Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
J Biochem ; 160(1): 59-68, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26896488

ABSTRACT

The binding properties of Ca(2+) to EF-hand I of aequorin (AQ) were characterized by replacing the loop sequence of EF-hand I (AQ[I]) with other known loop sequences of Ca(2+)-binding proteins, including photoproteins (aequorin, clytin-I, clytin-II and mitrocomin), Renilla luciferin-binding protein (RLBP) and calmodulin (CaM). For evaluation of the binding affinity of Ca(2+) to AQ[I] mutants, the half-decay time of the maximum intensity in the luminescence reaction triggered by Ca(2+) was used as an indicator and 22 kinds of AQ[I] mutants were expressed in Escherichia coli cells. AQ[I] mutants replaced with the EF-hand I and EF-hand III from photoproteins showed sufficient luminescence activity, but it was not shown by other EF-hands from RLBP and CaM. An AQ[I] mutant with a lysine or arginine residue at the second position of the non-conserved amino acid residue showed a slow-decay pattern of luminescence, indicating that the Ca(2+)-binding affinity to aequorin was reduced by a positive charge at the second position of the loop sequence. The specific loop sequence of the EF-hand I motif in aequorin caused the specific Ca(2+)-triggered luminescence pattern.


Subject(s)
Aequorin , Calcium-Binding Proteins , Gene Expression , Aequorin/biosynthesis , Aequorin/chemistry , Aequorin/genetics , Amino Acid Motifs , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Mutation , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
2.
Biochim Biophys Acta ; 1853(9): 2173-82, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25892494

ABSTRACT

Caveolae are plasma membrane invaginations enriched in sterols and sphingolipids. Sphingosine kinase 1 (SK1) is an oncogenic protein that converts sphingosine to sphingosine 1-phosphate (S1P), which is a messenger molecule involved in calcium signaling. Caveolae contain calcium responsive proteins, but the effects of SK1 or S1P on caveolar calcium signaling have not been investigated. We generated a Caveolin-1-Aequorin fusion protein (Cav1-Aeq) that can be employed for monitoring the local calcium concentration at the caveolae ([Ca²âº]cav). In HeLa cells, Cav1-Aeq reported different [Ca²âº] as compared to the plasma membrane [Ca²âº] in general (reported by SNAP25-Aeq) or as compared to the cytosolic [Ca²âº] (reported by cyt-Aeq). The Ca²âº signals detected by Cav1-Aeq were significantly attenuated when the caveolar structures were disrupted by methyl-ß-cyclodextrin, suggesting that the caveolae are specific targets for Ca²âº signaling. HeLa cells overexpressing SK1 showed increased [Ca²âº]cav during histamine-induced Ca²âº mobilization in the absence of extracellular Ca²âº as well as during receptor-operated Ca²âº entry (ROCE). The SK1-induced increase in [Ca²âº]cav during ROCE was reverted by S1P receptor antagonists. In accordance, pharmacologic inhibition of SK1 reduced the [Ca²âº]cav during ROCE. S1P treatment stimulated the [Ca²âº]cav upon ROCE. The Ca²âº responses at the plasma membrane in general were not affected by SK1 expression. In summary, our results show that SK1/S1P-signaling regulates Ca²âº signals at the caveolae. This article is part of a Special Issue entitled: 13th European Symposium on Calcium.


Subject(s)
Aequorin/biosynthesis , Calcium Signaling/physiology , Caveolae/metabolism , Caveolin 1/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Recombinant Fusion Proteins/biosynthesis , Aequorin/genetics , Calcium/metabolism , Calcium Signaling/drug effects , Caveolin 1/genetics , HeLa Cells , Humans , Lysophospholipids/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Recombinant Fusion Proteins/genetics , Sphingosine/analogs & derivatives , Sphingosine/pharmacology
3.
Regul Toxicol Pharmacol ; 69(2): 243-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24768935

ABSTRACT

Calcium-binding proteins are ubiquitous modulators of cellular activity and function. Cells possess numerous calcium-binding proteins that regulate calcium concentration in the cytosol by buffering excess free calcium ion. Disturbances in intracellular calcium homeostasis are at the heart of many age-related conditions making these proteins targets for therapeutic intervention. A calcium-binding protein, apoaequorin, has shown potential utility in a broad spectrum of applications for human health and well-being. Large-scale recombinant production of the protein has been successful; enabling further research and development and commercialization efforts. Previous work reported a 90-day subchronic toxicity test that demonstrated this protein has no toxicity by oral exposure in Sprague-Dawley rodents. The current study assesses the allergenic potential of the purified protein using bioinformatic analysis and simulated gastric digestion. The results from the bioinformatics searches with the apoaequorin sequence show the protein is not a known allergen and not likely to cross-react with known allergens. Apoaequorin is easily digested by pepsin, a characteristic commonly exhibited by many non-allergenic dietary proteins. From these data, there is no added concern of safety due to unusual stability of the protein by ingestion.


Subject(s)
Aequorin/genetics , Aequorin/toxicity , Apoproteins/genetics , Apoproteins/toxicity , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/toxicity , Escherichia coli/genetics , Safety , Aequorin/administration & dosage , Aequorin/biosynthesis , Aequorin/immunology , Allergens/immunology , Amino Acid Sequence , Animals , Apoproteins/administration & dosage , Apoproteins/biosynthesis , Apoproteins/immunology , Calcium-Binding Proteins/administration & dosage , Calcium-Binding Proteins/immunology , Computational Biology , Escherichia coli/metabolism , Gastric Mucosa/metabolism , Molecular Sequence Data , Pepsin A/metabolism , Protein Stability , Rats , Recombinant Proteins/administration & dosage , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/toxicity , Risk Assessment , Toxicity Tests, Subchronic
4.
J Exp Bot ; 63(4): 1751-61, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22213817

ABSTRACT

Calcium has long been acknowledged as one of the most important signalling components in plants. Many abiotic and biotic stimuli are transduced into a cellular response by temporal and spatial changes in cellular calcium concentration and the calcium-sensitive protein aequorin has been exploited as a genetically encoded calcium indicator for the measurement of calcium in planta. The objective of this work was to generate a compatible set of aequorin expression plasmids for the generation of transgenic plant lines to measure changes in calcium levels in different cellular subcompartments. Aequorin was fused to different targeting peptides or organellar proteins as a means to localize it to the cytosol, the nucleus, the plasma membrane, and the mitochondria. Furthermore, constructs were designed to localize aequorin in the stroma as well as the inner and outer surface of the chloroplast envelope membranes. The modular set-up of the plasmids also allows the easy replacement of targeting sequences to include other compartments. An additional YFP-fusion was included to verify the correct subcellular localization of all constructs by laser scanning confocal microscopy. For each construct, pBin19-based binary expression vectors driven by the 35S or UBI10 promoter were made for Agrobacterium-mediated transformation. Stable Arabidopsis lines were generated and initial tests of several lines confirmed their feasibility to measure calcium signals in vivo.


Subject(s)
Aequorin/biosynthesis , Aequorin/genetics , Arabidopsis/genetics , Arabidopsis/metabolism , Calcium/metabolism , Brassica rapa/genetics , Calcium Signaling , Gene Expression Regulation, Plant , Genetic Variation , Plants, Genetically Modified , Plasmids/genetics , Signal Transduction , Nicotiana/genetics
5.
Biochim Biophys Acta ; 1820(8): 1160-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22198462

ABSTRACT

BACKGROUND: In recent years, as our understanding of the various roles played by Ca2+ signaling in development and differentiation has expanded, the challenge of imaging Ca2+ dynamics within living cells, tissues, and whole animal systems has been extended to include specific signaling activity in organelles and non-membrane bound sub-cellular domains. SCOPE OF REVIEW: In this review we outline how recent advances in genetics and molecular biology have contributed to improving and developing current bioluminescence-based Ca2+ imaging techniques. Reporters can now be targeted to specific cell types, or indeed organelles or domains within a particular cell. MAJOR CONCLUSIONS: These advances have contributed to our current understanding of the specificity and heterogeneity of developmental Ca2+ signaling. The improvement in the spatial resolution that results from specifically targeting a Ca2+ reporter has helped to reveal how a ubiquitous signaling messenger like Ca2+ can regulate coincidental but different signaling events within an individual cell; a Ca2+ signaling paradox that until now has been hard to explain. GENERAL SIGNIFICANCE: Techniques used to target specific reporters via genetic means will have applications beyond those of the Ca2+ signaling field, and these will, therefore, make a significant contribution in extending our understanding of the signaling networks that regulate animal development. This article is part of a Special Issue entitled Biochemical, biophysical and genetic approaches to intracellular calcium signalling.


Subject(s)
Aequorin/genetics , Calcium Signaling , Aequorin/biosynthesis , Animals , Animals, Genetically Modified , Embryonic Development , Gene Expression , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Microscopy, Fluorescence , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Spectrometry, Fluorescence
6.
Int J Dev Biol ; 55(2): 153-74, 2011.
Article in English | MEDLINE | ID: mdl-21553383

ABSTRACT

Intact zebrafish embryos were used as an in vivo animal model to investigate the role of Ca2+ signaling during the differentiation of slow muscle cells (SMCs) within forming skeletal muscle. Transgenic zebrafish were generated using an a-actin promoter that targeted apoaequorin expression specifically to muscle cells. Two distinct Ca2+ signaling periods (CSPs) were visualized in the developing SMCs: between ~17.5-19.5 hours post-fertilization (hpf) and after ~23 hpf, separated by a ~3.5 h Ca2+ signaling quiet period. Further spatial characterization of these Ca2+ signals using confocal fluorescent microscopy and calcium green-1 dextran as a reporter, indicated that the earlier CSP displayed distinct nuclear and cytoplasmic components, whereas the later CSP was predominantly cytoplasmic. Both CSPs consisted of a series of oscillating Ca2+ waves generated at distinct frequencies, while the earlier CSP also displayed a slow rise then fall in the Ca2+ baseline-level. Imaging of cyclopamine- and forskolin-treated wild-type, or smo-/- mutant embryos, where SMCs do not form, confirmed the specific cell population generating the signals. Treating embryos with antagonists indicated that both IP3Rs and RyRs are responsible for generating the temporal characteristics of the Ca2+ signaling signature, and that the latter plays a necessary role in SMC differentiation and subsequent myotome patterning. Together, these data support and extend the proposition that specific spatiotemporal patterns of spontaneous Ca2+ signals might be used for different as well as combinatorial regulation of both nuclear and cytosolic signal transduction cascades, resulting in myofibrillogenesis in SMCs as well as myotome patterning.


Subject(s)
Calcium Signaling , Muscle Development/physiology , Muscle, Skeletal/embryology , Zebrafish/embryology , Zebrafish/metabolism , Actins/metabolism , Aequorin/biosynthesis , Aequorin/genetics , Animals , Animals, Genetically Modified , Apoproteins/biosynthesis , Apoproteins/genetics , Bungarotoxins/pharmacology , Calcium/metabolism , Colforsin/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Muscle Development/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myosins/metabolism , Nifedipine/pharmacology , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Veratrum Alkaloids/pharmacology
7.
Microsc Res Tech ; 73(8): 785-96, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20169618

ABSTRACT

The bioluminescent hydromedusan jellyfish, Aequorea victoria, emits a greenish light (lambda(max) = 508 nm) when stimulated electrically or mechanically. The light comes from photocytes located along the margin of its umbrella. The greenish light depends on two intracellular proteins working in consort: aequorin (21.4 kDa) and a green fluorescent protein (27 kDa). An excited state green fluorescent protein molecule results, which, on returning to the ground state, emits a greenish light. Similarly, a green light emission may be induced in the green fluorescent protein by exposing it to ultraviolet or blue light. Because the green light can be readily detected under a fluorescence microscope, the green fluorescent protein, tagged to a protein of interest, has been used widely as a marker to locate proteins in cells and to monitoring gene expression. This article reviews the work that took place leading to the discovery, cloning, and expression of the green fluorescent protein, with a note on an unfinished experiment.


Subject(s)
Aequorin/history , Aequorin/biosynthesis , Aequorin/isolation & purification , Animals , History, 20th Century , Luminescence , Scyphozoa , Stereoisomerism , United States
8.
Biochem Biophys Res Commun ; 386(4): 617-22, 2009 Sep 04.
Article in English | MEDLINE | ID: mdl-19549504

ABSTRACT

Blue fluorescent protein of aequorin (BFP) is a complex of Ca(2+)-bound apoaequorin with coelenteramide and is a bifunctional protein, which shows blue fluorescence and the luminescence activity like a luciferase. To reconstitute synthetic BFP (syn-BFP) from apoaequorin and coelenteramide, we established new synthetic route of coelenteramide and prepared highly purified recombinant aequorin using the histidine-tagged secretion system in Escherichia coli cells. As a result, we succeeded in reconstituting syn-BFP quantitatively and the fluorescence and luminescence properties of syn-BFP were identical to that of BFP obtained from aequorin.


Subject(s)
Aequorin/biosynthesis , Aequorin/chemistry , Apoproteins/biosynthesis , Apoproteins/chemistry , Benzeneacetamides/chemistry , Luminescent Proteins/biosynthesis , Luminescent Proteins/chemistry , Pyrazines/chemistry , Aequorin/isolation & purification , Apoproteins/isolation & purification , Benzeneacetamides/chemical synthesis , Escherichia coli/metabolism , Luminescent Proteins/isolation & purification , Pyrazines/chemical synthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Spectrometry, Fluorescence
9.
Anal Chem ; 80(22): 8470-6, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18937418

ABSTRACT

Progress in the miniaturization and automation of complex analytical processes depends largely on increasing the sensitivity, diversity, and robustness of current labels. Because of their ubiquity and ease of use, fluorescent, enzymatic, and bioluminescent labels are often employed in such miniaturized and multiplexed formats, with each type of label having its own unique advantages and drawbacks. The ultrasensitive detection limits of bioluminescent reporters are especially advantageous when dealing with very small sample volumes and biological fluids. However, bioluminescent reporters currently do not have the multiplexing capability that fluorescent labels do. In an effort to address this limitation, we have developed a method of discriminating two semisynthetic aequorin variants from one another using time resolution. In this work we paired two aequorin conjugates with different coelenterazine analogues and then resolved the two signals from one another using the difference in decay kinetics and half-life times. Utilizing this time-resolution, we then developed a simultaneous, dual-analyte, single well assay for 6-keto-prostaglandin-FI-alpha and angiotensin II, two important cardiovascular molecules.


Subject(s)
Aequorin/genetics , Aequorin/metabolism , Mutation , 6-Ketoprostaglandin F1 alpha/analysis , 6-Ketoprostaglandin F1 alpha/metabolism , Aequorin/biosynthesis , Angiotensin II/analysis , Angiotensin II/metabolism , Animals , Escherichia coli/metabolism , Half-Life , Imidazoles/chemistry , Imidazoles/metabolism , Immunoassay , Kinetics , Protein Engineering , Pyrazines/chemistry , Pyrazines/metabolism
10.
Biochem Biophys Res Commun ; 376(3): 448-53, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-18789309

ABSTRACT

We constructed a cold induced expression vector in Escherichia coli cells that consists of a histidine tag sequence for nickel chelate affinity purification, IgG-binding domain of protein A (ZZ-domain) and the multiple cloning sites. The role of ZZ-domain as a solubilizing partner at 15 degrees C was demonstrated by expressing the imidazopyrazinone-type luciferases of Renilla, Oplophorus, Gaussia, and Vargula (Cypridina) as well as the calcium-binding photoproteins and firefly luciferase. The fused protein with ZZ-domain was expressed efficiently as a soluble form in the cytoplasm of E. coli cells at low temperature.


Subject(s)
Cold Temperature , Genetic Vectors , Protein Biosynthesis , Recombinant Fusion Proteins/biosynthesis , Aequorin/biosynthesis , Aequorin/chemistry , Aequorin/genetics , Amino Acid Sequence , Animals , Apoproteins/biosynthesis , Apoproteins/chemistry , Apoproteins/genetics , CCAAT-Enhancer-Binding Proteins/genetics , Escherichia coli/genetics , Heat-Shock Proteins/genetics , Humans , Immunoglobulin G/immunology , Luciferases/genetics , Molecular Sequence Data , Protein Structure, Tertiary/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Solubility , Staphylococcal Protein A/chemistry , Staphylococcal Protein A/genetics , Staphylococcal Protein A/immunology
11.
Article in Russian | MEDLINE | ID: mdl-18819405

ABSTRACT

Results of development of shuttle expressing plasmid vector Escherichia coli-Lactobacillus which allowed high level expression of heterologous genes in lactobacilli are represented. Vector pTRKH2 which is able to replicate in E. coli and in wide range of Gram-positive bacteria was used as the base. In order to provide high level of cloned gene expression constitutive-active synthetic promoter, site of initiation of translation, and terminator of transcription were introduced in the vector. Functional activity of this vector was confirmed using green fluorescent protein (GFP) gene from Aequoria victoria. Transformation of model strain by gfp gene-carrying plasmid resulted in appearance of typical fluorescent phenotype.


Subject(s)
Genetic Vectors , Lactobacillus plantarum/metabolism , Recombinant Proteins/biosynthesis , Aequorin/biosynthesis , Aequorin/genetics , Animals , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Hydrozoa/genetics , Lactobacillus plantarum/genetics , Promoter Regions, Genetic , Recombinant Proteins/genetics , Transformation, Genetic
12.
Br J Pharmacol ; 151(5): 647-54, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17471180

ABSTRACT

BACKGROUND AND PURPOSE: The thiourea derivative KB-R7943, originally developed as inhibitor of the plasma membrane Na(+)/Ca(2+) exchanger, has been shown to protect against myocardial ischemia-reperfusion injury. We have studied here its effects on mitochondrial Ca(2+) fluxes. EXPERIMENTAL APPROACH: [Ca(2+)] in cytosol, mitochondria and endoplasmic reticulum (ER), and mitochondrial membrane potential were monitored using both luminescent (targeted aequorins) and fluorescent (fura-2, tetramethylrhodamine ethyl ester) probes in HeLa cells. KEY RESULTS: KB-R7943 was also a potent inhibitor of the mitochondrial Ca(2+) uniporter (MCU). In permeabilized HeLa cells, KB-R7943 inhibited mitochondrial Ca(2+) uptake with a Ki of 5.5+/-1.3 microM (mean+/-S.D.). In intact cells, 10 microM KB-R7943 reduced by 80% the mitochondrial [Ca(2+)] peak induced by histamine. KB-R7943 did not modify the mitochondrial membrane potential and had no effect on the mitochondrial Na(+)/Ca(2+) exchanger. KB-R7943 inhibited histamine-induced ER-Ca(2+) release in intact cells, but not in cells loaded with a Ca(2+)-chelator to damp cytosolic [Ca(2+)] changes. Therefore, inhibition of ER-Ca(2+)-release by KB-R7943 was probably due to the increased feedback Ca(2+)-inhibition of inositol 1,4,5-trisphosphate receptors after MCU block. This mechanism also explains why KB-R7943 reversibly blocked histamine-induced cytosolic [Ca(2+)] oscillations in the same range of concentrations required to inhibit MCU. CONCLUSIONS AND IMPLICATIONS: Inhibition of MCU by KB-R7943 may contribute to its cardioprotective activity by preventing mitochondrial Ca(2+)-overload during ischemia-reperfusion. In addition, the effects of KB-R7943 on Ca(2+) homeostasis provide new evidence for the role of mitochondria modulating Ca(2+)-release and regenerative Ca(2+)-oscillations. Search for permeable and selective MCU inhibitors may yield useful pharmacological tools in the future.


Subject(s)
Calcium Channels/metabolism , Mitochondria/metabolism , Sodium-Calcium Exchanger/antagonists & inhibitors , Thiourea/analogs & derivatives , Aequorin/biosynthesis , Calcium/metabolism , Calcium Channels/drug effects , Cell Membrane/drug effects , Cell Membrane/enzymology , Cells, Cultured , Cytosol/drug effects , Cytosol/metabolism , Fluorescent Dyes , Fura-2 , HeLa Cells , Histamine/pharmacology , Homeostasis/drug effects , Humans , Inositol 1,4,5-Trisphosphate/physiology , Inositol 1,4,5-Trisphosphate Receptors/drug effects , Membrane Potentials/drug effects , Mitochondria/drug effects , Thiourea/pharmacology
13.
Int J Dev Biol ; 50(6): 561-9, 2006.
Article in English | MEDLINE | ID: mdl-16741871

ABSTRACT

When aequorin is microinjected into cleavage-stage zebrafish embryos, it is largely used up by ~24 hours. Thus, it is currently not possible to image Ca(2+) signals from later stages of zebrafish development using this approach. We have, therefore, developed protocols to express apoaequorin, i.e., the protein component of aequorin, transiently in zebrafish embryos and then reconstitute intact aequorin in vivo by loading the coelenterazine co-factor into the embryos separately. Two types of apoaequorin mRNA, aeq-mRNA and aeq::EGFP-mRNA, the latter containing the enhanced green fluorescent protein (EGFP) sequence, were in vitro transcribed and when these were microinjected into embryos, they successfully translated apoaequorin and a fusion protein of apoaequorin and EGFP (apoaequorin-EGFP), respectively. We show that aeq::EGFP -mRNA was more toxic to embryos than equivalent amounts of aeq-mRNA. In addition, in an in vitro reconstitution assay, apoaequorin-EGFP produced less luminescence than apoaequorin, after reconstitution with coelenterazine and with the addition of Ca(2+). Furthermore, when imaging intact coelenterazine-loaded embryos that expressed apoaequorin, Ca(2+ )signals from ~2.5 to 48 hpf were observed, with the spatio-temporal pattern of these signals up to 24 hpf, being comparable to that observed with aequorin. This transient aequorin expression approach using aeq-mRNA provides a valuable tool for monitoring Ca(2+ )signaling during the 2448 hpf period of zebrafish development. Thus, it effectively extends the aequorin-based Ca(2+) imaging window by an additional 24 hours.


Subject(s)
Aequorin/genetics , Apoproteins/genetics , Calcium Signaling/physiology , Zebrafish/embryology , Aequorin/biosynthesis , Animals , Apoproteins/biosynthesis , Imidazoles , Microscopy, Fluorescence , Pyrazines , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Transfection , Zebrafish/metabolism
14.
Br J Pharmacol ; 145(7): 862-71, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15912132

ABSTRACT

Ca(2+) uptake by mitochondria is a key element in the control of cellular Ca(2+) homeostasis and Ca(2+)-dependent phenomena. It has been known for many years that this Ca(2+) uptake is mediated by the mitochondrial Ca(2+) uniporter, a specific Ca(2+) channel of the inner mitochondrial membrane. We have shown previously that this channel is strongly activated by a series of natural phytoestrogenic flavonoids. We show here that several agonists and antagonists of estrogen receptors (ERs) also modulate the activity of the uniporter. The specific alpha-ER agonist 4,4',4''-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT) was the strongest activator, increasing the rate of mitochondrial Ca(2+) uptake in permeabilized HeLa cells by 10-fold at 2 microM. Consistently, PPT largely increased the histamine-induced mitochondrial [Ca(2+)] peak and reduced the cytosolic one. Diethylstilbestrol and 17-beta-estradiol (but not 17-alpha-estradiol) were active at pharmacological concentrations while the beta-estrogen-receptor agonist 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN) was little effective. The ER modulators tamoxifen and 4-hydroxy-tamoxifen inhibited mitochondrial Ca(2+) uptake (IC(50) 2.5+/-1.5 and 2.5+/-1.4 microM, mean+/-s.d., respectively) both in the presence and in the absence of PPT, but raloxifene and the pure estrogen antagonist ICI 182,780 produced no effect. Activation by PPT was immediate and inhibition by tamoxifen or 4-hydroxy-tamoxifen required only 5 min to reach maximum. Tamoxifen did not modify mitochondrial membrane potential and PPT induced a slow mitochondrial depolarization at higher concentrations than those required to activate mitochondrial Ca(2+) uptake. These results suggest that some kind of ER or related protein located in mitochondria controls the activity of the Ca(2+) uniporter by a nongenomic mechanism. This novel mechanism of action of estrogen agonists and antagonists can provide a new interpretation for several previously reported effects of these compounds.


Subject(s)
Calcium Channels/drug effects , Calcium/metabolism , Mitochondria/metabolism , Phenols/pharmacology , Pyrazoles/pharmacology , Receptors, Estrogen/agonists , Selective Estrogen Receptor Modulators/pharmacology , Aequorin/biosynthesis , Aequorin/genetics , Calcium/analysis , Cell Line, Tumor , Diethylstilbestrol/pharmacology , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Estrogens, Non-Steroidal/pharmacology , Female , HeLa Cells , Humans , Luminescent Agents , Membrane Potentials/drug effects , Mitochondria/drug effects , Organometallic Compounds , Receptors, Estrogen/antagonists & inhibitors , Tamoxifen/analogs & derivatives , Tamoxifen/metabolism , Tamoxifen/pharmacology , Transfection
15.
Cell Calcium ; 36(6): 479-87, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15488597

ABSTRACT

Extracellular agonists mobilize Ca2+ from SERCA-comprising intracellular Ca2+ stores located in both the Golgi apparatus and the endoplasmic reticulum. Ca2+ release from both these compartments was studied in HeLa cells stably expressing the luminescent Ca2+ indicator aequorin specifically targeted to these compartments. Changes in lumenal [Ca2+] as detected by the aequorin measurements were correlated with parallel changes in total Ca2+ content of the stores. The latencies and initial rates of Ca2+ release from the Golgi apparatus and the endoplasmic reticulum were quite similar. However, maximal Ca2+ release measured with Golgi-targeted aequorin terminated faster than that from the endoplasmic reticulum. The rate and extent of Ca2+ depletion from both compartments correlated well with the peak amplitude of the cytosolic [Ca2+] rise. Time-course experiments further revealed that the peak of the cytosolic Ca2+ response occurred before the lumenal [Ca2+] reached its lowest level. We conclude that both the Golgi apparatus and the endoplasmic reticulum contribute to the rise in cytosolic [Ca2+] upon agonist stimulation, but the kinetics of the Ca2+ release are different.


Subject(s)
Aequorin/biosynthesis , Aequorin/genetics , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Aequorin/metabolism , Dose-Response Relationship, Drug , Endoplasmic Reticulum/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Golgi Apparatus/drug effects , HeLa Cells , Humans , Inositol 1,4,5-Trisphosphate/pharmacology
16.
Anal Biochem ; 324(2): 258-68, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14690690

ABSTRACT

We have expressed aequorin in mitochondria of the yeast Saccharomyces cerevisiae and characterized the resulting strain with respect to mitochondrial Ca(2+) transport in vivo and in vitro. When intact cells are suspended in water containing 1.4 mM ethanol and 14 mM CaCl(2), the matrix free Ca(2+) concentration is 200 nM, similar to the values expected in cytoplasm. Addition of ionophore ETH 129 allows an active accumulation of Ca(2+) and promptly increases the value to 1.2 microM. Elevated Ca(2+) concentrations are maintained for periods of 6 min or longer under these conditions. Isolated yeast mitochondria oxidizing ethanol also accumulate Ca(2+) when ETH 129 is present, but the cation is not retained depending on the medium conditions. This finding confirms the presence of a Ca(2+) release mechanism that requires free fatty acids as previously described [P.C. Bradshaw et al. (2001) J. Biol. Chem. 276, 40502-40509]. When a respiratory substrate is not present, Ca(2+) enters and leaves yeast mitochondria slowly, at a specific activity near 0.2 nmol/min/mg protein. Transport under these conditions equilibrates the internal and external concentrations of Ca(2+) and is not affected by ruthenium red, uncouplers, or ionophores that perturb transmembrane gradients of charge and pH. This activity displays sigmoid kinetics and a K(1/2) value for Ca(2+) that is near to 900 nM, in the absence of ethanol or when it is present. It is furthermore shown that the activity coefficient of Ca(2+) in yeast mitochondria is a function of the matrix Ca(2+) content and is substantially larger than that in mammalian mitochondria. Characteristics of the aequorin-expressing strain appear suitable for its use in expression-based methods directed at cloning Ca(2+) transporters from mammalian mitochondria and for further examining the interrelationships between mitochondrial and cytoplasmic Ca(2+) in yeast.


Subject(s)
Aequorin/biosynthesis , Calcium/metabolism , Mitochondria/metabolism , Saccharomyces cerevisiae/ultrastructure , Biological Transport , Cation Transport Proteins , Kinetics , Mitochondrial Proteins/metabolism , Recombinant Proteins , Saccharomyces cerevisiae/genetics
17.
Mikrobiologiia ; 73(6): 734-40, 2004.
Article in Russian | MEDLINE | ID: mdl-15688931

ABSTRACT

Using the mutant strain Aspergillus awamori 66A producing a recombinant Ca2+-dependent photosensitive protein aequorin, the dynamics of Ca2+ was studied for the first time in the cytosol of the micromycetes exposed to stressful factors, such as an increase in extracellular Ca2+ to 50 mM, hypoosmotic shock, and mechanical shock. Cell response to stress proved to involve an increase in the Ca2+ concentration in the cytosol, which was determined from the amplitude of aequorin luminescence and the time of the amplitude enhancement and relaxation. The level of Ca2+ response depended on the physiological stimulus. Inhibitory analysis with various agents that block Ca2+ channels and with agonists that specifically enhance the activity of the channels suggested that (1) the level of Ca2+ in the cytosol of micromycetes increases in response to stress because of the ion influx from both the growth medium and intracellular reservoirs and (2) the potential-dependent transport systems play the major role in the Ca2+ influx into the cytosol of the micromycete cells.


Subject(s)
Aspergillus/physiology , Aequorin/biosynthesis , Aequorin/genetics , Aspergillus/genetics , Aspergillus/metabolism , Calcium Channels/metabolism , Calcium Chloride/pharmacology , Cytosol/metabolism , Osmotic Pressure , Recombinant Proteins/biosynthesis
19.
Science ; 301(5630): 213-5, 2003 Jul 11.
Article in English | MEDLINE | ID: mdl-12855808

ABSTRACT

We report the use of genetically engineered cells in a pathogen identification sensor. This sensor uses B lymphocytes that have been engineered to emit light within seconds of exposure to specific bacteria and viruses. We demonstrated rapid screening of relevant samples and identification of a variety of pathogens at very low levels. Because of its speed, sensitivity, and specificity, this pathogen identification technology could prove useful for medical diagnostics, biowarfare defense, food- and water-quality monitoring, and other applications.


Subject(s)
B-Lymphocytes , Bacteria/isolation & purification , Bacteriological Techniques , Biosensing Techniques , Viruses/isolation & purification , Aequorin/biosynthesis , Antibodies, Bacterial/immunology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Bacillus anthracis/immunology , Bacillus anthracis/isolation & purification , Bacteria/immunology , Cell Line , Colony Count, Microbial , Encephalitis Virus, Venezuelan Equine/immunology , Encephalitis Virus, Venezuelan Equine/isolation & purification , Escherichia coli O157/immunology , Escherichia coli O157/isolation & purification , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease Virus/isolation & purification , Immunoglobulin Variable Region/immunology , Light , Receptors, Antigen, B-Cell/immunology , Sensitivity and Specificity , Time Factors , Transfection , Viruses/immunology , Yersinia pestis/immunology , Yersinia pestis/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...