor expression. Decreased or absent VILIP-1 expression in SCC cell subpopulations may lead to a more advanced malignant phenotype characterized by changes in adhesive ability and increased cell motility, suggestive of a tumor suppressor function.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cell Adhesion/physiology , Cell Movement/physiology , Nerve Tissue Proteins/physiology , Receptors, Calcium-Sensing/physiology , Animals , Antibodies/immunology , Base Sequence , Blotting, Western , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , DNA Primers , Integrins/antagonists & inhibitors , Integrins/immunology , Mice , Neurocalcin
SELECTION OF CITATIONS
SEARCH DETAIL
...