Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
1.
Paleoceanogr Paleoclimatol ; 34(7): 1057-1073, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31598586

ABSTRACT

Producing independent and accurate chronologies for marine sediments is a prerequisite to understand the sequence of millennial-scale events and reveal potential temporal offsets between marine and continental records, or between different marine records, possibly from different regions. The last 40 ky is a generally well-constrained period since radiocarbon (14C) can be used as an absolute dating tool. However, in the northern North Atlantic, calendar ages cannot be directly derived from 14C ages, due to temporal and spatial variations of surface reservoir ages. Alternatively, chronologies can be derived by aligning Greenland ice-core time series with marine surface records. Yet this approach suffers from the lack of clearly defined climatic events between 14.7 and 23.3 cal ky BP (hereafter ka), a crucial period encompassing Heinrich Stadial 1 and the onset of the last deglaciation. In this study, (i) we assess the benefits of 230Th normalization to refine the sedimentation history between surface temperature alignment tie points and (ii) revisit the chronologies of three North Atlantic marine records. Our study supports the contention that the marked increase in the Greenland Ca2+ record at 17.48 ka ± 0.21 ky (1σ) occurred within dating uncertainty of sea surface temperature cooling in the North Atlantic at the onset of Heinrich Stadial 1. This sharp feature might be useful for future chronostratigraphic alignments to remedy the lack of chronological constraint between 14.7 and 23.3 ka for North Atlantic marine records that are subject to large changes in 14C surface reservoir age.

2.
Cell Death Dis ; 4: e632, 2013 May 16.
Article in English | MEDLINE | ID: mdl-23681227

ABSTRACT

Disrupting inositol 1,4,5-trisphosphate (IP3) receptor (IP3R)/B-cell lymphoma 2 (Bcl-2) complexes using a cell-permeable peptide (stabilized TAT-fused IP3R-derived peptide (TAT-IDP(S))) that selectively targets the BH4 domain of Bcl-2 but not that of B-cell lymphoma 2-extra large (Bcl-Xl) potentiated pro-apoptotic Ca(2+) signaling in chronic lymphocytic leukemia cells. However, the molecular mechanisms rendering cancer cells but not normal cells particularly sensitive to disrupting IP3R/Bcl-2 complexes are poorly understood. Therefore, we studied the effect of TAT-IDP(S) in a more heterogeneous Bcl-2-dependent cancer model using a set of 'primed to death' diffuse large B-cell lymphoma (DL-BCL) cell lines containing elevated Bcl-2 levels. We discovered a large heterogeneity in the apoptotic responses of these cells to TAT-IDP(S) with SU-DHL-4 being most sensitive and OCI-LY-1 being most resistant. This sensitivity strongly correlated with the ability of TAT-IDP(S) to promote IP3R-mediated Ca(2+) release. Although total IP3R-expression levels were very similar among SU-DHL-4 and OCI-LY-1, we discovered that the IP3R2-protein level was the highest for SU-DHL-4 and the lowest for OCI-LY-1. Strikingly, TAT-IDP(S)-induced Ca(2+) rise and apoptosis in the different DL-BCL cell lines strongly correlated with their IP3R2-protein level, but not with IP3R1-, IP3R3- or total IP3R-expression levels. Inhibiting or knocking down IP3R2 activity in SU-DHL-4-reduced TAT-IDP(S)-induced apoptosis, which is compatible with its ability to dissociate Bcl-2 from IP3R2 and to promote IP3-induced pro-apoptotic Ca(2+) signaling. Thus, certain chronically activated B-cell lymphoma cells are addicted to high Bcl-2 levels for their survival not only to neutralize pro-apoptotic Bcl-2-family members but also to suppress IP3R hyperactivity. In particular, cancer cells expressing high levels of IP3R2 are addicted to IP3R/Bcl-2 complex formation and disruption of these complexes using peptide tools results in pro-apoptotic Ca(2+) signaling and cell death.


Subject(s)
Apoptosis/drug effects , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Peptides/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Calcium/metabolism , Cell Line, Tumor , Humans , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Inositol 1,4,5-Trisphosphate Receptors/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Peptides/chemistry , Protein Binding , Protein Isoforms/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/chemistry , RNA Interference , RNA, Small Interfering/metabolism
3.
Cell Mol Life Sci ; 70(15): 2697-712, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23076254

ABSTRACT

The cystic phenotype in autosomal dominant polycystic kidney disease is characterized by a profound dysfunction of many cellular signaling patterns, ultimately leading to an increase in both cell proliferation and apoptotic cell death. Disturbance of normal cellular Ca(2+) signaling seems to be a primary event and is clearly involved in many pathways that may lead to both types of cellular responses. In this review, we summarize the current knowledge about the molecular and functional interactions between polycystins and multiple components of the cellular Ca(2+)-signaling machinery. In addition, we discuss the relevant downstream responses of the changed Ca(2+) signaling that ultimately lead to increased proliferation and increased apoptosis as observed in many cystic cell types.


Subject(s)
Apoptosis/physiology , Calcium Signaling/physiology , Calcium/metabolism , Cell Proliferation , Models, Biological , Polycystic Kidney, Autosomal Dominant/physiopathology , TRPP Cation Channels/metabolism , Cyclic AMP/metabolism , Humans , Intracellular Membranes/metabolism , Polycystic Kidney, Autosomal Dominant/metabolism , Proto-Oncogene Proteins B-raf/metabolism , TOR Serine-Threonine Kinases/metabolism
5.
Cell Calcium ; 51(6): 452-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22456092

ABSTRACT

Autosomal dominant polycystic kidney disease is caused by loss-of-function mutations in the PKD1 or PKD2 genes encoding respectively polycystin-1 and polycystin-2. Polycystin-2 stimulates the inositol trisphosphate (IP(3)) receptor (IP(3)R), a Ca(2+)-release channel in the endoplasmic reticulum (ER). The effect of ER-located polycystin-1 is less clear. Polycystin-1 has been reported both to stimulate and to inhibit the IP(3)R. We now studied the effect of polycystin-1 and of polycystin-2 on the IP(3)R activity under conditions where the cytosolic Ca(2+) concentration was kept constant and the reuptake of released Ca(2+) was prevented. We also studied the interdependence of the interaction of polycystin-1 and polycystin-2 with the IP(3)R. The experiments were done in conditionally immortalized human proximal-tubule epithelial cells in which one or both polycystins were knocked down using lentiviral vectors containing miRNA-based short hairpins. The Ca(2+) release was induced in plasma membrane-permeabilized cells by various IP(3) concentrations at a fixed Ca(2+) concentration under unidirectional (45)Ca(2+)-efflux conditions. We now report that knock down of polycystin-1 or of polycystin-2 inhibited the IP(3)-induced Ca(2+) release. The simultaneous presence of the two polycystins was required to fully amplify the IP(3)-induced Ca(2+) release, since the presence of polycystin-1 alone or of polycystin-2 alone did not result in an increased Ca(2+) release. These novel findings indicate that ER-located polycystin-1 and polycystin-2 operate as a functional complex. They are compatible with the view that loss-of-function mutations in PKD1 and in PKD2 both cause autosomal dominant polycystic kidney disease.


Subject(s)
Calcium Signaling , Calcium/metabolism , Inositol 1,4,5-Trisphosphate/pharmacology , TRPP Cation Channels/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane Permeability , Cytosol/metabolism , Epithelium/metabolism , Epithelium/pathology , Feeder Cells , Gene Knockdown Techniques , Genetic Vectors/genetics , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Lentivirus/genetics , Mice , MicroRNAs/genetics , NIH 3T3 Cells , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Primary Cell Culture , Protein Interaction Mapping , TRPP Cation Channels/genetics
6.
Cell Death Differ ; 19(2): 295-309, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21818117

ABSTRACT

Antiapoptotic B-cell lymphoma 2 (Bcl-2) targets the inositol 1,4,5-trisphosphate receptor (IP(3)R) via its BH4 domain, thereby suppressing IP(3)R Ca(2+)-flux properties and protecting against Ca(2+)-dependent apoptosis. Here, we directly compared IP(3)R inhibition by BH4-Bcl-2 and BH4-Bcl-Xl. In contrast to BH4-Bcl-2, BH4-Bcl-Xl neither bound the modulatory domain of IP(3)R nor inhibited IP(3)-induced Ca(2+) release (IICR) in permeabilized and intact cells. We identified a critical residue in BH4-Bcl-2 (Lys17) not conserved in BH4-Bcl-Xl (Asp11). Changing Lys17 into Asp in BH4-Bcl-2 completely abolished its IP(3)R-binding and -inhibitory properties, whereas changing Asp11 into Lys in BH4-Bcl-Xl induced IP(3)R binding and inhibition. This difference in IP(3)R regulation between BH4-Bcl-2 and BH4-Bcl-Xl controls their antiapoptotic action. Although both BH4-Bcl-2 and BH4-Bcl-Xl had antiapoptotic activity, BH4-Bcl-2 was more potent than BH4-Bcl-Xl. The effect of BH4-Bcl-2, but not of BH4-Bcl-Xl, depended on its binding to IP(3)Rs. In agreement with the IP(3)R-binding properties, the antiapoptotic activity of BH4-Bcl-2 and BH4-Bcl-Xl was modulated by the Lys/Asp substitutions. Changing Lys17 into Asp in full-length Bcl-2 significantly decreased its binding to the IP(3)R, its ability to inhibit IICR and its protection against apoptotic stimuli. A single amino-acid difference between BH4-Bcl-2 and BH4-Bcl-Xl therefore underlies differential regulation of IP(3)Rs and Ca(2+)-driven apoptosis by these functional domains. Mutating this residue affects the function of Bcl-2 in Ca(2+) signaling and apoptosis.


Subject(s)
Apoptosis , Calcium Signaling , Calcium/metabolism , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-X Protein/metabolism , Amino Acid Sequence , Amino Acids/metabolism , Animals , Cytoprotection , Immobilized Proteins/metabolism , Mice , Molecular Sequence Data , Mutation/genetics , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/chemistry , Rats , Sequence Alignment , Structure-Activity Relationship , bcl-X Protein/chemistry
7.
Aliment Pharmacol Ther ; 34(7): 702-13, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21790682

ABSTRACT

BACKGROUND: Polycystic liver diseases (PCLD) represent a group of genetic disorders in which cysts occur solely in the liver, or together with renal cysts. Most of the patients with PCLD are asymptomatic, however, in some patients, expansion of liver cysts causes invalidating abdominal symptoms. AIM: To provide a systemic review on the pathophysiology and management of PCLD. METHODS: A PubMed search was undertaken to identify relevant literature using search terms including polycystic liver disease, pathophysiology, surgical and medical management. RESULTS: The most common complication in patients with PCLD is extensive hepatomegaly, which may lead to malnutrition and can be lethal. Conservative surgical approaches are only partially effective and do not change the natural course of the disease. Liver transplantation has been successfully performed in PCLD, however, in an era of organ shortage, medical management needs to be evaluated. A better understanding of the pathophysiology and the availability of animal models have already identified promising drugs. Abnormalities in cholangiocyte proliferation/apoptosis and enhanced fluid secretion are key factors in the pathophysiology. It has been demonstrated in rodents and in humans that somatostatin analogues diminish liver volume. The role of the inhibitors of the mammalian target of rapamycin (mTOR) in the management of PCLD is still under investigation. CONCLUSIONS: The exact pathophysiology of polycystic liver disease still remains unclear. In symptomatic patients, none of the currently available surgical options except liver transplantation have been shown to change the natural course of the disease. The use of somatostatin analogues has been shown to diminish liver volume.


Subject(s)
Cysts/physiopathology , Cysts/therapy , Liver Diseases/physiopathology , Liver Diseases/therapy , Animals , Cysts/genetics , Gene Expression Regulation , Humans , Liver Diseases/genetics , Liver Transplantation , Randomized Controlled Trials as Topic
8.
Subcell Biochem ; 45: 385-404, 2007.
Article in English | MEDLINE | ID: mdl-18193645

ABSTRACT

Secretory-pathway Ca2(+)-transport ATPases (SPCA) provide the Golgi apparatus with Ca2+ and Mn2+ needed for the normal functioning of this organelle. Loss of one functional copy of the human SPCA1 gene (ATP2C1) causes Hailey-Hailey disease, a rare skin disorder characterized by recurrent blisters and erosions in the flexural areas. Here, we will review the properties and functional role of the SPCAs. The relationship between Hailey-Hailey disease and its defective gene (ATP2C1) will be adressed as well.


Subject(s)
Calcium-Transporting ATPases/physiology , Golgi Apparatus/enzymology , Pemphigus, Benign Familial/physiopathology , Alternative Splicing , Animals , Calcium/physiology , Calcium Signaling , Calcium-Transporting ATPases/genetics , Female , Humans , Male , Manganese/physiology , Pemphigus, Benign Familial/genetics
9.
Cell Calcium ; 38(5): 489-95, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16122795

ABSTRACT

The Golgi apparatus is, like the endoplasmic reticulum, an inositol-1,4,5-trisphosphate-sensitive Ca2+ store, but its role in setting up Ca2+ signals is not well understood. We have now measured histamine-induced Ca2+ signals in HeLa cells pretreated with brefeldin A, a fungal metabolite that leads to the fragmentation and subsequent disappearance of the Golgi apparatus by its reabsorption within the endoplasmic reticulum. Ca2+ responses in which the free cytoplasmic Ca2+ concentration returned to resting levels during the histamine stimulation (mainly baseline Ca2+ oscillations or a single Ca2+ peak) occurred more often in brefeldin A pretreated cells, resulting in a lower Ca2+ plateau in population measurements. The latencies before the onset of the Ca2+ signals were longer after brefeldin A pretreatment. These results suggest that the integrity of the Golgi apparatus contributes to the shaping of intracellular Ca2+ signals.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Cytosol/physiology , Golgi Apparatus/physiology , Brefeldin A/pharmacology , Calcium Signaling/drug effects , Fluorescent Antibody Technique , Golgi Apparatus/drug effects , HeLa Cells/ultrastructure , Histamine/pharmacology , Humans , Reaction Time/drug effects
10.
Verh K Acad Geneeskd Belg ; 66(4): 277-303, 2004.
Article in Dutch | MEDLINE | ID: mdl-15553100

ABSTRACT

In this study, the FKBP12-binding properties of IP3Rs and RyRs were compared. Although the primary sequence of IP3Rs en RyRs contained a putative FKBP12-binding site, the functional, molecular and structural properties of these sites appeared to be completely different. For RyRs, FKBPs appear to function as associated proteins that are important for the functional regulation of the channel, thereby stabilizing the RyR complex. For IP3Rs, FKBPs might be involved in the de novo protein synthesis of the IP3Rs and the folding of the peptide chain to a functional IP3R protein, thereby functioning as helper enzymes. Hence, it is very unlikely that they function as associated regulatory proteins of the IP3R. In addition, we provided evidence that FKBP 12 is also an important regulating protein of the Ca(2+)-flux properties of the RyR3. FKBP12 clearly modulated both RyR3-mediated global and local Ca(2+)-responses.


Subject(s)
Calcium Channels/metabolism , Calcium/physiology , Immunophilins/metabolism , Amino Acid Sequence , Animals , Calcium/metabolism , Calcium Channels/chemistry , Humans , Inositol 1,4,5-Trisphosphate Receptors , Molecular Sequence Data , Mutation , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/metabolism , Sequence Alignment , Tacrolimus Binding Protein 1A/metabolism
11.
Cell Calcium ; 36(6): 479-87, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15488597

ABSTRACT

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


Subject(s)
Aequorin/biosynthesis , Aequorin/genetics , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Aequorin/metabolism , Dose-Response Relationship, Drug , Endoplasmic Reticulum/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Golgi Apparatus/drug effects , HeLa Cells , Humans , Inositol 1,4,5-Trisphosphate/pharmacology
12.
Cell Calcium ; 35(2): 115-21, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14706285

ABSTRACT

Non-mitochondrial intracellular Ca2+ stores contain both thapsigargin-sensitive sarco(endo)plasmic-reticulum Ca2+-ATPases (SERCA) and thapsigargin-insensitive secretory-pathway Ca2+-ATPases (SPCA1). We now have studied the Ca2+-release properties of the compartments associated with these pumps in intact, i.e. non-permeabilized, cells of different origin (HeLa, keratinocytes, 16HBE14o-, COS-1, A7r5) and with different approaches (45Ca2+ fluxes, Ca2+ imaging and measurements of the free luminal [Ca2+] in the endoplasmic-reticulum and the Golgi apparatus using targeted aequorin). Application of an extracellular agonist in the absence of thapsigargin induced in all cells a Ca2+ release from both the endoplasmic-reticulum and the Golgi apparatus. The agonists were not able to release Ca2+ in the presence of 10 microM thapsigargin, except in COS-1 cells overexpressing SPCA1, where this pump not only appeared in the Golgi compartment but also overflowed into the agonist-sensitive part of the endoplasmic-reticulum. We conclude that the subcompartments of the endoplasmic-reticulum and of the Golgi complex that endogenously express SPCA1 are insensitive to agonist stimulation.


Subject(s)
Calcium-Transporting ATPases/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Inositol 1,4,5-Trisphosphate/agonists , Inositol 1,4,5-Trisphosphate/biosynthesis , Thapsigargin/pharmacology , Animals , COS Cells , Calcium/metabolism , Cells, Cultured , Chlorocebus aethiops , Endoplasmic Reticulum/drug effects , Golgi Apparatus/drug effects , HeLa Cells , Humans , Male , Sarcoplasmic Reticulum Calcium-Transporting ATPases
13.
Biochem Biophys Res Commun ; 311(4): 1181-93, 2003 Nov 28.
Article in English | MEDLINE | ID: mdl-14623304

ABSTRACT

The Ca(2+)- and calmodulin-dependent phosphatase calcineurin was reported to interact with the inositol 1,4,5-trisphosphate receptor (IP(3)R) and the ryanodine receptor (RyR) and to modulate their phosphorylation status and activity. However, controversial data on the molecular mechanisms involved and on the functional relevance of calcineurin for these channel-complexes have been described. Hence, we will focus on the functional importance of calcineurin for IP(3)R and RyR function and on the different mechanisms by which Ca(2+)-dependent dephosphorylation can affect the gating of those intracellular Ca(2+)-release channels. Since many studies made use of immunosuppressive drugs that are inhibiting calcineurin activity, we will also have to take the different side effects of these drugs into account for the proper interpretation of the effects of calcineurin on intracellular Ca(2+)-release channels. In addition, it became recently known that various other phosphatases and kinases can associate with these channels, thereby forming macromolecular complexes. The relevance of these enzymes for IP(3)R and RyR functioning will be reviewed since in some cases they could interfere with the effects ascribed to calcineurin. Finally, we will discuss the downstream effects of calcineurin on the regulation of the expression levels of intracellular Ca(2+)-release channels as well as the relation between IP(3)R- and RyR-mediated Ca(2+) release and calcineurin-dependent gene expression.


Subject(s)
Calcineurin/metabolism , Calcium Channels/physiology , Calcium Signaling/physiology , Homeostasis/physiology , Intracellular Space/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Inositol 1,4,5-Trisphosphate Receptors , Macromolecular Substances , Phosphorylation
14.
Cell Calcium ; 34(2): 157-62, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12810057

ABSTRACT

Mutations in the ubiquitously expressed secretory-pathway Ca(2+)-ATPase (SPCA1) Ca(2+) pump result in Hailey-Hailey disease, which almost exclusively affects the epidermal part of the skin. We have studied Ca(2+) signaling in human keratinocytes by measuring the free Ca(2+) concentration in the cytoplasm and in the lumen of both the Golgi apparatus and the endoplasmic reticulum. These signals were compared with those recorded in SPCA1-overexpressing and control COS-1 cells. Both the sarco(endo)plasmic-reticulum Ca(2+)-ATPase (SERCA) and SPCA1 can mediate Ca(2+) uptake into the Golgi stacks. Our results indicate that keratinocytes mainly used the SPCA1 Ca(2+) pump to load the Golgi complex with Ca(2+) whereas the SERCA Ca(2+) pump was mainly used in control COS-1 cells. Cytosolic Ca(2+) signals in keratinocytes induced by extracellular ATP or capacitative Ca(2+) entry were characterized by an unusually long latency reflecting extra Ca(2+) buffering by an SPCA1-containing Ca(2+) store, similarly as in SPCA1-overexpressing COS-1 cells. Removal of extracellular Ca(2+) elicited spontaneous cytosolic Ca(2+) transients in keratinocytes, similarly as in SPCA1-overexpressing COS-1 cells. With respect to Ca(2+) signaling keratinocytes and SPCA1-overexpressing COS-1 cells therefore behaved similarly but differed from control COS-1 cells. The relatively large contribution of the SPCA1 pumps for loading the Golgi stores with Ca(2+) in keratinocytes may, at least partially, explain why mutations in the SPCA1 gene preferentially affect the skin in Hailey-Hailey patients.


Subject(s)
Calcium Signaling , Calcium-Transporting ATPases/metabolism , Calcium/metabolism , Keratinocytes/metabolism , Animals , COS Cells , Calcium-Transporting ATPases/genetics , Cells, Cultured , Cytoplasm/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Humans , Transfection
15.
Pflugers Arch ; 445(6): 629-42, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12632182

ABSTRACT

In almost all cells, cytosolic Ca(2+) is a crucial intracellular messenger, regulating many cellular processes. In non-excitable as well as in some excitable cells, Ca(2+) release from the intracellular stores into the cytoplasm is primarily initiated by the second messenger inositol 1,4,5-trisphosphate (IP(3)), which interacts with the IP(3) receptor (IP(3)R), a tetrameric intracellular Ca(2+)-release channel. This review focuses on the pharmacological modulation of the various functionally important sub-domains of the IP(3)R, including the IP(3)-binding domain, calmodulin-binding sites, adenine nucleotide-binding sites and the sites for interaction for FK506-binding proteins and other regulators. We will particularly focus on the pharmacological tools that interfere with these domains and discuss their relative specificity for the IP(3)R, thereby indicating their potential usefulness for unraveling the complex functional regulation of the IP(3)R.


Subject(s)
Calcium Channels , Calcium Signaling/physiology , Receptors, Cytoplasmic and Nuclear , Animals , Calcium Channels/physiology , Humans , Inositol 1,4,5-Trisphosphate Receptors , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/physiology
16.
Cell Calcium ; 32(2): 71-81, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12161107

ABSTRACT

Human neuroblastoma SH-SY5Y cells, predominantly expressing type 1 inositol 1,4,5-trisphosphate (IP(3)) receptor (IP(3)R), were stably transfected with IP(3)R type 3 (IP(3)R3) cDNA. Immunocytochemistry experiments showed a homogeneous cytoplasmic distribution of type 3 IP(3)Rs in transfected and selected high expression cloned cells. Using confocal Ca(2+) imaging, carbachol (CCh)-induced Ca(2+) release signals were studied. Low CCh concentrations (< or = 750 nM) evoked baseline Ca(2+) oscillations. Transfected cells displayed a higher CCh responsiveness than control or cloned cells. Ca(2+) responses varied between fast, large Ca(2+) spikes and slow, small Ca(2+) humps, while in the clone only Ca(2+) humps were observed. Ca(2+) humps in the transfected cells were associated with a high expression level of IP(3)R3. At high CCh concentrations (10 microM) Ca(2+) transients in transfected and cloned cells were similar to those in control cells. In the clone exogenous IP(3)R3 lacked the C-terminal channel domain but IP(3)-binding capacity was preserved. Transfected cells mainly expressed intact type 3 IP(3)Rs but some protein degradation was also observed. We conclude that in transfected cells expression of functional type 3 IP(3)Rs causes an apparent higher affinity for IP(3). In the clone, the presence of degraded receptors leads to an efficient cellular IP(3) buffer and attenuated IP(3)-evoked Ca(2+) release.


Subject(s)
Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Signaling/genetics , Calcium/metabolism , Eukaryotic Cells/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Binding Sites/drug effects , Binding Sites/genetics , Calcium Signaling/drug effects , Carbachol/pharmacology , Cell Division/genetics , Cholinergic Agonists/pharmacology , Clone Cells/cytology , Clone Cells/drug effects , Clone Cells/metabolism , Eukaryotic Cells/cytology , Eukaryotic Cells/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Immunohistochemistry , Inositol 1,4,5-Trisphosphate Receptors , Neuroblastoma , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transfection , Tumor Cells, Cultured
17.
Cell Calcium ; 31(5): 229-33, 2002 May.
Article in English | MEDLINE | ID: mdl-12098225

ABSTRACT

We have investigated how the immunosuppressant drug FK506 affected the basal Ca(2+) leak in permeabilized A7r5 cells. Non-mitochondrial Ca(2+) stores loaded to steady state with Ca(2+) slowly lost their accumulated Ca(2+) during incubation in a Ca(2+)-free efflux medium. FK506 up to 100 microM had no effect on the basal Ca(2+) leak. In contrast, the rate of Ca(2+) release proceeded much faster immediately after washing out FK506. The increase in rate of Ca(2+) release after washing out of this compound depended on both its initial concentration and on the time of pre-incubation. A similar effect was also observed after removing another immunosuppressant drug (rapamycin) and after removing the inositol 1,4,5-trisphosphate receptor inhibitor xestospongin C. Since all these substances have a high octanol/H(2)O partition coefficient and accumulate in the endoplasmic reticulum membrane, we suggest that the transient increase in the basal Ca(2+) leak is due to the sudden removal of these lipophilic substances from the membrane.


Subject(s)
Calcium/metabolism , Cell Membrane Permeability/drug effects , Endoplasmic Reticulum/drug effects , Immunosuppressive Agents/pharmacology , Intracellular Membranes/drug effects , Membrane Lipids/metabolism , Tacrolimus/pharmacology , Animals , Aorta , Cell Membrane Permeability/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Macrocyclic Compounds , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Oxazoles/pharmacology , Rats , Sirolimus/pharmacology , Up-Regulation/drug effects , Up-Regulation/physiology
18.
Neuropharmacology ; 42(5): 706-13, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11985829

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by selective motor neuron death. The exact mechanism responsible for this selectivity is not clear, although it is known that motor neurons are very sensitive to excitotoxicity. This high sensitivity is due to a high density of alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors on their surface and to a limited Ca(2+) buffering capacity. Ca(2+) can enter the cell upon stimulation through voltage-operated Ca(2+) channels and through the Ca(2+)-permeable portion of AMPA receptors. How this Ca(2+) kills motor neurons is incompletely understood. In the present study, we report that kainate (KA)-induced motor neuron death is purely mediated through Ca(2+) entering motor neurons through Ca(2+)-permeable AMPA receptors and that voltage-operated Ca(2+) channels play no significant role. In contrast to what has been observed in other neuronal models or after N-methyl-D-aspartate stimulation, NO synthase inhibition and a number of antioxidants did not protect motor neurons from KA-induced death. Only PD150606, derived from alpha-mercaptoacrylic acid and considered as a selective calpain antagonist, inhibited dose-dependently the KA-induced motor neuron death. However, other calmodulin and calpain inhibitors were not effective. At least part of the inhibitory effect of PD150606 is due to an irreversible inhibition of the Ca(2+) influx through the Ca(2+)-permeable AMPA receptor. These results demonstrate the interesting property of PD150606 to interfere with excitotoxicity-dependent motor neuron death and show that PD150606 is not an exclusive calpain/calmodulin antagonist.


Subject(s)
Acrylates/pharmacology , Calcium/metabolism , Calpain/metabolism , Kainic Acid/antagonists & inhibitors , Motor Neurons/drug effects , Neural Inhibition/drug effects , Animals , Calcium/antagonists & inhibitors , Calmodulin/antagonists & inhibitors , Calmodulin/metabolism , Calpain/antagonists & inhibitors , Cell Death/drug effects , Cell Death/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Kainic Acid/pharmacology , Motor Neurons/cytology , Motor Neurons/physiology , Neural Inhibition/physiology , Rats , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/metabolism
19.
Exp Neurol ; 174(2): 150-61, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11922657

ABSTRACT

The mechanism responsible for the selective vulnerability of motor neurons in amyotrophic lateral sclerosis (ALS) is poorly understood. Several lines of evidence indicate that susceptibility of motor neurons to Ca(2+) overload induced by excitotoxic stimuli is involved. In this study, we investigated whether the high density of Ca(2+)-permeable AMPA receptors on motor neurons gives rise to higher Ca(2+) transients in motor neurons compared to dorsal horn neurons. Dorsal horn neurons were chosen as controls as these cells do not degenerate in ALS. In cultured spinal motor neurons, the rise of the cytosolic Ca(2+) concentration induced by kainic acid (KA) and mediated by the AMPA receptor was almost twice as high as in spinal neurons from the dorsal horn. Furthermore, we investigated whether increasing the motor neuron's cytosolic Ca(2+)-buffering capacity protects them from excitotoxic death. To obtain motor neurons with increased Ca(2+) buffering capacity, we generated transgenic mice overexpressing parvalbumin (PV). These mice have no apparent phenotype. PV overexpression was present in the central nervous system, kidney, thymus, and spleen. Motor neurons from these transgenic mice expressed PV in culture and were partially protected from KA-induced death as compared to those isolated from nontransgenic littermates. PV overexpression also attenuated KA-induced Ca(2+) transients, but not those induced by depolarization. We conclude that the high density of Ca(2+)-permeable AMPA receptors on the motor neuron's surface results in high Ca(2+) transients upon stimulation and that the low cytosolic Ca(2+)-buffering capacity of motor neurons may contribute to the selective vulnerability of these cells in ALS. Overexpression of a high-affinity Ca(2+) buffer such as PV protects the motor neuron from excitotoxicity and this protective effect depends upon the mode of Ca(2+) entry into the cell.


Subject(s)
Excitatory Amino Acid Agonists/toxicity , Motor Neurons/drug effects , Neurotoxins/toxicity , Parvalbumins/pharmacology , Amyotrophic Lateral Sclerosis/etiology , Animals , Blotting, Western , Calcium/metabolism , Calcium Signaling/drug effects , Cell Death/drug effects , Cells, Cultured , Cytoprotection/drug effects , Cytoprotection/genetics , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Kainic Acid/toxicity , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/cytology , Motor Neurons/metabolism , Organ Specificity , Parvalbumins/genetics , Parvalbumins/metabolism , Phenotype , Posterior Horn Cells/cytology , Posterior Horn Cells/drug effects , Posterior Horn Cells/metabolism , Receptors, AMPA/metabolism
20.
Neurogastroenterol Motil ; 14(1): 63-73, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11874555

ABSTRACT

To characterize further the Ca2+ signalling mechanisms of myenteric neurones, we studied the effect of thapsigargin, a blocker of the Ca2+-store ATPase, and the mechanisms involved in restoring the intracellular Ca2+ concentration ([Ca2+]i) after activation. Thapsigargin (5 x 10(-6) mol L(-1)) induced an oscillatory [Ca2+]i response in 86.6% of the neurones (n=276), which was blocked by the removal of extracellular Ca2+ and by omega-conotoxin MVIIA (5 x 10(-7) mol L(-1)). The IP3-blocker, 2-aminoethyl-diphenyl-borate (75 x 10(-6) mol L(-1)), blocked or reduced the responses in 74.5% of the neurones. The oscillatory responses induced by the depletion of Ca2+ stores suggest that myenteric neurones might recruite N-type Ca2+ channels as a refill mechanism. Thapsigargin pretreatment increased the amplitude, the upstroke and duration of the K+-induced [Ca2+]i responses. Mitochondrial blockers (rotenone and antimycin/oligomycin) also prolonged the responses, but without affecting the amplitude. Furthermore, it was found that for high [Ca2+]i, the thapsigargin-sensitive Ca2+ uptake was crucial, while mitochondrial blockade affected the Ca2+ uptake over a wide range of concentrations. The Ca2+-sequestering components might also have been compensating for each other, as most drugs only delayed and not inhibited Ca2+ removal.


Subject(s)
Calcium Signaling/physiology , Myenteric Plexus/physiology , Neurons/physiology , Animals , Calcium/metabolism , Calcium/physiology , Calcium Channels/physiology , Calcium Signaling/drug effects , Cells, Cultured , Cytosol/chemistry , Cytosol/drug effects , Cytosol/metabolism , Cytosol/physiology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/physiology , Enzyme Inhibitors/pharmacology , Guinea Pigs , Intestine, Small/cytology , Intestine, Small/drug effects , Intestine, Small/physiology , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Intracellular Fluid/physiology , Microtubules/drug effects , Microtubules/metabolism , Microtubules/physiology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Myenteric Plexus/cytology , Myenteric Plexus/drug effects , Neurons/cytology , Neurons/drug effects , Thapsigargin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...