Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 131(15)2018 08 03.
Article in English | MEDLINE | ID: mdl-29930088

ABSTRACT

The cardiac muscle ryanodine receptor-Ca2+ release channel (RyR2) constitutes the sarcoplasmic reticulum (SR) Ca2+ efflux mechanism that initiates myocyte contraction, while cardiac myosin-binding protein-C (cMyBP-C; also known as MYBPC3) mediates regulation of acto-myosin cross-bridge cycling. In this paper, we provide the first evidence for the presence of direct interaction between these two proteins, forming a RyR2-cMyBP-C complex. The C-terminus of cMyBP-C binds with the RyR2 N-terminus in mammalian cells and the interaction is not mediated by a fibronectin-like domain. Notably, we detected complex formation between both recombinant cMyBP-C and RyR2, as well as between the native proteins in cardiac tissue. Cellular Ca2+ dynamics in HEK293 cells is altered upon co-expression of cMyBP-C and RyR2, with lowered frequency of RyR2-mediated spontaneous Ca2+ oscillations, suggesting that cMyBP-C exerts a potential inhibitory effect on RyR2-dependent Ca2+ release. Discovery of a functional RyR2 association with cMyBP-C provides direct evidence for a putative mechanistic link between cytosolic soluble cMyBP-C and SR-mediated Ca2+ release, via RyR2. Importantly, this interaction may have clinical relevance to the observed cMyBP-C and RyR2 dysfunction in cardiac pathologies, such as hypertrophic cardiomyopathy.


Subject(s)
Carrier Proteins/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Calcium/metabolism , Calcium Signaling/physiology , Cytosol/metabolism , HEK293 Cells , Humans , Protein Binding , Sarcoplasmic Reticulum/metabolism
2.
Cell Calcium ; 59(1): 41-53, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26794932

ABSTRACT

The precise role of hormones binding to Gαq protein-coupled receptors (H-GαqPCRs) in chronic heart diseases remains poorly understood. To address this, we used a model of cultured adult rat ventricular myocytes stimulated with endothelin-1 (ET-1) or phenylephrine (PE) over a period of 8 days in vitro (DIV). Chronically treated cells showed an increased number of arrhythmogenic Ca(2+) transients when electrically paced at 0.5 Hz. While their post-rest behaviour was preserved, from DIV6 onwards the amplitude of caffeine-evoked Ca(2+) transients was increased in hormone-treated cells, suggesting an elevated sarcoplasmic reticulum Ca(2+) load. The duration of electrically evoked global Ca(2+) transients gradually increased over the culturing time indicating decreased activity of processes removing cytosolic Ca(2+). In treated cells, spontaneous Ca(2+) sparks displayed smaller amplitudes from DIV6 onwards, and a slower decay period for PE (from DIV3) and for ET-1 (from DIV6). This cellular functional remodelling was associated with changes in gene expression: chronic ET-1 treatment decreased PKCγ transcripts, whereas PE increased PKCγ and SERCA2a transcripts as probed by qPCR. Western blot analysis confirmed the upregulation of PKCγ with PE. To study ET-1 receptor desensitization in vivo, osmotic minipumps containing either NaCl or ET-1 were implanted in mice and Ca(2+) signalling was studied in acutely isolated ventricular myocytes after 2 weeks of chronic treatment. Interestingly, while cellular responses to isoproterenol stimulation were preserved in ET-1 treated animals, the inotropic response of myocytes to ET-1 stimulation was abrogated. We therefore conclude that chronic stimulation of cardiac myocytes by H-GαqPCRs induces cellular remodelling of Ca(2+) cycling with altered PKCγ expression and promotion of arrhythmogenic cellular responses.


Subject(s)
Calcium/metabolism , Endothelin-1/administration & dosage , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Animals , Cells, Cultured , Infusion Pumps, Implantable , Male , Rats , Rats, Wistar
4.
Front Cell Dev Biol ; 2: 36, 2014.
Article in English | MEDLINE | ID: mdl-25364743

ABSTRACT

The burden of neurodegenerative disorders in an aging population has become a challenge for the modern world. While the biomarkers available and the methods of diagnosis have improved to detect the onset of these diseases at early stages, the question of adapted and efficient therapies is still a major issue. The prospect of replacing the loss of functional neural cells remains an attractive but still audacious approach. A huge progress has been made in the generation of neurons derived from human stem cell lines and transplantation assays are tested in animals for a wide range of pathologies of the central nervous system. Here we take one step back and examine neuronal differentiation and the characterization of neural progenitors derived from human embryonic stem cells. We gather results from our previous studies and present a cell model that was successfully used in functional analyses and engraftment experiments. These neuronal precursors exhibit spontaneous and evoked activity, indicating that their electrophysiological and calcium handling properties are similar to those of matured neurons. Hence this summarized information will serve as a basis to design better stem cell-based therapies to improve neural regeneration.

5.
J Cell Sci ; 126(Pt 21): 5042-51, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23943880

ABSTRACT

The ryanodine receptor (RyR) is an ion channel composed of four identical subunits mediating calcium efflux from the endo/sarcoplasmic reticulum of excitable and non-excitable cells. We present several lines of evidence indicating that the RyR2 N-terminus is capable of self-association. A combination of yeast two-hybrid screens, co-immunoprecipitation analysis, chemical crosslinking and gel filtration assays collectively demonstrate that a RyR2 N-terminal fragment possesses the intrinsic ability to oligomerize, enabling apparent tetramer formation. Interestingly, N-terminus tetramerization mediated by endogenous disulfide bond formation occurs in native RyR2, but notably not in RyR1. Disruption of N-terminal inter-subunit interactions within RyR2 results in dysregulation of channel activation at diastolic Ca(2+) concentrations from ryanodine binding and single channel measurements. Our findings suggest that the N-terminus interactions mediating tetramer assembly are involved in RyR channel closure, identifying a crucial role for this structural association in the dynamic regulation of intracellular Ca(2+) release.


Subject(s)
Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/metabolism , Amino Acid Motifs , Animals , Calcium/metabolism , Humans , Myocytes, Cardiac/chemistry , Protein Multimerization , Rabbits , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum/metabolism , Swine
6.
J Biol Chem ; 288(23): 16671-16679, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23632022

ABSTRACT

Ryanodine receptor channels (RyR) are key components of striated muscle excitation-contraction coupling, and alterations in their function underlie both inherited and acquired disease. A full understanding of the disease process will require a detailed knowledge of the mechanisms and structures involved in RyR function. Unfortunately, high-resolution structural data, such as exist for K(+)-selective channels, are not available for RyR. In the absence of these data, we have used modeling to identify similarities in the structural elements of K(+) channel pore-forming regions and postulated equivalent regions of RyR. This has identified a sequence of residues in the cytosolic cavity-lining transmembrane helix of RyR (G(4864)LIIDA(4869) in RyR2) analogous to the glycine hinge motif present in many K(+) channels. Gating in these K(+) channels can be disrupted by substitution of residues for the hinge glycine. We investigated the involvement of glycine 4864 in RyR2 gating by monitoring properties of recombinant human RyR2 channels in which this glycine is replaced by residues that alter gating in K(+) channels. Our data demonstrate that introducing alanine at position 4864 produces no significant change in RyR2 function. In contrast, function is altered when glycine 4864 is replaced by either valine or proline, the former preventing channel opening and the latter modifying both ion translocation and gating. Our studies reveal novel information on the structural basis of RyR gating, identifying both similarities with, and differences from, K(+) channels. Glycine 4864 is not absolutely required for channel gating, but some flexibility at this point in the cavity-lining transmembrane helix is necessary for normal RyR function.


Subject(s)
Ion Channel Gating/physiology , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/metabolism , Amino Acid Substitution , Crystallography, X-Ray , Glycine/chemistry , Glycine/genetics , Glycine/metabolism , HEK293 Cells , Humans , Ion Transport/physiology , Mutation, Missense , Protein Structure, Tertiary , Ryanodine Receptor Calcium Release Channel/genetics
7.
BMC Genomics ; 14 Suppl 6: S5, 2013.
Article in English | MEDLINE | ID: mdl-24564251

ABSTRACT

Lung cancer accounts for the highest number of cancer-related deaths worldwide. Early diagnosis significantly increases the disease-free survival rate and a large amount of effort has been expended in screening trials and the development of early molecular diagnostics. However, a gold standard diagnostic strategy is not yet available. Here, based on miRNA expression profile in lung cancer and using a novel in silico reverse-transcriptomics approach, followed by analysis of the interactome; we have identified potential transcription factor (TF) markers that would facilitate diagnosis of subtype specific lung cancer. A subset of seven TF markers has been used in a microarray screen and was then validated by blood-based qPCR using stage-II and IV non-small cell lung carcinomas (NSCLC). Our results suggest that overexpression of HMGA1, E2F6, IRF1, and TFDP1 and downregulation or no expression of SUV39H1, RBL1, and HNRPD in blood is suitable for diagnosis of lung adenocarcinoma and squamous cell carcinoma sub-types of NSCLC. Here, E2F6 was, for the first time, found to be upregulated in NSCLC blood samples. The miRNA-TF-miRNA interaction based molecular mechanisms of these seven markers in NSCLC revealed that HMGA1 and TFDP1 play vital roles in lung cancer tumorigenesis. The strategy developed in this work is applicable to any other cancer or disease and can assist in the identification of potential biomarkers.


Subject(s)
Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Computer Simulation , Gene Expression Profiling/methods , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Reverse Transcription/genetics , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Lung Neoplasms/blood , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Sequence Annotation , Polymerase Chain Reaction , Reproducibility of Results , Small Cell Lung Carcinoma/blood , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Transcription Factors/metabolism
8.
Brain Res ; 1483: 1-12, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-22975133

ABSTRACT

Arginine-vasopressin (AVP) is a nonapeptide of hypothalamic origin that has been shown to exert many important cognitive and physiological functions in neurons and terminals of both the central and peripheral nervous system (CNS and PNS). Here we report for the first time that AVP induced an increase in intracellular Ca²âº concentration ([Ca²âº](i)) in non-neuronal cells isolated from the rat dorsal root ganglion (DRG) and cultured in vitro. The ratiometric [Ca²âº](i) measurements showed that AVP evoked [Ca²âº](i) responses in the non-neuronal cells and these concentration-dependent (100 pM to 1 µM) responses increased with days in vitro in culture, reaching a maximum amplitude after 4-5 day. Immunostaining by anti-S-100 antibody revealed that more than 70% of S-100 positive cells were AVP-responsive, indicating that glial cells responded to AVP and increased their [Ca²âº](i). The responses were inhibited by depletion of the intracellular Ca²âº stores or in the presence of inhibitors of phospholipase C, indicating a metabotropic response involving inositol trisphosphate, and were mediated by the V1 subclass of AVP receptors, as evidenced by the use of the specific blockers for V1 and OT receptors, (d(CH2)5¹,Tyr(Me)²,Arg8)-Vasopressin and (d(CH2)5¹,Tyr(Me)²,Thr4,Orn8,des-Gly-NH29)-Vasotocin, respectively. V(1a) but not V(1b) receptor mRNA was expressed sustainably through the culture period in cultured DRG cells. These results suggest that AVP modulates the activity of DRG glial cells via activation of V(1a) receptor.


Subject(s)
Calcium/metabolism , Ganglia, Spinal/cytology , Intracellular Fluid/metabolism , Neuroglia/drug effects , Vasoconstrictor Agents/pharmacology , Vasopressins/pharmacology , Animals , Arginine Vasopressin/analogs & derivatives , Arginine Vasopressin/pharmacology , Calcium Signaling/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Intracellular Fluid/drug effects , Male , Potassium Chloride/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism , S100 Proteins/metabolism , Time Factors
9.
J Gen Physiol ; 140(3): 325-39, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22930804

ABSTRACT

Although no high-resolution structural information is available for the ryanodine receptor (RyR) channel pore-forming region (PFR), molecular modeling has revealed broad structural similarities between this region and the equivalent region of K(+) channels. This study predicts that, as is the case in K(+) channels, RyR has a cytosolic vestibule lined with predominantly hydrophobic residues of transmembrane helices (TM10). In K(+) channels, this vestibule is the binding site for blocking tetraalkylammonium (TAA) cations and Shaker B inactivation peptides (ShBPs), which are stabilized by hydrophobic interactions involving specific residues of the lining helices. We have tested the hypothesis that the cytosolic vestibule of RyR fulfils a similar role and that TAAs and ShBPs are stabilized by hydrophobic interactions with residues of TM10. Both TAAs and ShBPs block RyR from the cytosolic side of the channel. By varying the composition of TAAs and ShBPs, we demonstrate that the affinity of both species is determined by their hydrophobicity, with variations reflecting alterations in the dissociation rate of the bound blockers. We investigated the role of TM10 residues of RyR by monitoring block by TAAs and ShBPs in channels in which the hydrophobicity of individual TM10 residues was lowered by alanine substitution. Although substitutions changed the kinetics of TAA interaction, they produced no significant changes in ShBP kinetics, indicating the absence of specific hydrophobic sites of interactions between RyR and these peptides. Our investigations (a) provide significant new information on both the mechanisms and structural components of the RyR PFR involved in block by TAAs and ShBPs, (b) highlight important differences in the mechanisms and structures determining TAA and ShBP block in RyR and K(+) channels, and (c) demonstrate that although the PFRs of these channels contain analogous structural components, significant differences in structure determine the distinct ion-handling properties of the two species of channel.


Subject(s)
Calcium Channel Blockers/pharmacology , Peptides/pharmacology , Quaternary Ammonium Compounds/pharmacology , Ryanodine Receptor Calcium Release Channel/chemistry , Alanine/genetics , Amino Acid Sequence , Animals , Calcium Channel Blockers/chemistry , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Intracellular Signaling Peptides and Proteins , Ion Channel Gating , Mice , Molecular Sequence Data , Mutation, Missense , Protein Structure, Tertiary , Quaternary Ammonium Compounds/chemistry , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/physiology
10.
Adv Exp Med Biol ; 740: 183-215, 2012.
Article in English | MEDLINE | ID: mdl-22453943

ABSTRACT

In excitable tissues, the ryanodine receptor Ca(2+) release channel (RyR) protein complex regulates excitation-contraction coupling, exocytosis, gene expression and apoptosis. Defects in RyR function, in genetic or acquired pathologies, lead to massive disruptions of Ca(2+) release that can be lethal. Therefore, RyR has emerged as a putative therapeutic target and an increasing number of RyR-targeting drugs are currently being tested.Nonetheless this large-size channel is still a mystery in terms of structure, which hinders full characterization of the properties of this central protein. This chapter is dedicated to the methods available to examine RyR structure and function. The aim of the article is to concentrate on contemporary methodologies rather than focusing overtly on the progress that has been achieved using these techniques. Here we review a series of reliable approaches that are routinely employed to investigate this channel. Technical limitations are discussed, and technological developments are presented. This work is not a handbook, but it can be used as a resource and a starting point for the investigation of RyR at different levels of resolution.


Subject(s)
Ryanodine Receptor Calcium Release Channel/physiology , Animals , Binding Sites , Calcium/metabolism , Humans , Lipid Bilayers/chemistry , Protein Structure, Tertiary , Ryanodine Receptor Calcium Release Channel/analysis , Ryanodine Receptor Calcium Release Channel/chemistry
11.
J Integr Bioinform ; 7(1)2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20693604

ABSTRACT

MicroRNAs (miRNAs/miRs) are important cellular components that regulate gene expression at posttranscriptional level. Various upstream components regulate miR expression and any deregulation causes disease conditions. Therefore, understanding of miR regulatory network both at upstream and downstream level is crucial and a resource on this aspect will be helpful. Currently available miR databases are mostly related to downstream targets, sequences, or diseases. But as of now, no database is available that provides a complete picture of miR regulation in a specific condition. Our miR regulation web resource (miReg) is a manually curated one that represents validated upstream regulators (transcription factor, drug, physical, and chemical) along with downstream targets, associated biological process, experimental condition or disease state, up or down regulation of the miR in that condition, and corresponding PubMed references in a graphical and user friendly manner, browseable through 5 browsing options. We have presented exact facts that have been described in the corresponding literature in relation to a given miR, whether it's a feed-back/feed-forward loop or inhibition/activation. Moreover we have given various links to integrate data and to get a complete picture on any miR listed. Current version (Version 1.0) of miReg contains 47 important human miRs with 295 relations using 190 absolute references. We have also provided an example on usefulness of miReg to establish signalling pathways involved in cardiomyopathy. We believe that miReg will be an essential miRNA knowledge base to research community, with its continuous upgrade and data enrichment. This HTML based miReg can be accessed from: www.iioab-mireg.webs.com or www.iioab.webs.com/mireg.htm.


Subject(s)
Computational Biology/methods , Gene Expression Regulation , Internet , MicroRNAs/genetics , Animals , Cardiomyopathies/genetics , Humans , Models, Genetic , Software
12.
CNS Neurosci Ther ; 16(5): e138-56, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20626426

ABSTRACT

Is oxytocin the hormone of happiness? Probably not. However, this small nine amino acid peptide is involved in a wide variety of physiological and pathological functions such as sexual activity, penile erection, ejaculation, pregnancy, uterus contraction, milk ejection, maternal behavior, osteoporosis, diabetes, cancer, social bonding, and stress, which makes oxytocin and its receptor potential candidates as targets for drug therapy. In this review, we address the issues of drug design and specificity and focus our discussion on recent findings on oxytocin and its heterotrimeric G protein-coupled receptor OTR. In this regard, we will highlight the following topics: (i) the role of oxytocin in behavior and affectivity, (ii) the relationship between oxytocin and stress with emphasis on the hypothalamo-pituitary-adrenal axis, (iii) the involvement of oxytocin in pain regulation and nociception, (iv) the specific action mechanisms of oxytocin on intracellular Ca²(+) in the hypothalamo neurohypophysial system (HNS) cell bodies, (v) newly generated transgenic rats tagged by a visible fluorescent protein to study the physiology of vasopressin and oxytocin, and (vi) the action of the neurohypophysial hormone outside the central nervous system, including the myometrium, heart and peripheral nervous system. As a short nine amino acid peptide, closely related to its partner peptide vasopressin, oxytocin appears to be ideal for the design of agonists and antagonists of its receptor. In addition, not only the hormone itself and its binding to OTR, but also its synthesis, storage and release can be endogenously and exogenously regulated to counteract pathophysiological states. Understanding the fundamental physiopharmacology of the effects of oxytocin is an important and necessary approach for developing a potential pharmacotherapy.


Subject(s)
Brain/metabolism , Mental Disorders/drug therapy , Oxytocin/metabolism , Affect/physiology , Analgesics/therapeutic use , Animals , Diabetes Mellitus/drug therapy , Humans , Neoplasms/drug therapy , Osteoarthritis/drug therapy , Receptors, Oxytocin/agonists , Receptors, Oxytocin/antagonists & inhibitors , Receptors, Oxytocin/metabolism , Sexual Dysfunction, Physiological/drug therapy , Signal Transduction/drug effects , Signal Transduction/physiology , Social Behavior
13.
J Mol Cell Cardiol ; 49(3): 427-37, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20540947

ABSTRACT

It is well known that for cardiomyocytes, isolation and culturing induce largely unknown remodelling processes. We analysed changes in the structure of cell compartments with optical techniques such as confocal microscopy and fluorescence redistribution after photobleaching employing adenoviral-mediated transduction of targeted fluorescent proteins and small molecule dyes. We identified characteristic remodelling processes: the T-tubular membrane system was gradually lost by a process referred to as "sequential pinching off", in an outward direction. Mitochondria fell in one of three classes, very small (0.9 microm length), medium long (1.8 microm) or extended shape (3.6 microm) organelles. Over the culturing time mitochondria gradually fused. Bleaching of individual mitochondria revealed association between apparently separated mitochondria by "tunnelling" via sub-resolution organelle-tubes. This tunnelling process was increasing over the culturing time. A gradual loss of the cross-striation arrangement in the endoplasmic/sarcoplasmic reticulum was visualised. Analysis of large populations of Ca(2+) sparks by video-rate confocal 2D-scanning revealed significant albeit small changes of these elementary SR-Ca(2+) release events in adult cardiomyocytes that could be related to changes in SR-Ca(2+) content rather than resting Ca(2+) concentration. In conclusion, primary isolated cardiomyocytes from adult hearts undergo a well-defined, but reproducible subcellular remodelling during optimised long term culture.


Subject(s)
Calcium/metabolism , Heart Ventricles/metabolism , Myocytes, Cardiac/metabolism , Organelles/metabolism , Animals , Cell Culture Techniques , Cell Membrane Permeability , Endoplasmic Reticulum/metabolism , Heart Ventricles/cytology , Microscopy, Confocal , Mitochondria, Heart/metabolism , Myocytes, Cardiac/cytology , Rats
14.
Ophthalmic Genet ; 31(2): 53-65, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20450306

ABSTRACT

PURPOSE: The regulation of Ca(2+) entry and removal is a fine-tuned process which remains not well understood in mouse retinal ganglion cells (RGCs). The latter are known to be sensitive to dysfunctions of mitochondria, organelles playing a pivotal role in Ca(2+) reuptake. METHODS: We first described the Ca(2+) signals of RGCs in response to varied drugs with Fura-2 imaging, and secondly tested the role of optic atrophy 1 or OPA1, the gene responsible for Autosomal Dominant Optic Atrophy, on mitochondrial ability to capture intracellular Ca(2+) in cells transfected with the OPA1 small interfering ribonucleic acids (siRNAs). RESULTS: In control RGCs, K(+)-evoked [Ca(2+)](i) increase was blocked by the Ca(2+) channel antagonists (Ni(2+)+ Cd(2+)) and GABA(A) receptor agonist muscimol-induced [Ca(2+)](i) responses were attenuated by the GABA(A) receptor antagonists, picrotoxin and gabazine. We also prove the presence of NMDA and AMPA/Kainate (glutamate receptor agonists) responsive receptors in this model. Application of cyclopiazonic acid, an inhibitor of Ca(2+)-ATPase pumps of the intracellular Ca(2+) stores, induced an increase in [Ca(2+)](i) while ryanodine or caffeine had no effect on resting [Ca(2+)](i). Spontaneous Ca(2+) oscillations in contacting neurons highlighted the importance of cross-talks between RGCs during maturation. The mitochondrial respiration uncoupler, carbonyl cyanide 3-chlorophenylhydrazone (CCCP), induced robust raises of intracellular Ca(2+) after K(+) application, with a more pronounced effect in cells silenced for OPA1, which could lead to cell death. CONCLUSIONS: Our results indicate an important role of OPA1 in mitochondrial dependent Ca(2+) homeostasis and cell survival in RGCs, suggesting a possible patho-physiological mechanism involved in inherited optic neuropathies.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , GTP Phosphohydrolases/physiology , Retinal Ganglion Cells/metabolism , Animals , Animals, Newborn , Blotting, Western , Calcium Channel Blockers/pharmacology , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/metabolism , Cell Survival , Cells, Cultured , Cytosol/metabolism , Fura-2/metabolism , GABA-A Receptor Antagonists , Homeostasis , Indoles/pharmacology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Mitochondria/metabolism , Potassium/pharmacology , RNA, Small Interfering/genetics , Receptors, AMPA/metabolism , Receptors, GABA-A/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Retinal Ganglion Cells/drug effects , Transfection
16.
J Mol Biol ; 387(2): 320-34, 2009 Mar 27.
Article in English | MEDLINE | ID: mdl-19356589

ABSTRACT

Chloride intracellular channel 2 (CLIC2), a newly discovered small protein distantly related to the glutathione transferase (GST) structural family, is highly expressed in cardiac and skeletal muscle, although its physiological function in these tissues has not been established. In the present study, [3H]ryanodine binding, Ca2+ efflux from skeletal sarcoplasmic reticulum (SR) vesicles, single channel recording, and cryo-electron microscopy were employed to investigate whether CLIC2 can interact with skeletal ryanodine receptor (RyR1) and modulate its channel activity. We found that: (1) CLIC2 facilitated [3H]ryanodine binding to skeletal SR and purified RyR1, by increasing the binding affinity of ryanodine for its receptor without significantly changing the apparent maximal binding capacity; (2) CLIC2 reduced the maximal Ca2+ efflux rate from skeletal SR vesicles; (3) CLIC2 decreased the open probability of RyR1 channel, through increasing the mean closed time of the channel; (4) CLIC2 bound to a region between domains 5 and 6 in the clamp-shaped region of RyR1; (5) and in the same clamp region, domains 9 and 10 became separated after CLIC2 binding, indicating CLIC2 induced a conformational change of RyR1. These data suggest that CLIC2 can interact with RyR1 and modulate its channel activity. We propose that CLIC2 functions as an intrinsic stabilizer of the closed state of RyR channels.


Subject(s)
Chloride Channels/metabolism , Chloride Channels/ultrastructure , Cryoelectron Microscopy , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine Receptor Calcium Release Channel/ultrastructure , Animals , Calcium/metabolism , Crystallography, X-Ray , Ion Channel Gating , Models, Molecular , Muscle, Skeletal/metabolism , Protein Binding , Protein Conformation , Rabbits , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/isolation & purification , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/ultrastructure , Surface Properties , Tritium
17.
Int J Cell Biol ; 2009: 340346, 2009.
Article in English | MEDLINE | ID: mdl-20300593

ABSTRACT

Mutations of Ca(2+)-activated proteases (calpains) cause muscular dystrophies. Nevertheless, the specific role of calpains in Ca(2+) signalling during the onset of dystrophies remains unclear. We investigated Ca(2+) handling in skeletal cells from calpain 3-deficient mice. [Ca(2+)](i) responses to caffeine, a ryanodine receptor (RyR) agonist, were decreased in -/- myotubes and absent in -/- myoblasts. The -/- myotubes displayed smaller amplitudes of the Ca(2+) transients induced by cyclopiazonic acid in comparison to wild type cells. Inhibition of L-type Ca(2+) channels (LCC) suppressed the caffeine-induced [Ca(2+)](i) responses in -/- myotubes. Hence, the absence of calpain 3 modifies the sarcoplasmic reticulum (SR) Ca(2+) release, by a decrease of the SR content, an impairment of RyR signalling, and an increase of LCC activity. We propose that calpain 3-dependent proteolysis plays a role in activating support proteins of intracellular Ca(2+) signalling at a stage of cellular differentiation which is crucial for skeletal muscle regeneration.

18.
Prog Brain Res ; 170: 177-92, 2008.
Article in English | MEDLINE | ID: mdl-18655882

ABSTRACT

Neuroactive steroids synthesized from the brain or peripheral sources are called neurosteroids. Beside their common nuclear effects, they are considered to be potent neuromodulators, acting rapidly mainly in a non-genomic manner, either through allosteric regulation of ionic channels, or through membrane-bound steroid receptors. In contrast to the situation in the adult, the neurotransmitter GABA is excitatory during development and plays a trophic role, in particular inducing calcium signals necessary for the regulation of excitability and neuronal maturation. We demonstrated that the primary metabolite of progesterone (Proges), allopregnanolone (Allo), evoked a robust Ca(2+) influx in foetal hypothalamic neurons and in postnatal supraoptic nucleus (SON) neurons. In the latter, this led to oxytocin and arginine vasopressin release. Interestingly, these responses were GABA(A) and oxytocin-receptor-dependent. Allo is a well-known positive allosteric modulator of GABA(A) receptors. It is noteworthy that two other steroids, Proges and 17-beta-estradiol, displayed the same effect on Ca(2+) and oxytocin release but to a lesser extent. Importantly, no effect was observed in adult neurons from the SON, or in neurohypophysial axon terminals, regardless of the stage. The molecular mechanisms of the neurosteroid actions are multifaceted and depend on the type of cells, and are thus extremely interesting and challenging. In the peripheral nervous system, Allo and Proges surprisingly inhibited the GABA-induced Ca(2+) increases in embryonic dorsal root ganglion neurons. We propose that this rapid, reversible and dose-dependent phenomenon (at very low concentrations) was mediated by membrane Proges receptors, since transcripts for a newly discovered receptor protein, 25-Dx, were detected in our model. Recently, novel families of membrane steroid receptors, activating intracellular-signalling pathways such as MAP kinases, have been identified and described. This opens new perspectives to understand the intracellular machinery involved in the interaction between neuropeptides and neurosteroids, two major regulators of hypothalamo-neurohypophysial system development.


Subject(s)
Hypothalamo-Hypophyseal System/physiology , Neurons/physiology , Oxytocin/physiology , Peripheral Nervous System/physiology , Receptors, Progesterone/physiology , Supraoptic Nucleus/physiology , Animals , Arginine Vasopressin/metabolism , Calcium/metabolism , Calcium/pharmacology , Cell Membrane/physiology , Ganglia, Spinal/physiology , Hormones/physiology , Humans , Hypothalamus/embryology , Hypothalamus/physiology , Ion Channels/physiology , Neuropeptides/physiology , Progesterone/metabolism , Progestins/physiology , Receptors, Cell Surface/physiology , Receptors, Steroid/physiology
19.
Cell Calcium ; 43(1): 59-71, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17822759

ABSTRACT

For studying heart pathologies on the cellular level, cultured adult cardiac myocytes represent an important approach. We aimed to explore a novel adult rat ventricular myocyte culture system with minimised dedifferentiation allowing extended experimental manipulation of the cells such as expression of exogenous proteins. Various culture conditions were investigated including medium supplement, substrate coating and electrical pacing for one week. Adult myocytes were probed for (i) viability, (ii) morphology, (iii) frequency dependence of contractions, (iv) Ca(2+) transients, and (v) their tolerance towards adenovirus-mediated expression of the Ca(2+) sensor "inverse pericam". Conventionally, in either serum supplemented or serum-free medium, myocytes dedifferentiated into flat cells within 3 days or cell physiology and morphology were impaired, respectively. In contrast, myocytes cultured in medium supplemented with an insulin-transferrin-selenite mixture on substrates coated with extracellular matrix proteins showed an increased cell attachment and a conserved cross-striation. Moreover, these myocytes displayed optimised preservation of their contractile behaviour and Ca(2+) signalling even under conditions of continuous electrical pacing. Sustained expression of inverse pericam did not alter myocyte function and allowed long lasting high speed Ca(2+) imaging of electrically driven adult myocytes. Our single-cell model thus provides a new advance for high-content screening of these highly specialised cells.


Subject(s)
Calcium/analysis , Cell Culture Techniques , Fluorescent Dyes/analysis , Myocytes, Cardiac/cytology , Animals , Calcium Signaling , Cell Survival , Cells, Cultured , Electric Stimulation , Fluorescent Dyes/metabolism , Heart Ventricles/cytology , Luminescent Proteins/analysis , Luminescent Proteins/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Rats
20.
Cell Calcium ; 40(4): 383-91, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16769113

ABSTRACT

The non-genomic role of neuroactive steroids on [Ca2+]i transients induced by GABA receptor activation was investigated in cultured dorsal root ganglia (DRG) neurones at embryonic stage E13. [Ca2+]i measurements were performed with Fura-2 fast fluorescence microfluorimetry. Application of the GABAA receptor agonist muscimol (Musci) evoked an increase in [Ca2+]i, confirming the excitatory effect of GABA at this embryonic stage. The muscimol-induced [Ca2+]i response was inhibited by progesterone (Proges) and its primary metabolite allopregnanolone (Allo) in a rapid, reversible and dose-dependent manner. These calcium transients were suppressed in the absence of external Ca2+ or in the presence of Ni2+ + Cd2+ suggesting an involvement of voltage-activated Ca2+ channels. In contrast, none of these steroids affected the resting [Ca2+]i nor exhibited any inhibitory effect on 50 mM KCl-induced [Ca2+]i increases. In view of the well-established potentiation of GABAA receptor by direct binding of neurosteroids, the inhibitory effects described in this study seem to involve distinct mechanisms. This new inhibitory effect of progesterone is observed at low and physiological concentrations, is rapid and independent of RU38486, an antagonist of the classic progesterone receptor, probably involving a membrane receptor. Using RT-PCR, we demonstrated the expression of progesterone receptor membrane component 1 (Pgrmc1), encoding 25-Dx, a membrane-associated progesterone binding protein in DRG neurones at different stages of development. In conclusion, we describe for the first time a rapid effect of progestins on embryonic DRG neurones involving an antagonistic effect of progesterone and allopregnanolone on GABAA receptors.


Subject(s)
Calcium/metabolism , Embryo, Mammalian/cytology , Ganglia, Spinal/cytology , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Pregnanolone/pharmacology , Progesterone/pharmacology , Animals , Calcium Channel Blockers/metabolism , Cells, Cultured , Chelating Agents/metabolism , Fluorescent Dyes/metabolism , Fura-2/metabolism , GABA Agonists/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Muscimol/metabolism , Neurons, Afferent/cytology , Potassium Channel Blockers/metabolism , Receptors, GABA-A/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Sodium Channel Blockers/metabolism , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...