Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters










Publication year range
1.
Arterioscler Thromb Vasc Biol ; 44(4): 772-783, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38385293

ABSTRACT

Airway epithelial cells play an indispensable role in protecting the lung from inhaled pathogens and allergens by releasing an array of mediators that orchestrate inflammatory and immune responses when confronted with harmful environmental triggers. While this process is undoubtedly important for containing the effects of various harmful insults, dysregulation of the inflammatory response can cause lung diseases including asthma, chronic obstructive pulmonary disease, and pulmonary fibrosis. A key cellular mechanism that underlies the inflammatory responses in the airway is calcium signaling, which stimulates the production and release of chemokines, cytokines, and prostaglandins from the airway epithelium. In this review, we discuss the role of major Ca2+ signaling pathways found in airway epithelial cells and their contributions to airway inflammation, mucociliary clearance, and surfactant production. We highlight the importance of store-operated Ca2+ entry as a major signaling hub in these processes and discuss therapeutic implications of targeting Ca2+ signaling for airway inflammation.


Subject(s)
Asthma , Calcium Signaling , Humans , Asthma/metabolism , Lung , Epithelial Cells/metabolism , Inflammation/metabolism
2.
J Physiol ; 602(8): 1449-1462, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37029630

ABSTRACT

Store operated Ca2+ entry (SOCE) is a ubiquitous signalling module with established roles in the immune system, secretion and muscle development. Recent evidence supports a complex role for SOCE in the nervous system. In this review we present an update of the current knowledge on SOCE function in the brain with a focus on its role as a regulator of brain activity and excitability.

3.
Neuron ; 112(2): 247-263.e6, 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-37924811

ABSTRACT

Oligodendrocytes are the primary producers of many extracellular matrix (ECM)-related proteins found in the CNS. Therefore, oligodendrocytes play a critical role in the determination of brain stiffness, node of Ranvier formation, perinodal ECM deposition, and perineuronal net formation, all of which depend on the ECM. Nevertheless, the transcription factors that control ECM-related gene expression in oligodendrocytes remain unknown. Here, we found that the transcription factor Osterix (also known as Sp7) binds in proximity to genes important for CNS ECM and node of Ranvier formation and mediates their expression. Oligodendrocyte-specific ablation of Sp7 changes ECM composition and brain stiffness and results in aberrant node of Ranvier formation. Sp7 is known to control osteoblast maturation and bone formation. Our comparative analyses suggest that Sp7 plays a conserved biological role in oligodendrocytes and in bone-forming cells, where it mediates brain and bone tissue stiffness by controlling expression of ECM components.


Subject(s)
Oligodendroglia , Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Oligodendroglia/physiology , Extracellular Matrix/metabolism , Bone and Bones/metabolism , Extracellular Matrix Proteins/metabolism , Gene Expression
4.
Nat Commun ; 14(1): 5500, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37679321

ABSTRACT

Astrocytes contribute to brain inflammation in neurological disorders but the molecular mechanisms controlling astrocyte reactivity and their relationship to neuroinflammatory endpoints are complex and poorly understood. In this study, we assessed the role of the calcium channel, Orai1, for astrocyte reactivity and inflammation-evoked depression behaviors in mice. Transcriptomics and metabolomics analysis indicated that deletion of Orai1 in astrocytes downregulates genes in inflammation and immunity, metabolism, and cell cycle pathways, and reduces cellular metabolites and ATP production. Systemic inflammation by peripheral lipopolysaccharide (LPS) increases hippocampal inflammatory markers in WT but not in astrocyte Orai1 knockout mice. Loss of Orai1 also blunts inflammation-induced astrocyte Ca2+ signaling and inhibitory neurotransmission in the hippocampus. In line with these cellular changes, Orai1 knockout mice showed amelioration of LPS-evoked depression-like behaviors including anhedonia and helplessness. These findings identify Orai1 as an important signaling hub controlling astrocyte reactivity and astrocyte-mediated brain inflammation that is commonly observed in many neurological disorders.


Subject(s)
Astrocytes , Encephalitis , Animals , Mice , Depression/genetics , Lipopolysaccharides , Inflammation/genetics , Calcium Channels/genetics , Mice, Knockout , ORAI1 Protein/genetics
5.
Proc Natl Acad Sci U S A ; 120(17): e2217396120, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37068235

ABSTRACT

Octopamine is a well-established invertebrate neurotransmitter involved in fight or flight responses. In mammals, its function was replaced by epinephrine. Nevertheless, it is present at trace amounts and can modulate the release of monoamine neurotransmitters by a yet unidentified mechanism. Here, through a multidisciplinary approach utilizing in vitro and in vivo models of α-synucleinopathy, we uncovered an unprecedented role for octopamine in driving the conversion from toxic to neuroprotective astrocytes in the cerebral cortex by fostering aerobic glycolysis. Physiological levels of neuron-derived octopamine act on astrocytes via a trace amine-associated receptor 1-Orai1-Ca2+-calcineurin-mediated signaling pathway to stimulate lactate secretion. Lactate uptake in neurons via the monocarboxylase transporter 2-calcineurin-dependent pathway increases ATP and prevents neurodegeneration. Pathological increases of octopamine caused by α-synuclein halt lactate production in astrocytes and short-circuits the metabolic communication to neurons. Our work provides a unique function of octopamine as a modulator of astrocyte metabolism and subsequent neuroprotection with implications to α-synucleinopathies.


Subject(s)
Octopamine , alpha-Synuclein , Animals , alpha-Synuclein/metabolism , Astrocytes/metabolism , Calcineurin/metabolism , Lactates/metabolism , Mammals/metabolism , Neuroprotection , Neurotransmitter Agents/metabolism , Octopamine/metabolism
6.
Elife ; 122023 02 20.
Article in English | MEDLINE | ID: mdl-36806330

ABSTRACT

Ca2+ release-activated Ca2+ (CRAC) channels are activated by direct physical interactions between Orai1, the channel protein, and STIM1, the endoplasmic reticulum Ca2+ sensor. A hallmark of CRAC channels is fast Ca2+-dependent inactivation (CDI) which provides negative feedback to limit Ca2+ entry through CRAC channels. Although STIM1 is thought to be essential for CDI, its molecular mechanism remains largely unknown. Here, we examined a poorly understood gain-of-function (GOF) human Orai1 disease mutation, L138F, that causes tubular aggregate myopathy. Through pairwise mutational analysis, we determine that large amino acid substitutions at either L138 or the neighboring T92 locus located on the pore helix evoke highly Ca2+-selective currents in the absence of STIM1. We find that the GOF phenotype of the L138 pathogenic mutation arises due to steric clash between L138 and T92. Surprisingly, strongly activating L138 and T92 mutations showed CDI in the absence of STIM1, contradicting prevailing views that STIM1 is required for CDI. CDI of constitutively open T92W and L138F mutants showed enhanced intracellular Ca2+ sensitivity, which was normalized by re-adding STIM1 to the cells. Truncation of the Orai1 C-terminus reduced T92W CDI, indicating a key role for the Orai1 C-terminus for CDI. Overall, these results identify the molecular basis of a disease phenotype with broad implications for activation and inactivation of Orai1 channels.


Subject(s)
Calcium Channels , Calcium Release Activated Calcium Channels , Humans , Calcium Channels/metabolism , ORAI1 Protein/genetics , Mutation , Calcium Release Activated Calcium Channels/genetics , Gain of Function Mutation , Stromal Interaction Molecule 1/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism
7.
Sci Adv ; 9(4): eade7002, 2023 01 27.
Article in English | MEDLINE | ID: mdl-36706180

ABSTRACT

Microglia are important mediators of neuroinflammation, which underlies neuropathic pain. However, the molecular checkpoints controlling microglial reactivity are not well-understood. Here, we investigated the role of Orai1 channels for microglia-mediated neuroinflammation following nerve injury and find that deletion of Orai1 in microglia attenuates Ca2+ signaling and the production of inflammatory cytokines by proalgesic agonists. Conditional deletion of Orai1 attenuated microglial proliferation in the dorsal horn, spinal cytokine levels, and potentiation of excitatory neurotransmission following peripheral nerve injury. These cellular effects were accompanied by mitigation of pain hyperalgesia in microglial Orai1 knockout mice. A small-molecule Orai1 inhibitor, CM4620, similarly mitigated allodynia in male mice. Unexpectedly, these protective effects were not seen in female mice, revealing sexual dimorphism in Orai1 regulation of microglial reactivity and hyperalgesia. Together, these findings indicate that Orai1 channels are key regulators of the sexually dimorphic role of microglia for the neuroinflammation that underlies neuropathic pain.


Subject(s)
Microglia , Neuralgia , Mice , Male , Female , Animals , Microglia/metabolism , Hyperalgesia/genetics , Neuroinflammatory Diseases , Neuralgia/genetics , Mice, Knockout , Cytokines/metabolism , Spinal Cord , ORAI1 Protein/genetics
8.
EMBO Mol Med ; 14(9): e15687, 2022 09 07.
Article in English | MEDLINE | ID: mdl-35919953

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by dysregulated intestinal immune responses. Using mass cytometry (CyTOF) to analyze the immune cell composition in the lamina propria (LP) of patients with ulcerative colitis (UC) and Crohn's disease (CD), we observed an enrichment of CD4+ effector T cells producing IL-17A and TNF, CD8+ T cells producing IFNγ, T regulatory (Treg) cells, and innate lymphoid cells (ILC). The function of these immune cells is regulated by store-operated Ca2+ entry (SOCE), which results from the opening of Ca2+ release-activated Ca2+ (CRAC) channels formed by ORAI and STIM proteins. We observed that the pharmacologic inhibition of SOCE attenuated the production of proinflammatory cytokines including IL-2, IL-4, IL-6, IL-17A, TNF, and IFNγ by human colonic T cells and ILCs, reduced the production of IL-6 by B cells and the production of IFNγ by myeloid cells, but had no effect on the viability, differentiation, and function of intestinal epithelial cells. T cell-specific deletion of CRAC channel genes in mice showed that Orai1, Stim1, and Stim2-deficient T cells have quantitatively distinct defects in SOCE, which correlate with gradually more pronounced impairment of cytokine production by Th1 and Th17 cells and the severity of IBD. Moreover, the pharmacologic inhibition of SOCE with a selective CRAC channel inhibitor attenuated IBD severity and colitogenic T cell function in mice. Our data indicate that SOCE inhibition may be a suitable new approach for the treatment of IBD.


Subject(s)
Calcium Release Activated Calcium Channels , Inflammatory Bowel Diseases , Animals , CD8-Positive T-Lymphocytes/metabolism , Calcium/metabolism , Calcium Channels/metabolism , Humans , Immunity, Innate , Interleukin-17/metabolism , Interleukin-6/metabolism , Mice , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/genetics , Th17 Cells/metabolism
9.
J Biol Chem ; 298(8): 102157, 2022 08.
Article in English | MEDLINE | ID: mdl-35724962

ABSTRACT

Stromal interaction molecule 1 (STIM1) is a widely expressed protein that functions as the endoplasmic reticulum (ER) Ca2+ sensor and activator of Orai1 channels. In resting cells with replete Ca2+ stores, an inhibitory clamp formed by the coiled-coil 1 (CC1) domain interacting with the CRAC-activation domain (CAD) of STIM1 helps keep STIM1 in a quiescent state. Following depletion of ER Ca2+ stores, the brake is released, allowing CAD to extend away from the ER membrane and enabling it to activate Orai1 channels. However, the molecular determinants of CC1-CAD interactions that enforce the inhibitory clamp are incompletely understood. Here, we performed Ala mutagenesis in conjunction with live-cell FRET analysis to examine residues in CC1 and CAD that regulate the inhibitory clamp. Our results indicate that in addition to previously identified hotspots in CC1⍺1 and CC3, several hydrophobic residues in CC2 and the apex region of CAD are critical for CC1-CAD interactions. Mutations in these residues loosen the CC1-CAD inhibitory clamp to release CAD from CC1 in cells with replete Ca2+ stores. By contrast, altering the hydrophobic residues L265 and L273 strengthens the clamp to prevent STIM1 activation. Inclusion of the inactivation domain of STIM1 helps stabilize CC1-CAD interaction in several mutants to prevent spontaneous STIM1 activation. In addition, R426C, a human disease-linked mutation in CC3, affects the clamp but also impairs Orai1 binding to inhibit CRAC channel activation. These results identify the CC2, apex, and inactivation domain regions of STIM1 as important determinants of STIM1 activation.


Subject(s)
Calcium Signaling , Endoplasmic Reticulum , Stromal Interaction Molecule 1 , Calcium/metabolism , Calcium Channels/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Protein Domains , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism
10.
J Immunol ; 208(10): 2390-2402, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35459743

ABSTRACT

Respiratory viruses stimulate the release of antiviral IFNs from the airway epithelium. Previous studies have shown that asthmatic patients show diminished release of type I and type III IFNs from bronchial epithelia. However, the mechanism of this suppression is not understood. In this study, we report that extracellular nucleotides and histamine, which are elevated in asthmatic airways, strongly inhibit release of type I and type III IFNs from human bronchial airway epithelial cells (AECs). Specifically, ATP, UTP, and histamine all inhibited the release of type I and type III IFNs from AECs induced by activation of TLR3, retinoic acid-inducible gene I (RIG-I), or cyclic GMP-AMP synthase-STING. This inhibition was at least partly mediated by Gq signaling through purinergic P2Y2 and H1 receptors, but it did not involve store-operated calcium entry. Pharmacological blockade of protein kinase C partially reversed inhibition of IFN production. Conversely, direct activation of protein kinase C with phorbol esters strongly inhibited TLR3- and RIG-I-mediated IFN production. Inhibition of type I and type III IFNs by ATP, UTP, histamine, and the proteinase-activated receptor 2 (PAR2) receptor agonist SLIGKV also occurred in differentiated AECs grown at an air-liquid interface, indicating that the suppression is conserved following mucociliary differentiation. Importantly, histamine and, more strikingly, ATP inhibited type I IFN release from human airway cells infected with live influenza A virus or rhinovirus 1B. These results reveal an important role for extracellular nucleotides and histamine in attenuating the induction of type I and III IFNs from AECs and help explain the molecular basis of the suppression of IFN responses in asthmatic patients.


Subject(s)
DEAD Box Protein 58 , Histamine , Interferons , Nucleotides , Receptors, Immunologic , Respiratory Mucosa , Toll-Like Receptor 3 , Adenosine Triphosphate/immunology , DEAD Box Protein 58/immunology , Epithelial Cells/immunology , Histamine/immunology , Humans , Interferons/immunology , Nucleotides/immunology , Protein Kinase C/immunology , Receptors, Immunologic/immunology , Respiratory Mucosa/immunology , Toll-Like Receptor 3/immunology , Uridine Triphosphate/metabolism , Uridine Triphosphate/pharmacology
11.
Nat Commun ; 13(1): 2033, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440113

ABSTRACT

TCR stimulation triggers Ca2+ signals that are critical for T cell function and immunity. Several pore-forming α and auxiliary ß subunits of voltage-gated Ca2+ channels (VGCC) were reported in T cells, but their mechanism of activation remains elusive and their contribution to Ca2+ signaling in T cells is controversial. We here identify CaVß1, encoded by Cacnb1, as a regulator of T cell function. Cacnb1 deletion enhances apoptosis and impairs the clonal expansion of T cells after lymphocytic choriomeningitis virus (LCMV) infection. By contrast, Cacnb1 is dispensable for T cell proliferation, cytokine production and Ca2+ signaling. Using patch clamp electrophysiology and Ca2+ recordings, we are unable to detect voltage-gated Ca2+ currents or Ca2+ influx in human and mouse T cells upon depolarization with or without prior TCR stimulation. mRNAs of several VGCC α1 subunits are detectable in human (CaV3.3, CaV3.2) and mouse (CaV2.1) T cells, but they lack transcription of many 5' exons, likely resulting in N-terminally truncated and non-functional proteins. Our findings demonstrate that although CaVß1 regulates T cell function, these effects are independent of VGCC channel activity.


Subject(s)
Apoptosis , T-Lymphocytes , Animals , Apoptosis/genetics , Calcium Channels, L-Type , Cell Proliferation/genetics , Mice , Receptors, Antigen, T-Cell
12.
J Immunol ; 207(5): 1275-1287, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34389624

ABSTRACT

The airway epithelial cells (AECs) lining the conducting passageways of the lung secrete a variety of immunomodulatory factors. Among these, PGE2 limits lung inflammation and promotes bronchodilation. By contrast, IL-6 drives intense airway inflammation, remodeling, and fibrosis. The signaling that differentiates the production of these opposing mediators is not understood. In this study, we find that the production of PGE2 and IL-6 following stimulation of human AECs by the damage-associated molecular pattern extracellular ATP shares a common requirement for Ca2+ release-activated Ca2+ (CRAC) channels. ATP-mediated synthesis of PGE2 required activation of metabotropic P2Y2 receptors and CRAC channel-mediated cytosolic phospholipase A2 signaling. By contrast, ATP-evoked synthesis of IL-6 occurred via activation of ionotropic P2X receptors and CRAC channel-mediated calcineurin/NFAT signaling. In contrast to ATP, which elicited the production of both PGE2 and IL-6, the uridine nucleotide, UTP, stimulated PGE2 but not IL-6 production. These results reveal that human AECs employ unique receptor-specific signaling mechanisms with CRAC channels as a signaling nexus to regulate release of opposing immunomodulatory mediators. Collectively, our results identify P2Y2 receptors, CRAC channels, and P2X receptors as potential intervention targets for airway diseases.


Subject(s)
Dinoprostone/metabolism , Inflammation/immunology , Interleukin-6/metabolism , Respiratory Mucosa/metabolism , Adenosine Triphosphate/pharmacokinetics , Alarmins/metabolism , Calcium Release Activated Calcium Channels/metabolism , Cells, Cultured , Humans , Immunomodulation , Interleukin-6/genetics , NFATC Transcription Factors/metabolism , Phospholipases A2/metabolism , Receptors, Purinergic P2X/metabolism , Respiratory Mucosa/pathology , Signal Transduction , Uracil Nucleotides/metabolism
13.
Methods Enzymol ; 652: 213-239, 2021.
Article in English | MEDLINE | ID: mdl-34059283

ABSTRACT

Chemical modification of ion channels using the substituted cysteine accessibility method has a rich and successful history in elucidating the structural basis of ion channel function. In this approach, cysteine residues are introduced in regions of interest into the protein and their accessibility to water soluble thiol-reactive reagents is determined by monitoring ion channel activity. Because a wide range of these reagents are available with differing size, charge, and membrane solubility, the physio-chemical environment of the introduced cysteine residue and therefore the protein domain of interest can be probed with great precision. The approach has been widely employed for determining the secondary structure of specific ion channel domains, the location and nature of the channel gate, and the conformational rearrangements in the channel pore that underlie the opening/closing of the pore. In this chapter, we describe the use of these and related approaches to probe the functional architecture and gating of store-operated Orai1 channels.


Subject(s)
Cysteine , Ion Channel Gating , Calcium/metabolism , ORAI1 Protein/metabolism , Protein Domains
16.
Cell Calcium ; 94: 102366, 2021 03.
Article in English | MEDLINE | ID: mdl-33581587

ABSTRACT

Store-operated Orai channels are a primary mechanism for mobilizing Ca2+ signals in both non-excitable cells and excitable cells. The structure of the open channel, vital for understanding the mechanism of channel opening, is incompletely understood. We highlight a new study that unveils the structure of a constitutively active Orai mutant and takes us closer towards understanding the molecular basis of Orai channel activation.


Subject(s)
ORAI1 Protein/metabolism , Animals , Drosophila melanogaster/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Ion Channel Gating , ORAI1 Protein/chemistry , ORAI1 Protein/ultrastructure , Protein Conformation
17.
Cell Rep ; 33(9): 108464, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33264616

ABSTRACT

Store-operated Orai1 calcium channels function as highly Ca2+-selective ion channels and are broadly expressed in many tissues including the central nervous system, but their contributions to cognitive processing are largely unknown. Here, we report that many measures of synaptic, cellular, and behavioral models of learning are markedly attenuated in mice lacking Orai1 in forebrain excitatory neurons. Results with focal glutamate uncaging in hippocampal neurons support an essential role of Orai1 channels in amplifying NMDA-receptor-induced dendritic Ca2+ transients that drive activity-dependent spine morphogenesis and long-term potentiation at Schaffer collateral-CA1 synapses. Consistent with these signaling roles, mice lacking Orai1 in pyramidal neurons (but not interneurons) exhibit striking deficits in working and associative memory tasks. These findings identify Orai1 channels as essential regulators of dendritic spine Ca2+ signaling, synaptic plasticity, and cognition.


Subject(s)
Calcium Signaling , Calcium/metabolism , Dendritic Spines/metabolism , Glutamic Acid/metabolism , Animals , Hippocampus/metabolism , Memory , Mice , ORAI1 Protein , Pyramidal Cells/metabolism , Signal Transduction
18.
Elife ; 92020 10 30.
Article in English | MEDLINE | ID: mdl-33124982

ABSTRACT

Sulfur-aromatic interactions occur in the majority of protein structures, yet little is known about their functional roles in ion channels. Here, we describe a novel molecular motif, the M101 gate latch, which is essential for gating of human Orai1 channels via its sulfur-aromatic interactions with the F99 hydrophobic gate. Molecular dynamics simulations of different Orai variants reveal that the gate latch is mostly engaged in open but not closed channels. In experimental studies, we use metal-ion bridges to show that promoting an M101-F99 bond directly activates Orai1, whereas disrupting this interaction triggers channel closure. Mutational analysis demonstrates that the methionine residue at this position has a unique combination of length, flexibility, and chemistry to act as an effective latch for the phenylalanine gate. Because sulfur-aromatic interactions provide additional stabilization compared to purely hydrophobic interactions, we infer that the six M101-F99 pairs in the hexameric channel provide a substantial energetic contribution to Orai1 activation.


Subject(s)
Ion Channel Gating/physiology , ORAI1 Protein/metabolism , Sulfur/metabolism , HEK293 Cells , Humans , Models, Molecular , Molecular Dynamics Simulation , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ORAI1 Protein/genetics , Protein Conformation , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Sulfur/chemistry
19.
Cell Calcium ; 92: 102287, 2020 12.
Article in English | MEDLINE | ID: mdl-32932145

ABSTRACT

Cardiolipin (CL) is a cone-shaped lipid found nearly exclusively in the inner mitochondrial membrane of animal cells. Disruption of CL synthesis leads to abnormalities in mitochondrial shape and function, but the underlying causes are incompletely understood. We highlight a new study that reveals that the activity of the mitochondrial calcium uniporter (MCU) is regulated by CL, advancing our understanding of the mechanisms of CL-linked human disease.


Subject(s)
Calcium , Cardiolipins , Animals , Calcium/metabolism , Calcium Channels , Humans , Mitochondrial Membranes/metabolism
20.
J Physiol ; 598(23): 5391-5409, 2020 12.
Article in English | MEDLINE | ID: mdl-32851638

ABSTRACT

KEY POINTS: Temporal lobe epilepsy is a complex neurological disease caused by imbalance of excitation and inhibition in the brain. Growing literature implicates altered Ca2+ signalling in many aspects of epilepsy but the diversity of Ca2+ channels that regulate this syndrome are not well-understood. Here, we report that mice lacking the store-operated Ca2+ channel, Orai1, in the brain show markedly stronger seizures in response to the chemoconvulsants, kainic acid and pilocarpine. Electrophysiological analysis reveals that selective deletion of Orai1 channels in inhibitory neurons disables chemoconvulsant-induced excitation of GABAergic neurons in the CA1 hippocampus. Likewise, deletion of Orai1 in GABAergic neurons abrogates the chemoconvulsant-induced burst of spontaneous inhibitory postsynaptic currents (sIPSCs) on CA1 pyramidal neurons in the hippocampus. This loss of chemoconvulsant inhibition likely aggravates status epilepticus in Orai1 KO mice. These results identify Orai1 channels as regulators of hippocampal interneuron excitability and seizures. ABSTRACT: Store-operated Orai1 channels are a major mechanism for Ca2+ entry in many cells and mediate numerous functions including gene expression, cytokine production and gliotransmitter release. Orai1 is expressed in many regions of the mammalian brain; however, its role in regulating neuronal excitability, synaptic function and brain disorders has only now begun to be investigated. To investigate a potential role of Orai1 channels in status epilepticus induced by chemoconvulsants, we examined acute seizures evoked by intraperitoneal injections of kainic acid (KA) and pilocarpine in mice with a conditional deletion of Orai1 (or its activator STIM1) in the brain. Brain-specific Orai1 and STIM1 knockout (KO) mice exhibited significantly stronger seizures (P = 0.00003 and P < 0.00001), and higher chemoconvulsant-induced mortality (P = 0.02) compared with wildtype (WT) littermates. Electrophysiological recordings in hippocampal brain slices revealed that KA stimulated the activity of inhibitory interneurons in the CA1 hippocampus (P = 0.04) which failed to occur in Orai1 KO mice. Further, KA and pilocarpine increased the frequency of spontaneous IPSCs in CA1 pyramidal neurons >twofold (KA: P = 0.04; pilocarpine: P = 0.0002) which was abolished in Orai1 KO mice. Mice with selective deletion of Orai1 in GABAergic neurons alone also showed stronger seizures to KA (P = 0.001) and pilocarpine (P < 0.00001) and loss of chemoconvulsant-induced increases in sIPSC responses compared with WT controls. We conclude that Orai1 channels regulate chemoconvulsant-induced excitation in GABAergic neurons and that destabilization of the excitatory/inhibitory balance in Orai1 KO mice aggravates chemoconvulsant-mediated seizures. These results identify Orai1 channels as novel molecular regulators of hippocampal neuronal excitability and seizures.


Subject(s)
Hippocampus , Seizures , Animals , Kainic Acid/toxicity , Mice , ORAI1 Protein/genetics , Pilocarpine/toxicity , Pyramidal Cells , Seizures/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...