Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 537
1.
Biochem J ; 478(4): 895-910, 2021 02 26.
Article En | MEDLINE | ID: mdl-33635336

Thirty-six years after the publication of the important article by Busa and Nuccitelli on the variability of intracellular pH (pHi) and the interdependence of pHi and intracellular Ca2+ concentration ([Ca2+]i), little research has been carried out on pHi and calcium signaling. Moreover, the results appear to be contradictory. Some authors claim that the increase in [Ca2+]i is due to a reduction in pHi, others that it is caused by an increase in pHi. The reasons for these conflicting results have not yet been discussed and clarified in an exhaustive manner. The idea that variations in pHi are insignificant, because cellular buffers quickly stabilize the pHi, may be a limiting and fundamentally wrong concept. In fact, it has been shown that protons can move and react in the cell before they are neutralized. Variations in pHi have a remarkable impact on [Ca2+]i and hence on some of the basic biochemical mechanisms of calcium signaling. This paper focuses on the possible triggering role of protons during their short cellular cycle and it suggests a new hypothesis for an IP3 proton dependent mechanism of action.


Calcium Signaling/physiology , Protons , Animals , Calcium/chemistry , Feedback, Physiological , Humans , Hydrogen/chemistry , Hydrogen-Ion Concentration , Inositol 1,4,5-Trisphosphate/physiology , Inositol Polyphosphate 5-Phosphatases/physiology , Models, Chemical , Phospholipases/physiology , Second Messenger Systems/physiology
2.
Biochim Biophys Acta Biomembr ; 1862(6): 183240, 2020 06 01.
Article En | MEDLINE | ID: mdl-32119862

The IP3 receptor binding protein released with inositol 1,4,5-trisphosphate (IRBIT) plays important roles in the regulation of intracellular Ca2+ signaling and intracellular pH. The mammals express two IRBIT paralogs, i.e., IRBIT1 (encoded by AHCYL1) and IRBIT2 (encoded by AHCYL2). The clawed frog Xenopus laevis oocyte is widely used for biophysical studies on ion channels and transporters. It remains unknown whether endogenous IRBIT is expressed in Xenopus oocytes. Here, we cloned from frog oocyte irbit2.L and irbit2.S, orthologs of mammalian IRBIT2. When over-expressed, the frog IRBITs powerfully stimulate the electrogenic Na+/HCO3- cotransporter NBCe1-B as mouse IRBIT2-V2 does. Expression of an isolated Nt fragment of NBCe1-B containing the IRBIT-binding domain greatly decreases NBCe1-B activity in oocytes, suggesting that the basal activity of NBCe1-B contains a large component derived from the stimulation by endogenous frog IRBIT. The frog IRBITs are highly homologous to the mammalian ones in the carboxyl-terminal region, but varies greatly in the amino-terminal (Nt) appendage. Interestingly, truncation study showed that the Nt appendage of IRBIT1 and the long Nt appendage of IRBIT2-V2 modestly enhance, whereas the short Nt appendage of IRBIT2-V4 greatly inhibits the functional interaction between IRBIT and NBCe1-B. Finally, Ala-substitution of Ser68, a key phosphorylation site in the PEST domain of IRBIT, causes distinct functional consequences depending on the structural context of the Nt appendage in different IRBIT isoforms. We conclude that the Nt appendage of IRBITs is not necessary for, but plays an important regulatory role in the functional interaction between IRBIT and NBCe1-B.


Inositol 1,4,5-Trisphosphate/physiology , Sodium-Bicarbonate Symporters/metabolism , Animals , Inositol 1,4,5-Trisphosphate/metabolism , Mice , Peptide Fragments/pharmacology , Protein Binding/drug effects , Sodium-Bicarbonate Symporters/drug effects , Xenopus Proteins/metabolism , Xenopus laevis
3.
Annu Rev Physiol ; 82: 151-176, 2020 02 10.
Article En | MEDLINE | ID: mdl-31730387

In the body, extracellular stimuli produce inositol 1,4,5-trisphosphate (IP3), an intracellular chemical signal that binds to the IP3 receptor (IP3R) to release calcium ions (Ca2+) from the endoplasmic reticulum. In the past 40 years, the wide-ranging functions mediated by IP3R and its genetic defects causing a variety of disorders have been unveiled. Recent cryo-electron microscopy and X-ray crystallography have resolved IP3R structures and begun to integrate with concurrent functional studies, which can explicate IP3-dependent opening of Ca2+-conducting gates placed ∼90 Šaway from IP3-binding sites and its regulation by Ca2+. This review highlights recent research progress on the IP3R structure and function. We also propose how protein plasticity within IP3R, which involves allosteric gating and assembly transformations accompanied by rapid and chronic structural changes, would enable it to regulate diverse functions at cellular microdomains in pathophysiological states.


Inositol 1,4,5-Trisphosphate Receptors/physiology , Allosteric Regulation , Animals , Calcium Channels/metabolism , Calcium Signaling , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate/physiology , Inositol 1,4,5-Trisphosphate Receptors/genetics
4.
J Diabetes Res ; 2019: 7602427, 2019.
Article En | MEDLINE | ID: mdl-31950065

BACKGROUND AND PURPOSE: Metformin, a widely used antidiabetic drug, has been shown to have anti-inflammatory properties; nevertheless, its influence on ß-cell meta-inflammation remains unclear. The following study investigated the effects of metformin on meta-inflammatory in ß-cells and whether the underlying mechanisms were associated with the G protein-coupled receptor 40-phospholipase C-inositol 1, 4, 5-trisphosphate (GPR40-PLC-IP3) pathway. MATERIALS AND METHODS: Lipotoxicity-induced ß-cells and the high-fat diet-induced obese rat model were used in the study. RESULTS: Metformin-reduced lipotoxicity-induced ß-cell meta-inflammatory injury was associated with the expression of GPR40. GPR40 was involved in metformin reversing metabolic inflammation key marker TLR4 activation-mediated ß-cell injury. Furthermore, downstream signaling protein PLC-IP3 of GPR40 was involved in the protective effect of metformin on meta-inflammation, and the above process of metformin was partially regulated by AMPK activity. In addition, the anti-inflammatory effects of metformin were observed in obese rats. CONCLUSION: Metformin can reduce lipotoxicity-induced meta-inflammation in ß-cells through the regulation of the GPR40-PLC-IP3 pathway and partially via the regulation of AMPK activity.


Diet, High-Fat/adverse effects , Inflammation/prevention & control , Inositol 1,4,5-Trisphosphate/physiology , Insulin-Secreting Cells/drug effects , Metformin/pharmacology , Receptors, G-Protein-Coupled/physiology , Type C Phospholipases/physiology , Adenylate Kinase/physiology , Animals , Cells, Cultured , Insulin-Secreting Cells/metabolism , Male , Mice , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Toll-Like Receptor 4/physiology
6.
Metab Brain Dis ; 29(4): 1083-93, 2014 Dec.
Article En | MEDLINE | ID: mdl-24748364

Thiamine dependent enzymes are diminished in Alzheimer's disease (AD). Thiamine deficiency in vitro and in rodents is a useful model of this reduction. Thiamine interacts with cellular calcium stores. To directly test the relevance of the thiamine dependent changes to dynamic processes in AD, the interactions must be studied in cells from patients with AD. These studies employed fibroblasts. Mitochondrial dysfunction including reductions in thiamine dependent enzymes and abnormalities in calcium homeostasis and oxidative processes occur in fibroblasts from Alzheimer's Disease (AD) patients. Bombesin-releasable calcium stores (BRCS) from the endoplasmic reticulum (ER) are exaggerated in fibroblasts from patients with AD bearing a presenilin-1 (PS-1) mutation and in control fibroblasts treated with oxidants. ER calcium regulates calcium entry into the cell through capacitative calcium entry (CCE), which is reduced in fibroblasts and neurons from mice bearing PS-1 mutations. Under physiological conditions, mitochondria and ER play important and interactive roles in the regulation of Ca(2+) homeostasis. Thus, the interactions of mitochondria and oxidants with CCE were tested. Inhibition of ER Ca(2+)-ATPase by cyclopiazonic acid (CPA) stimulates CCE. CPA-induced CCE was diminished by inhibition of mitochondrial Ca(2+) export (-60%) or import (-40%). Different aspects of mitochondrial Ca(2+) coupled to CPA-induced-CCE were sensitive to select oxidants. The effects were very different when CCE was examined in the presence of InsP3, a physiological regulator of ER calcium release, and subsequent CCE. CCE under these conditions was only mildly reduced (20-25%) by inhibition of mitochondrial Ca(2+) export, and inhibition of mitochondrial Ca(2+) uptake exaggerated CCE (+53%). However, t-BHP reversed both abnormalities. The results suggest that in the presence of InsP3, mitochondria buffer the local Ca(2+) released from ER following rapid activation of InsP3R and serve as a negative feedback to the CCE. The results suggest that mitochondrial Ca(2+) modifies the depletion and refilling mechanism of ER Ca(2+) stores.


Calcium Signaling , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Thiamine/physiology , Alzheimer Disease/pathology , Animals , Cells, Cultured , Fibroblasts/metabolism , Homeostasis , Humans , Indoles/pharmacology , Inositol 1,4,5-Trisphosphate/physiology , Inositol 1,4,5-Trisphosphate Receptors/physiology , Ion Transport , Male , Mice , Mice, Neurologic Mutants , Neurons/metabolism
7.
Adv Exp Med Biol ; 991: 141-57, 2013.
Article En | MEDLINE | ID: mdl-23775694

Work over the recent years has greatly expanded our understanding of the specific molecules involved in plant phosphoinositide signaling. Physiological approaches, combined with analytical techniques and genetic mutants have provided tools to understand how individual genes function in this pathway. Several key differences between plants and animals have become apparent. This chapter will highlight the key areas where major differences between plants and animals occur. In particular, phospholipase C and levels of phosphatidylinositol phosphates differ between plants and animals, and may influence how inositol second messengers form and function in plants. Whether inositol 1,4,5-trisphosphate and/or inositol hexakisphosphate (InsP6) function as second messengers in plants is discussed. Recent data on potential, novel roles of InsP6 in plants is considered, along with the existence of a unique InsP6 synthesis pathway. Lastly, the complexity of myo-inositol synthesis in plants is discussed in reference to synthesis of phosphoinositides and impact on plant growth and development.


Inositol Phosphates/physiology , Phosphatidylinositols/physiology , Plant Cells/physiology , Signal Transduction/physiology , Diglycerides/physiology , Inositol/biosynthesis , Inositol 1,4,5-Trisphosphate/physiology , Phosphatidylinositols/analysis , Phytic Acid/physiology , Type C Phospholipases/physiology
8.
Am J Physiol Heart Circ Physiol ; 304(10): H1328-36, 2013 May 15.
Article En | MEDLINE | ID: mdl-23479262

The cardiac hormone atrial natriuretic factor (ANF or ANP) plays significant, well-established roles in a large number of physiological and pathophysiological processes, including water and electrolyte balance, blood pressure regulation, and cardiovascular growth. Understanding the regulation of its production and secretion by atrial cardiomyocytes is incomplete. We have previously established a significant role of G(i/o) protein signaling in modulating ANF secretion as promoted by stretch of the atrial myocardium. In the present study, we investigated the role of G(q) protein signaling and its relationship to G(i/o) protein signaling using pharmacological manipulation of proximal effectors of G(αq) in an ex vivo model of spontaneously beating rat atria. Phospholipase C (PLC) and protein kinase C (PKC) inhibitors dramatically increased basal secretion of ANF. Furthermore, although atrial wall stretch is a potent stimulus for secretion, stretch unexpectedly reduced ANF secretion to basal levels under PLC and PKC inhibitory conditions. Inhibition of the inositol triphosphate receptor did not appear to affect basal secretion but dose-dependently blocked stretch-secretion coupling. The results obtained demonstrate that the PLC and PKC signaling cascades play important albeit unexpected roles in the regulation of basal and stimulated ANF secretion and suggest interplay between the G(q) and G(i/o) protein signaling pathways.


Atrial Natriuretic Factor/metabolism , Myocardium/metabolism , Signal Transduction/physiology , Type C Phospholipases/physiology , Animals , Blood Pressure/physiology , Dose-Response Relationship, Drug , Heart Atria/metabolism , In Vitro Techniques , Inositol 1,4,5-Trisphosphate/physiology , Inositol 1,4,5-Trisphosphate Receptors/physiology , Male , Mechanoreceptors/physiology , Myocardium/enzymology , Protein Kinase C/antagonists & inhibitors , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Second Messenger Systems/physiology
9.
Pharmacol Rev ; 65(2): 641-69, 2013 Apr.
Article En | MEDLINE | ID: mdl-23429059

Studies of inositol polyphosphates in the pancreatic ß-cell have led to an exciting synergism between new discoveries regarding their cellular roles and new insights into ß-cell function. Because the loss or malfunction of the ß-cell is central to diabetes, these studies open the possibility of new pharmacological interventions in a disease that has reached epidemic proportions worldwide. Using the ß-cell as our prime but not exclusive example, we examine the inositol polyphosphates in three main groups: 1) inositol 1,4,5-trisphosphate and its influence on Ca(2+) signaling, specifically in a cell in which cytoplasmic-free Ca(2+) concentration is principally increased by plasma membrane standing voltage-gated Ca(2+) channels; 2) higher inositol polyphosphates including a novel second messenger inositol 3,4,5,6-tetrakisphosphate and a regulatory role for inositol hexakisphosphate in ß-cell Ca(2+) homeostasis and exo- and endocytosis; and 3) inositol pyrophosphates and their role in ß-cell exocytosis, together with the exciting possibility of being novel targets for therapy in diabetes. We conclude with some of the new perspectives that are likely to become apparent in the next few years.


Inositol Phosphates/physiology , Insulin-Secreting Cells/physiology , Insulin/metabolism , Animals , Calcium/metabolism , Exocytosis/physiology , Glucose/metabolism , Humans , Inositol 1,4,5-Trisphosphate/chemistry , Inositol 1,4,5-Trisphosphate/physiology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol Phosphates/chemistry , Insulin Secretion , Insulin-Secreting Cells/metabolism , Phytic Acid/chemistry , Phytic Acid/physiology
10.
J Mol Cell Cardiol ; 54: 19-24, 2013 Jan.
Article En | MEDLINE | ID: mdl-23137780

Autophagy is a process that removes damaged proteins and organelles and is of particular importance in terminally differentiated cells such as cardiomyocytes, where it has primarily a protective role. We investigated the involvement of inositol(1,4,5)trisphosphate (Ins(1,4,5)P(3)) and its receptors in autophagic responses in neonatal rat ventricular myocytes (NRVM). Treatment with the IP(3)-receptor (IP(3)-R) antagonist 2-aminoethoxydiphenyl borate (2-APB) at 5 or 20 µmol/L resulted in an increase in autophagosome content, defined as puncta labeled by antibody to microtubule associated light chain 3 (LC3). 2-APB also increased autophagic flux, indicated by heightened LC3II accumulation, which was further enhanced by bafilomycin (10nmol/L). Expression of Ins(1,4,5)P(3) 5-phosphatase (IP(3)-5-Pase) to deplete Ins(1,4,5)P(3) also increased LC3-labeled puncta and LC3II content, suggesting that Ins(1,4,5)P(3) inhibits autophagy. The IP(3)-R can act as an inhibitory scaffold sequestering the autophagic effector, beclin-1 to its ligand binding domain (LBD). Expression of GFP-IP(3)-R-LBD inhibited autophagic signaling and furthermore, beclin-1 co-immunoprecipitated with the IP(3)-R-LBD. A mutant GFP-IP(3)-R-LBD with reduced ability to bind Ins(1,4,5)P(3) bound beclin-1 and inhibited autophagy similarly to the wild type sequence. These data provide evidence that Ins(1,4,5)P(3) and IP(3)-R act as inhibitors of autophagic responses in cardiomyocytes. By suppressing autophagy, IP(3)-R may contribute to cardiac pathology.


Autophagy , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate/physiology , Myocytes, Cardiac/physiology , Animals , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Binding Sites , Boron Compounds/pharmacology , Cells, Cultured , Cytoplasmic Granules/metabolism , Forkhead Transcription Factors/metabolism , Heart Ventricles/cytology , Heat-Shock Proteins/metabolism , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Lysosomes/metabolism , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Phagosomes/metabolism , Phosphatidylethanolamines/physiology , Protein Binding , Protein Structure, Tertiary , Proteolysis , Rats , Rats, Sprague-Dawley , Sequestosome-1 Protein , Signal Transduction
11.
Annu Rev Physiol ; 75: 127-54, 2013.
Article En | MEDLINE | ID: mdl-23140367

Phospholipase C (PLC) converts phosphatidylinositol 4,5-bisphosphate (PIP(2)) to inositol 1,4,5-trisphosphate (IP(3)) and diacylglycerol (DAG). DAG and IP(3) each control diverse cellular processes and are also substrates for synthesis of other important signaling molecules. PLC is thus central to many important interlocking regulatory networks. Mammals express six families of PLCs, each with both unique and overlapping controls over expression and subcellular distribution. Each PLC also responds acutely to its own spectrum of activators that includes heterotrimeric G protein subunits, protein tyrosine kinases, small G proteins, Ca(2+), and phospholipids. Mammalian PLCs are autoinhibited by a region in the catalytic TIM barrel domain that is the target of much of their acute regulation. In combination, the PLCs act as a signaling nexus that integrates numerous signaling inputs, critically governs PIP(2) levels, and regulates production of important second messengers to determine cell behavior over the millisecond to hour timescale.


Second Messenger Systems/physiology , Signal Transduction/physiology , Type C Phospholipases/physiology , Animals , Diglycerides/physiology , Humans , Inositol 1,4,5-Trisphosphate/physiology , Phosphatidylinositol 4,5-Diphosphate/physiology , Type C Phospholipases/chemistry
12.
Cell Physiol Biochem ; 30(6): 1398-412, 2012.
Article En | MEDLINE | ID: mdl-23154210

BACKGROUND/AIMS: The mechanism underlying transmitter release from astrocytes is not fully understood. The present study examined A(2a) adenosine receptor-mediated glutamate release and intracellular Ca(2+) rise in cultured rat hippocampal astrocytes. METHODS: Intracellular amino acids were measured with HPLC. Glutamate release from astrocytes and intracellular Ca(2+) mobilizations were monitored in the NADH imaging, FM1-43 imaging, and fura-2 imaging. The siRNA to silence the A(2a) adenosine receptor-targeted gene was constructed and transfected into cells. RESULTS: Glutamate was condensed in 'synaptic-like vesicle' fractions. In the NADH imaging, CGS21680, an agonist of A2a adenosine receptors, increased NADH fluorescent signals, that reflects glutamate release, and the effect was inhibited by DMPX, an inhibitor of A(2a) adenosine receptors, H-89, a PKA inhibitor, vesicular transport inhibitors, or botulinum toxin-A, an exocytosis inhibitor. In the FM1-43 imaging to see vesicular recycling, CGS21680 decreased FM1-43 fluorescent signals, that was also prevented by DMPX, H-89, vesicular transport inhibitors, or botulinum toxin-A. CGS21680 increased intracellular Ca(2+) concentrations both in Ca(2+)-containing and -free extracellular solution. The Ca(2+) rise was inhibited by DMPX, H-89, or the vesicular transport inhibitor brefeldin A, but it was not affected by inhibitors for phospholipase C, IP(3) receptor, and ryanodine receptor. CGS21680-induced glutamate release and intracellular Ca(2+) rise were prevented by knocking-down A(2a) adenosine receptor. CONCLUSION: The results of the present study show that A(2a) adenosine receptor/PKA promotes glutamate release from synaptic-like vesicles and stimulates Ca(2+) efflux from an IP(3)- and ryanodine-insensitive intracellular calcium store.


Astrocytes/metabolism , Calcium Signaling , Cyclic AMP-Dependent Protein Kinases/metabolism , Glutamic Acid/metabolism , Receptor, Adenosine A2A/metabolism , Secretory Vesicles/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Amino Acids/metabolism , Animals , Cell Fractionation , Cells, Cultured , Hippocampus/cytology , Inositol 1,4,5-Trisphosphate/physiology , Phenethylamines/pharmacology , Rats , Rats, Wistar , Receptor, Adenosine A2A/genetics , Ryanodine/pharmacology , SNARE Proteins/metabolism , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Glutamate Transport Proteins/metabolism
13.
J Physiol ; 590(23): 5993-6011, 2012 Dec 01.
Article En | MEDLINE | ID: mdl-22966160

Electrogenic Na(+)-bicarbonate cotransporter NBCe1 variants contribute to pH(i) regulation, and promote ion reabsorption or secretion by many epithelia. Most Na(+)-coupled bicarbonate transporter (NCBT) families such as NBCe1 contain variants with differences primarily at the cytosolic N and/or C termini that are likely to impart on the transporters different modes of regulation. For example, N-terminal regions of NBCe1 autoregulate activity. Our group previously reported that cytosolic phosphatidylinositol 4,5-bisphosphate (PIP(2)) stimulates heterologously expressed rat NBCe1-A in inside-out macropatches excised from Xenopus laevis oocytes. In the current study on whole oocytes, we used the two-electrode voltage-clamp technique, as well as pH- and voltage-sensitive microelectrodes, to characterize the effect of injecting PIP(2) on the activity of heterologously expressed NBCe1-A, -B, or -C. Injecting PIP(2) (10 µM estimated final) into voltage-clamped oocytes stimulated NBC-mediated, HCO(3)(-)-induced outward currents by >100% for the B and C variants, but not for the A variant. The majority of this stimulation involved PIP(2) hydrolysis and endoplasmic reticulum (ER) Ca(2+) release. Stimulation by PIP(2) injection was mimicked by injecting IP(3), but inhibited by either applying the phospholipase C (PLC) inhibitor U73112 or depleting ER Ca(2+) with prolonged thapsigargin/EGTA treatment. Stimulating the activity of store-operated Ca(2+) channels (SOCCs) to trigger a Ca(2+) influx mimicked the PIP(2)/IP(3) stimulation of the B and C variants. Activating the endogenous G(q) protein-coupled receptor in oocytes with lysophosphatidic acid (LPA) also stimulated the B and C variants in a Ca(2+)-dependent manner, although via an increase in surface expression for the B variant. In simultaneous voltage-clamp and pH(i) studies on NBCe1-C-expressing oocytes, LPA increased the NBC-mediated pH(i)-recovery rate from a CO(2)-induced acid load by ∼80%. Finally, the general kinase inhibitor staurosporine completely inhibited the IP(3)-induced stimulation of NBCe1-C. In summary, injecting PIP(2) stimulates the activity of NBCe1-B and -C expressed in oocytes through an increase in IP(3)/Ca(2+) that involves a staurosporine-sensitive kinase. In conjunction with our previous macropatch findings, PIP(2) regulates NBCe1 through a dual pathway involving both a direct stimulatory effect of PIP(2) on at least NBCe1-A, as well as an indirect stimulatory effect of IP(3)/Ca(2+) on the B and C variants.


Phosphatidylinositol 4,5-Diphosphate/physiology , Sodium-Bicarbonate Symporters/physiology , Animals , Calcium/physiology , Female , Hydrolysis , Inositol 1,4,5-Trisphosphate/physiology , Oocytes/physiology , Patch-Clamp Techniques , Rats , Xenopus laevis
14.
J Neurosci Res ; 90(10): 1913-23, 2012 Oct.
Article En | MEDLINE | ID: mdl-22714707

Group I metabotropic glutamate receptors (mGluRs) are linked to intracellular Ca(2+) signalling and play important roles related to synaptic plasticity and development. In neurons from the central nucleus of the inferior colliculus (CIC), the activation of these receptors evokes large [Ca(2+) ](i) responses. By using optical imaging of the fluorescent Ca(2+) -sensitive dye Fura-2, we have explored which [Ca(2+) ](i) routes are triggered by group I mGluR activation in young CIC neurons and whether mGluR-induced [Ca(2+) ](i) responses are regulated during postnatal development. In addition, real-time quantitative RT-PCR was used to study the developmental expression of both group I mGluR subtypes, mGluR1 and mGluR5. Application of DHPG, a specific agonist of group I mGluRs, was used on CIC slices from young rats to elicit [Ca(2+) ](i) responses. A majority of responses consisted of an initial thapsigargin-sensitive Ca(2+) peak, related to store depletion, followed by a plateau phase, sensitive to the store-operated Ca(2+) entry blocker 2-APB. During postnatal development, from P6 to P17, DHPG-induced [Ca(2+) ](i) responses changed. The largest Ca(2+) responses were reached at P6, whereas lower peak and plateau responses were found after hearing onset, at P13-P14 and P17. qRT-PCR analysis also revealed important differences in the expression of both mGluR1 and mGluR5 subtypes during development, with the highest levels of both subtypes at P7 and a developmental decrease of both transcripts. Our results suggest both intra- and extracellular routes for [Ca(2+) ](i) increases linked to group I mGluRs in CIC neurons and a regulation of group I mGluR activity and expression during auditory development.


Auditory Cortex/physiology , Mesencephalon/physiology , Neurons/physiology , Receptors, Metabotropic Glutamate/physiology , Signal Transduction/physiology , Aging/physiology , Animals , Auditory Cortex/cytology , Auditory Cortex/drug effects , Calcium Channels/physiology , Calcium Signaling/physiology , Cell Membrane/metabolism , Cell Membrane/physiology , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Down-Regulation/drug effects , In Vitro Techniques , Inferior Colliculi/physiology , Inositol 1,4,5-Trisphosphate/physiology , Male , Mesencephalon/cytology , Mesencephalon/drug effects , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/metabolism , Neurons/drug effects , Polymerase Chain Reaction , RNA/biosynthesis , RNA/genetics , RNA/isolation & purification , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
15.
Adv Exp Med Biol ; 740: 255-79, 2012.
Article En | MEDLINE | ID: mdl-22453946

Activation of cells by many extracellular agonists leads to the production of inositol 1,4,5-trisphosphate (IP3). IP3 is a global messenger that easily diffuses in the cytosol. Its receptor (IP3R) is a Ca(2+)-release channel located on intracellular membranes, especially the endoplasmic reticulum (ER). The IP3R has an affinity for IP(3) in the low nanomolar range. A prime regulator of the IP3R is the Ca(2+) ion itself. Cytosolic Ca(2+) is considered as a co-agonist of the IP3R, as it strongly increases IP(3)R activity at concentrations up to about 300 nM. In contrast, at higher concentrations, cytosolic Ca(2+) inhibits the IP3R. Also the luminal Ca(2+) sensitizes the IP3R. In higher organisms three genes encode for an IP3R and additional diversity exists as a result of alternative splicing mechanisms and the formation of homo- and heterotetramers. The various IP3R isoforms have a similar structure and a similar function, but due to differences in their affinity for IP3, their variable sensitivity to regulatory parameters, their differential interaction with associated proteins, and the variation in their subcellular localization, they participate differently in the formation of intracellular Ca(2+) signals and this affects therefore the physiological consequences of these signals.


Inositol 1,4,5-Trisphosphate Receptors/physiology , Inositol 1,4,5-Trisphosphate/physiology , Animals , Calcium/metabolism , Calcium Signaling , Cytosol/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors/analysis , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Inositol 1,4,5-Trisphosphate Receptors/chemistry
16.
Am J Physiol Endocrinol Metab ; 302(11): E1419-30, 2012 Jun 01.
Article En | MEDLINE | ID: mdl-22414808

The role of 3,5,3'-triiodo-l-thyronine (T3) and its metabolite 3,5-diiodo-l-thyronine (T2) in modulating the intracellular Ca(2+) concentration ([Ca(2+)](i)) and endogenous nitric oxide (NO) synthesis was evaluated in pituitary GH(3) cells in the absence or presence of extracellular Ca(2+). When applied in Ca(2+)-free solution, T2 and T3 increased [Ca(2+)](i), in a dose-dependent way, and NO levels. Inhibition of neuronal NO synthase by N(G)-nitro-l-arginine methyl ester and l-n(5)-(1-iminoethyl)ornithine hydrochloride significantly reduced the [Ca(2+)](i) increase induced by T2 and T3. However, while depletion of inositol trisphosphate-dependent Ca(2+) stores did not interfere with the T2- and T3-induced [Ca(2+)](i) increases, the inhibition of phosphatidylinositol 3-kinase by LY-294002 and the dominant negative form of Akt mutated at the ATP binding site prevented these effects. Furthermore, the mitochondrial protonophore carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone prevented the increases in both [Ca(2+)](i) and NO elicited by T2 or T3. Interestingly, rotenone blocked the early [Ca(2+)](i) increases elicited by T2 and T3, while antimycin prevented only that elicited by T3. Inhibition of mitochondrial Na(+)/Ca(2+) exchanger by CGP37157 significantly reduced the [Ca(2+)](i) increases induced by T2 and T3. In the presence of extracellular calcium (1.2 mM), under carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone, T2 and T3 increased both [Ca(2+)](i) and intracellular Na(+) concentration; nimodipine reduced the [Ca(2+)](i) increases elicited by T2 and T3, but inhibition of NO synthase and blockade of the Na(+)/H(+) pump by 5-(N-ethyl-N-isopropyl)amiloride prevented only that elicited by T3; and CB-DMB, bisindolylmaleimide, and LY-294002 (inhibitors of the Na(+)/Ca(2+) exchanger, PKC, and phosphatidylinositol 3-kinase, respectively) failed to modify the T2- and T3-induced effects. Collectively, the present results suggest that T2 and T3 exert short-term nongenomic effects on intracellular calcium and NO by modulating plasma membrane and mitochondrial pathways that differ between these iodothyronines.


Calcium/metabolism , Cell Membrane/physiology , Diiodothyronines/pharmacology , Intracellular Membranes/physiology , Pituitary Gland/metabolism , Triiodothyronine/pharmacology , Calcium Signaling/drug effects , Carrier Proteins/metabolism , Cell Membrane/drug effects , Cells, Cultured , Fluorescein , Fluorescent Dyes , Homeostasis/drug effects , Humans , Inositol 1,4,5-Trisphosphate/physiology , Intracellular Membranes/drug effects , Ion Channels/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide/physiology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Pituitary Gland/cytology , Pituitary Gland/drug effects , Signal Transduction/drug effects , Sodium/metabolism , Transfection
17.
J Neurochem ; 121(4): 516-25, 2012 May.
Article En | MEDLINE | ID: mdl-22356163

Oxytocin (OT) is clinically important in gut motility and constitutively reduces duodenum contractility. Intrinsic primary afferent neurons (IPANs), whose physiological classification is as AH cells, are the 1st neurons of the peristaltic reflex pathway. We set out to investigate if this inhibitory effect is mediated by IPANs and to identify the ion channel(s) and intracellular signal transduction pathway that are involved in this effect. Myenteric neurons were isolated from the longitudinal muscle myenteric plexus (LMMP) preparation of rat duodenum and cultured for 16-24 h before electrophysiological recording in whole cell mode and AH cells identified by their electrophysiological characteristics. The cytoplasmic Ca²âº concentration ([Ca²âº](i) ) of isolated neurons was measured using calcium imaging. The concentration of IP(3) in the LMMP and the OT secreted from the LMMP were measured using ELISA. The oxytocin receptor (OTR) and large-conductance calcium-activated potassium (BK(Ca)) channels, as well as the expression of OT and the IPAN marker calbindin 28 K, on the myenteric plexus neurons were localized using double-immunostaining techniques. We found that administration of OT (10⁻7 to 10⁻5 M) dose dependently hyperpolarized the resting membrane potential and increased the total outward current. The OTR antagonist atosiban or the BK(Ca) channel blocker iberiotoxin (IbTX) blocked the effects of OT suggesting that the increased outward current resulted from BK(Ca) channel opening. OTR and the BK(Ca) α subunit were co-expressed on a subset of myenteric neurons at the LMMP. NS1619 (10⁻5 M, a BK(Ca) channel activator) increased the outward current similar to the effect of OT. OT administration also increased [Ca²âº](i) and the OT-evoked outward current was significantly attenuated by thapsigargin (10⁻6 M) or CdCl2. The effect of OT on the BK(Ca) current was also blocked by pre-treatment with the IP3 receptor antagonist 2-APB (10⁻4 M) or the PLC inhibitor U73122 (10⁻5 M). OT (10⁻6 M) also increased the IP3 concentration within the LMMP. Both of the spontaneous and KCl-induced secretion of OT was enhanced by atosiban. Most of OT-immunoreactive cells are also immunoreactive for calbindin 28 K. In summary, we concluded that OT hyperpolarized myenteric IPANs by activating BK(Ca) channels via the OTR-PLC-IP3-Ca²âº signal pathway. OT might modulate IPANs mediated ENS reflex by an autocrine and negative feedback manner.


Duodenum/innervation , Inositol 1,4,5-Trisphosphate/physiology , Large-Conductance Calcium-Activated Potassium Channels/drug effects , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Myenteric Plexus/drug effects , Neurons, Afferent/drug effects , Oxytocin/pharmacology , Signal Transduction/drug effects , Action Potentials/drug effects , Animals , Autocrine Communication/drug effects , Cell Separation , Cells, Cultured , Duodenum/drug effects , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , In Vitro Techniques , Male , Membrane Potentials/drug effects , Neurons, Afferent/ultrastructure , Patch-Clamp Techniques , Potassium Chloride/pharmacology , Rats , Receptors, Oxytocin/drug effects , Type C Phospholipases/physiology , Vasotocin/analogs & derivatives , Vasotocin/pharmacology
18.
Biochim Biophys Acta ; 1820(8): 1214-27, 2012 Aug.
Article En | MEDLINE | ID: mdl-22033379

BACKGROUND: Inositol 1,4,5-trisphosphate receptors (IP3R) are expressed in almost all animal cells. Three mammalian genes encode closely related IP3R subunits, which assemble into homo- or hetero-tetramers to form intracellular Ca2+ channels. SCOPE OF THE REVIEW: In this brief review, we first consider a variety of complementary methods that allow the links between IP3 binding and channel gating to be defined. How does IP3 binding to the IP3-binding core in each IP3R subunit cause opening of a cation-selective pore formed by residues towards the C-terminal? We then describe methods that allow IP3, Ca2+ signals and IP3R mobility to be examined in intact cells. A final section briefly considers genetic analyses of IP3R signalling. MAJOR CONCLUSIONS: All IP3R are regulated by both IP3 and Ca2+. This allows them to initiate and regeneratively propagate intracellular Ca2+ signals. The elementary Ca2+ release events evoked by IP3 in intact cells are mediated by very small numbers of active IP3R and the Ca2+-mediated interactions between them. The spatial organization of these Ca2+ signals and their stochastic dependence on so few IP3Rs highlight the need for methods that allow the spatial organization of IP3R signalling to be addressed with single-molecule resolution. GENERAL SIGNIFICANCE: A variety of complementary methods provide insight into the structural basis of IP3R activation and the contributions of IP3-evoked Ca2+ signals to cellular physiology. This article is part of a Special Issue entitled Biochemical, biophysical and genetic approaches to intracellular calcium signaling.


Inositol 1,4,5-Trisphosphate Receptors/metabolism , Animals , Binding Sites , Calcium Signaling , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate/physiology , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Inositol 1,4,5-Trisphosphate Receptors/genetics , Membrane Potentials , Microscopy, Fluorescence , Patch-Clamp Techniques , Protein Transport , Single-Cell Analysis
19.
J Physiol ; 590(5): 1093-100, 2012 Mar 01.
Article En | MEDLINE | ID: mdl-22183727

Photoactivated probes have gained interest as experimental tools to study intracellular signalling pathways all the way to the molecular level. However technical limitations of the means to activate such compounds have put constraints on their use in spatially highly restricted subcellular areas. The Mosaic digital illumination system uses a high-speed array of individually addressable, tiltable micromirrors to direct continuous-wave laser light onto a specimen with diffraction-limited precision. The system, integrated into a Nikon A1R confocal microscope, was used to uncage Ca²âº or IP3 and conduct photobleaching experiments from multiple geometrically complex subcellular regions while simultaneously measuring [Ca²âº]i with high-speed confocal imaging.


Calcium/physiology , Myocytes, Cardiac/physiology , Animals , Fluorescent Dyes , Inositol 1,4,5-Trisphosphate/physiology , Light , Microscopy, Confocal , Photobleaching , Rabbits , Signal Transduction
20.
J Physiol ; 589(Pt 20): 4903-20, 2011 Oct 15.
Article En | MEDLINE | ID: mdl-21844002

Postsynaptic [Ca(2+)](i) changes contribute to several kinds of plasticity in pyramidal neurons. We examined the effects of synaptically activated Ca(2+) waves and NMDA spikes on subsequent Ca(2+) signalling in CA1 pyramidal cell dendrites in hippocampal slices. Tetanic synaptic stimulation evoked a localized Ca(2+) wave in the primary apical dendrites. The [Ca(2+)](i) increase from a backpropagating action potential (bAP) or subthreshold depolarization was reduced if it was generated immediately after the wave. The suppression had a recovery time of 30-60 s. The suppression only occurred where the wave was generated and was not due to a change in bAP amplitude or shape. The suppression also could be generated by Ca(2+) waves evoked by uncaging IP(3), showing that other signalling pathways activated by the synaptic tetanus were not required. The suppression was proportional to the amplitude of the [Ca(2+)](i) change of the Ca(2+) wave and was not blocked by a spectrum of kinase or phosphatase inhibitors, consistent with suppression due to Ca(2+)-dependent inactivation of Ca(2+) channels. The waves also reduced the frequency and amplitude of spontaneous, localized Ca(2+) release events in the dendrites by a different mechanism, probably by depleting the stores at the site of wave generation. The same synaptic tetanus often evoked NMDA spike-mediated [Ca(2+)](i) increases in the oblique dendrites where Ca(2+) waves do not propagate. These NMDA spikes suppressed the [Ca(2+)](i) increase caused by bAPs in those regions. [Ca(2+)](i) increases by Ca(2+) entry through voltage-gated Ca(2+) channels also suppressed the [Ca(2+)](i) increases from subsequent bAPs in regions where the voltage-gated [Ca(2+)](i) increases were largest, showing that all ways of raising [Ca(2+)](i) could cause suppression.


Calcium Signaling/physiology , Calcium/physiology , Dendrites/physiology , N-Methylaspartate/physiology , Pyramidal Cells/physiology , Action Potentials/physiology , Animals , Calcium Channels/physiology , In Vitro Techniques , Inositol 1,4,5-Trisphosphate/physiology , Rats , Rats, Sprague-Dawley , Synapses/physiology
...