Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.114
Filter
1.
Life Sci ; 308: 120913, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36037871

ABSTRACT

AIMS: Lung type 2 alveolar cells, by secreting surfactant to lower surface tension, contribute to enhance lung compliance. Stretching, as a result of lung expansion, triggers type 1 alveolar cell to release ATP, which in turn stimulates Ca2+-dependent surfactant secretion by neighboring type 2 cells. In this report, we studied ATP-triggered Ca2+ signaling in human alveolar type 2 A549 cells. MAIN METHODS: Ca2+ signaling was examined using microfluorimetric measurement with fura-2 as fluorescent dye. KEY FINDINGS: Ca2+ oscillations triggered by ATP relied on inositol 1,4,5-trisphosphate-induced Ca2+ release and store-operated Ca2+ entry. Pathological conditions such as influenza virus infection and diabetes reportedly inhibit sarcoplasmic/endoplasmic reticulum Ca2+ ATPase (SERCA). We found that a very mild inhibition of SERCA by cyclopiazonic acid (CPA) sufficed to decrease Ca2+ oscillation frequency and the percentage of cells exhibiting Ca2+ oscillations. Ochratoxin A (OTA), an activator of SERCA, could prevent the suppressive effects by CPA. Inhibition of SERCA by hydrogen peroxide also suppressed Ca2+ oscillations. Interestingly, hydrogen peroxide-induced inhibition was prevented by OTA but aggravated by CDN1163, an allosteric activator of SERCA. CDN1163 also had an untoward effect of releasing intracellular Ca2+. SIGNIFICANCE: Different modes of activation of SERCA may determine the outcome of rescue of Ca2+ oscillations in case of SERCA inhibition in alveolar type 2 cells.


Subject(s)
Alveolar Epithelial Cells , Diabetes Mellitus, Type 2 , A549 Cells , Adenosine Triphosphate/metabolism , Alveolar Epithelial Cells/metabolism , Aminoquinolines , Benzamides , Calcium/metabolism , Calcium Signaling/physiology , Fluorescent Dyes , Fura-2/pharmacology , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Inositol 1,4,5-Trisphosphate/pharmacology , Ochratoxins , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Surface-Active Agents
2.
Cell Rep ; 37(5): 109932, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731613

ABSTRACT

Inositol 1,4,5-trisphosphate receptors (IP3Rs) are intracellular Ca2+ channels that link extracellular stimuli to Ca2+ signals. Ca2+ release from intracellular stores is "quantal": low IP3 concentrations rapidly release a fraction of the stores. Ca2+ release then slows or terminates without compromising responses to further IP3 additions. The mechanisms are unresolved. Here, we synthesize a high-affinity partial agonist of IP3Rs and use it to demonstrate that quantal responses do not require heterogenous Ca2+ stores. IP3Rs respond incrementally to IP3 and close after the initial response to low IP3 concentrations. Comparing functional responses with IP3 binding shows that only a tiny fraction of a cell's IP3Rs mediate incremental Ca2+ release; inactivation does not therefore affect most IP3Rs. We conclude, and test by simulations, that Ca2+ signals evoked by IP3 pulses arise from rapid activation and then inactivation of very few IP3Rs. This allows IP3Rs to behave as increment detectors mediating graded Ca2+ release.


Subject(s)
Calcium Signaling/drug effects , Calcium/metabolism , Endoplasmic Reticulum/drug effects , Inositol 1,4,5-Trisphosphate Receptors/agonists , Inositol 1,4,5-Trisphosphate/pharmacology , Animals , Chickens , Drug Partial Agonism , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol Phosphates/pharmacology , Time Factors
3.
Cell Calcium ; 92: 102284, 2020 12.
Article in English | MEDLINE | ID: mdl-32947181

ABSTRACT

In contrast to animal cells, the inositol 1,4,5-trisphosphate receptor of Trypanosoma cruzi (TcIP3R) localizes to acidocalcisomes instead of the endoplasmic reticulum. Here, we present evidence that TcIP3R is a Ca2+ release channel gated by IP3 when expressed in DT40 cells knockout for all vertebrate IP3 receptors, and is required for Ca2+ uptake by T. cruzi mitochondria, regulating pyruvate dehydrogenase dephosphorylation and mitochondrial O2 consumption, and preventing autophagy. Localization studies revealed its co-localization with an acidocalcisome marker in all life cycle stages of the parasite. Ablation of TcIP3R by CRISPR/Cas9 genome editing caused: a) a reduction in O2 consumption rate and citrate synthase activity; b) decreased mitochondrial Ca2+ transport without affecting the membrane potential; c) increased ammonia production and AMP/ATP ratio; d) stimulation of autophagosome formation, and e) marked defects in growth of culture forms (epimastigotes) and invasion of host cells by infective stages (trypomastigotes). Moreover, TcIP3R overexpressing parasites showed decreased metacyclogenesis, trypomastigote host cell invasion and intracellular amastigote replication. In conclusion, the results suggest a modulatory activity of TcIP3R-mediated acidocalcisome Ca2+ release on cell bioenergetics in T. cruzi.


Subject(s)
Autophagy , Calcium/metabolism , Energy Metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mitochondria/metabolism , Trypanosoma cruzi/metabolism , Animals , Autophagy/drug effects , Chickens , Chlorocebus aethiops , Energy Metabolism/drug effects , Inositol 1,4,5-Trisphosphate/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Inositol 1,4,5-Trisphosphate Receptors/genetics , Life Cycle Stages/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mutation/genetics , Phenotype , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/growth & development , Vero Cells
4.
Cell Calcium ; 89: 102224, 2020 07.
Article in English | MEDLINE | ID: mdl-32502904

ABSTRACT

Recent advances in imaging technology and fluorescent probes have made it possible to gain information about the dynamics of subcellular processes at unprecedented spatiotemporal scales. Unfortunately, a lack of automated tools to efficiently process the resulting imaging data encoding fine details of the biological processes remains a major bottleneck in utilizing the full potential of these powerful experimental techniques. Here we present a computational tool, called PunctaSpecks, that can characterize fluorescence signals arising from a wide range of biological molecules under normal and pathological conditions. Among other things, the program can calculate the number, areas, life-times, and amplitudes of fluorescence signals arising from multiple sources, track diffusing fluorescence sources like moving mitochondria, and determine the overlap probability of two processes or organelles imaged using indicator dyes of different colors. We have tested PunctaSpecks on synthetic time-lapse movies containing mobile fluorescence objects of various sizes, mimicking the activity of biomolecules. The robustness of the software is tested by varying the level of noise along with random but known pattern of appearing, disappearing, and movement of these objects. Next, we use PunctaSpecks to characterize protein-protein interaction involved in store-operated Ca2+ entry through the formation and activation of plasma membrane-bound ORAI1 channel and endoplasmic reticulum membrane-bound stromal interaction molecule (STIM), the evolution of inositol 1,4,5-trisphosphate (IP3)-induced Ca2+ signals from sub-micrometer size local events into global waves in human cortical neurons, and the activity of Alzheimer's disease-associated ß amyloid pores in the plasma membrane. The tool can also be used to study other dynamical processes imaged through fluorescence molecules. The open source algorithm allows for extending the program to analyze more than two types of biomolecules visualized using markers of different colors.


Subject(s)
Fluorescent Dyes/chemistry , Software , Algorithms , Amyloid beta-Peptides/metabolism , Automation , Calcium/metabolism , Calcium Signaling/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cerebral Cortex/cytology , Diffusion , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate/pharmacology , Kinetics , Neurons/drug effects , Neurons/metabolism , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/metabolism
5.
J Med Chem ; 63(10): 5442-5457, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32286062

ABSTRACT

Chiral sugar derivatives are potential cyclitol surrogates of the Ca2+-mobilizing intracellular messenger d-myo-inositol 1,4,5-trisphosphate [Ins(1,4,5)P3]. Six novel polyphosphorylated analogues derived from both d- and l-glucose were synthesized. Binding to Ins(1,4,5)P3 receptors [Ins(1,4,5)P3R] and the ability to release Ca2+ from intracellular stores via type 1 Ins(1,4,5)P3Rs were investigated. ß-d-Glucopyranosyl 1,3,4-tris-phosphate, with similar phosphate regiochemistry and stereochemistry to Ins(1,4,5)P3, and α-d-glucopyranosyl 1,3,4-tris-phosphate are full agonists, being equipotent and 23-fold less potent than Ins(1,4,5)P3, respectively, in Ca2+-release assays and similar to Ins(1,4,5)P3 and 15-fold weaker in binding assays. They can be viewed as truncated analogues of adenophostin A and refine understanding of structure-activity relationships for this Ins(1,4,5)P3R agonist. l-Glucose-derived ligands, methyl α-l-glucopyranoside 2,3,6-trisphosphate and methyl α-l-glucopyranoside 2,4,6-trisphosphate, are also active, while their corresponding d-enantiomers, methyl α-d-glucopyranoside 2,3,6-trisphosphate and methyl α-d-glucopyranoside 2,4,6-trisphosphate, are inactive. Interestingly, both l-glucose-derived ligands are partial agonists: they are among the least efficacious agonists of Ins(1,4,5)P3R yet identified, providing new leads for antagonist development.


Subject(s)
Drug Partial Agonism , Glucose/chemistry , Inositol 1,4,5-Trisphosphate Receptors/agonists , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Inositol 1,4,5-Trisphosphate/chemistry , Molecular Mimicry/drug effects , Polyphosphates/chemistry , Animals , Dose-Response Relationship, Drug , Glucose/pharmacology , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate/pharmacology , Molecular Docking Simulation/methods , Molecular Mimicry/physiology , Polyphosphates/pharmacology , Protein Structure, Secondary , Rats , Rats, Wistar
6.
Sci Rep ; 9(1): 3487, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30837563

ABSTRACT

Schwann cells (SCs) constitute a crucial element of the peripheral nervous system, by structurally supporting the formation of myelin and conveying vital trophic factors to the nervous system. However, the functions of SCs in developmental and regenerative stages remain unclear. Here, we investigated how optogenetic stimulation (OS) of SCs regulates their development. In SC monoculture, OS substantially enhanced SC proliferation and the number of BrdU+-S100ß+-SCs over time. In addition, OS also markedly promoted the expression of both Krox20 and myelin basic protein (MBP) in SC culture medium containing dBcAMP/NRG1, which induced differentiation. We found that the effects of OS are dependent on the intracellular Ca2+ level. OS induces elevated intracellular Ca2+ levels through the T-type voltage-gated calcium channel (VGCC) and mobilization of Ca2+ from both inositol 1,4,5-trisphosphate (IP3)-sensitive stores and caffeine/ryanodine-sensitive stores. Furthermore, we confirmed that OS significantly increased expression levels of both Krox20 and MBP in SC-motor neuron (MN) coculture, which was notably prevented by pharmacological intervention with Ca2+. Taken together, our results demonstrate that OS of SCs increases the intracellular Ca2+ level and can regulate proliferation, differentiation, and myelination, suggesting that OS of SCs may offer a new approach to the treatment of neurodegenerative disorders.


Subject(s)
Cell Differentiation , Cell Proliferation , Light , Myelin Basic Protein/metabolism , Animals , Calcium/metabolism , Calcium Channels, T-Type/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Culture Media/chemistry , Culture Media/pharmacology , Early Growth Response Protein 2/metabolism , Inositol 1,4,5-Trisphosphate/pharmacology , Mice , Motor Neurons/cytology , Motor Neurons/metabolism , Optogenetics , Schwann Cells/cytology , Schwann Cells/metabolism
7.
Mol Cell Endocrinol ; 473: 17-30, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29275169

ABSTRACT

Asthma symptoms have been associated with sex steroids. During childhood, this illness seems more frequent in boys than in girls and this tendency reverts in puberty when it is more severe in women. Testosterone (TES), at supraphysiological concentrations, relaxed pre-contracted airway smooth muscle, but its effects at physiological concentrations have not been thoroughly studied. We explored this possibility in guinea pig tracheal smooth muscle. In myocytes TES (10 nM) abolished carbachol (CCh)-induced intracellular Ca2+ concentration ([Ca2+]i) increment. Ca2+ responses to ATP were partially modified by TES while histamine's were not. These results indicate that inositol 1,4,5-trisphosphate (IP3) signaling pathway might be involved. Photolysis of caged-IP3 increased [Ca2+]i and TES abolished this effect. TES diminished reactivity of the smooth muscle to CCh and this effect was non-genomic since it was unchanged by flutamide. In tracheal smooth muscle, mRNA for each IP3 receptor (ITPR) isoform was found and, by immunofluorescence, ITPR1 and ITPR3 seems to be the main isoforms observed while ITPR2 was less prominent. Comparing the amino acid sequence of ITPR1 and the sequence of the TES binding site on the androgen receptor, we found that they share a short sequence. This domain could be responsible for the TES binding to the ITPR1 and probably for its blocking effect. We conclude that TES modifies ITPR1 function in airway smooth muscle, turning this tissue less reactive to contractile agonists that act through PLCß-IP3 signaling cascade. These results might be related to the low asthma prevalence in males from puberty to adulthood.


Subject(s)
Inositol 1,4,5-Trisphosphate Receptors/metabolism , Muscle, Smooth/physiology , Testosterone/pharmacology , Trachea/physiology , Amino Acid Sequence , Animals , Calcium/metabolism , Calcium Channels/metabolism , Carbachol/pharmacology , Genome , Guinea Pigs , Histamine/pharmacology , Humans , Inositol 1,4,5-Trisphosphate/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Intracellular Space/metabolism , Male , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Protein Isoforms/metabolism , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Trachea/drug effects
8.
J Neurosci ; 37(8): 2032-2044, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28115480

ABSTRACT

Hyperalgesic priming, a model of pain chronification in the rat, is mediated by ryanodine receptor-dependent calcium release. Although ryanodine induces priming in both sexes, females are 5 orders of magnitude more sensitive, by an estrogen receptor α (EsRα)-dependent mechanism. An inositol 1,4,5-triphosphate (IP3) receptor inhibitor prevented the induction of priming by ryanodine. For IP3 induced priming, females were also more sensitive. IP3-induced priming was prevented by pretreatment with inhibitors of the sarcoendoplasmic reticulum calcium ATPase and ryanodine receptor. Antisense to EsRα prevented the induction of priming by low-dose IP3 in females. The induction of priming by an EsRα agonist was ryanodine receptor-dependent and prevented by the IP3 antagonist. Thus, an EsRα-dependent bidirectional interaction between endoplasmic reticulum IP3 and ryanodine receptor-mediated calcium signaling is present in the induction of hyperalgesic priming, in females. In cultured male DRG neurons, IP3 (100 µm) potentiated depolarization-induced transients produced by extracellular application of high-potassium solution (20 mm, K20), in nociceptors incubated with ß-estradiol. This potentiation of depolarization-induced calcium transients was blocked by the IP3 antagonist, and not observed in the absence of IP3 IP3 potentiation was also blocked by ryanodine receptor antagonist. The application of ryanodine (2 nm), instead of IP3, also potentiated K20-induced calcium transients in the presence of ß-estradiol, in an IP3 receptor-dependent manner. Our results point to an EsRα-dependent, reciprocal interaction between IP3 and ryanodine receptors that contributes to sex differences in hyperalgesic priming.SIGNIFICANCE STATEMENT The present study demonstrates a mechanism that plays a role in the marked sexual dimorphism observed in a model of the transition to chronic pain, hyperalgesic priming. This mechanism involves a reciprocal interaction between the endoplasmic reticulum receptors, IP3 and ryanodine, in the induction of priming, regulated by estrogen receptor α in the nociceptor of female rats. The presence of this signaling pathway modulating the susceptibility of nociceptors to develop plasticity may contribute to our understanding of sex differences observed clinically in chronic pain syndromes.


Subject(s)
Hyperalgesia/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Pain Threshold/physiology , Ryanodine Receptor Calcium Release Channel/metabolism , Sex Characteristics , Animals , Cells, Cultured , Dinoprostone/adverse effects , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Ganglia, Spinal/cytology , Hyperalgesia/chemically induced , Inositol 1,4,5-Trisphosphate/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Macrocyclic Compounds/pharmacology , Male , Oligodeoxyribonucleotides, Antisense/pharmacology , Oxazoles/pharmacology , Pain Measurement , Pain Threshold/drug effects , Rats , Rats, Sprague-Dawley , Ryanodine/adverse effects , Sensory Receptor Cells/drug effects , Thapsigargin/pharmacology
10.
Proc Natl Acad Sci U S A ; 113(23): E3280-9, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27217559

ABSTRACT

An important consequence of gliotransmission, a signaling mechanism that involves glial release of active transmitter molecules, is its manifestation as N-methyl-d-aspartate receptor (NMDAR)-dependent slow inward currents in neurons. However, the intraneuronal spatial dynamics of these events or the role of active dendrites in regulating their amplitude and spatial spread have remained unexplored. Here, we used somatic and/or dendritic recordings from rat hippocampal pyramidal neurons and demonstrate that a majority of NMDAR-dependent spontaneous slow excitatory potentials (SEP) originate at dendritic locations and are significantly attenuated through their propagation across the neuronal arbor. We substantiated the astrocytic origin of SEPs through paired neuron-astrocyte recordings, where we found that specific infusion of inositol trisphosphate (InsP3) into either distal or proximal astrocytes enhanced the amplitude and frequency of neuronal SEPs. Importantly, SEPs recorded after InsP3 infusion into distal astrocytes exhibited significantly slower kinetics compared with those recorded after proximal infusion. Furthermore, using neuron-specific infusion of pharmacological agents and morphologically realistic conductance-based computational models, we demonstrate that dendritically expressed hyperpolarization-activated cyclic-nucleotide-gated (HCN) and transient potassium channels play critical roles in regulating the strength, kinetics, and compartmentalization of neuronal SEPs. Finally, through the application of subtype-specific receptor blockers during paired neuron-astrocyte recordings, we provide evidence that GluN2B- and GluN2D-containing NMDARs predominantly mediate perisomatic and dendritic SEPs, respectively. Our results unveil an important role for active dendrites in regulating the impact of gliotransmission on neurons and suggest astrocytes as a source of dendritic plateau potentials that have been implicated in localized plasticity and place cell formation.


Subject(s)
Dendrites/physiology , Hippocampus/physiology , Pyramidal Cells/physiology , Animals , Astrocytes/drug effects , Astrocytes/physiology , Cell Communication/physiology , Computer Simulation , Hippocampus/cytology , In Vitro Techniques , Inositol 1,4,5-Trisphosphate/pharmacology , Ion Channels/antagonists & inhibitors , Ion Channels/physiology , Male , Models, Neurological , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
11.
Cell Rep ; 15(3): 563-573, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27068462

ABSTRACT

Dopamine action in the nucleus accumbens (NAc) is thought to drive appetitive behavior and Pavlovian reward learning. However, it remains controversial how dopamine achieves these behavioral effects by regulating medium spiny projection neurons (MSNs) of the NAc, especially on a behaviorally relevant timescale. Metabotropic glutamate receptor (mGluR)-induced Ca(2+) signaling dependent on the Ca(2+)- releasing messenger inositol 1,4,5-triphosphate (IP3) plays a critical role in controlling neuronal excitability and synaptic plasticity. Here, we show that transient dopamine application facilitates mGluR/IP3-induced Ca(2+) signals within a time window of ∼2-10 s in a subpopulation of MSNs in the NAc core. Dopamine facilitation of IP3-induced Ca(2+) signaling is mediated by D1 dopamine receptors. In dopamine-insensitive MSNs, activation of A2A adenosine receptors causes enhancement of IP3-evoked Ca(2+) signals, which is reversed by D2 dopamine receptor activation. These results show that dopamine differentially regulates Ca(2+) signaling on the order of seconds in two distinct MSN subpopulations.


Subject(s)
Calcium Signaling/drug effects , Dopamine/pharmacology , Nucleus Accumbens/metabolism , Action Potentials/drug effects , Animals , Calcium/metabolism , Inositol 1,4,5-Trisphosphate/pharmacology , Male , Mice, Inbred C57BL , Nucleus Accumbens/drug effects , Receptor, Adenosine A2A/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Metabotropic Glutamate/metabolism , Time Factors
12.
Cell Calcium ; 58(2): 196-207, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26077147

ABSTRACT

Cocaine promotes addictive behavior primarily by blocking the dopamine transporter, thus increasing dopamine transmission in the nucleus accumbens (nAcc); however, additional mechanisms are continually emerging. Sigma-1 receptors (σ1Rs) are known targets for cocaine, yet the mechanisms underlying σ1R-mediated effects of cocaine are incompletely understood. The present study examined direct effects of cocaine on dissociated nAcc neurons expressing phosphatidylinositol-linked D1 receptors. Endoplasmic reticulum-located σ1Rs and inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs) were targeted using intracellular microinjection. IP3 microinjection robustly elevated intracellular Ca(2+) concentration, [Ca(2+)]i. While cocaine alone was devoid of an effect, the IP3-induced response was σ1R-dependently enhanced by cocaine co-injection. Likewise, cocaine augmented the [Ca(2+)]i increase elicited by extracellularly applying an IP3-generating molecule (ATP), via σ1Rs. The cocaine-induced enhancement of the IP3/ATP-mediated Ca(2+) elevation occurred at pharmacologically relevant concentrations and was mediated by transient receptor potential canonical channels (TRPC). IP3 microinjection elicited a slight, transient depolarization, further converted to a greatly enhanced, prolonged response, by cocaine co-injection. The cocaine-triggered augmentation was σ1R-dependent, TRPC-mediated and contingent on [Ca(2+)]i elevation. ATP-induced depolarization was similarly enhanced by cocaine. Thus, we identify a novel mechanism by which cocaine promotes activation of D1-expressing nAcc neurons: enhancement of IP3R-mediated responses via σ1R activation at the endoplasmic reticulum, resulting in augmented Ca(2+) release and amplified depolarization due to subsequent stimulation of TRPC. In vivo, intra-accumbal blockade of σ1R or TRPC significantly diminished cocaine-induced hyperlocomotion and locomotor sensitization, endorsing a physio-pathological significance of the pathway identified in vitro.


Subject(s)
Cocaine/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Neurons/drug effects , Nucleus Accumbens/cytology , Receptors, sigma/metabolism , Adenosine Triphosphate/pharmacology , Animals , Behavior, Animal/drug effects , Calcium/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Endoplasmic Reticulum/metabolism , Imidazoles/pharmacology , Inositol 1,4,5-Trisphosphate/pharmacology , Locomotion/drug effects , Male , Membrane Potentials/drug effects , Neurons/cytology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , TRPC Cation Channels/metabolism , Sigma-1 Receptor
13.
Biochem Pharmacol ; 96(2): 143-50, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25986882

ABSTRACT

As ectopic expression of the neuronal inositol-1,4,5-trisphosphate-3-kinase A (InsP3Kinase) in tumor cells increases the metastatic potential, InsP3Kinase is an interesting target for tumor therapy. Recently, we have identified a membrane-permeable InsP3Kinase inhibitor (BAMB-4) exhibiting an IC50-value of 20 µM. Here we characterized a new InsP3Kinase inhibitor which shows a 130-fold lower IC50 value (157 ± 57 nM) as compared to BAMB-4. We demonstrate that this nitrophenolic compound, BIP-4, is non-competitive to ATP but competitive to InsP3, thus exhibits a high selectivity for inhibition of InsP3Kinase activity. Docking analysis suggested a putative binding mode of this molecule into the InsP3Kinase active site. Determination of cellular uptake in lung cancer cells (H1299) revealed that 6% of extracellular BIP-4 is internalized by non-endosomal uptake, showing that BIP-4 is not trapped inside endo/lysosomes but is available to inhibit cellular InsP3Kinase activity. Interestingly, we found that BIP-4 mediated inhibition of InsP3Kinase activity in the two lung cancer cell lines H1299 and LN4323 inhibited proliferation and adhesion at IC50 values of 3 µM or 2 µM, respectively. InsP3Kinase inhibition did not alter ATP-induced calcium signals but significantly reduced the level of Ins(1,3,4,5,6)P5. From these data we conclude that the inhibitory effect of BIP-4 on proliferation and adhesion of lung cancer cells does not result from alterations of calcium but from alterations of inositol phosphate signals. In summary, we reveal that inhibition of cellular InsP3Kinase by BIP-4 impairs proliferation and adhesion and therefore BIP-4 might be a promising compound to reduce the metastatic potential of lung carcinoma cells.


Subject(s)
Antineoplastic Agents/pharmacology , Inositol 1,4,5-Trisphosphate/pharmacology , Lung Neoplasms/pathology , Naphthalimides/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Pyrazoles/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Benzamides/chemistry , Benzamides/pharmacology , Benzoxazoles/chemistry , Benzoxazoles/pharmacology , Calcium Signaling , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , High-Throughput Screening Assays , Humans , Molecular Docking Simulation , Naphthalimides/chemistry , Pyrazoles/chemistry
14.
J Theor Biol ; 371: 102-16, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25698230

ABSTRACT

This paper explores the influence of burst properties of the sympathetic nervous system on arterial contractility. Specifically, a mathematical model is constructed of the pathway from action potential generation in a sympathetic postganglionic neurone to contraction of an arterial smooth muscle cell. The differential equation model is a synthesis of models of the individual physiological processes, and is shown to be consistent with physiological data. The model is found to be unresponsive to tonic (regular) stimulation at typical frequencies recorded in sympathetic efferents. However, when stimulated at the same average frequency, but with repetitive respiratory-modulated burst patterns, it produces marked contractions. Moreover, the contractile force produced is found to be highly dependent on the number of spikes in each burst. In particular, when the model is driven by preganglionic spike trains recorded from wild-type and spontaneously hypertensive rats (which have increased spiking during each burst) the contractile force was found to be 10-fold greater in the hypertensive case. An explanation is provided in terms of the summative increased release of noradrenaline. Furthermore, the results suggest the marked effect that hypertensive spike trains had on smooth muscle cell tone can provide a significant contribution to the pathology of hypertension.


Subject(s)
Models, Cardiovascular , Neurons/physiology , Sympathetic Nervous System/physiology , Animals , Calcium/pharmacology , GTP-Binding Proteins/metabolism , Inositol 1,4,5-Trisphosphate/pharmacology , Muscle Contraction/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Neurons/drug effects , Norepinephrine/pharmacology , Rats, Inbred SHR , Reproducibility of Results , Signal Transduction/drug effects , Sympathetic Nervous System/drug effects
15.
J Biol Chem ; 290(15): 9823-41, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25713146

ABSTRACT

Regulated secretion is a central issue for the specific function of many cells; for instance, mammalian sperm acrosomal exocytosis is essential for egg fertilization. ARF6 (ADP-ribosylation factor 6) is a small GTPase implicated in exocytosis, but its downstream effectors remain elusive in this process. We combined biochemical, functional, and microscopy-based methods to show that ARF6 is present in human sperm, localizes to the acrosomal region, and is required for calcium and diacylglycerol-induced exocytosis. Results from pulldown assays show that ARF6 exchanges GDP for GTP in sperm challenged with different exocytic stimuli. Myristoylated and guanosine 5'-3-O-(thio)triphosphate (GTPγS)-loaded ARF6 (active form) added to permeabilized sperm induces acrosome exocytosis even in the absence of extracellular calcium. We explore the ARF6 signaling cascade that promotes secretion. We demonstrate that ARF6 stimulates a sperm phospholipase D activity to produce phosphatidic acid and boosts the synthesis of phosphatidylinositol 4,5-bisphosphate. We present direct evidence showing that active ARF6 increases phospholipase C activity, causing phosphatidylinositol 4,5-bisphosphate hydrolysis and inositol 1,4,5-trisphosphate-dependent intra-acrosomal calcium release. We show that active ARF6 increases the exchange of GDP for GTP on Rab3A, a prerequisite for secretion. We propose that exocytic stimuli activate ARF6, which is required for acrosomal calcium efflux and the assembly of the membrane fusion machinery. This report highlights the physiological importance of ARF6 as a key factor for human sperm exocytosis and fertilization.


Subject(s)
ADP-Ribosylation Factors/metabolism , Acrosome/physiology , Exocytosis/physiology , Lipid Metabolism/physiology , rab3A GTP-Binding Protein/metabolism , ADP-Ribosylation Factor 6 , Acrosome/drug effects , Acrosome Reaction/drug effects , Acrosome Reaction/physiology , Calcium/metabolism , Cells, Cultured , Diglycerides/pharmacology , Exocytosis/drug effects , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Humans , Immunoblotting , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate/pharmacology , Male , Microscopy, Confocal , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase D/metabolism , Spermatozoa/drug effects , Spermatozoa/metabolism , Spermatozoa/physiology , Type C Phospholipases/metabolism
16.
Chembiochem ; 15(18): 2774-82, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25399672

ABSTRACT

Inositol 1,4,5-trisphosphate (IP3 ) is a universal signalling molecule that releases calcium from stores within cells by activating the IP3 receptor. Although chemical tools that modulate the IP3 receptor exist, none is ideal due to trade offs between potency, selectivity and cell permeability, and their chemical properties make them challenging starting points for optimisation. Therefore, to find new leads, we used virtual screening to scaffold hop from IP3 by using the program ROCS to perform a 3D ligand-based screen of the ZINC database of purchasable compounds. We then used the program FRED to dock the top-ranking hits into the IP3 binding pocket of the receptor. We tested the 12 highest-scoring hits in a calcium-release bioassay and identified SI-9 as a partial agonist. SI-9 competed with [(3) H]IP3 binding, and reduced histamine-induced calcium signalling in HeLa cells. SI-9 has a novel 2D scaffold that represents a tractable lead for designing improved IP3 receptor modulators.


Subject(s)
Inositol 1,4,5-Trisphosphate Receptors/agonists , Inositol 1,4,5-Trisphosphate/analogs & derivatives , Inositol 1,4,5-Trisphosphate/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Calcium/metabolism , Drug Design , HeLa Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Ligands , Molecular Docking Simulation
17.
Acta Pharmacol Sin ; 35(12): 1514-20, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25345744

ABSTRACT

AIM: Congo red, a secondary diazo dye, is usually used as an indicator for the presence of amyloid fibrils. Recent studies show that congo red exerts neuroprotective effects in a variety of models of neurodegenerative diseases. However, its pharmacological profile remains unknown. In this study, we investigated the effects of congo red on ACh-induced Ca(2+) oscillations in mouse pancreatic acinar cells in vitro. METHODS: Acutely dissociated pancreatic acinar cells of mice were prepared. A U-tube drug application system was used to deliver drugs into the bath. Intracellular Ca(2+) oscillations were monitored by whole-cell recording of Ca(2+)-activated Cl(-) currents and by using confocal Ca(2+) imaging. For intracellular drug application, the drug was added in pipette solution and diffused into cell after the whole-cell configuration was established. RESULTS: Bath application of ACh (10 nmol/L) induced typical Ca(2+) oscillations in dissociated pancreatic acinar cells. Addition of congo red (1, 10, 100 µmol/L) dose-dependently enhanced Ach-induced Ca(2+) oscillations, but congo red alone did not induce any detectable response. Furthermore, this enhancement depended on the concentrations of ACh: congo red markedly enhanced the Ca(2+) oscillations induced by ACh (10-30 nmol/L), but did not alter the Ca(2+) oscillations induced by ACh (100-10000 nmol/L). Congo red also enhanced the Ca(2+) oscillations induced by bath application of IP3 (30 µmol/L). Intracellular application of congo red failed to alter ACh-induced Ca(2+) oscillations. CONCLUSION: Congo red significantly modulates intracellular Ca(2+) signaling in pancreatic acinar cells, and this pharmacological effect should be fully considered when developing congo red as a novel therapeutic drug.


Subject(s)
Acetylcholine/pharmacology , Calcium Signaling/drug effects , Congo Red/pharmacology , Pancreas, Exocrine/drug effects , Animals , Calcium Channel Blockers/pharmacology , Dose-Response Relationship, Drug , Inositol 1,4,5-Trisphosphate/pharmacology , Male , Membrane Potentials , Mice , Pancreas, Exocrine/cytology , Pancreas, Exocrine/metabolism , Time Factors
18.
Cold Spring Harb Protoc ; 2014(9): 988-92, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25183812

ABSTRACT

NAADP and other Ca(2+)-mobilizing messengers are membrane impermeant and thus must be added directly to cell-free or broken-cell preparations to effect Ca(2+) release. The sea urchin egg homogenate, where the biological activity of NAADP was first reported, remains the gold standard cell-free system for studying NAADP-mediated Ca(2+) release. Here we describe how to prepare sea urchin egg homogenate and use it to measure NAADP-mediated Ca(2+) release.


Subject(s)
Calcium/metabolism , Cell Fractionation/methods , NADP/analogs & derivatives , Ovum/drug effects , Ovum/metabolism , Adenosine Triphosphate/pharmacology , Aniline Compounds , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Female , Inositol 1,4,5-Trisphosphate/pharmacology , Male , NADP/metabolism , NADP/pharmacology , Ovum/ultrastructure , Sea Urchins , Xanthenes
19.
Dev Biol ; 392(1): 80-92, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24809798

ABSTRACT

Jellyfish eggs neither undergo apparent cortical reaction nor show any significant change in the membrane potential at fertilization, but nevertheless show monospermy. Utilizing the perfectly transparent eggs of the hydrozoan jellyfish Cytaeis uchidae, here we show that the polyspermy block is accomplished via a novel mechanism: a collaboration between Ca(2+) and mitogen-activated protein kinase (MAPK). In Cytaeis, adhesion of a sperm to the animal pole surface of an egg was immediately followed by sperm-egg fusion and initiation of an intracellular Ca(2+) rise from this site. The elevated Ca(2+) levels lasted for several minutes following the sperm-egg fusion. The Ca(2+) rise proved to be necessary and sufficient for a polyspermy block, as inhibiting a Ca(2+) rise with EGTA promoted polyspermy, and conversely, triggering a Ca(2+) rise by inositol 1,4,5-trisphosphate (IP3) or excess K(+) immediately abolished the egg's capacity for sperm-egg fusion. A Ca(2+) rise at fertilization or by artificial stimulations evoked dephosphorylation of MAPK in eggs. The eggs in which phosphorylated MAPK was maintained by injection of mRNA for MAPK kinase kinase (Mos), like intact eggs, exhibited a Ca(2+) rise at fertilization or by IP3 injection, and shut down the subsequent sperm-egg fusion. However, the Mos-expressing eggs became capable of accepting sperm following the arrest of Ca(2+) rise. In contrast, addition of inhibitors of MAPK kinase (MEK) to unfertilized eggs caused MAPK dephosphorylation without elevating Ca(2+) levels, and prevented sperm-egg fusion. Rephosphorylation of MAPK by injecting Mos mRNA after fertilization recovered sperm attraction, which is known to be another MAPK-dependent event, but did not permit subsequent sperm-egg fusion. Thus, it is possible that MAPK dephosphorylation irreversibly blocks sperm-egg fusion and reversibly suppresses sperm attraction. Collectively, our data suggest that both the fast and late mechanisms dependent on Ca(2+) and MAPK, respectively, ensure a polyspermy block in jellyfish eggs.


Subject(s)
Calcium/metabolism , Fertilization/physiology , Hydrozoa/physiology , Mitogen-Activated Protein Kinases/metabolism , Ovum/physiology , Sperm-Ovum Interactions/drug effects , Animals , Calcium/pharmacology , Hydrozoa/metabolism , Inositol 1,4,5-Trisphosphate/pharmacology , Phosphorylation , Potassium/pharmacology , Proto-Oncogene Proteins c-mos/genetics , RNA, Messenger/genetics
20.
Hippocampus ; 24(7): 869-76, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24710998

ABSTRACT

Rise in Ca(2+) concentration in the nucleus affects gene transcription and has been implicated in neuroprotection, transcription-dependent neuronal plasticity, and pain modulation, but the mechanism of regulation of nuclear Ca(2+) remains poorly understood. The nuclear envelope is a part of the endoplasmic reticulum and may be one of the sources of nuclear Ca(2+) . Here, we studied ion channels in the nuclear membrane of hippocampal neurons using the patch-clamp technique. We have found that the nuclear membrane of CA1 pyramidal and dentate gyrus granule (DG), but not CA3 pyramidal neurons, was enriched in functional inositol 1,4,5-trisphosphate receptors/Ca(2+) -release channels (IP3 Rs) localized mainly in the inner nuclear membrane. A single nuclear ryanodine receptor (RyR) has been detected only in DG granule neurons. Nuclei of the hippocampal neurons also expressed a variety of spontaneously active cation and anion channels specific for each type of neuron. In particular, large-conductance ion channels selective for monovalent cations (LCC) were coexpressed with IP3 Rs. These data suggest that: (1) the nuclear membranes of hippocampal neurons contain distinct sets of ion channels, which are specific for each type of neuron; (2) IP3 Rs, but not RyRs are targeted to the inner nuclear membrane of CA1 pyramidal and DG granule, but they were not found in the nuclear membranes of CA3 pyramidal neurons; (3) the nuclear envelope of these neurons is specialized to release Ca(2+) into the nucleoplasm which may amplify Ca(2+) signals entering the nucleus from the cytoplasm or generate Ca(2+) transients on its own; (4) LCC channels are an integral part the of Ca(2+) -releasing machinery providing a route for counterflow of К(+) and thereby facilitating Ca(2+) movement in and out of the Ca(2+) store.


Subject(s)
Calcium Signaling/physiology , Hippocampus/cytology , Ion Channels/physiology , Neurons/physiology , Nuclear Envelope/physiology , Adenosine Triphosphate/pharmacology , Animals , Calcium/metabolism , Gene Expression Regulation , Hippocampus/physiology , Inositol 1,4,5-Trisphosphate/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/agonists , Inositol 1,4,5-Trisphosphate Receptors/physiology , Ion Transport , Male , Patch-Clamp Techniques , Pyramidal Cells/physiology , Rats , Rats, Wistar , Ryanodine Receptor Calcium Release Channel/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...