Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
J Nutr Health Aging ; 23(7): 641-647, 2019.
Article in English | MEDLINE | ID: mdl-31367729

ABSTRACT

BACKGROUND: The number of older adults attending emergency department (ED) is increasing all over the world. Usually, those patients are potentially more complex due to their greater number of comorbidities, cognitive disorders, and functional or physical disabilities. Frailty is a vulnerable state that could predict adverse outcomes of those patients. There are very few studies that addressed this topic in the ED, and none of them used a simple instrument for frailty assessment. OBJECTIVES: The primary outcome was to evaluate the association between frailty identified through the FRAIL questionnaire at baseline and death after a 6-month follow-up period after hospital discharge from the ED. Secondary outcomes were readmission to the ED and disability after 6 months. METHODS: A 6-month follow-up prospective study (FASES study) was conducted at a university-based trauma-center ED in Jundiaí, southwestern of Brazil. A total of 316 older adults aged 60 or older were randomly included based on a lottery of their medical record admission number. Frailty was evaluated through the FRAIL questionnaire. The association between frailty and death was estimated through a binary logistic regression adjusted for age, sex, and cognitive performance. RESULTS: From the total sample, the mean age was 72.11±8.0 years, and 51.6% were women. Participants presented 2.28±1.4 comorbidities and 25.6% were frail. Mean hospital stay was 5.43±5.6 days. Death occurred in 52 participants, readmission to the emergency in 55, and new disability in 16 after 6 months. Frailty was associated with an odds ratio of 2.18 for death after 6 months (95% CI = 1.10-4.31; p = 0.024). This association lost significance after multivariate analysis taking into account cognitive performance. There was no association between frailty status at baseline and readmission to the ED or disability. CONCLUSION: The identification of frailty using the FRAIL at admission was not predictive of death after a 6-month period after discharge from the ED. Simple frailty assessment could identify patients at higher risk for death in the follow-up.


Subject(s)
Comorbidity , Frail Elderly/statistics & numerical data , Frailty/mortality , Geriatric Assessment/statistics & numerical data , Aged , Aged, 80 and over , Brazil , Emergency Service, Hospital/statistics & numerical data , Female , Follow-Up Studies , Hospitalization/statistics & numerical data , Humans , Income , Length of Stay/statistics & numerical data , Logistic Models , Male , Odds Ratio , Patient Discharge/statistics & numerical data , Prospective Studies , Risk Assessment , Surveys and Questionnaires
2.
Br J Pharmacol ; 167(8): 1712-22, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22862290

ABSTRACT

BACKGROUND AND PURPOSE: Pyrazole derivatives have recently been suggested as selective blockers of transient receptor potential cation (TRPC) channels but their ability to distinguish between the TRPC and Orai pore complexes is ill-defined. This study was designed to characterize a series of pyrazole derivatives in terms of TRPC/Orai selectivity and to delineate consequences of selective suppression of these pathways for mast cell activation. EXPERIMENTAL APPROACH: Pyrazoles were generated by microwave-assisted synthesis and tested for effects on Ca(2+) entry by Fura-2 imaging and membrane currents by patch-clamp recording. Experiments were performed in HEK293 cells overexpressing TRPC3 and in RBL-2H3 mast cells, which express classical store-operated Ca(2+) entry mediated by Orai channels. The consequences of inhibitory effects on Ca(2+) signalling in RBL-2H3 cells were investigated at the level of both degranulation and nuclear factor of activated T-cells activation. KEY RESULTS: Pyr3, a previously suggested selective inhibitor of TRPC3, inhibited Orai1- and TRPC3-mediated Ca(2+) entry and currents as well as mast cell activation with similar potency. By contrast, Pyr6 exhibited a 37-fold higher potency to inhibit Orai1-mediated Ca(2+) entry as compared with TRPC3-mediated Ca(2+) entry and potently suppressed mast cell activation. The novel pyrazole Pyr10 displayed substantial selectivity for TRPC3-mediated responses (18-fold) and the selective block of TRPC3 channels by Pyr10 barely affected mast cell activation. CONCLUSIONS AND IMPLICATIONS: The pyrazole derivatives Pyr6 and Pyr10 are able to distinguish between TRPC and Orai-mediated Ca(2+) entry and may serve as useful tools for the analysis of cellular functions of the underlying Ca(2+) channels.


Subject(s)
Calcium Channels/physiology , Calcium/metabolism , Pyrazoles/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Cell Degranulation/drug effects , Cell Line, Tumor , HEK293 Cells , Humans , Mast Cells/drug effects , Mast Cells/physiology , Rats
3.
J Biomed Mater Res B Appl Biomater ; 100(1): 170-6, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21948557

ABSTRACT

We present a method for laser lithography of cell-adhesive arrays on a fluoropolymer surface. The method is based on 172 nm excimer-lamp photomodification in ammonia atmosphere followed by microstructuring by laser ablation. The improved wettability of the polymer is caused by new chemical groups on the surface after the UV treatment that we proved by Fourier transform infrared spectroscopy and X-ray photoelectron spectroscopy analyses. The cell adhesion properties of micropatterned structures were tested by cultivation of mammalian cells. We show that single elongated cells can grow confined to lines with sharply defined boundaries of the cell-covered areas. In preliminary experiments, we also demonstrate that the described technique allows the production of single-cell arrays with variable cell shape.


Subject(s)
Fluorine/chemistry , Lasers , Microarray Analysis , Polyethylene/chemistry , Polypropylenes/chemistry , Ultraviolet Rays , Animals , CHO Cells , Cell Adhesion , Cell Separation , Cricetinae , Cricetulus , Photochemistry/methods , Wettability
4.
Cell Death Dis ; 1: e75, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-21364678

ABSTRACT

The molecular nature of calcium (Ca(2+))-dependent mechanisms and the ion channels having a major role in the apoptosis of cancer cells remain a subject of debate. Here, we show that the recently identified Orai1 protein represents the major molecular component of endogenous store-operated Ca(2+) entry (SOCE) in human prostate cancer (PCa) cells, and constitutes the principal source of Ca(2+) influx used by the cell to trigger apoptosis. The downregulation of Orai1, and consequently SOCE, protects the cells from diverse apoptosis-inducing pathways, such as those induced by thapsigargin (Tg), tumor necrosis factor α, and cisplatin/oxaliplatin. The transfection of functional Orai1 mutants, such as R91W, a selectivity mutant, and L273S, a coiled-coil mutant, into the cells significantly decreased both SOCE and the rate of Tg-induced apoptosis. This suggests that the functional coupling of STIM1 to Orai1, as well as Orai1 Ca(2+)-selectivity as a channel, is required for its pro-apoptotic effects. We have also shown that the apoptosis resistance of androgen-independent PCa cells is associated with the downregulation of Orai1 expression as well as SOCE. Orai1 rescue, following Orai1 transfection of steroid-deprived cells, re-established the store-operated channel current and restored the normal rate of apoptosis. Thus, Orai1 has a pivotal role in the triggering of apoptosis, irrespective of apoptosis-inducing stimuli, and in the establishment of an apoptosis-resistant phenotype in PCa cells.


Subject(s)
Apoptosis , Calcium Channels/metabolism , Prostatic Neoplasms/metabolism , Amino Acid Substitution , Antineoplastic Agents/therapeutic use , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/physiology , Cell Line, Tumor , Cisplatin/therapeutic use , Humans , Male , Membrane Proteins/metabolism , Mutation , Neoplasm Proteins/metabolism , ORAI1 Protein , Phenotype , Prostatic Neoplasms/drug therapy , Stromal Interaction Molecule 1 , Thapsigargin/therapeutic use , Tumor Necrosis Factor-alpha/therapeutic use
5.
Cell Calcium ; 46(4): 227-32, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19733393

ABSTRACT

Ca(2+) entry in non-excitable cells is mainly carried by store-operated channels among which the CRAC channel is best characterized. Its two limiting molecular components are represented by the Ca(2+) sensor protein STIM1 located in the endoplasmic reticulum and Orai1 in the plasma membrane. STIM1 senses a decrease of the Ca(2+) content in internal stores and triggers its accumulation into puncta like structures resulting in coupling to as well as activation of Orai1 channels. The STIM1-Orai coupling process is determined by an interaction via their C-termini. This review highlights recent developments on domains particularly within the cytosolic part of STIM1 that govern this interaction.


Subject(s)
Calcium Channels/chemistry , Calcium Channels/metabolism , Calcium Channels/ultrastructure , Calcium/chemistry , Calcium/metabolism , Ion Channel Gating , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Neoplasm Proteins/ultrastructure , Protein Interaction Domains and Motifs , Animals , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Humans , ORAI1 Protein , Protein Binding , Stromal Interaction Molecule 1
6.
Biochem Soc Trans ; 35(Pt 1): 84-5, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17233607

ABSTRACT

The large family of mammalian TRP (transient receptor potential) ion channels encompasses diverse sensory functions. TRP proteins consist of six transmembrane domains, with a pore-loop motif between the fifth and sixth domains and cytosolic N- and C-termini. The intracellular strands not only interact with various proteins and lipids, but also include essential multimerization regions. This review summarizes the current knowledge of the intrinsic assembly domains that assure tetrameric TRP channel formation.


Subject(s)
TRPC Cation Channels/chemistry , TRPC Cation Channels/physiology , Amino Acid Motifs , Animals , Ankyrins/chemistry , Calcium Channels/chemistry , Cytosol/metabolism , Drosophila , Humans , Models, Biological , Protein Binding , Protein Structure, Tertiary , TRPV Cation Channels/chemistry
7.
Cardiovasc Res ; 73(1): 111-9, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17129578

ABSTRACT

OBJECTIVE: Members of the classical transient receptor potential protein (TRPC) family are considered as key components of phospholipase C (PLC)-dependent Ca2+ signaling. Previous results obtained in the HEK 293 expression system suggested a physical and functional coupling of TRPC3 to the cardiac-type Na+/Ca2+ exchanger, NCX1 (sodium calcium exchanger 1). This study was designed to test for expression of TRPC3 (transient receptor potential channel 3) and for the existence of a native TRPC3/NCX1 signaling complex in rat cardiac myocytes. METHODS: Protein expression and cellular distribution were determined by Western blot and immunocytochemistry. Protein-protein interactions were investigated by reciprocal co-immunoprecipitation and glutathione S-transferase (GST)-pulldown experiments. Recruitment of protein complexes into the plasma membrane was assayed by surface biotinylation. The functional role of TRPC3 was investigated by fluorimetric recording of angiotensin II-induced calcium signals employing a dominant negative knockdown strategy. RESULTS: TRPC3 immunoreactivity was observed in surface plasma membrane regions and in an intracellular membrane system. Co-immunolabeling of TRPC3 and NCX1 indicated significant co-localization of the two proteins. Both co-immunoprecipitation and GST-pulldown experiments demonstrated association of TRPC3 with NCX1. PLC stimulation was found to trigger NCX-mediated Ca2+ entry, which was dependent on TRPC3-mediated Na+ loading of myocytes. This NCX-mediated Ca2+ signaling was significantly suppressed by expression of a dominant negative fragment of TRPC3. PLC stimulation was associated with increased membrane presentation of both TRPC3 and NCX1. CONCLUSION: These results suggest a PLC-dependent recruitment of a TRPC3-NCX1 complex into the plasma membrane as a pivotal mechanism for the control of cardiac Ca2+ homeostasis.


Subject(s)
Calcium/metabolism , Myocytes, Cardiac/metabolism , Signal Transduction/physiology , Sodium-Calcium Exchanger/metabolism , TRPC Cation Channels/metabolism , Type C Phospholipases/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Homeostasis , Humans , Immunohistochemistry/methods , Immunoprecipitation/methods , Microscopy, Confocal , Myocytes, Cardiac/chemistry , Rats , Rats, Sprague-Dawley , TRPC Cation Channels/analysis , TRPC Cation Channels/genetics , Transfection
8.
J Biomed Mater Res A ; 67(1): 130-7, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14517870

ABSTRACT

We report on the modification of polytetrafluoroethylene (PTFE) by exposure to the ultraviolet (UV) light of a Xe(2)*-excimer lamp at a wavelength of 172 nm in an ammonia atmosphere. Typical treatment times were up to 30 min. Subsequently, the samples were grafted with the amino acid alanine from an aqueous solution. The samples were characterized by means of optical transmission spectroscopy, laser-induced fluorescence and contact-angle measurements. We studied the adhesion of rat aortic smooth muscle cells (SMC) and mouse fibroblasts (3T3 cells) to the modified polymer samples using an in vitro technique, where the population density and spread of adhering cells is determined 24 h after seeding by image analysis. For both cell types the exposure of PTFE to UV-light in an ammonia atmosphere resulted in a significant increase in the number of adhering cells and in the size of their spreading area. The grafting with alanine enhanced this effect. Additional experiments with human endothelial cells (HEC) also demonstrated improved adhesion to modified PTFE. Thus, PTFE modified by our method appears to be a promising material for fabrication of artificial vascular prostheses and implants or for cultivation of skin substitutes.


Subject(s)
Ammonia , Cell Adhesion/physiology , Polytetrafluoroethylene , Ultraviolet Rays , 3T3 Cells , Animals , Biocompatible Materials/radiation effects , Humans , Mice , Polytetrafluoroethylene/radiation effects
9.
Biomaterials ; 24(28): 5139-44, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14568430

ABSTRACT

We studied the adhesion and proliferation of human endothelial cells on photochemically modified polytetrafluoroethylene samples. The polymer surfaces were modified by exposure to the ultraviolet light of a Xe(2)(*)-excimer lamp at a wavelength of 172 nm in an ammonia atmosphere. Treatment times were between 10 and 20 min. The endothelial cell density was determined 1, 3 and 8 days after seeding by image analysis. Surface modification of the samples resulted in a significant increase in the number of adhering cells and in the formation of a confluent cell layer after 3-8 days. The results were comparable than those obtained on polystyrene Petri dishes, which are used as standard substrates in cell cultivation. Thus modified PTFE appears to be a promising material for the fabrication of artificial vascular prostheses coated with endothelial cells.


Subject(s)
Biocompatible Materials/chemistry , Cell Culture Techniques/methods , Endothelial Cells/cytology , Endothelial Cells/physiology , Polytetrafluoroethylene/chemistry , Tissue Engineering/methods , Cell Adhesion/physiology , Cell Count , Cell Culture Techniques/instrumentation , Cell Division/physiology , Cells, Cultured , Humans , Materials Testing , Membranes, Artificial , Photochemistry/methods , Polytetrafluoroethylene/radiation effects , Surface Properties , Tissue Engineering/instrumentation , Ultraviolet Rays , Umbilical Veins/cytology , Umbilical Veins/physiology
10.
J Biol Chem ; 276(51): 48149-58, 2001 Dec 21.
Article in English | MEDLINE | ID: mdl-11600493

ABSTRACT

The role of Trp3 in cellular regulation of Ca(2+) entry by NO was studied in human embryonic kidney (HEK) 293 cells. In vector-transfected HEK293 cells (controls), thapsigargin (TG)-induced (capacitative Ca(2+) entry (CCE)-mediated) intracellular Ca(2+) signals and Mn(2+) entry were markedly suppressed by the NO donor 2-(N,N-diethylamino)diazenolate-2-oxide sodium salt (3 microm) or by authentic NO (100 microm). In cells overexpressing Trp3 (T3-9), TG-induced intracellular Ca(2+) signals exhibited an amplitude similar to that of controls but lacked sensitivity to inhibition by NO. Consistently, NO inhibited TG-induced Mn(2+) entry in controls but not in T3-9 cells. Moreover, CCE-mediated Mn(2+) entry into T3-9 cells exhibited a striking sensitivity to inhibition by extracellular Ca(2+), which was not detectable in controls. Suppression of mitochondrial Ca(2+) handling with the uncouplers carbonyl cyanide m-chlorophenyl hydrazone (300 nm) or antimycin A(1) (-AA(1)) mimicked the inhibitory effect of NO on CCE in controls but barely affected CCE in T3-9 cells. T3-9 cells exhibited enhanced carbachol-stimulated Ca(2+) entry and clearly detectable cation currents through Trp3 cation channels. NO as well as carbonyl cyanide m-chlorophenyl hydrazone slightly promoted carbachol-induced Ca(2+) entry into T3-9 cells. Simultaneous measurement of cytoplasmic Ca(2+) and membrane currents revealed that Trp3 cation currents are inhibited during Ca(2+) entry-induced elevation of cytoplasmic Ca(2+), and that this negative feedback regulation is blunted by NO. Our results demonstrate that overexpression of Trp3 generates phospholipase C-regulated cation channels, which exhibit regulatory properties different from those of endogenous CCE channels. Moreover, we show for the first time that Trp3 expression determines biophysical properties as well as regulation of CCE channels by NO and mitochondrial Ca(2+) handling. Thus, we propose Trp3 as a subunit of CCE channels.


Subject(s)
Anthranilate Synthase/metabolism , Antimycin A/analogs & derivatives , Calcium Channels/metabolism , Calcium/metabolism , Indole-3-Glycerol-Phosphate Synthase/metabolism , Mitochondria/metabolism , Multienzyme Complexes/metabolism , Nitric Oxide/metabolism , Saccharomyces cerevisiae Proteins , Antimycin A/pharmacology , Calcium Channels/chemistry , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Cell Line , Diethylamines/pharmacology , Humans , Mitochondria/drug effects , Nitrogen Oxides , Thapsigargin/pharmacology
11.
Biophys J ; 81(5): 2639-46, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11606277

ABSTRACT

L-type Ca(2+) channels are an important means by which a cell regulates the Ca(2+) influx into the cytosol on electrical stimulation. Their structure and dynamics in the plasma membrane, including their molecular mobility and aggregation, is of key interest for the in-depth understanding of their function. Construction of a fluorescent variant by fusion of the yellow-fluorescent protein to the ion channel and expression in a human cell line allowed us to address its dynamic embedding in the membrane at the level of individual channels in vivo. We report on the observation of individual fluorescence-labeled human cardiac L-type Ca(2+) channels using wide-field fluorescence microscopy in living cells. Our fluorescence and electrophysiological data indicate that L-type Ca(2+) channels tend to form larger aggregates which are mobile in the plasma membrane.


Subject(s)
Bacterial Proteins/chemistry , Calcium Channels, L-Type/analysis , Diagnostic Imaging/methods , Luminescent Proteins/chemistry , Recombinant Fusion Proteins/analysis , Bacterial Proteins/genetics , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cell Line/cytology , Cell Membrane/metabolism , Electrophysiology/methods , Humans , Kidney/cytology , Luminescent Proteins/genetics , Microscopy, Fluorescence/methods , Movement/physiology , Myocardium/cytology , Protein Binding/physiology
12.
Crit Rev Oncol Hematol ; 39(3): 235-45, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11500265

ABSTRACT

Since cancer incidence tends to increase with age, health professionals will encounter ever-greater numbers of older people with cancer. Elderly cancer patients present complex problems that need comprehensive physical and psychosocial support. In order to give specialised care to this segment of the population, a multidisciplinary approach must be used; only in this way can an individualised treatment program be provided. Oncology nurses are an important component of this team and can contribute significantly to the panorama of needs of this segment of the population, which include the prevention and early detection of cancer, the use of state-of-the-art treatments, patient education, care during and after hospitalisation and quality of life (QOL) issues. In this way, the older person with cancer can be treated in an optimal manner and survival can hopefully be improved in a meaningful way.


Subject(s)
Neoplasms/nursing , Aged , Aged, 80 and over , Geriatric Assessment , Geriatric Nursing , Humans , Neoplasms/complications , Neoplasms/diagnosis , Neoplasms/therapy , Oncology Nursing
13.
J Biol Chem ; 276(18): 14797-803, 2001 May 04.
Article in English | MEDLINE | ID: mdl-11278396

ABSTRACT

Modulation of smooth muscle, L-type Ca(2+) channels (class C, Ca(V)1.2b) by thionitrite S-nitrosoglutathione (GSNO) was investigated in the human embryonic kidney 293 expression system at the level of whole-cell and single-channel currents. Extracellular administration of GSNO (2 mM) rapidly reduced whole-cell Ba(2+) currents through channels derived either by expression of alpha1C-b or by coexpression of alpha1C-b plus beta2a and alpha2-delta. The non-thiol nitric oxide (NO) donors 2,2-diethyl-1-nitroso-oxhydrazin (2 mM) and 3-morpholinosydnonimine-hydrochloride (2 mM), which elevated cellular cGMP levels to a similar extent as GSNO, failed to affect Ba(2+) currents significantly. Intracellular administration of copper ions, which promote decomposition of the thionitrite, antagonized its inhibitory effect, and loading of cells with high concentrations of dithiothreitol (2 mM) prevented the effect of GSNO on alpha1C-b channels. Intracellular loading of cells with oxidized glutathione (2 mM) affected neither alpha1C-b channel function nor their modulation by GSNO. Analysis of single-channel behavior revealed that GSNO inhibited Ca(2+) channels mainly by reducing open probability. The development of GSNO-induced inhibition was associated with the transient occurrence of a reduced conductance state of the channel. Our results demonstrate that GSNO modulates the alpha1 subunit of smooth muscle L-type Ca(2+) channels by an intracellular mechanism that is independent of NO release and stimulation of guanylyl cyclase. We suggest S-nitrosation of intracellularly located sulfhydryl groups as an important determinant of Ca(2+) channel gating and conductance.


Subject(s)
Calcium Channels, L-Type/physiology , Ion Channel Gating , Nitroso Compounds/chemistry , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/chemistry , Cell Line , Cyclic GMP/metabolism , Humans
14.
J Physiol ; 529 Pt 1: 119-30, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11080256

ABSTRACT

1. The role of the sequence 1572-1651 in the C-terminal tail of the alpha1C subunit in run-down of Ca2+ channels was studied by comparing functional properties of the conventional alpha1C,77 channel with those of three isoforms carrying alterations in this motif. 2. The pore-forming alpha1C subunits were co-expressed with alpha2delta and beta2a subunits in HEK-tsA201 cells, a subclone of the human embryonic kidney cell line, and studied by whole-cell and single-channel patch-clamp techniques. 3. Replacement of amino acids 1572-1651 in alpha1C,77 with 81 different amino acids leading to alpha1C,86 significantly altered run-down behaviour. Run-down of Ba2+ currents was rapid with alpha1C,77 channels, but was slow with alpha1C,86. 4. Transfer of the alpha1C,86 segments L (amino acids 1572-1598) or K (amino acids 1595-1652) into the alpha1C,77 channel yielded alpha1C,77L and alpha1C,77K channels, respectively, the run-down of which resembled more that of alpha1C,77. These results demonstrate that a large stretch of sequence between residues 1572 and 1652 of alpha1C,86 renders Ca2+ channels markedly resistant to run-down. 5. The protease inhibitor calpastatin added together with ATP was able to reverse the run-down of alpha1C,77 channels. Calpastatin expression was demonstrated in the HEK-tsA cells by Western blot analysis. 6. These results indicate a significant role of the C-terminal sequence 1572-1651 of the alpha1C subunit in run-down of L-type Ca2+ channels and suggest this sequence as a target site for a modulatory effect by endogenous calpastatin.


Subject(s)
Calcium Channels, L-Type/metabolism , Amino Acid Sequence , Amino Acid Substitution/physiology , Barium/metabolism , Blotting, Western , Calcium Channels, L-Type/genetics , Calcium-Binding Proteins/pharmacology , Calcium-Binding Proteins/physiology , Calpain/antagonists & inhibitors , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , Electrophysiology , Green Fluorescent Proteins , Humans , Isomerism , Kidney/metabolism , Luminescent Proteins , Molecular Sequence Data , Patch-Clamp Techniques , Plasmids/genetics
15.
FEBS Lett ; 477(3): 161-9, 2000 Jul 21.
Article in English | MEDLINE | ID: mdl-10908714

ABSTRACT

The role of the 80-amino acid motif 1572-1651 in the C-terminal tail of alpha(1C) Ca(2+) channel subunits was studied by comparing properties of the conventional alpha(1C,77) channel expressed in HEK-tsA201 cells to three isoforms carrying alterations in this motif. Replacement of amino acids 1572-1651 in alpha(1C,77) with 81 non-identical residues leading to alpha(1C,86) impaired membrane targeting and cluster formation of the channel. Similar to alpha(1C, 86), substitution of its 1572-1598 (alpha(1C,77L)) or 1595-1652 (alpha(1C,77K)) segments into the alpha(1C,77) channel yielded single-channel Ba(2+) currents with increased inactivation, reduced open probability and unitary conductance, when compared to the alpha(1C,77) channel. Thus, the C-terminal sequence 1572-1651 of the alpha(1C) subunit is important for membrane targeting, permeation and open probability of L-type Ca(2+) channels.


Subject(s)
Calcium Channels, L-Type/physiology , Protein Isoforms/physiology , Amino Acid Sequence , Calcium Channels, L-Type/chemistry , Cell Line , Humans , Ion Channel Gating , Membrane Potentials , Molecular Sequence Data , Probability , Protein Isoforms/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Subcellular Fractions/metabolism
16.
J Biol Chem ; 275(36): 27799-805, 2000 Sep 08.
Article in English | MEDLINE | ID: mdl-10882720

ABSTRACT

To analyze the functional consequences of coassembly of transient receptor potential 1 (Trp1) and Trp3 channel proteins, we characterized membrane conductances and divalent cation entry derived by separate overexpression and by coexpression of both Trp isoforms. Trp1 expression generated a 1-oleoyl-2-acetyl-sn-glycerol (OAG)-activated conductance that was detectable only in Ca(2+)-free extracellular solution. Trp3 expression gave rise to an OAG-activated conductance that was suppressed but clearly detectable at physiological Ca(2+) concentrations. Coexpression of both species resulted in a constitutively active, OAG-sensitive conductance, which exhibited distinctive cation selectivity and high sensitivity to inhibition by intracellular Ca(2+). Trp1-expressing cells displayed only modest carbachol-induced Ca(2+) entry and lacked OAG-induced Sr(2+) entry, whereas Trp3-expressing cells responded to both agents with a substantial divalent cation entry. Coexpression of Trp1 plus Trp3 suppressed carbachol-induced Ca(2+) entry compared with Trp3 expression and abolished OAG-induced Sr(2+) entry signals. We concluded that coassembly of Trp1 and Trp3 resulted in the formation of oligomeric Trp channels that are subject to regulation by phospholipase C and Ca(2+). The distinguished Ca(2+) sensitivity of these Trp1/Trp3 hetero-oligomers appeared to limit Trp-mediated Ca(2+) signals and may be of importance for negative feedback control of Trp function in mammalian cells.


Subject(s)
Calcium Channels/physiology , Calcium/physiology , Diglycerides/pharmacology , Fungal Proteins/physiology , Animals , Calcium/pharmacology , Calcium Channels/genetics , Cell Line , Diglycerides/physiology , Drosophila , Egtazic Acid/pharmacology , Fungal Proteins/genetics , Humans , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Recombinant Proteins/metabolism , Signal Transduction , Strontium/pharmacology , TRPC Cation Channels , Transfection
17.
Biochem J ; 348 Pt 3: 657-65, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10839999

ABSTRACT

Modulation of the smooth-muscle Ca(2+) channel alpha1C-b subunit by the auxiliary beta2a subunit was studied in the HEK 293 (cell line from human embryonic kidney cells) expression system. In addition, we tested whether the alpha1-beta interaction in functional channels is sensitive to an 18-amino-acid synthetic peptide that corresponds to the sequence of the defined major interaction domain in the cytoplasmic I-II linker of alpha1C (AID-peptide). Ca(2+) channels derived by co-expression of alpha1C-b and beta2a subunits exhibited an about 3-fold higher open probability (P(o)) than alpha1C-b channels. High-P(o) gating of alpha1C-b.beta2a channels was associated with the occurrence of long-lasting channel openings [mean open time (tau)>10 ms] which were rarely observed in alpha1C-b channels. Modulation of fast gating by the beta2a subunit persisted in the cell-free, inside-out recording configuration. Biochemical experiments showed that the AID-peptide binds with appreciable affinity to beta2 subunits of native Ca(2+) channels. Binding of the beta2 protein to immobilized AID-peptide was specifically inhibited (K(i) of 100 nM) by preincubation with free (uncoupled) AID-peptide, but not by a corresponding scrambled peptide. Administration of the AID-peptide (10 microM) to the cytoplasmic side of inside-out patches induced a substantial reduction of P(o) of alpha1C-b.beta2a channels. The scrambled control peptide failed to affect alpha1C-b. beta2a channels, and the AID-peptide (10 microM) did not modify alpha1C-b channel function in the absence of expressed beta2a subunit. Our results demonstrate that the beta2a subunit controls fast gating of alpha1C-b channels, and suggest the alpha1-beta interaction domain in the cytoplasmic I-II linker of alpha1C (AID) as a possible target of modulation of the channel. Moreover, our data are consistent with a model of alpha1-beta interaction that is based on multiple interaction sites, including AID as a determinant of the affinity of the alpha1-beta interaction.


Subject(s)
Calcium Channels, L-Type/metabolism , Ion Channel Gating , Muscle, Smooth/metabolism , Peptide Fragments/metabolism , Animals , Calcium Channels, L-Type/chemistry , Cell Line , Cytoplasm/metabolism , Humans , Protein Binding , Rats , Rats, Wistar
18.
FEBS Lett ; 471(2-3): 256-60, 2000 Apr 14.
Article in English | MEDLINE | ID: mdl-10767434

ABSTRACT

Ca(2+) channel properties of the mink ryanodine receptor type 3 (RyR3), expressed in HEK293 cells, were studied in planar lipid bilayers to which RyR3 rich membrane fragments had been fused. RyR3 channels were not active at resting levels of Ca(2+)(free) but were gated by an additional 1 mM ATP, exhibiting long open times. The second major finding was the absence of channel inactivation at millimolar Ca(2+)(free). Insertion of a myc tag at the N-terminus of RyR3 did not affect the channel properties. As to skeletal muscle, the observed type 3 channel properties appear physiologically meaningful by assisting type 1 channels in calcium release.


Subject(s)
Adenosine Triphosphate/pharmacology , Calcium/metabolism , Ion Channel Gating/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcium/pharmacology , Cell Line , Electric Conductivity , Humans , Kinetics , Lipid Bilayers/metabolism , Microsomes/drug effects , Microsomes/metabolism , Mink , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Transfection
19.
FEBS Lett ; 487(2): 301-6, 2000 Dec 29.
Article in English | MEDLINE | ID: mdl-11150529

ABSTRACT

Ca(2+)-induced inactivation of L-type Ca(2+) is differentially mediated by two C-terminal motifs of the alpha(1C) subunit, L (1572-1587) and K (1599-1651) implicated for calmodulin binding. We found that motif L is composed of a highly selective Ca(2+) sensor and an adjacent Ca(2+)-independent tethering site for calmodulin. The Ca(2+) sensor contributes to higher Ca(2+) sensitivity of the motif L complex with calmodulin. Since only combined mutation of both sites removes Ca(2+)-dependent current decay, the two-site modulation by Ca(2+) and calmodulin may underlie Ca(2+)-induced inactivation of the channel.


Subject(s)
Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/physiology , Calcium/metabolism , Calmodulin/metabolism , Amino Acid Sequence , Animals , Binding Sites , Calcium Channels, L-Type/genetics , Cell Line , Glutathione Transferase/genetics , Kinetics , Membrane Potentials/physiology , Molecular Sequence Data , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Transfection
20.
FEBS Lett ; 437(1-2): 101-6, 1998 Oct 16.
Article in English | MEDLINE | ID: mdl-9804180

ABSTRACT

Members of the Trp protein family have been suggested as the structural basis of store-operated cation conductances. With this study, we provide evidence for the expression of three isoforms of Trp (hTrp1, 3 and 4) in human umbilical vein endothelial cells (HUVEC). The role of Trp proteins in store regulation of endothelial membrane conductances was tested by expression of an N-terminal fragment of hTrp3 (N-TRP) which exerts a dominant negative effect on Trp channel function presumably due to suppression of channel assembly. Depletion of intracellular Ca2+ stores with IP3 (100 microM) or thapsigargin (100 nM) induced a substantial cation conductance in sham-transfected HUVEC as well as in HUVEC transfected with hTrp3. In contrast, HUVEC transfected with N-TRP failed to exhibit store-operated currents. Our results suggest the involvement of Trp related proteins in the store-operated cation conductance of human vascular endothelial cells.


Subject(s)
Calcium Channels/metabolism , Endothelium, Vascular/metabolism , Calcium Channels/genetics , Cells, Cultured , Humans , Patch-Clamp Techniques , TRPC Cation Channels , Thapsigargin/pharmacology , Transfection , Umbilical Veins
SELECTION OF CITATIONS
SEARCH DETAIL
...