1
|
Michelangeli F, Mohammed NA, Jones B, Tairu M, Al‐Mousa F. Cytotoxicity by endocrine disruptors through effects on ER Ca 2+ transporters, aberrations in Ca 2+ signalling pathways and ER stress. FEBS Open Bio 2024; 14:1384-1396. [PMID: 39138623 PMCID: PMC11492318 DOI: 10.1002/2211-5463.13880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/24/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
Concerns regarding man-made organic chemicals pervading our ecosystem and having adverse and detrimental effects upon organisms, including man, have now been studied for several decades. Since the 1970s, some environmental pollutants were identified as having endocrine disrupting affects. These endocrine disrupting chemicals (EDC) were initially shown to have estrogenic or anti-estrogenic properties and some were also shown to bind to a variety of hormone receptors. However, since the 1990s it has also been identified that many of these EDC additionally, have the ability of causing abnormal alterations in Ca2+ signalling pathways (also commonly involved in hormone signalling), leading to exaggerated elevations in cytosolic [Ca2+] levels, that is known to cause activation of a number of cell death pathways. The major emphasis of this review is to present a personal perspective of the evidence for some types of EDC, specifically alkylphenols and brominated flame retardants (BFRs), causing direct effects on Ca2+ transporters (mainly the SERCA Ca2+ ATPases), culminating in acute cytotoxicity and cell death. Evidence is also presented to indicate that this Ca2+ATPase inhibition, which leads to abnormally elevated cytosolic [Ca2+], as well as a decreased luminal ER [Ca2+], which triggers the ER stress response, are both involved in acute cytotoxicity.
Collapse
Affiliation(s)
- Francesco Michelangeli
- Chester Medical SchoolUniversity of ChesterUK
- School of BiosciencesUniversity of BirminghamUK
| | - Noor A. Mohammed
- School of BiosciencesUniversity of BirminghamUK
- Department of BiologyUniversity of DuhokIraq
| | | | | | - Fawaz Al‐Mousa
- General Directorate of Poison Control CentreMinistry of HealthRiyadhSaudi Arabia
| |
Collapse
|
2
|
Tao S, Hulpiau P, Wagner LE, Witschas K, Yule DI, Bultynck G, Leybaert L. IP3RPEP6, a novel peptide inhibitor of IP 3 receptor channels that does not affect connexin-43 hemichannels. Acta Physiol (Oxf) 2024; 240:e14086. [PMID: 38240350 DOI: 10.1111/apha.14086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/24/2023] [Accepted: 01/01/2024] [Indexed: 02/24/2024]
Abstract
AIM Inositol 1,4,5-trisphosphate receptors (IP3 Rs) are intracellular Ca2+ -release channels with crucial roles in cell function. Current IP3 R inhibitors suffer from off-target effects and poor selectivity towards the three distinct IP3 R subtypes. We developed a novel peptide inhibitor of IP3 Rs and determined its effect on connexin-43 (Cx43) hemichannels, which are co-activated by IP3 R stimulation. METHODS IP3RPEP6 was developed by in silico molecular docking studies and characterized by on-nucleus patch-clamp experiments of IP3 R2 channels and carbachol-induced IP3 -mediated Ca2+ responses in IP3 R1, 2 or 3 expressing cells, triple IP3 R KO cells and astrocytes. Cx43 hemichannels were studied by patch-clamp and ATP-release approaches, and by inhibition with Gap19 peptide. IP3RPEP6 interactions with IP3 Rs were verified by co-immunoprecipitation and affinity pull-down assays. RESULTS IP3RPEP6 concentration-dependently reduced the open probability of IP3 R2 channels and competitively inhibited IP3 Rs in an IC50 order of IP3 R2 (~3.9 μM) < IP3 R3 (~4.3 μM) < IP3 R1 (~9.0 μM), without affecting Cx43 hemichannels or ryanodine receptors. IP3RPEP6 co-immunoprecipitated with IP3 R2 but not with IP3 R1; interaction with IP3 R3 varied between cell types. The IC50 of IP3RPEP6 inhibition of carbachol-induced Ca2+ responses decreased with increasing cellular Cx43 expression. Moreover, Gap19-inhibition of Cx43 hemichannels significantly reduced the amplitude of the IP3 -Ca2+ responses and strongly increased the EC50 of these responses. Finally, we identified palmitoyl-8G-IP3RPEP6 as a membrane-permeable IP3RPEP6 version allowing extracellular application of the IP3 R-inhibiting peptide. CONCLUSION IP3RPEP6 inhibits IP3 R2/R3 at concentrations that have limited effects on IP3 R1. IP3 R activation triggers hemichannel opening, which strongly affects the amplitude and concentration-dependence of IP3 -triggered Ca2+ responses.
Collapse
Affiliation(s)
- Siyu Tao
- Department of Basic and Applied Medical Sciences-Physiology Group, Ghent University, Ghent, Belgium
| | - Paco Hulpiau
- Department of Bio-Medical Sciences, HOWEST University of Applied Sciences (Hogeschool West-Vlaanderen), Bruges, Belgium
| | - Larry E Wagner
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, New York, USA
| | - Katja Witschas
- Department of Basic and Applied Medical Sciences-Physiology Group, Ghent University, Ghent, Belgium
| | - David I Yule
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, New York, USA
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signaling, Department of Molecular Cell Biology, KU Leuven, Leuven, Belgium
| | - Luc Leybaert
- Department of Basic and Applied Medical Sciences-Physiology Group, Ghent University, Ghent, Belgium
| |
Collapse
|
3
|
Song X, Kirtipal N, Lee S, Malý P, Bharadwaj S. Current therapeutic targets and multifaceted physiological impacts of caffeine. Phytother Res 2023; 37:5558-5598. [PMID: 37679309 DOI: 10.1002/ptr.8000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/04/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023]
Abstract
Caffeine, which shares consubstantial structural similarity with purine adenosine, has been demonstrated as a nonselective adenosine receptor antagonist for eliciting most of the biological functions at physiologically relevant dosages. Accumulating evidence supports caffeine's beneficial effects against different disorders, such as total cardiovascular diseases and type 2 diabetes. Conversely, paradoxical effects are also linked to caffeine ingestion in humans including hypertension-hypotension and tachycardia-bradycardia. These observations suggest the association of caffeine action with its ingested concentration and/or concurrent interaction with preferential molecular targets to direct explicit events in the human body. Thus, a coherent analysis of the functional targets of caffeine, relevant to normal physiology, and disease pathophysiology, is required to understand the pharmacology of caffeine. This review provides a broad overview of the experimentally validated targets of caffeine, particularly those of therapeutic interest, and the impacts of caffeine on organ-specific physiology and pathophysiology. Overall, the available empirical and epidemiological evidence supports the dose-dependent functional activities of caffeine and advocates for further studies to get insights into the caffeine-induced changes under specific conditions, such as asthma, DNA repair, and cancer, in view of its therapeutic applications.
Collapse
Affiliation(s)
- Xinjie Song
- Zhejiang Provincial Key Lab for Chemical and Biological Processing Technology of Farm Product, School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China
| | - Nikhil Kirtipal
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Sunjae Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Petr Malý
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences v.v.i, BIOCEV Research Center, Vestec, Czech Republic
| | - Shiv Bharadwaj
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences v.v.i, BIOCEV Research Center, Vestec, Czech Republic
| |
Collapse
|
4
|
Augmented K Ca2.3 Channel Feedback Regulation of Oxytocin Stimulated Uterine Strips from Nonpregnant Mice. Int J Mol Sci 2021; 22:ijms222413585. [PMID: 34948381 PMCID: PMC8709448 DOI: 10.3390/ijms222413585] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/07/2021] [Accepted: 12/11/2021] [Indexed: 11/16/2022] Open
Abstract
Uterine contractions prior to 37 weeks gestation can result in preterm labor with significant risk to the infant. Current tocolytic therapies aimed at suppressing premature uterine contractions are largely ineffective and cause serious side effects. Calcium (Ca2+) dependent contractions of uterine smooth muscle are physiologically limited by the opening of membrane potassium (K+) channels. Exploiting such inherent negative feedback mechanisms may offer new strategies to delay labor and reduce risk. Positive modulation of small conductance Ca2+-activated K+ (KCa2.3) channels with cyclohexyl-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-pyrimidin-4-yl]-amine (CyPPA), effectively decreases uterine contractions. This study investigates whether the receptor agonist oxytocin might solicit KCa2.3 channel feedback that facilitates CyPPA suppression of uterine contractions. Using isometric force myography, we found that spontaneous phasic contractions of myometrial tissue from nonpregnant mice were suppressed by CyPPA and, in the presence of CyPPA, oxytocin failed to augment contractions. In tissues exposed to oxytocin, depletion of internal Ca2+ stores with cyclopiazonic acid (CPA) impaired CyPPA relaxation, whereas blockade of nonselective cation channels (NSCC) using gadolinium (Gd3+) had no significant effect. Immunofluorescence revealed close proximity of KCa2.3 channels and ER inositol trisphosphate receptors (IP3Rs) within myometrial smooth muscle cells. The findings suggest internal Ca2+ stores play a role in KCa2.3-dependent feedback control of uterine contraction and offer new insights for tocolytic therapies.
Collapse
|
5
|
Sherwood MW, Arizono M, Panatier A, Mikoshiba K, Oliet SHR. Astrocytic IP 3Rs: Beyond IP 3R2. Front Cell Neurosci 2021; 15:695817. [PMID: 34393726 PMCID: PMC8363081 DOI: 10.3389/fncel.2021.695817] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/30/2021] [Indexed: 12/31/2022] Open
Abstract
Astrocytes are sensitive to ongoing neuronal/network activities and, accordingly, regulate neuronal functions (synaptic transmission, synaptic plasticity, behavior, etc.) by the context-dependent release of several gliotransmitters (e.g., glutamate, glycine, D-serine, ATP). To sense diverse input, astrocytes express a plethora of G-protein coupled receptors, which couple, via Gi/o and Gq, to the intracellular Ca2+ release channel IP3-receptor (IP3R). Indeed, manipulating astrocytic IP3R-Ca2+ signaling is highly consequential at the network and behavioral level: Depleting IP3R subtype 2 (IP3R2) results in reduced GPCR-Ca2+ signaling and impaired synaptic plasticity; enhancing IP3R-Ca2+ signaling affects cognitive functions such as learning and memory, sleep, and mood. However, as a result of discrepancies in the literature, the role of GPCR-IP3R-Ca2+ signaling, especially under physiological conditions, remains inconclusive. One primary reason for this could be that IP3R2 has been used to represent all astrocytic IP3Rs, including IP3R1 and IP3R3. Indeed, IP3R1 and IP3R3 are unique Ca2+ channels in their own right; they have unique biophysical properties, often display distinct distribution, and are differentially regulated. As a result, they mediate different physiological roles to IP3R2. Thus, these additional channels promise to enrich the diversity of spatiotemporal Ca2+ dynamics and provide unique opportunities for integrating neuronal input and modulating astrocyte–neuron communication. The current review weighs evidence supporting the existence of multiple astrocytic-IP3R isoforms, summarizes distinct sub-type specific properties that shape spatiotemporal Ca2+ dynamics. We also discuss existing experimental tools and future refinements to better recapitulate the endogenous activities of each IP3R isoform.
Collapse
Affiliation(s)
- Mark W Sherwood
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| | - Misa Arizono
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Aude Panatier
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| | - Katsuhiko Mikoshiba
- ShanghaiTech University, Shanghai, China.,Faculty of Science, Toho University, Funabashi, Japan.,RIKEN CLST, Kobe, Japan
| | - Stéphane H R Oliet
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| |
Collapse
|
6
|
Improving fertilization rate in ICSI cycles by adding myoinositol to the semen preparation procedures: a prospective, bicentric, randomized trial on sibling oocytes. J Assist Reprod Genet 2015; 32:387-94. [PMID: 25601322 DOI: 10.1007/s10815-014-0401-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 12/04/2014] [Indexed: 10/24/2022] Open
Abstract
PURPOSE To evaluate whether the in vitro incubation of spermatozoa with myoinositol may improve the fertilization rate in ICSI cycles. METHODS This is a prospective, bicentric, randomized study on 500 MII sibling oocytes injected in 78 ICSI cycles performed between March and October 2013. Randomization of the oocytes into two groups was performed at the time of the denudation. Fertilization rates (per oocyte injected with spermatozoa treated with myoinositol versus per oocyte injected with spermatozoa treated with placebo) were measured as primary outcome and embryo morphology as secondary outcome. Clinical outcomes were also documented. RESULT (S) Fertilization rate (78.9 ± 28.6% vs 63.2 ± 36.7, P = 0.002) and percentage of grade A embryos on day 3 (59.8 ± 35.6% vs 43.5 ± 41.5, P = 0.019) were significantly higher when spermatozoa were treated in vitro with myoinositol versus placebo. No differences were found for the expanded blastocyst formation rate. CONCLUSION (S) In vitro treatment of spermatozoa with myoinositol may optimize ICSI outcomes by improving the fertilization rate and embryo quality on day 3. The improvement of the number and the quality of embryos available in an ICSI cycle may have clinical utility if these findings can be confirmed.
Collapse
|
7
|
Vasudevan SR, Singh N, Churchill GC. Scaffold hopping with virtual screening from IP3 to a drug-like partial agonist of the inositol trisphosphate receptor. Chembiochem 2014; 15:2774-82. [PMID: 25399672 DOI: 10.1002/cbic.201402440] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Indexed: 11/06/2022]
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.
Collapse
Affiliation(s)
- Sridhar R Vasudevan
- Department of Pharmacology, University of Oxford, Mansfield Road, OX1 3QT (UK).
| | | | | |
Collapse
|
8
|
Saleem H, Tovey SC, Molinski TF, Taylor CW. Interactions of antagonists with subtypes of inositol 1,4,5-trisphosphate (IP3) receptor. Br J Pharmacol 2014; 171:3298-312. [PMID: 24628114 PMCID: PMC4080982 DOI: 10.1111/bph.12685] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 02/26/2014] [Accepted: 03/05/2014] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND AND PURPOSE Inositol 1,4,5-trisphosphate receptors (IP3 Rs) are intracellular Ca(2+) channels. Interactions of the commonly used antagonists of IP3Rs with IP3R subtypes are poorly understood. EXPERIMENTAL APPROACH IP3-evoked Ca(2+) release from permeabilized DT40 cells stably expressing single subtypes of mammalian IP3R was measured using a luminal Ca(2+) indicator. The effects of commonly used antagonists on IP3-evoked Ca(2+) release and (3) H-IP3 binding were characterized. KEY RESULTS Functional analyses showed that heparin was a competitive antagonist of all IP3R subtypes with different affinities for each (IP3R3 > IP3R1 ≥ IP3R2). This sequence did not match the affinities for heparin binding to the isolated N-terminal from each IP3R subtype. 2-aminoethoxydiphenyl borate (2-APB) and high concentrations of caffeine selectively inhibited IP3R1 without affecting IP3 binding. Neither Xestospongin C nor Xestospongin D effectively inhibited IP3-evoked Ca(2+) release via any IP3R subtype. CONCLUSIONS AND IMPLICATIONS Heparin competes with IP3, but its access to the IP3-binding core is substantially hindered by additional IP3R residues. These interactions may contribute to its modest selectivity for IP3R3. Practicable concentrations of caffeine and 2-APB inhibit only IP3R1. Xestospongins do not appear to be effective antagonists of IP3Rs.
Collapse
Affiliation(s)
- Huma Saleem
- Department of Pharmacology, University of CambridgeCambridge, UK
| | - Stephen C Tovey
- Department of Pharmacology, University of CambridgeCambridge, UK
| | | | - Colin W Taylor
- Department of Pharmacology, University of CambridgeCambridge, UK
| |
Collapse
|
9
|
Abstract
The Ca(2) (+) signals evoked by inositol 1,4,5-trisphosphate (IP(3)) are built from elementary Ca(2) (+) release events involving progressive recruitment of IP(3) receptors (IP(3)R), intracellular Ca(2) (+) channels that are expressed in almost all animal cells. The smallest events ('blips') result from opening of single IP(3)R. Larger events ('puffs') reflect the near-synchronous opening of a small cluster of IP(3)R. These puffs become more frequent as the stimulus intensity increases and they eventually trigger regenerative Ca(2) (+) waves that propagate across the cell. This hierarchical recruitment of IP(3)R is important in allowing Ca(2) (+) signals to be delivered locally to specific target proteins or more globally to the entire cell. Co-regulation of IP(3)R by Ca(2) (+) and IP(3), the ability of a single IP(3)R rapidly to mediate a large efflux of Ca(2) (+) from the endoplasmic reticulum, and the assembly of IP(3)R into clusters are key features that allow IP(3)R to propagate Ca(2) (+) signals regeneratively. We review these properties of IP(3)R and the structural basis of IP(3)R behavior.
Collapse
Affiliation(s)
- Colin W Taylor
- Department of Pharmacology, Tennis Court Road, CB2 1PD, Cambridge, UK,
| | | |
Collapse
|
10
|
Yeung-Yam-Wah V, Lee AK, Tse A. Arachidonic acid mobilizes Ca2+ from the endoplasmic reticulum and an acidic store in rat pancreatic β cells. Cell Calcium 2011; 51:140-8. [PMID: 22197025 DOI: 10.1016/j.ceca.2011.11.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 11/30/2011] [Accepted: 11/30/2011] [Indexed: 11/17/2022]
Abstract
In rat pancreatic β cells, arachidonic acid (AA) triggered intracellular Ca(2+) release. This effect could be mimicked by eicosatetraynoic acid, indicating that AA metabolism is not required. The AA-mediated Ca(2+) signal was not affected by inhibition of ryanodine receptors or emptying of ryanodine-sensitive store but was reduced by ∼70% following the disruption of acidic stores (treatment with bafilomycin A1 or glycyl-phenylalanyl-β-naphthylamide (GPN)). The action of AA did not involve TRPM2 channels or NAADP receptors because intracellular dialysis of adenosine diphosphoribose (ADPR; an activator of TRPM2 channels) or NAADP did not affect the AA response. In contrast, stimulation of IP(3) receptors via intracellular dialysis of adenophostin A, or exogenous application of ATP largely abolished the AA-mediated Ca(2+) signal. Intracellular dialysis of heparin abolished the ATP-mediated Ca(2+) signal but not the AA response, suggesting that the action of AA did not involve the IP(3)-binding site. Treatment with the SERCA pump inhibitor, thapsigargin, reduced the amplitude of the AA-mediated Ca(2+) signal by ∼70%. Overall, our finding suggests that AA mobilizes Ca(2+) from the endoplasmic reticulum as well as an acidic store and both stores could be depleted by IP(3) receptor agonist. The possibility of secretory granules as targets of AA is discussed.
Collapse
|
11
|
Abstract
Inositol 1,4,5-trisphosphate receptors (IP(3)R) and their relatives, ryanodine receptors, are the channels that most often mediate Ca(2+) release from intracellular stores. Their regulation by Ca(2+) allows them also to propagate cytosolic Ca(2+) signals regeneratively. This brief review addresses the structural basis of IP(3)R activation by IP(3) and Ca(2+). IP(3) initiates IP(3)R activation by promoting Ca(2+) binding to a stimulatory Ca(2+)-binding site, the identity of which is unresolved. We suggest that interactions of critical phosphate groups in IP(3) with opposite sides of the clam-like IP(3)-binding core cause it to close and propagate a conformational change toward the pore via the adjacent N-terminal suppressor domain. The pore, assembled from the last pair of transmembrane domains and the intervening pore loop from each of the four IP(3)R subunits, forms a structure in which a luminal selectivity filter and a gate at the cytosolic end of the pore control cation fluxes through the IP(3)R.
Collapse
Affiliation(s)
- Colin W Taylor
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, United Kingdom.
| | | |
Collapse
|
12
|
Costello S, Michelangeli F, Nash K, Lefievre L, Morris J, Machado-Oliveira G, Barratt C, Kirkman-Brown J, Publicover S. Ca2+-stores in sperm: their identities and functions. Reproduction 2009; 138:425-37. [PMID: 19542252 DOI: 10.1530/rep-09-0134] [Citation(s) in RCA: 150] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Intracellular Ca2+ stores play a central role in the regulation of cellular [Ca2+](i) and the generation of complex [Ca2+] signals such as oscillations and waves. Ca2+ signalling is of particular significance in sperm cells, where it is a central regulator in many key activities (including capacitation, hyperactivation, chemotaxis and acrosome reaction) yet mature sperm lack endoplasmic reticulum and several other organelles that serve as Ca2+ stores in somatic cells. Here, we review i) the evidence for the expression in sperm of the molecular components (pumps and channels) which are functionally significant in the activity of Ca2+ stores of somatic cells and ii) the evidence for the existence of functional Ca2+ stores in sperm. This evidence supports the existence of at least two storage organelles in mammalian sperm, one in the acrosomal region and another in the region of the sperm neck and midpiece. We then go on to discuss the probable identity of these organelles and their discrete functions: regulation by the acrosome of its own secretion and regulation by membranous organelles at the sperm neck (and possibly by the mitochondria) of flagellar activity and hyperactivation. Finally, we consider the ability of the sperm discretely to control mobilisation of these stores and the functional interaction of stored Ca2+ at the sperm neck/midpiece with CatSper channels in the principal piece in regulation of the activities of mammalian sperm.
Collapse
Affiliation(s)
- Sarah Costello
- School of Biosciences, University of Birmingham, Birmingham B152TT, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Jimenez-Gonzalez C, Michelangeli F, Harper CV, Barratt CLR, Publicover SJ. Calcium signalling in human spermatozoa: a specialized 'toolkit' of channels, transporters and stores. Hum Reprod Update 2005; 12:253-67. [PMID: 16338990 DOI: 10.1093/humupd/dmi050] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Ca(2+) is a ubiquitous intracellular messenger which encodes information by temporal and spatial patterns of concentration. In spermatozoa, several key functions, including acrosome reaction and motility, are regulated by cytoplasmic Ca(2+) concentration. Despite the very small size and apparent structural simplicity of spermatozoa, evidence is accumulating that they possess sophisticated mechanisms for regulation of cytoplasmic Ca(2+) concentration and generation of complex Ca(2+) signals. In this review, we consider the various components of the Ca(2+)-signalling 'toolkit' that have been characterized in somatic cells and summarize the evidence for their presence and activity in spermatozoa. In particular, data accumulated over the last few years show that spermatozoa possess one (and probably two) Ca(2+) stores as well as a range of plasma membrane pumps and channels. Selective regulation of the various components of the 'toolkit' by agonists probably allows spermatozoa to generate localized Ca(2+) signals despite their very small cytoplasmic volume, permitting the discrete and selective activation of cell functions.
Collapse
|
14
|
Pattni K, Banting G. Ins(1,4,5)P3 metabolism and the family of IP3-3Kinases. Cell Signal 2005; 16:643-54. [PMID: 15093605 DOI: 10.1016/j.cellsig.2003.10.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2003] [Accepted: 10/24/2003] [Indexed: 11/17/2022]
Abstract
The release of Ca2+ from intracellular stores is triggered by the second messenger inositol (1,4,5)-trisphosphate (Ins(1,4,5)P3). The regulation of this process is critically important for cellular homeostasis. Ins(1,4,5)P3 is rapidly metabolised, either to inositol (1,4)-bisphosphate (Ins(1,4)P2) by inositol polyphosphate 5-phosphatases or to inositol (1,3,4,5)-tetrakisphosphate (Ins(1,3,4,5)P4) by one of a family of inositol (1,4,5)P3 3-kinases (IP3-3Ks). Three isoforms of IP3-3K have now been identified in mammals; they have a conserved C-terminal catalytic domain, but divergent N-termini. This review discusses the metabolism of Ins(1,4,5)P3, compares the IP3-3K isoforms and addresses potential mechanisms by which their activity might be regulated.
Collapse
Affiliation(s)
- Krupa Pattni
- Department of Biochemistry, University of Bristol, Bristol, BS8 1TD, UK
| | | |
Collapse
|
15
|
Chang YT, Rosania GR, Chung SK. Inositol phospholipid pathway inhibitors and regulators Inositol phospholipid pathway inhibitors and regulators. Expert Opin Ther Pat 2005. [DOI: 10.1517/13543776.11.1.45] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
16
|
Laporte R, Hui A, Laher I. Pharmacological modulation of sarcoplasmic reticulum function in smooth muscle. Pharmacol Rev 2004; 56:439-513. [PMID: 15602008 DOI: 10.1124/pr.56.4.1] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The sarco/endoplasmic reticulum (SR/ER) is the primary storage and release site of intracellular calcium (Ca2+) in many excitable cells. The SR is a tubular network, which in smooth muscle (SM) cells distributes close to cellular periphery (superficial SR) and in deeper aspects of the cell (deep SR). Recent attention has focused on the regulation of cell function by the superficial SR, which can act as a buffer and also as a regulator of membrane channels and transporters. Ca2+ is released from the SR via two types of ionic channels [ryanodine- and inositol 1,4,5-trisphosphate-gated], whereas accumulation from thecytoplasm occurs exclusively by an energy-dependent sarco-endoplasmic reticulum Ca2+-ATPase pump (SERCA). Within the SR, Ca2+ is bound to various storage proteins. Emerging evidence also suggests that the perinuclear portion of the SR may play an important role in nuclear transcription. In this review, we detail the pharmacology of agents that alter the functions of Ca2+ release channels and of SERCA. We describe their use and selectivity and indicate the concentrations used in investigating various SM preparations. Important aspects of cell regulation and excitation-contractile activity coupling in SM have been uncovered through the use of such activators and inhibitors of processes that determine SR function. Likewise, they were instrumental in the recent finding of an interaction of the SR with other cellular organelles such as mitochondria. Thus, an appreciation of the pharmacology and selectivity of agents that interfere with SR function in SM has greatly assisted in unveiling the multifaceted nature of the SR.
Collapse
Affiliation(s)
- Régent Laporte
- Ferring Research Institute, Inc., Ferring Pharmaceuticals, San Diego, California, USA
| | | | | |
Collapse
|
17
|
Pattni K, Millard TH, Banting G. Calpain cleavage of the B isoform of Ins(1,4,5)P3 3-kinase separates the catalytic domain from the membrane anchoring domain. Biochem J 2003; 375:643-51. [PMID: 12906709 PMCID: PMC1223724 DOI: 10.1042/bj20030505] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2003] [Revised: 07/24/2003] [Accepted: 08/07/2003] [Indexed: 11/17/2022]
Abstract
Inositol (1,4,5)-trisphosphate [Ins(1,4,5)P3] is one of the key intracellular second messengers in cells and mobilizes Ca2+ stores in the ER (endoplasmic reticulum). Ins(1,4,5)P3 has a short half-life within the cell, and is rapidly metabolized through one of two pathways, one of which involves further phosphorylation of the inositol ring: Ins(1,4,5)P3 3-kinase (IP3-3K) phosphorylates Ins(1,4,5)P3, resulting in the formation of inositol (1,3,4,5)-tetrakisphosphate [Ins(1,3,4,5)P4]. There are three known isoforms of IP3-3K, designated IP3-3KA, IP3-3KB and IP3-3KC. These have differing N-termini, but highly conserved C-termini harbouring the catalytic domain. The three IP3-3K isoforms have different subcellular locations and the B-kinase is uniquely present in both cytosolic and membrane-bound pools. As it is the N-terminus of the B-kinase that differs most from the A- and C-kinases, we have hypothesized that this portion of the protein may be responsible for membrane localization. Although there are no known membrane-targeting protein motifs within the sequence of IP3-3KB, it is found to be tightly associated with the ER membrane. Here, we show that specific regions of the N-terminus of IP3-3KB are necessary and sufficient for efficient membrane localization of the protein. We also report that, in the presence of Ca2+, the kinase domain of IP3-3KB is cleaved from the membrane-anchoring region by calpain.
Collapse
Affiliation(s)
- Krupa Pattni
- Department of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | | | | |
Collapse
|
18
|
Khan SZ, Kirk CJ, Michelangeli F. Alkylphenol endocrine disrupters inhibit IP3-sensitive Ca2+ channels. Biochem Biophys Res Commun 2003; 310:261-6. [PMID: 14521904 DOI: 10.1016/j.bbrc.2003.09.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We have investigated the influence of alkylphenol endocrine disrupters and the synthetic estrogen diethylstilbestrol (DES) on inositol-1,4,5-trisphosphate (IP(3))-sensitive Ca(2+) channels from porcine cerebellum and rat testicular membranes. All alkylphenols and DES inhibited the extent of IP(3)-induced Ca(2+) release (IICR) from both cerebellar and testicular microsomes. 4-n-nonylphenol was the most potent compound tested (IC(50), 8 microM). Inhibition of IICR was directly related to the length and hydrophobicity of the alkylphenol side chain. None of the alkylphenols or DES appeared to influence the concentration dependence of IICR nor did they have a significant effect on [3H]IP(3) binding to the membranes. An investigation of the effects of nonylphenol on the transient kinetics of IICR showed that it inhibited the rate constants for both the fast and the slow phases of IICR and also the extent of Ca(2+) release. These results illustrate another mechanism by which these environmental pollutants can disrupt endocrine function without the involvement of estrogen receptors.
Collapse
Affiliation(s)
- Shahla Zafar Khan
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | | | | |
Collapse
|
19
|
White C, McGeown JG. Inositol 1,4,5-trisphosphate receptors modulate Ca2+ sparks and Ca2+ store content in vas deferens myocytes. Am J Physiol Cell Physiol 2003; 285:C195-204. [PMID: 12620813 DOI: 10.1152/ajpcell.00374.2002] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Spontaneous Ca2+ sparks were observed in fluo 4-loaded myocytes from guinea pig vas deferens with line-scan confocal imaging. They were abolished by ryanodine (100 microM), but the inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) blockers 2-aminoethoxydiphenyl borate (2-APB; 100 microM) and intracellular heparin (5 mg/ml) increased spark frequency, rise time, duration, and spread. Very prolonged Ca2+ release events were also observed in approximately 20% of cells treated with IP3R blockers but not under control conditions. 2-APB and heparin abolished norepinephrine (10 microM; 0 Ca2+)-evoked Ca2+ transients but increased caffeine (10 mM; 0 Ca2+) transients in fura 2-loaded myocytes. Transients evoked by ionomycin (25 microM; 0 Ca2+) were also enhanced by 2-APB. Ca2+ sparks and transients evoked by norepinephrine and caffeine were abolished by thimerosal (100 microM), which sensitizes the IP3R to IP3. In cells voltage clamped at -40 mV, spontaneous transient outward currents (STOCs) were increased in frequency, amplitude, and duration in the presence of 2-APB. These data are consistent with a model in which the Ca2+ store content in smooth muscle is limited by tonic release of Ca2+ via an IP3-dependent pathway. Blockade of IP3Rs elevates sarcoplasmic reticulum store content, promoting Ca2+ sparks and STOC activity.
Collapse
Affiliation(s)
- Carl White
- Smooth Muscle Research Group, School of Medicine, Queen's University of Belfast, Belfast BT9 7BL, UK
| | | |
Collapse
|
20
|
Dyer JL, Khan SZ, Bilmen JG, Hawtin SR, Wheatley M, Javed MUH, Michelangeli F. Curcumin: a new cell-permeant inhibitor of the inositol 1,4,5-trisphosphate receptor. Cell Calcium 2002; 31:45-52. [PMID: 11990299 DOI: 10.1054/ceca.2001.0259] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Curcumin (diferuoylmethane or 1,7-bis (4-hydroxy-3-methoxyphenol)-1,6-hepatadiene-3,5-dione) is the active ingredient of the spice turmeric. Curcumin has been shown to have a number of pharmacological and therapeutic uses. This study shows that curcumin is a potent inhibitor of the inositol 1,4,5-trisphosphate-sensitive Ca2+ channel (InsP3 receptor). In porcine cerebellar microsomes, the extent of InsP3-induced Ca2+ release (IICR) is almost completely inhibited by 50 microM curcumin (IC50 = 10 microM). As the extent of IICR cannot be restored back to control levels by the addition of excess InsP3 and since it has little effect on [3H]InsP3 binding to cerebellar microsomes, this inhibition is likely to be non-competitive in nature. IICR in cerebellar microsomes is biphasic consisting of a fast and slow component. The rate constants for the two components are both reduced by curcumin to similar extents (by about 70% of control values at 40 microM curcumin). In addition, curcumin also reduces agonist (ATP)-stimulated Ca2+ mobilization from intact HL-60 cells, indicating that curcumin is cell permeant. However, since it also affects intracellular Ca2+ pumps and possibly ryanodine receptors, it may lead to complex Ca2+ transient responses within cells, which may well explain some of its putative therapeutic properties.
Collapse
Affiliation(s)
- J L Dyer
- School of Biosciences, University of Birmingham, Edgbaston, UK
| | | | | | | | | | | | | |
Collapse
|
21
|
Dyer JL, Michelangeli F. Inositol 1,4,5-trisphosphate receptor isoforms show similar Ca2+ release kinetics. Cell Calcium 2001; 30:245-50. [PMID: 11587548 DOI: 10.1054/ceca.2001.0231] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The inositol 1,4,5-trisphosphate receptor (InsP3R) is an intracellular Ca2+ release channel which upon activation initiates many cellular functions. Multiple InsP3R subtypes are expressed in most cell types but the physiological significance of this heterogeneity is poorly understood. This study has directly compared the functional properties of the three different InsP3R isoforms by analyzing their InsP3-induced Ca2+ release (IICR) properties in cell lines which predominantly express each isoform subtype. The InsP3-dependence of the amount or extent of IICR was InsP3R isoform-specific, with the type III isoform having the lowest affinity with respect to Ca2+ release. The transient kinetics of IICR, measured using stopped-flow spectrofluorimetry, however, were similar for all three InsP3R isoforms. At maximal InsP3 concentrations (20 microM) the rate constants where between 0.8 and 1.0 s(-1) for the fast phase and 0.25-0.45 s(-1) for the slow phase. The concentration of InsP3 required to induce half-maximal rates of Ca2+ release (EC50) were also similar for the three isoforms (0.2-0.4 microM for the fast phase and 0.75-0.95 microM for the slow phase). These results indicate the InsP3R channel does not significantly differ functionally in terms of Ca2+ release rates between isoforms. The temporal and spatial features of intracellular Ca2+ signals are thus probably achieved through InsP3R isoform-specific regulation or localization rather than their intrinsic Ca2+ efflux properties.
Collapse
Affiliation(s)
- J L Dyer
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, UK
| | | |
Collapse
|
22
|
Khan SZ, Dyer JL, Michelangeli F. Inhibition of the type 1 inositol 1,4,5-trisphosphate-sensitive Ca2+ channel by calmodulin antagonists. Cell Signal 2001; 13:57-63. [PMID: 11257448 DOI: 10.1016/s0898-6568(00)00140-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study describes the effects of a number of calmodulin antagonists on the cerebellar type 1 inositol 1,4,5-trisphosphate (InsP3) receptor. All the antagonists tested (trifluoperazine, fluphenazine, chlorpromazine and calmidazolium) inhibited the extent of InsP3-induced Ca2+ release (IICR) with similar IC(50) values (between 60 and 85 microM). They did not affect the efficacy of InsP3 to release Ca2+, since the concentrations of InsP3 required to cause half-maximal release was little affected in the presence of these agents. In addition, these agents did not affect InsP3 binding to its receptor. Stopped-flow studies to determine the rate constants of IICR showed this process to be biphasic with a fast and slow component. All the calmodulin antagonists appeared to reduce the rate constants for Ca2+ release in a phase-specific manner, preferentially reducing the fast phase component. Chlorpromazine (75 microM) appeared to have the most potent effect on the fast phase rate constant, reducing it from 1.0 to 0.08 s(-1), while only reducing the rate constant for the slow phase about twofold (0.2-0.08 s(-1)). The fact that calmodulin itself inhibits both IICR and InsP3 binding, while these calmodulin antagonists also reduce Ca2+ release and do not affect InsP3 binding, suggests that the mechanism of action of these agents is unlikely to be due to the reversal of the modulatory action of calmodulin on this receptor.
Collapse
Affiliation(s)
- S Z Khan
- School of Biosciences, University of Birmingham, Edgbaston, B15 2TT, Birmingham, UK
| | | | | |
Collapse
|
23
|
Tovey SC, Dyer JL, Godfrey RE, Khan SZ, Bilmen JG, Mezna M, Michelangeli F. Subtype identification and functional properties of inositol 1,4, 5-trisphosphate receptors in heart and aorta. Pharmacol Res 2000; 42:581-90. [PMID: 11058412 DOI: 10.1006/phrs.2000.0733] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
One of the major mechanisms by which hormones elevate intracellular Ca(2+)levels is by generating the second messenger inositol 1,4, 5-trisphosphate (InsP(3)), which activates a Ca(2+)channel (InsP(3)receptor) located in the endoplasmic reticulum (ER). This study undertakes to identify the InsP(3)receptor subtypes (isoforms) in heart and aorta and to characterize their functional properties. The InsP(3)receptor isoforms were identified from rat heart and aorta tissues using both reverse-transcriptase polymerase chain reaction (RT-PCR) to assess the presence of mRNA for the different isoforms and immunochemistry using InsP(3)receptor isoform-specific antibodies. Functional studies included ligand binding experiments using [(3)H]InsP(3)and InsP(3)-induced Ca(2+)release studies using Fluo-3 as the Ca(2+)sensing dye. All three isoforms of the InsP(3)receptor were identified using RT-PCR and immunochemical analyses. [(3)H]InsP(3)binding studies using microsomes derived from these tissues showed that heart had a 3-fold lower abundance of InsP(3)receptors than aorta, while both have considerably lower abundance than the well characterized cerebellar microsomes. The affinity of the InsP(3)binding to the receptor was also different in the three tissues. In cerebellum the K(d)was 60 nM, while aorta had a much higher K(d)of 220 nM. Heart microsomes, appeared to show two classes of binding affinity with K(d)s of 150 nM and 60 nM. Furthermore, the effects of free [Ca(2+)] on [(3)H]InsP(3)binding levels were also different for the three tissues. InsP(3)binding to both cerebellar and aorta microsomes decreased by 90% and 60%, respectively, above 30 nM free [Ca(2+)], while InsP(3)binding to heart was relatively insensitive to changes in [Ca(2+)]. At maximal InsP(3)concentrations, aorta microsomes were able to release about 5% of the accumulated Ca(2+), compared to 25% by cerebellar microsomes. Heart microsomes, however, showed only very little InsP(3)-induced Ca(2+)release ( <0.5%). The EC(50)concentration for InsP(3)-induced Ca(2+)release was 1.2 micro M for aorta while that for cerebellum was 0.3 micro M. Known agonists of the cerebellar InsP(3)receptor such as 3-deoxy InsP(3)and adenophostin A were also able to mobilize Ca(2+)from aorta microsomes. In addition, the competitive antagonist heparin and the non-competitive antagonists of the cerebellar InsP(3)receptor, tetracaine and tetrahexylammonium chloride, were also able to block InsP(3)-induced Ca(2+)release from aorta microsomes.
Collapse
Affiliation(s)
- S C Tovey
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | | | | | | | | | | | | |
Collapse
|
24
|
Longland CL, Dyer JL, Michelangeli F. The mycotoxin paxilline inhibits the cerebellar inositol 1,4, 5-trisphosphate receptor. Eur J Pharmacol 2000; 408:219-25. [PMID: 11090637 DOI: 10.1016/s0014-2999(00)00775-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Paxilline, a tremorgenic alkaloid mycotoxin produced by Penicillium paxilline, is a reversible inhibitor of the cerebellar inositol 1,4, 5-trisphophate (InsP(3)) receptor. It inhibits the amount or extent of InsP(3)-induced Ca(2+) release (IICR), at sub-maximal concentrations of InsP(3), in a biphasic manner consistent with two inhibition constants (K(i)'s 6.7 and > or =400 microM). As paxilline does not affect InsP(3) binding to the receptor, it can be considered a non-competitive inhibitor. The fact that IICR is biphasic has been interpreted as there being two populations of InsP(3)-sensitive Ca(2+) stores, which release Ca(2+) in either a fast or slow fashion. This study has shown that the rate constants for Ca(2+) release from both the fast and slow populations are reduced by paxilline (100 microM) by about 70% and 60%, respectively. Detailed analysis of the way different concentrations of paxilline inhibit the rate constants for Ca(2+) release indicates that the population of Ca(2+) stores that contribute to the slower phase of Ca(2+) release is more sensitive to the inhibitory action of paxilline.
Collapse
MESH Headings
- Animals
- Binding, Competitive
- Calcium/metabolism
- Calcium Channels/metabolism
- Cerebellum/drug effects
- Cerebellum/metabolism
- Dose-Response Relationship, Drug
- Indoles/pharmacology
- Inositol 1,4,5-Trisphosphate/metabolism
- Inositol 1,4,5-Trisphosphate/pharmacology
- Inositol 1,4,5-Trisphosphate Receptors
- Microsomes/drug effects
- Microsomes/metabolism
- Mycotoxins/pharmacology
- Rats
- Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
- Receptors, Cytoplasmic and Nuclear/metabolism
Collapse
Affiliation(s)
- C L Longland
- School of Biosciences, University of Birmingham, Edgbaston, B152TT, Birmingham, UK
| | | | | |
Collapse
|
25
|
Krause M, Pedarzani P. A protein phosphatase is involved in the cholinergic suppression of the Ca(2+)-activated K(+) current sI(AHP) in hippocampal pyramidal neurons. Neuropharmacology 2000; 39:1274-83. [PMID: 10760369 DOI: 10.1016/s0028-3908(99)00227-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The slow calcium-activated potassium current sI(AHP) underlies spike-frequency adaptation and has a substantial impact on the excitability of hippocampal CA1 pyramidal neurons. Among other neuromodulatory substances, sI(AHP) is modulated by acetylcholine acting via muscarinic receptors. The second-messenger systems mediating the suppression of sI(AHP) by muscarinic agonists are largely unknown. Both protein kinase C and A do not seem to be involved, whereas calcium calmodulin kinase II has been shown to take part in the muscarinic action on sI(AHP). We re-examined the mechanism of action of muscarinic agonists on sI(AHP) combining whole-cell recordings with the use of specific inhibitors or activators of putative constituents of the muscarinic pathway. Our results suggest that activation of muscarinic receptors reduces sI(AHP) in a G-protein-mediated and phospholipase C-independent manner. Furthermore, we obtained evidence for the involvement of the cGMP-cGK pathway and of a protein phosphatase in the cholinergic suppression of sI(AHP), whereas release of Ca(2+) from IP(3)-sensitive stores seems to be relevant neither for maintenance nor for modulation of sI(AHP).
Collapse
Affiliation(s)
- M Krause
- Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Str. 3, D-37075, Göttingen, Germany
| | | |
Collapse
|
26
|
Kirischuk S, Kirchhoff F, Matyash V, Kettenmann H, Verkhratsky A. Glutamate-triggered calcium signalling in mouse bergmann glial cells in situ: role of inositol-1,4,5-trisphosphate-mediated intracellular calcium release. Neuroscience 1999; 92:1051-9. [PMID: 10426545 DOI: 10.1016/s0306-4522(99)00067-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The mechanisms of glutamate-induced changes in intracellular free calcium concentration in Bergmann glial cells in mouse cerebellar slices were investigated by Fura-2-based microfluorimetry. Extracellular applications of glutamate, quisqualate and kainate triggered an increase in cytoplasmic calcium concentration, whereas N-methyl-D-aspartate and alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate were ineffective. The calcium elevation triggered by kainate was completely blocked by removal of calcium ions from the external solutions or by slice incubation with 6-cyano-7-nitroquinoxaline-2,3-dione. Conversely, both glutamate- and quisqualate-induced intracellular calcium transients were only slightly attenuated by slice incubation with either 6-cyano-7-nitroquinoxaline-2,3-dione or calcium-free solution, suggesting the intracellular origin for calcium ions. The glutamate-triggered cytosolic calcium increases were inhibited by slice incubation with thapsigargin, the inhibitor of intracellular calcium pumps, or by intracellular perfusion of Bergmann glial cells with heparin, the antagonist of inositol-1,4,5-trisphosphate-gated calcium release channels. Therefore the calcium release from inositol-1,4,5-trisphosphate-sensitive intracellular stores plays the major role in glutamate-induced calcium signalling. We concluded that Bergmann glial cells express calcium permeable ionotropic glutamate receptors, which might be important for generation of fast calcium signals. However, slow glutamate-evoked calcium signals are mostly determined by inositol-1,4,5-trisphosphate-dependent intracellular signalling chain.
Collapse
Affiliation(s)
- S Kirischuk
- Max-Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
| | | | | | | | | |
Collapse
|
27
|
Taylor CW. Inositol trisphosphate receptors: Ca2+-modulated intracellular Ca2+ channels. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1436:19-33. [PMID: 9838027 DOI: 10.1016/s0005-2760(98)00122-2] [Citation(s) in RCA: 117] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The three subtypes of inositol trisphosphate (InsP3) receptor expressed in mammalian cells are each capable of forming intracellular Ca2+ channels that are regulated by both InsP3 and cytosolic Ca2+. The InsP3 receptors of many, though perhaps not all, tissues are biphasically regulated by cytosolic Ca2+: a rapid stimulation of the receptors by modest increases in Ca2+ concentration is followed by a slower inhibition at higher Ca2+ concentrations. Despite the widespread occurrence of this form of regulation and the belief that it is an important element of the mechanisms responsible for the complex Ca2+ signals evoked by physiological stimuli, the underlying mechanisms are not understood. Both accessory proteins and Ca2+-binding sites on InsP3 receptors themselves have been proposed to mediate the effects of cytosolic Ca2+ on InsP3 receptor function, but the evidence is equivocal. The effects of cytosolic Ca2+ on InsP3 binding and channel opening, and the possible means whereby the effects are mediated are discussed in this review.
Collapse
Affiliation(s)
- C W Taylor
- Department of Pharmacology, Tennis Court Road, Cambridge CB2 1QJ, UK.
| |
Collapse
|
28
|
Mezna M, Michelangeli F. Effects of thimerosal on the transient kinetics of inositol 1,4,5-trisphosphate-induced Ca2+ release from cerebellar microsomes. Biochem J 1997; 325 ( Pt 1):177-82. [PMID: 9224644 PMCID: PMC1218543 DOI: 10.1042/bj3250177] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Thimerosal, a thiol-reactive reagent, has been shown to increase the cytosolic Ca2+ concentration in a variety of cells by sensitizing inositol 1,4,5-trisphosphate (InsP3) receptors. Thimerosal can have both sensitizing (at concentrations of <2 microM) and inhibitory (at concentrations of >2 microM) effects on InsP3-induced Ca2+ release (IICR) from cerebellar microsomes. Transient kinetic studies were performed by employing a fluorimetric stopped-flow approach using fluo-3. IICR was found to be a bi-exponential process with a fast and a slow component. At a maximal InsP3 concentration (20 microM), the fast phase had a rate constant of 0.9 s-1 and the slow phase had a rate constant of 0.4 s-1. The amplitudes of the two phases were 60% and 40% respectively. When the rate constants for the two phases were plotted as Hill plots, the processes were found to be non-co-operative in both cases (Hill coefficient of 1.0), thus arguing for a simple mechanism linking InsP3 binding to channel opening. At a submaximal InsP3 concentration (0.2 microM), where the sensitizing effects of thimerosal are most pronounced, thimerosal increased the rate constants of both phases in a sigmoidal fashion, with a Hill coefficient of 4.0, suggesting that several cysteine residues (up to four) need to be modified in order for maximum sensitization to occur. The rate constants remained elevated even at thimerosal concentrations that inhibited IICR. The amplitude or extent of Ca2+ release was, however, elevated to a much greater extent in the slow phase, suggesting that the two phases respond differently. At maximal InsP3 concentrations, thimerosal has no effect upon the rate constants but inhibits the amplitude of Ca2+ release.
Collapse
Affiliation(s)
- M Mezna
- School of Biochemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
| | | |
Collapse
|
29
|
Genazzani AA, Mezna M, Dickey DM, Michelangeli F, Walseth TF, Galione A. Pharmacological properties of the Ca2+-release mechanism sensitive to NAADP in the sea urchin egg. Br J Pharmacol 1997; 121:1489-95. [PMID: 9257932 PMCID: PMC1564845 DOI: 10.1038/sj.bjp.0701295] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
1. The sea urchin egg homogenate is an ideal model to characterize Ca2+-release mechanisms because of its reliability and high signal-to-noise-ratio. Apart from the InsP3- and ryanodine-sensitive Ca2+-release mechanisms, it has been recently demonstrated that this model is responsive to a third independent mechanism, that has the pyridine nucleotide, nicotinic acid adenine dinucleotide phosphate (NAADP), as an endogenous agonist. 2. The sea urchin egg homogenate was used to characterize the pharmacological and biochemical characteristics of the novel Ca2+-releasing agent, NAADP, compared to inositol trisphosphate (InsP3) and cyclic ADP ribose (cyclic ADPR), an endogenous activator of ryanodine receptors. 3. NAADP-induced Ca2+-release was blocked by L-type Ca2+-channel blockers and by Bay K 8644, while InsP3- and cyclic ADPR-induced Ca2+-release were insensitive to these agents. L-type Ca2+-channel blockers did not displace [32P]-NAADP binding, suggesting that their binding site was different. Moreover, stopped-flow kinetic studies revealed that these agents blocked NAADP in a all-or-none fashion. 4. Similarly, a number of K+-channel antagonists blocked NAADP-induced Ca2+-release selectively over InsP3- and cyclic ADPR-induced Ca2+-release. Radioligand studies showed that these agents were not competitive antagonists. 5. As has been shown for InsP3 and ryanodine receptors, NAADP receptors were sensitive to calmodulin antagonists, suggesting that this protein could be a common regulatory feature of intracellular Ca2+-release mechanisms. 6. The presence of K+ was not essential for NAADP-induced Ca2+-release, since substitution of K+ with other monovalent cations in the experimental media did not significantly alter Ca2+ release by NAADP. On the contrary, cyclic ADPR and InsP3-sensitive mechanisms were affected profoundly, although to a different extent depending on the monovalent cation which substituted for K+. Similarly, modifications of the pH in the experimental media from 7.2 to 6.7 or 8.0 only slightly affected NAADP-induced Ca2+-release. While the alkaline condition permitted InsP3 and cyclic ADPR-induced Ca2+-release, the acidic condition completely hampered both Ca2+-release mechanisms. 7. The present results characterize pharmacologically and biochemically the novel Ca2+-release mechanism sensitive to NAADP. Such characterization will help future research aimed at understanding the role of NAADP in mammalian systems.
Collapse
|
30
|
Svichar N, Shmigol A, Verkhratsky A, Kostyuk P. InsP3-induced Ca2+ release in dorsal root ganglion neurones. Neurosci Lett 1997; 227:107-10. [PMID: 9180215 DOI: 10.1016/s0304-3940(97)00313-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The intracellular calcium signalling was studied on subpopulation of freshly isolated adult mouse dorsal root ganglia (DRG) neurones with large somatas (30-45 microns in diameter). The cytoplasmic Ca2+ concentration ([Ca2+]i) was measured using indo-1 based microfluorimetry. The extracellular application of ATP (100 microM) triggered both inward current and [Ca2+]i elevation. Removal of extracellular Ca2+ had no effect on both ATP-induced current and [Ca2+]i transient. The ATP-induced Ca2+ elevation was inhibited by intracellular perfusion of DRG neurones with 20 microM heparin, or by cells incubation with thapsigargin or ryanodine. We conclude that mouse proprioceptive sensory neurones are endowed with Ca2+-impermeable ionotropic P2X purinoreceptors and metabotropic P2Y purinoreceptors, which, by means of phospholipase C-driven inositol-trisphosphate (InsP3) production, trigger the InsP3-induced Ca2+ release from intracellular stores.
Collapse
Affiliation(s)
- N Svichar
- Bogomoletz Institute of Physiology and International Center of Molecular Physiology, Kiev, Ukraine
| | | | | | | |
Collapse
|
31
|
Tovey SC, Godfrey RE, Hughes PJ, Mezna M, Minchin SD, Mikoshiba K, Michelangeli F. Identification and characterization of inositol 1,4,5-trisphosphate receptors in rat testis. Cell Calcium 1997; 21:311-9. [PMID: 9160167 DOI: 10.1016/s0143-4160(97)90119-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
PCR analysis and immunoblotting with isoform specific antibodies was used to identify the presence of type I, II and III inositol 1,4,5-trisphosphate receptors (InsP3Rs) in rat testis. PCR analysis also revealed that rat testis express both forms of the S1 splice variant (S1+ and S1-), but only the S2- from of the S2 splice variant of the type I InsP3 receptor. PCR analysis was also used to identify InsP3R isoform expression at a cellular level using myoid, Sertoli and germ cells derived from the testis of Wistar rats. The extent of [3H]-InsP3 binding was found to be 9 times lower for testicular microsomes than for cerebellar microsomes, with a Bmax of 1.4 pmoles/mg protein compared to 12.5 pmoles/mg protein for cerebellar microsomes. The Kd for InsP3 binding to its receptor in testicular microsomes was 60 +/- 10 nM which was similar to that found for cerebellar microsomes (80 +/- 20 nM). InsP3-induced Ca2+ release (IICR) in testicular microsomes was found to have an EC50 (concentration which causes a half-maximal response) of 0.5 +/- 0.03 microM, also similar to that seen for cerebellar microsomes (0.3 microM). Maximal IICR occurred at about 20 microM InsP3, with up to 4% of total intracellular Ca2+ stores being mobilized as compared to between 10-30% for cerebellar microsomes. Time resolved IICR using stopped-flow spectrofluorimetry, showed the kinetics of IICR for this testis preparation to be monophasic with a maximum rate constant of 0.15 s-1 at 30 microM InsP3. The rate constants are 7 times slower than values for cerebellar microsomes under similar conditions (approximately 1 s-1) and taken together with the binding data support the proposal that the receptor density/Ca2+ store is approximately 8 times lower than seen in cerebellar microsomal vesicles. The pharmacological properties as assessed using heparin and InsP3 analogues also confirmed similar behaviour for testicular InsP3Rs and cerebellar InsP3Rs.
Collapse
Affiliation(s)
- S C Tovey
- School of Biochemistry, University of Birmingham, UK
| | | | | | | | | | | | | |
Collapse
|
32
|
Mezna M, Michelangeli F. The effects of inositol 1,4,5-trisphosphate (InsP3) analogues on the transient kinetics of Ca2+ release from cerebellar microsomes. InsP3 analogues act as partial agonists. J Biol Chem 1996; 271:31818-23. [PMID: 8943223 DOI: 10.1074/jbc.271.50.31818] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
An investigation of the effects of a number of inositol trisphosphate analogues on the transient kinetics of Ca2+ release from cerebellar microsomes was undertaken. All the analogues investigated could release the total Ca2+ content of the inositol 1, 4,5-trisphosphate (Ins(1,4,5)P3) mobilizable Ca2+ store; however, their potencies were substantially reduced compared to Ins(1,4,5)P3. The concentration required to induce half-maximal Ca2+ mobilization was 0.14 microM for Ins(1,4,5)P3, 1.8 microM for 3-deoxyinositol 1,4, 5-trisphosphate (3-deoxyInsP3), 1.0 microM for 2,3-dideoxyinositol 1, 4,5-trisphosphate (2,3-dideoxyInsP3), 24 microM for 2,3, 6-trideoxyinositol 1,4,5-trisphopshate (2,3,6-trideoxyInsP3), and 2.9 microM for inositol 2,4,5-trisphosphate (Ins(2,4,5)P3). In all cases and for all concentrations tested, the inositol trisphosphate analogues induced biphasic transient release of Ca2+, which could fit to a biexponential equation assuming two independent processes. The rate constants calculated for the release process were much larger for Ins(1,4,5)P3 than the other inositol trisphosphates (the fast phase rate constant varying from 0.3 to 1.6 s-1 and the slow phase from 0.01-0.5 s-1, at concentrations between 0.03 and 20 microM Ins(1,4,5)P3). The rate constants for all other inositol trisphosphates did not appear to exceed 0.4 s-1 for the fast phase and 0.1 s-1 for the slow phase at their highest concentrations tested. The maximum amplitudes for Ca2+ release by the two phases appeared to be similar for all inositol trisphosphates (approximately 45% for the fast phase and approximately 55% for the slow phase). On comparing the rate constants for Ca2+ release at inositol trisphosphate concentrations for the analogues which all induced the same extent of Ca2+ release, it was apparent that the rates of release were independent of the extent of Ca2+ release. As the extent of Ca2+ release can be related to degree of occupancy of the binding sites, it is evident that different analogues which occupy the binding site of the receptor to the same extent can induce Ca2+ to be released at different rates. We explain this conclusion in terms of partial agonism where inositol phosphates can induce two (or more) occupied states of the channel.
Collapse
Affiliation(s)
- M Mezna
- School of Biochemistry, University of Birmingham, Edgbaston B15 2TT, United Kingdom.
| | | |
Collapse
|
33
|
Mezna M, Patchick T, Tovey S, Michelangeli F. Inhibition of the cerebellar inositol 1,4,5-trisphosphate-sensitive Ca2+ channel by ethanol and other aliphatic alcohols. Biochem J 1996; 314 ( Pt 1):175-9. [PMID: 8660280 PMCID: PMC1217022 DOI: 10.1042/bj3140175] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The effects of ethanol and other aliphatic alcohols on the endoplasmic reticulum Ca2+ pump and the inositol 1,4,5-trisphosphate (InsP3)-sensitive Ca2+ channel were studied in pig cerebellar microsomes. Methanol, ethanol and propanol all stimulated ATP-dependent Ca2+ uptake, whereas butanol inhibited this process. Ethanol inhibited InsP3-induced Ca2+ release [half-maximal inhibition at 3.5%, v/v (600 mM)]. However, ethanol affected only the amount of InsP3-releasable Ca2+, without affecting the concentration of InsP3 required to induce half-maximal release. Other alcohols of longer chain length were more potent than ethanol at inhibiting InsP3-induced Ca2+ release, but none of the alcohols tested affected [3H]InsP3 binding to its receptor. Using stopped-flow techniques, measurements of the rate of InsP3-induced Ca2+ release in the preparation of pig cerebellar microsomes used in this study showed the kinetics to be monophasic, with a rate constant of 0.93s-1 at 20 microM InsP3. This rate constant was dependent upon InsP3 concentration, decreasing to 0.38s-1 at 0.25 microM InsP3. Ethanol was shown to reduce the fractional amount of InsP3-induced Ca2+ release without significantly affecting the rate constant for this process.
Collapse
Affiliation(s)
- M Mezna
- School of Biochemistry, University of Birmingham, Edgbaston, U.K
| | | | | | | |
Collapse
|
34
|
Mezna M, Michelangeli F. Alkali metal ion dependence of inositol 1,4,5-trisphosphate-induced calcium release from rat cerebellar microsomes. J Biol Chem 1995; 270:28097-102. [PMID: 7499297 DOI: 10.1074/jbc.270.47.28097] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The effects of the alkali metal ions Na+, K+, Rb+, and Cs+ on ATP-dependent Ca2+ uptake, [3H]Inositol 1,4,5-trisphosphate (InsP3) binding, and quantal InsP3-induced Ca2+ release were investigated using rat cerebellar microsomes. Both the ion species and concentration affected the ability of the microsomes to support Ca2+ uptake with K+ being mot effective (3.8 nmol of Ca2+/min/mg at 100 mM K+). The order of efficacy of the other ions was as follows: K+ > Na+ > Rb+ = Cs+ >> Li+. The binding of [3H]InsP3 to cerebellar microsomes was, however, affected little by the presence of these ions. All these alkali metal ions (except Li+) supported InsP3-induced Ca2+ release at concentrations above 25 mM; however, the extent of Ca2+ release (expressed as a percent Ca2+ release compared with that released by the ionophore A23187) was dependent upon the ion species present. Again K+ was more potent than the other ions at facilitating InsP3-induced Ca2+ release (order of efficacy: K+ > Rb+ > Na+ > Cs+), although the concentration of InsP3 required to induce half-maximal Ca2+ release (IC50) was not significantly altered. Over the ion concentration range tested (25-100 mM), the extent of InsP3-induced Ca2+ release with both K+ and Rb+ increased in a linear fashion, while Na+ showed only a slight increase and Cs+ showed no increase over this range. The effect of K+ concentration on quantal Ca2+ release was to alter the extent of release rather than the IC50 InsP3 concentration. Using stopped-flow techniques, the effects of InsP3 and K+ concentrations on the kinetics of InsP3-induced Ca2+ release were shown to exhibit a monoexponential process in this microsomal preparation. The rate constants for Ca2+ release increased with InsP3 concentration (0.11 s-1 at 0.02 microM InsP3 to 0.5 s-1 at 40 microM InsP3); however, the relationship between the fractional extent of release and rate constants for release did not change in a similar way with InsP3 concentration. Although the fractional extent of Ca2+ release increased with K+ concentration, the rate constants for release over this K+ concentration range were unaffected. This observation leads us to question the role of K+ as a counter ion required for Ca2+ release, and we therefore postulate a role for K+ (and the other alkali metal ions) as a "co-factor" required for channel opening.
Collapse
Affiliation(s)
- M Mezna
- School of Biochemistry, University of Birmingham, Edgbaston, United Kingdom
| | | |
Collapse
|