Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Mol Cell ; 58(6): 1015-27, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-25959394

ABSTRACT

Blood pressure maintenance is vital for systemic homeostasis, and angiotensin II is a critical regulator. The upstream mechanisms that regulate angiotensin II are not completely understood. Here, we show that angiotensin II is regulated by ERp44, a factor involved in disulfide bond formation in the ER. In mice, genetic loss of ERp44 destabilizes angiotensin II and causes hypotension. We show that ERp44 forms a mixed disulfide bond with ERAP1, an aminopeptidase that cleaves angiotensin II. ERp44 controls the release of ERAP1 in a redox-dependent manner to control blood pressure. Additionally, we found that systemic inflammation triggers ERAP1 retention in the ER to inhibit hypotension. These findings suggest that the ER redox state calibrates serum angiotensin II levels via regulation of the ERp44-ERAP1 complex. Our results reveal a link between ER function and normotension and implicate the ER redox state as a potential risk factor in the development of cardiovascular disease.


Subject(s)
Aminopeptidases/metabolism , Blood Pressure , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Molecular Chaperones/metabolism , Amino Acid Sequence , Aminopeptidases/genetics , Angiotensin II/blood , Angiotensin II/metabolism , Animals , Blotting, Western , Cells, Cultured , HeLa Cells , Humans , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , Molecular Chaperones/genetics , Molecular Sequence Data , Oxidation-Reduction , Protein Binding , RNA Interference , Sequence Homology, Amino Acid
2.
J Neurosci ; 41(39): 8134-8149, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34417327

ABSTRACT

Tuberous sclerosis complex (TSC) is a multisystem developmental disorder characterized by hamartomas in various organs, such as the brain, lungs, and kidneys. Epilepsy, along with autism and intellectual disability, is one of the neurologic impairments associated with TSC that has an intimate relationship with developmental outcomes and quality of life. Sustained activation of the mammalian target of rapamycin (mTOR) via TSC1 or TSC2 mutations is known to be involved in the onset of epilepsy in TSC. However, the mechanism by which mTOR causes seizures remains unknown. In this study, we showed that, human induced pluripotent stem cell-derived TSC2-deficient (TSC2-/-) neurons exhibited elevated neuronal activity with highly synchronized Ca2+ spikes. Notably, TSC2-/- neurons presented enhanced Ca2+ influx via L-type Ca2+ channels (LTCCs), which contributed to the abnormal neurite extension and sustained activation of cAMP response element binding protein (CREB), a critical mediator of synaptic plasticity. Expression of Cav1.3, a subtype of LTCCs, was increased in TSC2-/- neurons, but long-term rapamycin treatment suppressed this increase and reversed the altered neuronal activity and neurite extensions. Thus, we identified Cav1.3 LTCC as a critical downstream component of TSC-mTOR signaling that would trigger enhanced neuronal network activity of TSC2-/- neurons. We suggest that LTCCs could be potential novel targets for the treatment of epilepsy in TSC.SIGNIFICANCE STATEMENT There is a close relationship between elevated mammalian target of rapamycin (mTOR) activity and epilepsy in tuberous sclerosis complex (TSC). However, the underlying mechanism by which mTOR causes epilepsy remains unknown. In this study, using human TSC2-/- neurons, we identified elevated Ca2+ influx via L-type Ca2+ channels as a critical downstream component of TSC-mTOR signaling and a potential cause of both elevated neuronal activity and neurite extension in TSC2-/- neurons. Our findings demonstrate a previously unrecognized connection between sustained mTOR activation and elevated Ca2+ signaling via L-type Ca2+ channels in human TSC neurons, which could cause epilepsy in TSC.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium/metabolism , Nerve Net/metabolism , Neurons/metabolism , Tuberous Sclerosis Complex 1 Protein/metabolism , Tuberous Sclerosis Complex 2 Protein/metabolism , Cell Differentiation/physiology , Humans , Induced Pluripotent Stem Cells/metabolism , Mutation , Neuronal Outgrowth/physiology , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/genetics
3.
Proc Natl Acad Sci U S A ; 114(26): E5256-E5265, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28607044

ABSTRACT

Dendritic spines of Purkinje cells form excitatory synapses with parallel fiber terminals, which are the primary sites for cerebellar synaptic plasticity. Nevertheless, how density and morphology of these spines are properly maintained in mature Purkinje cells is not well understood. Here we show an activity-dependent mechanism that represses excessive spine development in mature Purkinje cells. We found that CaMKIIß promotes spine formation and elongation in Purkinje cells through its F-actin bundling activity. Importantly, activation of group I mGluR, but not AMPAR, triggers PKC-mediated phosphorylation of CaMKIIß, which results in dissociation of the CaMKIIß/F-actin complex. Defective function of the PKC-mediated CaMKIIß phosphorylation promotes excess F-actin bundling and leads to abnormally numerous and elongated spines in mature IP3R1-deficient Purkinje cells. Thus, our data suggest that phosphorylation of CaMKIIß through the mGluR/IP3R1/PKC signaling pathway represses excessive spine formation and elongation in mature Purkinje cells.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dendritic Spines/metabolism , Protein Kinase C/metabolism , Purkinje Cells/metabolism , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/physiology , Actins/genetics , Actins/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Dendritic Spines/genetics , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mice , Mice, Knockout , Phosphorylation/genetics , Protein Kinase C/genetics , Purkinje Cells/cytology , Receptors, Metabotropic Glutamate/genetics
4.
Hum Mol Genet ; 25(16): 3491-3499, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27378687

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating progressive neurodegenerative disease characterized by the selective death of motor neurons. Disease pathophysiology is complex and not yet fully understood. Higher gene expression of the inositol 1,4,5-trisphosphate receptor 2 gene (ITPR2), encoding the IP3 receptor 2 (IP3R2), was detected in sporadic ALS patients. Here, we demonstrate that IP3R2 gene expression was also increased in spinal cords of ALS mice. Moreover, an increase of IP3R2 expression was observed in other models of chronic and acute neurodegeneration. Upregulation of IP3R2 gene expression could be induced by lipopolysaccharide (LPS) in murine astrocytes, murine macrophages and human fibroblasts indicating that it may be a compensatory response to inflammation. Preventing this response by genetic deletion of ITPR2 from SOD1G93A mice had a dose-dependent effect on disease duration, resulting in a significantly shorter lifespan of these mice. In addition, the absence of IP3R2 led to increased innate immunity, which may contribute to the decreased survival of the SOD1G93A mice. Besides systemic inflammation, IP3R2 knockout mice also had increased IFNγ, IL-6 and IL1α expression. Altogether, our data indicate that IP3R2 protects against the negative effects of inflammation, suggesting that the increase in IP3R2 expression in ALS patients is a protective response.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Inflammation/genetics , Inositol 1,4,5-Trisphosphate Receptors/genetics , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Models, Animal , Humans , Inflammation/pathology , Interferon-gamma/biosynthesis , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Lipopolysaccharides , Male , Mice , Mice, Knockout , Motor Neurons/metabolism , Motor Neurons/pathology , Spinal Cord/metabolism , Spinal Cord/pathology
5.
Proc Natl Acad Sci U S A ; 112(17): 5515-20, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25922519

ABSTRACT

Inositol 1,4,5-trisphosphate receptor (IP3R) binding protein released with IP3 (IRBIT) contributes to various physiological events (electrolyte transport and fluid secretion, mRNA polyadenylation, and the maintenance of genomic integrity) through its interaction with multiple targets. However, little is known about the physiological role of IRBIT in the brain. Here we identified calcium calmodulin-dependent kinase II alpha (CaMKIIα) as an IRBIT-interacting molecule in the central nervous system. IRBIT binds to and suppresses CaMKIIα kinase activity by inhibiting the binding of calmodulin to CaMKIIα. In addition, we show that mice lacking IRBIT present with elevated catecholamine levels, increased locomotor activity, and social abnormalities. The level of tyrosine hydroxylase (TH) phosphorylation by CaMKIIα, which affects TH activity, was significantly increased in the ventral tegmental area of IRBIT-deficient mice. We concluded that IRBIT suppresses CaMKIIα activity and contributes to catecholamine homeostasis through TH phosphorylation.


Subject(s)
Adenosylhomocysteinase/metabolism , Brain/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Catecholamines/metabolism , Homeostasis/physiology , Tyrosine 3-Monooxygenase/metabolism , Adenosylhomocysteinase/genetics , Animals , Brain/cytology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Catecholamines/genetics , HEK293 Cells , Humans , Mice , Mice, Knockout , Phosphorylation/physiology , Tyrosine 3-Monooxygenase/genetics
6.
Glia ; 65(3): 502-513, 2017 03.
Article in English | MEDLINE | ID: mdl-28063222

ABSTRACT

Astrocytes regulate hippocampal synaptic plasticity by the Ca2+ dependent release of the N-methyl d-aspartate receptor (NMDAR) co-agonist d-serine. Previous evidence indicated that d-serine release would be regulated by the intracellular Ca2+ release channel IP3 receptor (IP3 R), however, genetic deletion of IP3 R2, the putative astrocytic IP3 R subtype, had no impact on synaptic plasticity or transmission. Although IP3 R2 is widely believed to be the only functional IP3 R in astrocytes, three IP3 R subtypes (1, 2, and 3) have been identified in vertebrates. Therefore, to better understand gliotransmission, we investigated the functionality of IP3 R and the contribution of the three IP3 R subtypes to Ca2+ signalling. As a proxy for gliotransmission, we found that long-term potentiation (LTP) was impaired by dialyzing astrocytes with the broad IP3 R blocker heparin, and rescued by exogenous d-serine, indicating that astrocytic IP3 Rs regulate d-serine release. To explore which IP3 R subtypes are functional in astrocytes, we used pharmacology and two-photon Ca2+ imaging of hippocampal slices from transgenic mice (IP3 R2-/- and IP3 R2-/- ;3-/- ). This approach revealed that underneath IP3 R2-mediated global Ca2+ events are an overlooked class of IP3 R-mediated local events, occurring in astroglial processes. Notably, multiple IP3 Rs were recruited by high frequency stimulation of the Schaffer collaterals, a classical LTP induction protocol. Together, these findings show the dependence of LTP and gliotransmission on Ca2+ release by astrocytic IP3 Rs. GLIA 2017;65:502-513.


Subject(s)
Astrocytes/metabolism , Calcium Signaling/physiology , Hippocampus/cytology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Long-Term Potentiation/physiology , Age Factors , Animals , Animals, Newborn , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Electric Stimulation , In Vitro Techniques , Inositol 1,4,5-Trisphosphate Receptors/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Organ Culture Techniques , Patch-Clamp Techniques , Transfection
7.
J Neurochem ; 141(6): 790-807, 2017 06.
Article in English | MEDLINE | ID: mdl-28211945

ABSTRACT

The inositol 1,4,5-trisphosphate receptor (IP3 R) is a huge Ca2+ channel that is localized at the endoplasmic reticulum. The IP3 R releases Ca2+ from the endoplasmic reticulum upon binding to IP3 , which is produced by various extracellular stimuli through phospholipase C activation. All vertebrate organisms have three subtypes of IP3 R genes, which have distinct properties of IP3 -binding and Ca2+ sensitivity, and are differently regulated by phosphorylation and by their associated proteins. Each cell type expresses the three subtypes of IP3 R in a distinct proportion, which is important for creating and maintaining spatially and temporally appropriate intracellular Ca2+ level patterns for the regulation of specific physiological phenomena. Of the three types of IP3 Rs, the type 1 receptor (IP3 R1) is dominantly expressed in the brain and is important for brain function. Recent emerging evidence suggests that abnormal Ca2+ signals from the IP3 R1 are closely associated with human brain pathology. In this review, we focus on the recent advances in our knowledge of the regulation of IP3 R1 and its functional implication in human brain diseases, as revealed by IP3 R mutation studies and analysis of human disease-associated genes. This article is part of the mini review series "60th Anniversary of the Japanese Society for Neurochemistry".


Subject(s)
Brain/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Animals , Humans
8.
Proc Natl Acad Sci U S A ; 111(38): E3966-75, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25201980

ABSTRACT

The inositol 1,4,5-trisphosphate receptor (IP3R) in the endoplasmic reticulum mediates calcium signaling that impinges on intracellular processes. IP3Rs are allosteric proteins comprising four subunits that form an ion channel activated by binding of IP3 at a distance. Defective allostery in IP3R is considered crucial to cellular dysfunction, but the specific mechanism remains unknown. Here we demonstrate that a pleiotropic enzyme transglutaminase type 2 targets the allosteric coupling domain of IP3R type 1 (IP3R1) and negatively regulates IP3R1-mediated calcium signaling and autophagy by locking the subunit configurations. The control point of this regulation is the covalent posttranslational modification of the Gln2746 residue that transglutaminase type 2 tethers to the adjacent subunit. Modification of Gln2746 and IP3R1 function was observed in Huntington disease models, suggesting a pathological role of this modification in the neurodegenerative disease. Our study reveals that cellular signaling is regulated by a new mode of posttranslational modification that chronically and enzymatically blocks allosteric changes in the ligand-gated channels that relate to disease states.


Subject(s)
Autophagy , Calcium Signaling , GTP-Binding Proteins/metabolism , Huntington Disease/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Protein Processing, Post-Translational , Transglutaminases/metabolism , Allosteric Regulation/genetics , Animals , Disease Models, Animal , GTP-Binding Proteins/genetics , HeLa Cells , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Inositol 1,4,5-Trisphosphate Receptors/genetics , PC12 Cells , Protein Glutamine gamma Glutamyltransferase 2 , Protein Structure, Tertiary , Rats , Transglutaminases/genetics
9.
J Neurosci ; 33(30): 12186-96, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23884927

ABSTRACT

The structural maintenance of neural circuits is critical for higher brain functions in adulthood. Although several molecules have been identified as regulators for spine maintenance in hippocampal and cortical neurons, it is poorly understood how Purkinje cell (PC) spines are maintained in the mature cerebellum. Here we show that the calcium channel type 1 inositol trisphosphate receptor (IP3R1) in PCs plays a crucial role in controlling the maintenance of parallel fiber (PF)-PC synaptic circuits in the mature cerebellum in vivo. Significantly, adult mice lacking IP3R1 specifically in PCs (L7-Cre;Itpr1(flox/flox)) showed dramatic increase in spine density and spine length of PCs, despite having normal spines during development. In addition, the abnormally rearranged PF-PC synaptic circuits in mature cerebellum caused unexpectedly severe ataxia in adult L7-Cre;Itpr1(flox/flox) mice. Our findings reveal a specific role for IP3R1 in PCs not only as an intracellular mediator of cerebellar synaptic plasticity induction, but also as a critical regulator of PF-PC synaptic circuit maintenance in the mature cerebellum in vivo; this mechanism may underlie motor coordination and learning in adults.


Subject(s)
Cerebellar Ataxia/physiopathology , Cerebellum/physiology , Dendritic Spines/physiology , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Purkinje Cells/physiology , Animals , Cerebellar Ataxia/genetics , Cerebellar Ataxia/pathology , Cerebellum/cytology , Cerebellum/pathology , Chimera , Dendritic Spines/pathology , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Mutant Strains , Neuronal Plasticity/physiology , Nystagmus, Optokinetic/physiology , Purkinje Cells/cytology , Purkinje Cells/ultrastructure , Reflex, Vestibulo-Ocular/physiology , Synapses/physiology
10.
Transgenic Res ; 22(6): 1241-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23624740

ABSTRACT

We generated a transgenic (Tg) mouse line expressing Cre recombinase under the control of the Gpr88 promoter within a bacterial artificial chromosome clone. We crossed the established Tg mice with reporter mice (CAG-CAT-Z Tg), which express Escherichia coli lacZ in response to Cre-mediated excision of the loxP-flanked chloramphenicol acetyltransferase gene, and examined the Cre activity in the Tg mouse brains by assessing ß-galactosidase activity. Cre activity was specifically detected in the caudate-putamen, nucleus accumbens, and olfactory tubercle of the Gpr88-Cre Tg mouse brain. Medium spiny neurons within the caudate-putamen exhibited Cre activity. Thus, Gpr88-Cre Tg mice could be a useful tool for analyzing the function of the basal ganglia by using Cre/loxP systems.


Subject(s)
Integrases/genetics , Promoter Regions, Genetic , Receptors, G-Protein-Coupled/genetics , Animals , Basal Ganglia/metabolism , Brain/metabolism , Chromosomes, Artificial, Bacterial/genetics , Escherichia coli , Integrases/metabolism , Lac Operon , Mice , Mice, Transgenic , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
11.
J Neurosci ; 31(49): 18155-65, 2011 Dec 07.
Article in English | MEDLINE | ID: mdl-22159127

ABSTRACT

Global brain state dynamics regulate plasticity in local cortical circuits, but the underlying cellular and molecular mechanisms are unclear. Here, we demonstrate that astrocyte Ca(2+) signaling provides a critical bridge between cholinergic activation, associated with attention and vigilance states, and somatosensory plasticity in mouse barrel cortex in vivo. We investigated first whether a combined stimulation of mouse whiskers and the nucleus basalis of Meynert (NBM), the principal source of cholinergic innervation to the cortex, leads to enhanced whisker-evoked local field potential. This plasticity is dependent on muscarinic acetylcholine receptors (mAChR) and N-methyl-d-aspartic acid receptors (NMDARs). During the induction of this synaptic plasticity, we find that astrocytic [Ca(2+)](i) is pronouncedly elevated, which is blocked by mAChR antagonists. The elevation of astrocytic [Ca(2+)](i) is crucial in this type of synaptic plasticity, as the plasticity could not be induced in inositol-1,4,5-trisphosphate receptor type 2 knock-out (IP(3)R2-KO) mice, in which astrocytic [Ca(2+)](i) surges are diminished. Moreover, NBM stimulation led to a significant increase in the extracellular concentration of the NMDAR coagonist d-serine in wild-type mice when compared to IP(3)R2-KO mice. Finally, plasticity in IP(3)R2-KO mice could be rescued by externally supplying d-serine. Our data present coherent lines of in vivo evidence for astrocytic involvement in cortical plasticity. These findings suggest an unexpected role of astrocytes as a gate for cholinergic plasticity in the cortex.


Subject(s)
Astrocytes/physiology , Basal Nucleus of Meynert/physiology , Calcium Signaling/physiology , Cholinergic Agents/metabolism , Neuronal Plasticity/physiology , Somatosensory Cortex/physiology , Acetylcholine/pharmacology , Action Potentials/drug effects , Action Potentials/genetics , Afferent Pathways/physiology , Aniline Compounds/metabolism , Animals , Astrocytes/drug effects , Atropine/pharmacology , Calcium Signaling/drug effects , Calcium Signaling/genetics , Chromatography, High Pressure Liquid , Evoked Potentials, Somatosensory/drug effects , Evoked Potentials, Somatosensory/genetics , Excitatory Amino Acid Antagonists/pharmacology , Fluoresceins/metabolism , Inositol 1,4,5-Trisphosphate Receptors/deficiency , Male , Mice , Mice, Knockout , Microdialysis/methods , Muscarinic Antagonists/pharmacology , Nerve Growth Factors/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Neurons/drug effects , Neurons/metabolism , Physical Stimulation , Piperazines/pharmacology , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , Serine/pharmacology , Somatosensory Cortex/drug effects , Sulfonamides/metabolism , Thiazoles/metabolism , Vibrissae/innervation
12.
Biochem Biophys Res Commun ; 429(3-4): 210-3, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23131553

ABSTRACT

Amyotrophic Lateral Sclerosis (ALS) is a devastating neurodegenerative disease causing progressive paralysis of the patient followed by death on average 3-5 years after diagnosis. Disease pathology is multi-factorial including the process of excitotoxicity that induces cell death by cytosolic Ca(2+) overload. In this study, we increased the neuronal expression of an endoplasmic reticulum (ER) Ca(2+) release channel, inositol 1,4,5-trisphosphate receptor 2 (IP(3)R2), to assess whether increased cytosolic Ca(2+) originating from the ER is detrimental for neurons. Overexpression of IP(3)R2 in N2a cells using a Thy1.2-IP(3)R2 construct increases cytosolic Ca(2+) concentrations evoked by bradykinin. In addition, mice generated from this construct have increased expression of IP(3)R2 in the spinal cord and brain. This overexpression of IP(3)R2 does not affect symptom onset, but decreases disease duration and shortens the lifespan of the ALS mice significantly. These data suggest that ER Ca(2+) released by IP(3) receptors may be detrimental in ALS and that motor neurons are vulnerable to impaired Ca(2+) metabolism.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Inositol 1,4,5-Trisphosphate Receptors/biosynthesis , Neurons/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Superoxide Dismutase/genetics , Superoxide Dismutase-1
13.
Proc Natl Acad Sci U S A ; 105(25): 8643-8, 2008 Jun 24.
Article in English | MEDLINE | ID: mdl-18552177

ABSTRACT

Intercellular cross-talk between osteoblasts and osteoclasts is important for controlling bone remolding and maintenance. However, the precise molecular mechanism by which osteoblasts regulate osteoclastogenesis is still largely unknown. Here, we show that osteoblasts can induce Ca(2+) oscillation-independent osteoclastogenesis. We found that bone marrow-derived monocyte/macrophage precursor cells (BMMs) lacking inositol 1,4,5-trisphosphate receptor type2 (IP(3)R2) did not exhibit Ca(2+) oscillation or differentiation into multinuclear osteoclasts in response to recombinant receptor activator of NF-kappaB ligand/macrophage colony-stimulating factor stimulation. IP(3)R2 knockout BMMs, however, underwent osteoclastogenesis when they were cocultured with osteoblasts or in vivo in the absence of Ca(2+) oscillation. Furthermore, we found that Ca(2+) oscillation-independent osteoclastogenesis was insensitive to FK506, a calcineurin inhibitor. Taken together, we conclude that both Ca(2+) oscillation/calcineurin-dependent and -independent signaling pathways contribute to NFATc1 activation, leading to efficient osteoclastogenesis in vivo.


Subject(s)
Calcium/metabolism , NFATC Transcription Factors/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Animals , Cell Communication , Cell Differentiation , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mice , Mice, Knockout , Models, Biological , NF-kappa B/metabolism , RANK Ligand/metabolism , Signal Transduction
14.
J Biol Chem ; 284(42): 29158-69, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19706611

ABSTRACT

The inositol 1,4,5-trisphosphate (IP(3)) receptor (IP(3)R) is an intracellular IP(3)-gated calcium (Ca(2+)) release channel and plays important roles in regulation of numerous Ca(2+)-dependent cellular responses. Many intracellular modulators and IP(3)R-binding proteins regulate the IP(3)R channel function. Here we identified G-protein-coupled receptor kinase-interacting proteins (GIT), GIT1 and GIT2, as novel IP(3)R-binding proteins. We found that both GIT1 and GIT2 directly bind to all three subtypes of IP(3)R. The interaction was favored by the cytosolic Ca(2+) concentration and it functionally inhibited IP(3)R activity. Knockdown of GIT induced and accelerated caspase-dependent apoptosis in both unstimulated and staurosporine-treated cells, which was attenuated by wild-type GIT1 overexpression or pharmacological inhibitors of IP(3)R, but not by a mutant form of GIT1 that abrogates the interaction. Thus, we conclude that GIT inhibits apoptosis by modulating the IP(3)R-mediated Ca(2+) signal through a direct interaction with IP(3)R in a cytosolic Ca(2+)-dependent manner.


Subject(s)
Apoptosis , Calcium/metabolism , Gene Expression Regulation , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Cytosol/metabolism , HeLa Cells , Humans , Mice , Molecular Sequence Data , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Sequence Homology, Amino Acid , Signal Transduction
15.
Biochem Biophys Res Commun ; 369(1): 213-9, 2008 Apr 25.
Article in English | MEDLINE | ID: mdl-18241669

ABSTRACT

Inositol 1,4,5-trisphosphate receptor (IP(3)R) plays a crucial role in generating Ca(2+) signaling and three subtypes of IP(3)R have been identified. In spite of a high degree of similarity among these subtypes, their effects on spatio-temporal Ca(2+) patterns are specific and diverse; therefore the physiological significance of the differential expression levels of IP(3)R subtypes in various tissues remains unknown. Here, we examined the relative contribution of the specific subtype of IP(3)Rs to the agonist-induced Ca(2+) signaling and contraction in IP(3)R-deficient vascular smooth muscle cells and found that IP(3)R1 deficient cells exclusively showed less sensitivity to the agonist, compared to those from the other genotypes. We also found that IP(3)R1 dominantly expressed in vascular aortae on a consistent basis, and that phenylephrine (PE)-induced aortic muscle contraction was reduced specifically in IP(3)R1-deficient aortae. Taken together, we concluded that IP(3)R1 plays a predominant role in the function of the vascular smooth muscle in vivo.


Subject(s)
Aorta/physiology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Animals , Cells, Cultured , Mice , Mice, Knockout
16.
Biochim Biophys Acta Mol Cell Res ; 1865(11 Pt B): 1733-1744, 2018 11.
Article in English | MEDLINE | ID: mdl-29777722

ABSTRACT

Spinocerebellar ataxia (SCA) is a neural disorder, which is caused by degenerative changes in the cerebellum. SCA is primarily characterized by gait ataxia, and additional clinical features include nystagmus, dysarthria, tremors and cerebellar atrophy. Forty-four hereditary SCAs have been identified to date, along with >35 SCA-associated genes. Despite the great diversity and distinct functionalities of the SCA-related genes, accumulating evidence supports the occurrence of a common pathophysiological event among several hereditary SCAs. Altered calcium (Ca2+) homeostasis in the Purkinje cells (PCs) of the cerebellum has been proposed as a possible pathological SCA trigger. In support of this, signaling events that are initiated from or lead to aberrant Ca2+ release from the type 1 inositol 1,4,5-trisphosphate receptor (IP3R1), which is highly expressed in cerebellar PCs, seem to be closely associated with the pathogenesis of several SCA types. In this review, we summarize the current research on pathological hereditary SCA events, which involve altered Ca2+ homeostasis in PCs, through IP3R1 signaling.


Subject(s)
Calcium Signaling , Calcium/metabolism , Disease Susceptibility , Spinocerebellar Ataxias/etiology , Spinocerebellar Ataxias/metabolism , Animals , Cerebellum/metabolism , Cerebellum/physiopathology , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mutation , Purkinje Cells/metabolism
17.
J Neurosci ; 26(42): 10916-24, 2006 Oct 18.
Article in English | MEDLINE | ID: mdl-17050730

ABSTRACT

Here, we show that cultured Purkinje cells from inositol 1,4,5-trisphosphate receptor type 1 knock-out (IP3R1KO) mice exhibited abnormal dendritic morphology. Interestingly, despite the huge amount of IP3R1 expression in Purkinje cells, IP3R1 in granule cells, not in the Purkinje cells, was responsible for the shape of Purkinje cell dendrites. We also found that BDNF application rescued the dendritic abnormality of IP3R1KO Purkinje cells, and that the increase in BDNF expression in response to activation of AMPA receptor (AMPAR) and metabotropic glutamate receptor (mGluR) was impaired in IP3R1KO cerebellar granule cells. In addition, we observed abnormalities in the dendritic morphology of Purkinje cells and in the ultrastructure of parallel fiber-Purkinje cell (PF-PC) synapses in IP3R1KO mice in vivo. We concluded that activation of AMPAR and mGluR increases BDNF expression through IP3R1-mediated signaling in cerebellar granule cells, which contributes to the dendritic outgrowth of Purkinje cells intercellularly, possibly by modifying PF-PC synaptic efficacy.


Subject(s)
Brain-Derived Neurotrophic Factor/biosynthesis , Calcium Channels/physiology , Dendrites/metabolism , Purkinje Cells/cytology , Purkinje Cells/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/physiology , Calcium Channels/deficiency , Cells, Cultured , Dendrites/genetics , Inositol 1,4,5-Trisphosphate Receptors , Mice , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/deficiency
18.
Sci Rep ; 7(1): 10260, 2017 08 31.
Article in English | MEDLINE | ID: mdl-28860523

ABSTRACT

Signal transduction in sensory neurons of the mammalian vomeronasal organ (VNO) involves the opening of the canonical transient receptor potential channel Trpc2, a Ca2+-permeable cation channel that is activated by diacylglycerol and inhibited by Ca2+-calmodulin. There has been a long-standing debate about the extent to which the second messenger inositol 1,4,5-trisphosphate (InsP3) and type 3 InsP3 receptor (InsP3R3) are involved in the opening of Trpc2 channels and in sensory activation of the VNO. To address this question, we investigated VNO function of mice carrying a knockout mutation in the Itpr3 locus causing a loss of InsP3R3. We established a new method to monitor Ca2+ in the endoplasmic reticulum of vomeronasal sensory neurons (VSNs) by employing the GFP-aequorin protein sensor erGAP2. We also performed simultaneous InsP3 photorelease and Ca2+ monitoring experiments, and analysed Ca2+ dynamics, sensory currents, and action potential or field potential responses in InsP3R3-deficient VSNs. Disruption of Itpr3 abolished or minimized the Ca2+ transients evoked by photoactivated InsP3, but there was virtually no effect on sensory activation of VSNs. Therefore, InsP3R3 is dispensable for primary chemoelectrical transduction in mouse VNO. We conclude that InsP3R3 is not required for gating of Trpc2 in VSNs.


Subject(s)
Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mammals , Sensation , Vomeronasal Organ/physiology , Action Potentials , Animals , Calcium/metabolism , Calcium Signaling , Calmodulin/metabolism , Evoked Potentials , Extracellular Space/metabolism , Gene Expression , Intracellular Space/metabolism , Ligands , Mice , Molecular Imaging , Photolysis , Sensory Receptor Cells/physiology , Signal Transduction
19.
J Neurol ; 264(7): 1444-1453, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28620721

ABSTRACT

We report three affected members, a mother and her two children, of a non-consanguineous Irish family who presented with a suspected autosomal dominant spinocerebellar ataxia characterized by early motor delay, poor coordination, gait ataxia, and dysarthria. Whole exome sequencing identified a novel missense variant (c.106C>T; p.[Arg36Cys]) in the suppressor domain of type 1 inositol 1,4,5-trisphosphate receptor gene (ITPR1) as the cause of the disorder, resulting in a molecular diagnosis of spinocerebellar ataxia type 29. In the absence of grandparental DNA, microsatellite genotyping of healthy family members was used to confirm the de novo status of the ITPR1 variant in the affected mother, which supported pathogenicity. The Arg36Cys variant exhibited a significantly higher IP3-binding affinity than wild-type (WT) ITPR1 and drastically changed the property of the intracellular Ca2+ signal from a transient to a sigmoidal pattern, supporting a gain-of-function disease mechanism. To date, ITPR1 mutation has been associated with a loss-of-function effect, likely due to reduced Ca2+ release. This is the first gain-of-function mechanism to be associated with ITPR1-related SCA29, providing novel insights into how enhanced Ca2+ release can also contribute to the pathogenesis of this neurological disorder.


Subject(s)
Calcium Signaling/genetics , Calcium Signaling/physiology , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mutation, Missense , Spinocerebellar Degenerations/genetics , Spinocerebellar Degenerations/metabolism , Family , Female , Humans , Male , Spinocerebellar Degenerations/diagnostic imaging
20.
Sci STKE ; 2005(313): pe53, 2005 Dec 06.
Article in English | MEDLINE | ID: mdl-16333017

ABSTRACT

The endoplasmic reticulum (ER) is not simply a uniform continuous organelle, but is spatially and functionally heterogeneous with nonuniform distribution of endoplasmic Ca(2+)-handling proteins, such as Ca(2+)-binding proteins, Ca(2+) pumps, and Ca(2+)-release channels. Such nonuniform distribution of Ca(2+)-handling proteins is thought to create a spatially divided calcium store and to contribute to the generation of complex intracellular Ca(2+) dynamics. In addition to the particular distribution of these Ca(2+)-handling proteins within ER, extracellular stimuli may also stimulate the formation of dynamic new ER compartments containing Ca(2+)-handling proteins. These compartments containing Ca(2+)-handling proteins have potential roles in Ca(2+) signaling; specifically, they may function as "induced coupling domains" between the ER and plasma membrane, thereby allowing Ca(2+) entry into the ER.


Subject(s)
Calcium Signaling/physiology , Cell Compartmentation , Endoplasmic Reticulum/physiology , Animals , Calcium Channels/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/physiology , Endoplasmic Reticulum/ultrastructure , Humans , Inositol 1,4,5-Trisphosphate Receptors , Membrane Proteins/physiology , Multiprotein Complexes , Neoplasm Proteins/physiology , RNA Interference , Receptors, Cytoplasmic and Nuclear/physiology , Stromal Interaction Molecule 1 , Type C Phospholipases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL