Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters










Publication year range
1.
J Aging Res Clin Pract ; 7: 47-54, 2018 Mar 26.
Article in English | MEDLINE | ID: mdl-29755960

ABSTRACT

OBJECTIVES: Does excessive sun-exposure, non-use of sunscreen and/or high doses of vitamin-D3 supplements provoke exacerbation of asthma? DESIGN: Clinical examinations, retrospective records-access and questionnaire surveys were distributed to a convenience sample of allergic-asthma patient (n=183). SETTING: Patients (19-89 years) attending the outpatient respiratory clinics at Maidstone Hospital were enrolled. RESULTS: 90.3% of patients (total IgE levels ≥75 kU/L ; n=103) exposed to direct sunlight of ≥ 15 minutes per day continuously for 6-7 days presented with wheeze (χ2(1) = 7.46; p< 0.05) compared to only 9.7% patients of similar atopy-status, presenting with wheeze if exposed to sunlight of < 15 minutes per day for 6-7 days. 68.9% patients (with IgE levels ≥ 75 kU/L ; n=103), non-users of sunscreen (SPF 30 and above), exposed to direct sunlight of ≥ 15 minutes per day continuously for 6-7 days developed a wheeze, compared to fewer users of sunscreen (9.7%, n=103), exposed to the same duration of sunlight who developed asthma symptoms (p< 0.05). Vitamin-D3 supplementation in asthma-patients with clinical signs of hypovitaminosis-D (n=21), produced symptoms of morning chest-tightness (76.2%), allergic rhinitis (61.9%) and wheeze (100%), 2 weeks after initiation of treatment. CONCLUSIONS: Our results advocate direct sunlight exposure < 15 minutes per day and use of sunscreen as a novel approach to preventing atopic-asthma symptoms in allergic-asthma patients.. Activated vitamin-D3 is well-recognised to shift the immune-balance towards Th2 predominance, favouring allergic asthma. These results suggest that limiting subcutaneous synthesis of vitamin-D3 in asthma patients and re-addressing dosage of vitamin-D3 supplementation is necessary may contribute to prevent exacerbation of symptoms.

2.
Hum Reprod ; 23(2): 365-73, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18003622

ABSTRACT

BACKGROUND: A sperm-specific phospholipase C zeta (PLCzeta) has been shown to trigger Ca(2+) oscillations in mouse and human oocytes and appears to be the sperm factor responsible for activation at fertilization. Previously, complementary RNA (cRNA) injection was used to introduce PLCzeta into oocytes, but it was unclear how much PLCzeta protein is required for development. Here we have injected cRNA encoding luciferase-tagged human PLCzeta (hPLCzeta-luc) into mouse oocytes and established the relationship between hPLCzeta-luc expression, Ca(2+) oscillations and development. METHODS: Mouse oocytes were injected with hPLCzeta-luc cRNA and a fluorescent Ca(2+)dye to monitor hPLCzeta-luc expression and Ca(2+) oscillations, respectively. After inducing diploidy, development in vitro was monitored in hPLCzeta-luc cRNA microinjected oocytes and compared with parallel oocytes activated by incubation in Sr(2+). RESULTS: Repetitive Ca(2+) oscillations and oocyte activation were triggered by hPLCzeta over a wide range of luciferase expression levels. However, subsequent development of embryos to the blastocyst stage was observed only when expression of hPLCzeta-luc was optimized within a specific range. The blastocyst cell number was also affected by the level of hPLCzeta expression. CONCLUSIONS: Human PLCzeta can readily activate mouse oocytes, however, effective development to blastocyst stages is only achieved within a specific window of hPLCzeta-luc protein expression levels.


Subject(s)
Blastocyst/physiology , Oocytes/drug effects , Oocytes/physiology , Phosphoinositide Phospholipase C/pharmacology , Animals , Blastocyst/cytology , Calcium/metabolism , Cell Count , Female , Gene Transfer Techniques , Humans , Intracellular Membranes/metabolism , Luciferases/genetics , Luciferases/metabolism , Mice , Oocytes/metabolism , Oscillometry , Phosphoinositide Phospholipase C/genetics
3.
Biochem Soc Trans ; 35(Pt 5): 946-51, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17956252

ABSTRACT

The cardiac ryanodine receptor (RyR2) mediates rapid Ca(2+) efflux from intracellular stores to effect myocyte contraction during the process of EC (excitation-contraction) coupling. It is now known that mutations in this channel perturb Ca(2+) release function, leading to triggered arrhythmias that may cause SCD (sudden cardiac death). Resolving the precise molecular mechanisms by which SCD-linked RyR2 dysfunction occurs currently constitutes a burgeoning area of cardiac research. So far, defective channel phosphorylation, accessory protein binding, luminal/cytosolic Ca(2+) sensing, and the disruption of interdomain interactions represent the main candidate mechanisms for explaining aberrant SR (sarcoplasmic reticulum) Ca(2+) release via mutants of RyR2. It appears increasingly unlikely that a single exclusive common mechanism underlies every case of mutant channel dysfunction, and that each of these potential mechanisms may contribute to the resultant phenotype. The present review will consider very recent mechanistic developments in this field, including new observations from mutant RyR2 transgenic mouse models, peptide-probe studies, and the implications of functional and phenotypic heterogeneity of RyR2 mutations and polymorphisms.


Subject(s)
Arrhythmias, Cardiac/genetics , Ion Channels/physiology , Mutation , Ryanodine Receptor Calcium Release Channel/genetics , Animals , Arrhythmias, Cardiac/physiopathology , Humans , Phenotype , Phosphorylation , Ryanodine Receptor Calcium Release Channel/metabolism , Tacrolimus Binding Proteins/metabolism
4.
Biochem Soc Trans ; 34(Pt 5): 913-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17052226

ABSTRACT

The RyR (ryanodine receptor) mediates rapid Ca2+ efflux from the ER (endoplasmic reticulum) and is responsible for triggering numerous Ca2+-activated physiological processes. The most studied RyR-mediated process is excitation-contraction coupling in striated muscle, where plasma membrane excitation is transmitted to the cell interior and results in Ca2+ efflux that triggers myocyte contraction. Recently, single-residue mutations in the cardiac RyR (RyR2) have been identified in families that exhibit CPVT (catecholaminergic polymorphic ventricular tachycardia), a condition in which physical or emotional stress can trigger severe tachyarrhythmias that can lead to sudden cardiac death. The RyR2 mutations in CPVT are clustered in the N- and C-terminal domains, as well as in a central domain. Further, a critical signalling role for dysfunctional RyR2 has also been implicated in the generation of arrhythmias in the common condition of HF (heart failure). We have prepared cardiac RyR2 plasmids with various CPVT mutations to enable expression and analysis of Ca2+ release mediated by the wild-type and mutated RyR2. These studies suggest that the mutational locus may be important in the mechanism of Ca2+ channel dysfunction. Understanding the causes of aberrant Ca2+ release via RyR2 may assist in the development of effective treatments for the ventricular arrhythmias that often leads to sudden death in HF and in CPVT.


Subject(s)
Myocardium/pathology , Ryanodine Receptor Calcium Release Channel/genetics , Tachycardia, Ventricular/physiopathology , Calcium/metabolism , Endoplasmic Reticulum/physiology , Humans , Models, Molecular , Mutation , Protein Conformation , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/physiology , Tachycardia, Ventricular/genetics
5.
Biochem Soc Trans ; 34(Pt 5): 919-21, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17052227

ABSTRACT

The RyR (ryanodine receptor)/calcium release channel contains a number of highly reactive thiol groups that endow it with redox sensitivity. In general, oxidizing conditions favour channel opening, while reducing conditions have the opposite effect. Thiol modification affects the channel sensitivity to its principal effectors, Ca2+, Mg2+ and ATP, and alters RyR protein interactions. Here, we give a brief account of the major findings and prevailing views in the field.


Subject(s)
Ryanodine Receptor Calcium Release Channel/physiology , Adenosine Triphosphate/physiology , Animals , Calcium/physiology , Humans , Magnesium/physiology , Oxidation-Reduction , Ryanodine Receptor Calcium Release Channel/drug effects , Sulfhydryl Compounds/pharmacology
6.
Semin Cell Dev Biol ; 17(2): 264-73, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16730199

ABSTRACT

At fertilization in mammals, the sperm activates development by causing a prolonged series of intracellular Ca(2+) oscillations that are generated by increased production of inositol trisphosphate (InsP(3)). It appears that the sperm initiates InsP(3) generation via the introduction of a sperm factor into the egg after gamete membrane fusion. We recently identified a sperm-specific form of phospholipase C (PLC), referred to as PLCzeta(zeta). We review the evidence that PLCzeta represents the sperm factor that activates development of the egg and discuss the characteristics of PLCzeta that distinguish it from the somatic forms of PLC.


Subject(s)
Calcium Signaling , Sperm-Ovum Interactions , Spermatozoa/enzymology , Type C Phospholipases/physiology , Amino Acid Sequence , Animals , Cell Nucleus/enzymology , Humans , Male , Mice , Molecular Sequence Data , Ovum/metabolism , Phosphoinositide Phospholipase C , Species Specificity , Type C Phospholipases/chemistry
7.
Reproduction ; 128(6): 697-702, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15579586

ABSTRACT

At fertilization in mammals the sperm activates development of the oocyte by inducing a prolonged series of oscillations in the cytosolic free Ca2+ concentration. One theory of signal transduction at fertilization suggests that the sperm cause the Ca2+ oscillations by introducing a protein factor into the oocyte after gamete membrane fusion. We recently identified this sperm-specific protein as phospholipase Czeta (PLCzeta), and we showed that PLCzeta triggers Ca2+ oscillations in unfertilized mouse oocytes. Here we report that microinjection of the complementary RNA for human PLCzeta causes prolonged Ca2+ oscillations in aged human oocytes that had failed to fertilize during in vitro fertilization or intracytoplasmic sperm injection. The frequency of Ca2+ oscillations was related to the concentration of complementary RNA injected. At low concentrations, PLCzeta stimulated parthenogenetic activation of oocytes. These embryos underwent cleavage divisions and some formed blastocysts. These data show that PLCzeta is a novel parthenogenetic stimulus for human oocytes and that it is unique in its ability to mimic the repetitive nature of the Ca2+ stimulus provided by the sperm during human fertilization.


Subject(s)
Calcium/metabolism , Cytosol/metabolism , Oocytes/metabolism , RNA, Messenger/administration & dosage , Type C Phospholipases/genetics , Cells, Cultured , Female , Humans , Microinjections , Oocytes/drug effects , Parthenogenesis/drug effects , Phosphoinositide Phospholipase C , Type C Phospholipases/metabolism
8.
Reproduction ; 127(4): 431-9, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15047934

ABSTRACT

When sperm activate eggs at fertilization the signal for activation involves increases in the intracellular free Ca2+ concentration. In mammals the Ca2+ changes at fertilization consist of intracellular Ca2+ oscillations that are driven by the generation of inositol 1,4,5-trisphosphate (InsP3). It is not established how sperm trigger the increases in InsP3 and Ca2+ at fertilization. One theory suggests that sperm initiate signals to activate the egg by introducing a specific factor into the egg cytoplasm after membrane fusion. This theory has been mainly based upon the observation that injecting a cytosolic sperm protein factor into eggs can trigger the same pattern of Ca2+ oscillations induced by the sperm. We have recently shown that this soluble sperm factor protein is a novel form of phospholipase C (PLC), and it is referred to as PLCzeta(zeta). We describe the evidence that led to the identification of PLCzeta and discuss the issues relating to its potential role in fertilization.


Subject(s)
Calcium/metabolism , Cytosol/metabolism , Mammals/metabolism , Ovum/metabolism , Type C Phospholipases/metabolism , Animals , Female , Fertilization/physiology , Humans , Male , Mice , Phospholipase C gamma
9.
Reproduction ; 124(5): 611-23, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12416999

ABSTRACT

Fusion with a fertilizing spermatozoon induces the mammalian oocyte to undergo a remarkable series of oscillations in cytosolic Ca(2+), leading to oocyte activation and development of the embryo. The exact molecular mechanism for generating Ca(2+) oscillations has not been established. A sperm-specific zeta isoform of phospholipase C (PLCzeta) has been identified in mice. Mouse PLCzeta triggers Ca(2+) oscillations in mouse oocytes and exhibits properties synonymous with the 'sperm factor' that has been proposed to diffuse into the oocyte after gamete fusion. The present study isolated the PLCzeta homologue from human and cynomolgus monkey testes. Comparison with mouse and monkey PLCzeta protein sequences indicates a shorter X-Y linker region in human PLCzeta and predicts a distinctly different isoelectric point. Microinjection of complementary RNA for both human and cynomolgus monkey PLCzeta elicits Ca(2+) oscillations in mouse oocytes equivalent to those seen during fertilization in mice. Moreover, human PLCzeta elicits mouse egg activation and early embryonic development up to the blastocyst stage, and exhibits greater potency than PLCzeta from monkeys and mice. These results are consistent with the proposal that sperm PLCzeta is the molecular trigger for egg activation during fertilization and that the role and activity of PLCzeta is highly conserved across mammalian species.


Subject(s)
Calcium Signaling/physiology , Isoenzymes/pharmacology , Oocytes/metabolism , Sperm-Ovum Interactions/physiology , Spermatozoa/enzymology , Type C Phospholipases/physiology , Amino Acid Sequence , Animals , Calcium/metabolism , Cells, Cultured , Cloning, Molecular , Cytosol/metabolism , Female , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred Strains , Microinjections , Molecular Sequence Data , RNA, Complementary/pharmacology , Sequence Alignment , Testis/enzymology , Type C Phospholipases/genetics
10.
Biochem Biophys Res Commun ; 285(1): 52-7, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11437371

ABSTRACT

Although dissociation of the 12 kDa FK506 binding protein (FKBP12)-type 1 ryanodine receptor (RyR1) complex by macrolide immunosuppressants is well documented, effects of many solutes and drugs have not been quantitated. In the current study, the influence of these on binding between solubilised RyR1 and an FKBP12-glutathione-S-transferase fusion protein was analysed using a novel assay. Association between these two proteins is stable, and is not greatly altered by changes in temperature, pH, cations, and endogenous solutes over physiological ranges. Ascomycin, an FK506 analogue, was identified for the first time as a drug which can disrupt the FKBP12-RyR1 complex.


Subject(s)
Ryanodine Receptor Calcium Release Channel/metabolism , Tacrolimus Binding Proteins/metabolism , Animals , Glutathione Transferase/metabolism , Protein Binding , Rabbits , Recombinant Fusion Proteins/metabolism
11.
Cell Signal ; 13(5): 353-62, 2001 May.
Article in English | MEDLINE | ID: mdl-11369517

ABSTRACT

The plant alkaloid ryanodine (Ry) is a high-affinity modulator of ryanodine receptor (RyR) Ca(2+) release channels. Although these channels are present in a variety of cell types, their functional role in nerve cells is still puzzling. Here, a monosubstituted fluorescent Ry analogue, B-FL-X Ry, was used to reveal the distribution of RyRs in cultured rat sympathetic neurons. B-FL-X Ry competitively inhibited the binding of [3H]Ry to rabbit skeletal muscle SR membranes, with an IC(50) of 150 nM, compared to 7 nM of unlabeled Ry. Binding of B-FL-X Ry to the cytoplasm of sympathetic neurons is saturable, reversible and of high affinity. The pharmacology of B-FL-X Ry showed marked differences with unlabeled Ry, which are partially explained by its lower affinity: (1) use-dependent reversible inhibition of caffeine-induced intracellular Ca(2+) release; (2) diminished voltage-gated Ca(2+) influx, due to a positive shift in the activation of voltage gated Ca(2+) currents. B-FL-X Ry-stained sympathetic neurons, viewed under confocal microscopy, showed conspicuous labeling of crescent-shaped structures pertaining to the Golgi complex, a conclusion supported by experiments showing co-localization with Golgi-specific fluorescent probes and the breaking up of crescent-shaped staining after treatment with drugs that disassemble Golgi complex. The presence of RyRs to the Golgi could be confirmed with specific anti-RyR(2) antibodies, but evidence of caffeine-induced Ca(2+) release from this organelle could not be obtained using fast confocal microscopy. Rather, an apparent decrease of the cytosolic Ca(2+) signal was detected close to this organelle. In spite of that, short-term incubation with brefeldin A (BFA) suppressed the fast component of caffeine-induced Ca(2+) release, and the Ca(2+) release process lasted longer and appeared less organized. These observations, which suggest a possible role of the Golgi complex in Ca(2+) homeostasis and signaling in nerve cells, could be relevant to reports involving derangement of the Golgi complex as a probable cause of some forms of progressive neuronal degeneration, such as Alzheimer's disease and amyotrophic lateral sclerosis.


Subject(s)
Calcium Signaling/physiology , Golgi Apparatus/metabolism , Neurons/physiology , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine/analogs & derivatives , Ryanodine/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Antibody Specificity , Boron Compounds/metabolism , Brefeldin A/pharmacology , Caffeine/pharmacology , Calcium/metabolism , Calcium Channels/metabolism , Calcium Signaling/drug effects , Fluorescent Dyes , Golgi Apparatus/chemistry , Macrolides , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/chemistry , Neurons/ultrastructure , Phosphodiesterase Inhibitors/pharmacology , Rats , Ryanodine Receptor Calcium Release Channel/analysis , Ryanodine Receptor Calcium Release Channel/immunology , Superior Cervical Ganglion/cytology
12.
Ultramicroscopy ; 86(1-2): 129-37, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11215615

ABSTRACT

The skeletal muscle Ca2+ release channel (ryanodine receptor 1, RYR1) plays an important role in the excitation-contraction coupling process. We purified ryanodine receptor type 1 from rabbit white muscle and adsorbed it to mica sheets with the cytoplasmic side facing up. Single receptors of uniformly distributed size and shape of 10-12 nm height and 40-50 nm width, and occasionally some aggregates were seen in contact mode AFM images. These immobilized RYR1 were specifically recognized by rabbit anti-RYR1 (antibody#8) with at least 30% efficiency, as measured by an enzyme immunoassay with goat-anti-rabbit. Single specific antibody-antigen recognition events were detected with AFM tips to which an antibody#8 was tethered. In linear scans, the occurrence of antibody-antigen binding showed significant lateral dependence, which allowed for the localization of binding sites with nm resolution. Variation of the loading rate in force spectroscopy experiments revealed a logarithmic dependence of the unbinding forces, ranging from 42 to 73 pN. From this dependence, a bond width of the binding pocket of L = 0.2 nm and a kinetic off-rate of koff = 12.7s(-1) was determined.


Subject(s)
Microscopy, Atomic Force/methods , Muscle, Skeletal/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Animals , Antibodies/immunology , Antibodies/metabolism , Immunoenzyme Techniques , Ligands , Rabbits , Ryanodine Receptor Calcium Release Channel/isolation & purification
13.
Nat Neurosci ; 3(12): 1256-65, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11100146

ABSTRACT

The cellular mechanisms responsible for large miniature currents in some brain synapses remain undefined. In Purkinje cells, we found that large-amplitude miniature inhibitory postsynaptic currents (mIPSCs) were inhibited by ryanodine or by long-term removal of extracellular Ca2+. Two-photon Ca2+ imaging revealed random, ryanodine-sensitive intracellular Ca2+ transients, spatially constrained at putative presynaptic terminals. At high concentration, ryanodine decreased action-potential-evoked rises in intracellular Ca2+. Immuno-localization showed ryanodine receptors in these terminals. Our data suggest that large mIPSCs are multivesicular events regulated by Ca2+ release from ryanodine-sensitive presynaptic Ca2+ stores.


Subject(s)
Calcium Signaling/physiology , Calcium/deficiency , Neural Inhibition/physiology , Presynaptic Terminals/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Synaptic Vesicles/metabolism , Animals , Animals, Newborn , Calcium Signaling/drug effects , Cerebellum/drug effects , Cerebellum/metabolism , Cerebellum/ultrastructure , Extracellular Space/drug effects , Extracellular Space/metabolism , Interneurons/drug effects , Interneurons/metabolism , Interneurons/ultrastructure , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neural Inhibition/drug effects , Presynaptic Terminals/drug effects , Presynaptic Terminals/ultrastructure , Rats , Reaction Time/drug effects , Reaction Time/physiology , Ryanodine/pharmacology , Ryanodine Receptor Calcium Release Channel/drug effects , Synaptic Vesicles/drug effects , Synaptic Vesicles/ultrastructure , Tetrodotoxin/pharmacology
14.
Biol Cell ; 92(3-4): 267-75, 2000 Jul.
Article in English | MEDLINE | ID: mdl-11043414

ABSTRACT

At fertilisation in mammals the sperm initiates a series of Ca2+ oscillations that activate development. One theory of signalling at fertilisation suggests that the sperm contains a soluble protein factor that causes these Ca2+ oscillations by entering the egg after sperm-egg membrane fusion. This theory is supported by the finding that, in some species, injection of sperm protein extracts into eggs triggers a pattern of Ca2+ oscillations similar to those seen at fertilisation. So far, all the direct evidence for a sperm factor has been based upon the injection of soluble proteins from mature sperm. Here, we demonstrate that injection of mRNA extracted from hamster spermatogenic cells also leads to generation of prolonged Ca2+ oscillations in mouse eggs. The ability of spermatogenic cell mRNA to induce Ca2+ oscillations is dependent upon translation into protein and also appears to be specific to spermatogenic cells since injection of mRNA isolated from somatic tissues into eggs was ineffective. These data support the hypothesis that sperm contain a soluble, cytosolic protein factor that induces Ca2+ oscillations in eggs at fertilisation. These data are discussed in the light of our recent findings that suggest that the sperm factor possesses a phospholipase C activity.


Subject(s)
Calcium/metabolism , Ovum/metabolism , Sperm-Ovum Interactions/physiology , Spermatozoa/metabolism , Animals , Cricetinae , Female , Gene Expression , Male , Mammals , Mice , Microinjections , Ovum/physiology , Proteins/genetics , RNA, Messenger , Solubility , Spermatogenesis/physiology , Spermatozoa/physiology , Zygote/metabolism
15.
Biochem J ; 351(Pt 1): 265-71, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-10998370

ABSTRACT

Regulation of cytosolic Ca(2+) is important for a variety of cell functions. The ryanodine receptor (RyR) is a Ca(2+) channel that conducts Ca(2+) from internal pools to the cytoplasm. To demonstrate the presence of the RyR in the pancreatic acinar cell, we performed reverse transcriptase (RT)-PCR, Western blot, immunocytochemistry and microscopic Ca(2+)-release measurements on these cells. RT-PCR showed the presence of mRNA for RyR isoforms 1, 2 and 3 in both rat pancreas and dispersed pancreatic acini. Furthermore, mRNA expression for RyR isoforms 1 and 2 was demonstrated by RT-PCR in individual pancreatic acinar cells selected under the microscope. Western-blot analysis of acinar cell immunoprecipitates, using antibodies against RyR1 and RyR2, showed a high-molecular-mass (>250 kDa) protein band that was much less intense when immunoprecipitated in the presence of RyR peptide. Functionally, permeablized acinar cells stimulated with the RyR activator, palmitoyl-CoA, released Ca(2+) from both basolateral and apical regions. These data show that pancreatic acinar cells express multiple isoforms of the RyR and that there are functional receptors throughout the cell.


Subject(s)
Pancreas/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Blotting, Western , Calcium/metabolism , Cell Membrane Permeability , Cell Polarity , Cells, Cultured , Immunohistochemistry , Molecular Weight , Palmitoyl Coenzyme A/pharmacology , Pancreas/cytology , Pancreas/drug effects , Precipitin Tests , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/genetics
16.
Nat Cell Biol ; 2(9): 669-71, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10980710

ABSTRACT

Recent theoretical analysis of a model lattice of interacting transmembrane receptor proteins has indicated that such clustering in the membrane could provide a novel mechanism for regulating receptor signalling in cells. It has been calculated that cooperative interactions between receptors organized into a cluster, or array, in the membrane would dramatically increase their sensitivity to activation by ligand. Sensitivity to ligand would increase with the extent of spread of activity within the receptor lattice. Hence, formation of extensive receptor lattices in the membrane would allow a large population of receptors to be simultaneously switched on, or off, by a very small change in ligand concentration. We show here that lattice formation is an intrinsic property of an integral membrane protein, the ryanodine-sensitive calcium-release channel (RyR) of endoplasmic reticulum. The purified protein spontaneously assembled into two-dimensional lattices in solution, enabling the construction of a 25 A projection map that identifies the mode of interaction between RyR oligomers. Our observations on the RyR provide a new perspective on various properties of cell signalling via this and other receptors.


Subject(s)
Ryanodine Receptor Calcium Release Channel/ultrastructure , Animals , Protein Conformation , Rabbits
17.
Am J Physiol Renal Physiol ; 278(5): F784-91, 2000 May.
Article in English | MEDLINE | ID: mdl-10807590

ABSTRACT

We report the first biochemical and functional characterization of inositol trisphosphate receptors (IP(3)Rs) and ryanodine receptors (RyRs) in the nuclear membrane of bone-forming (MC3T3-E1) osteoblasts. Intact nuclei fluoresced intensely with anti-RyR (Ab(34)) and anti-IP(3)R (Ab(40)) antisera in a typically peripheral nuclear membrane pattern. Isolated nuclear membranes were next subjected to SDS-PAGE and blotted with isoform-specific anti-receptor antisera, notably Ab(40), anti-RyR-1, anti-RyR-2 (Ab(129)), and anti-RyR-3 (Ab(180)). Only anti-RyR-1 and Ab(40) showed bands corresponding, respectively, to full-length RyR-1 ( approximately 500 kDa) and IP(3)R-1 (approximately 250 kDa). Band intensity was reduced by just approximately 20% after brief tryptic proteolysis of intact nuclei; this confirmed that isolated nuclear membranes were mostly free of endoplasmic reticular contaminants. Finally, the nucleoplasmic Ca(2+) concentration ([Ca(2+)](np)) was measured in single nuclei by using fura-dextran. The nuclear envelope was initially loaded with Ca(2+) via Ca(2+)-ATPase activation (1 mM ATP and approximately 100 nM Ca(2+)). Adequate Ca(2+) loading was next confirmed by imaging the nuclear envelope (and nucleoplasm). Exposure of Ca(2+)-loaded nuclei to IP(3) or cADP ribose resulted in a rapid and sustained [Ca(2+)](np) elevation. Taken together, the results provide complementary evidence for nucleoplasmic Ca(2+) influx in osteoblasts through nuclear membrane-resident IP(3)Rs and RyRs. Our findings may conceivably explain the direct regulation of osteoblastic gene expression by hormones that use the IP(3)-Ca(2+) pathway.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Cell Nucleus/metabolism , Osteoblasts/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , 3T3 Cells , Animals , Immunohistochemistry , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors , Mice , Nuclear Envelope/metabolism , Protein Isoforms/metabolism
18.
Am J Physiol Renal Physiol ; 278(4): F576-84, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10751218

ABSTRACT

Herein we demonstrate that replicative cellular senescence in vitro results in sharply reduced inositol 1,4,5-trisphosphate (IP(3)) receptor levels, reduced mitogen-evoked IP(3) formation and Ca(2+) release, and Ca(2+) store depletion. Human diploid fibroblasts (HDFs) underwent either 30 mean population doublings [mean population doublings (MPDs) thymidine labeling index (TI) >92% ("young") or between 53 and 58 MPDs (TI < 28%; "senescent")]. We found that the cytosolic Ca(2+) release triggered by either ionomycin or by several IP(3)-generating mitogens, namely bradykinin, thrombin, platelet-derived growth factor (PDGF), and epidermal growth factor (EGF), was attenuated markedly in senescent HDFs. Notably, the triggered cytosolic Ca(2+) transients were of a smaller magnitude in senescent HDFs. However, the response latency seen with both PDGF and EGF was greater for senescent cells. Finally, a smaller proportion of senescent HDFs showed oscillations. In parallel, IP(3) formation in response to bradykinin or EGF was also attenuated in senescent HDFs. Furthermore, senescent HDFs displayed a sharply diminished Ca(2+) release response to intracellularly applied IP(3). Finally, to compare IP(3) receptor protein levels directly in young and senescent HDFs, their microsomal membranes were probed in Western blots with a highly specific anti-IP(3) receptor antiserum, Ab(40). A approximately 260-kDa band corresponding to the IP(3) receptor protein was noted; its intensity was reduced by approximately 50% in senescent cells. Thus, we suggest that reduced IP(3) receptor expression, lowered IP(3) formation, and Ca(2+) release, as well as Ca(2+) store depletion, all contribute to the deficient Ca(2+) signaling seen in HDFs undergoing replicative senescence.


Subject(s)
Calcium Channels/metabolism , Fibroblasts/physiology , Inositol 1,4,5-Trisphosphate/biosynthesis , Receptors, Cytoplasmic and Nuclear/metabolism , Bradykinin/pharmacology , Calcium/metabolism , Cells, Cultured , Cellular Senescence/physiology , Child , Cytosol/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Growth Substances/pharmacology , Humans , Inositol 1,4,5-Trisphosphate/pharmacology , Inositol 1,4,5-Trisphosphate Receptors , Mitogens/pharmacology , Thrombin/pharmacology
19.
Nat Cell Biol ; 1(7): 409-14, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10559984

ABSTRACT

Nucleoplasmic calcium ions (Ca2+) influence nuclear functions as critical as gene transcription, apoptosis, DNA repair, topoisomerase activation and polymerase unfolding. Although both inositol trisphosphate receptors and ryanodine receptors, types of Ca2+ channel, are present in the nuclear membrane, their role in the homeostasis of nuclear Ca2+ remains unclear. Here we report the existence in the inner nuclear membrane of a functionally active CD38/ADP-ribosyl cyclase that has its catalytic site within the nucleoplasm. We propose that the enzyme catalyses the intranuclear cyclization of nicotinamide adenine dinucleotide to cyclic adenosine diphosphate ribose. The latter activates ryanodine receptors of the inner nuclear membrane to trigger nucleoplasmic Ca2+ release.


Subject(s)
Antigens, CD , Antigens, Differentiation/metabolism , Calcium/metabolism , Cell Nucleus/metabolism , NAD+ Nucleosidase/metabolism , Nuclear Envelope/metabolism , 3T3 Cells , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/pharmacology , Animals , Cell Fractionation/methods , Cyclic ADP-Ribose , Genes, Reporter/genetics , Immunoblotting , Inositol 1,4,5-Trisphosphate/pharmacology , Membrane Glycoproteins , Mice , Microscopy, Confocal , Multienzyme Complexes , NAD/pharmacology , Recombinant Fusion Proteins/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism
20.
J Cell Biol ; 146(5): 1161-72, 1999 Sep 06.
Article in English | MEDLINE | ID: mdl-10477767

ABSTRACT

The multifunctional ADP-ribosyl cyclase, CD38, catalyzes the cyclization of NAD(+) to cyclic ADP-ribose (cADPr). The latter gates Ca(2+) release through microsomal membrane-resident ryanodine receptors (RyRs). We first cloned and sequenced full-length CD38 cDNA from a rabbit osteoclast cDNA library. The predicted amino acid sequence displayed 59, 59, and 50% similarity, respectively, to the mouse, rat, and human CD38. In situ RT-PCR revealed intense cytoplasmic staining of osteoclasts, confirming CD38 mRNA expression. Both confocal microscopy and Western blotting confirmed the plasma membrane localization of the CD38 protein. The ADP-ribosyl cyclase activity of osteoclastic CD38 was next demonstrated by its ability to cyclize the NAD(+) surrogate, NGD(+), to its fluorescent derivative cGDP-ribose. We then examined the effects of CD38 on osteoclast function. CD38 activation by an agonist antibody (A10) in the presence of substrate (NAD(+)) triggered a cytosolic Ca(2+) signal. Both ryanodine receptor modulators, ryanodine, and caffeine, markedly attenuated this cytosolic Ca(2+) change. Furthermore, the anti-CD38 agonist antibody expectedly inhibited bone resorption in the pit assay and elevated interleukin-6 (IL-6) secretion. IL-6, in turn, enhanced CD38 mRNA expression. Taken together, the results provide compelling evidence for a new role for CD38/ADP-ribosyl cyclase in the control of bone resorption, most likely exerted via cADPr.


Subject(s)
Antigens, CD , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Bone Resorption , NAD+ Nucleosidase/genetics , NAD+ Nucleosidase/metabolism , Osteoclasts/metabolism , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Antigens, Differentiation/chemistry , Base Sequence , Calcium Signaling , Cell Membrane/enzymology , Cells, Cultured , Cloning, Molecular , Cyclic ADP-Ribose , Enzyme Activation , Gene Expression Regulation, Enzymologic/drug effects , Humans , Interleukin-6/metabolism , Interleukin-6/pharmacology , Membrane Glycoproteins , Molecular Sequence Data , NAD/analogs & derivatives , NAD/metabolism , NAD+ Nucleosidase/chemistry , Osteoclasts/cytology , Osteoclasts/enzymology , RNA, Messenger/analysis , RNA, Messenger/genetics , Rabbits , Rats , Rats, Wistar , Ryanodine Receptor Calcium Release Channel/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL