Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 6411, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37828018

ABSTRACT

Progress in neuroscience research hinges on technical advances in visualizing living brain tissue with high fidelity and facility. Current neuroanatomical imaging approaches either require tissue fixation (electron microscopy), do not have cellular resolution (magnetic resonance imaging) or only give a fragmented view (fluorescence microscopy). Here, we show how regular light microscopy together with fluorescence labeling of the interstitial fluid in the extracellular space provide comprehensive optical access in real-time to the anatomical complexity and dynamics of living brain tissue at submicron scale. Using several common fluorescence microscopy modalities (confocal, light-sheet and 2-photon microscopy) in mouse organotypic and acute brain slices and the intact mouse brain in vivo, we demonstrate the value of this straightforward 'shadow imaging' approach by revealing neurons, microglia, tumor cells and blood capillaries together with their complete anatomical tissue contexts. In addition, we provide quantifications of perivascular spaces and the volume fraction of the extracellular space of brain tissue in vivo.


Subject(s)
Brain , Neurons , Mice , Animals , Brain/diagnostic imaging , Microscopy, Fluorescence/methods , Extracellular Space , Head
2.
Elife ; 122023 04 11.
Article in English | MEDLINE | ID: mdl-37039453

ABSTRACT

Fatty acid amide hydrolase (FAAH) degrades the endocannabinoid anandamide. A polymorphism in FAAH (FAAH C385A) reduces FAAH expression, increases anandamide levels, and increases the risk of obesity. Nevertheless, some studies have found no association between FAAH C385A and obesity. We investigated whether the environmental context governs the impact of FAAH C385A on metabolic outcomes. Using a C385A knock-in mouse model, we found that FAAH A/A mice are more susceptible to glucocorticoid-induced hyperphagia, weight gain, and activation of hypothalamic AMP-activated protein kinase (AMPK). AMPK inhibition occluded the amplified hyperphagic response to glucocorticoids in FAAH A/A mice. FAAH knockdown exclusively in agouti-related protein (AgRP) neurons mimicked the exaggerated feeding response of FAAH A/A mice to glucocorticoids. FAAH A/A mice likewise presented exaggerated orexigenic responses to ghrelin, while FAAH knockdown in AgRP neurons blunted leptin anorectic responses. Together, the FAAH A/A genotype amplifies orexigenic responses and decreases anorexigenic responses, providing a putative mechanism explaining the diverging human findings.


Subject(s)
AMP-Activated Protein Kinases , Endocannabinoids , Mice , Humans , Animals , Agouti-Related Protein , Endocannabinoids/metabolism , Amidohydrolases/metabolism , Obesity
3.
J Neurosci ; 43(4): 571-583, 2023 01 25.
Article in English | MEDLINE | ID: mdl-36460464

ABSTRACT

Repeated seizures result in a persistent maladaptation of endocannabinoid (eCB) signaling, mediated part by anandamide signaling deficiency in the basolateral amygdala (BLA) that manifests as aberrant synaptic function and altered emotional behavior. Here, we determined the effect of repeated seizures (kindling) on 2-arachidonoylglycerol (2-AG) signaling on GABA transmission by directly measuring tonic and phasic eCB-mediated retrograde signaling in an in vitro BLA slice preparation from male rats. We report that both activity-dependent and muscarinic acetylcholine receptor (mAChR)-mediated depression of GABA synaptic transmission was reduced following repeated seizure activity. These effects were recapitulated in sham rats by preincubating slices with the 2-AG synthesizing enzyme inhibitor DO34. Conversely, preincubating slices with the 2-AG degrading enzyme inhibitor KML29 rescued activity-dependent 2-AG signaling, but not mAChR-mediated synaptic depression, over GABA transmission in kindled rats. These effects were not attributable to a change in cannabinoid type 1 (CB1) receptor sensitivity or altered 2-AG tonic signaling since the application of the highly selective CB1 receptor agonist CP55,940 provoked a similar reduction in GABA synaptic activity in both sham and kindled rats, while no effect of either DO34 or of the CB1 inverse agonist AM251 was observed on frequency and amplitude of spontaneous IPSCs in either sham or kindled rats. Collectively, these data provide evidence that repeated amygdala seizures persistently alter phasic 2-AG-mediated retrograde signaling at BLA GABAergic synapses, probably by impairing stimulus-dependent 2-AG synthesis/release, which contributes to the enduring aberrant synaptic plasticity associated with seizure activity.SIGNIFICANCE STATEMENT The plastic reorganization of endocannabinoid (eCB) signaling after seizures and during epileptogenesis may contribute to the negative neurobiological consequences associated with seizure activity. Therefore, a deeper understanding of the molecular basis underlying the pathologic long-term eCB signaling remodeling following seizure activity will be crucial to the development of novel therapies for epilepsy that not only target seizure activity, but, most importantly, the epileptogenesis and the comorbid conditions associated with epilepsy.


Subject(s)
Endocannabinoids , Epilepsy , Rats , Male , Animals , Endocannabinoids/pharmacology , Drug Inverse Agonism , Cannabinoid Receptor Agonists/pharmacology , Receptors, Cannabinoid , Enzyme Inhibitors/pharmacology , Seizures , gamma-Aminobutyric Acid , Receptor, Cannabinoid, CB1
4.
Nat Commun ; 13(1): 7872, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36550102

ABSTRACT

Functional hyperemia occurs when enhanced neuronal activity signals to increase local cerebral blood flow (CBF) to satisfy regional energy demand. Ca2+ elevation in astrocytes can drive arteriole dilation to increase CBF, yet affirmative evidence for the necessity of astrocytes in functional hyperemia in vivo is lacking. In awake mice, we discovered that functional hyperemia is bimodal with a distinct early and late component whereby arteriole dilation progresses as sensory stimulation is sustained. Clamping astrocyte Ca2+ signaling in vivo by expressing a plasma membrane Ca2+ ATPase (CalEx) reduces sustained but not brief sensory-evoked arteriole dilation. Elevating astrocyte free Ca2+ using chemogenetics selectively augments sustained hyperemia. Antagonizing NMDA-receptors or epoxyeicosatrienoic acid production reduces only the late component of functional hyperemia, leaving brief increases in CBF to sensory stimulation intact. We propose that a fundamental role of astrocyte Ca2+ is to amplify functional hyperemia when neuronal activation is prolonged.


Subject(s)
Hyperemia , Neocortex , Neurovascular Coupling , Mice , Animals , Neurovascular Coupling/physiology , Wakefulness , Arterioles , Astrocytes/metabolism , Cerebrovascular Circulation/physiology
5.
Neuron ; 110(12): 1881-1884, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35709691

ABSTRACT

Glutamate excitotoxicity during ischemia triggers an intracellular signaling avalanche leading to cell death, yet blocking NMDA receptors directly in human stroke trials failed. In this issue of Neuron, Zong et al. (2022) disrupt downstream NMDAR-TRPM2 coupling to improve stroke outcomes, supporting intracellular NMDAR signaling as an alternate therapeutic target.


Subject(s)
Stroke , TRPM Cation Channels , Humans , Neurons/metabolism , Peptides/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Stroke/drug therapy , TRPM Cation Channels/metabolism
6.
Sci Signal ; 15(720): eabn2081, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35133865

ABSTRACT

Large-pore channels such as pannexin-1 (PANX1) typically lack pore-lining constriction points, leaving only speculations on how these channels functionally "close." In this issue of Science Signaling, Kuzuya et al. found that rearrangements in the PANX1 amino-terminal helix mediate channel gating by a surprising mechanism in which lipids block the ion conduction pathway, creating a hydrophobic gate.


Subject(s)
Lipids , Signal Transduction , Hydrophobic and Hydrophilic Interactions
7.
Cell Rep ; 32(12): 108169, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32966798

ABSTRACT

Neuronal hyperactivity is an early primary dysfunction in Alzheimer's disease (AD) in humans and animal models, but effective neuronal hyperactivity-directed anti-AD therapeutic agents are lacking. Here we define a previously unknown mode of ryanodine receptor 2 (RyR2) control of neuronal hyperactivity and AD progression. We show that a single RyR2 point mutation, E4872Q, which reduces RyR2 open time, prevents hyperexcitability, hyperactivity, memory impairment, neuronal cell death, and dendritic spine loss in a severe early-onset AD mouse model (5xFAD). The RyR2-E4872Q mutation upregulates hippocampal CA1-pyramidal cell A-type K+ current, a well-known neuronal excitability control that is downregulated in AD. Pharmacologically limiting RyR2 open time with the R-carvedilol enantiomer (but not racemic carvedilol) prevents and rescues neuronal hyperactivity, memory impairment, and neuron loss even in late stages of AD. These AD-related deficits are prevented even with continued ß-amyloid accumulation. Thus, limiting RyR2 open time may be a hyperactivity-directed, non-ß-amyloid-targeted anti-AD strategy.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Memory Disorders/complications , Memory Disorders/pathology , Neurons/pathology , Ryanodine Receptor Calcium Release Channel/metabolism , Alzheimer Disease/physiopathology , Animals , CA1 Region, Hippocampal/pathology , Carvedilol/pharmacology , Dendritic Spines/drug effects , Dendritic Spines/pathology , Ion Channel Gating , Long-Term Potentiation , Memory Disorders/physiopathology , Mice, Transgenic , Mutation/genetics , Neuroprotection/drug effects , Potassium Channels/metabolism , Pyramidal Cells/pathology , Ryanodine Receptor Calcium Release Channel/genetics , Time Factors , Up-Regulation
8.
ACS Chem Neurosci ; 11(15): 2163-2172, 2020 08 05.
Article in English | MEDLINE | ID: mdl-32639715

ABSTRACT

Pannexin 1 (Panx1) channels are transmembrane proteins that release adenosine triphosphate and play an important role in intercellular communication. They are widely expressed in somatic and nervous system tissues, and their activity has been associated with many pathologies such as stroke, epilepsy, inflammation, and chronic pain. While there are a variety of small molecules known to inhibit Panx1, currently little is known about the mechanism of channel inhibition, and there is a dearth of sufficiently potent and selective drugs targeting Panx1. Herein we provide a review of the current literature on Panx1 structural biology and known pharmacological agents that will help provide a basis for rational development of Panx1 chemical modulators.


Subject(s)
Connexins , Epilepsy , Adenosine Triphosphate , Humans , Nerve Tissue Proteins
9.
Nat Commun ; 11(1): 3064, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32528004

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

10.
Nat Commun ; 11(1): 2014, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32332733

ABSTRACT

Astrocytes support the energy demands of synaptic transmission and plasticity. Enduring changes in synaptic efficacy are highly sensitive to stress, yet whether changes to astrocyte bioenergetic control of synapses contributes to stress-impaired plasticity is unclear. Here we show in mice that stress constrains the shuttling of glucose and lactate through astrocyte networks, creating a barrier for neuronal access to an astrocytic energy reservoir in the hippocampus and neocortex, compromising long-term potentiation. Impairing astrocytic delivery of energy substrates by reducing astrocyte gap junction coupling with dominant negative connexin 43 or by disrupting lactate efflux was sufficient to mimic the effects of stress on long-term potentiation. Furthermore, direct restoration of the astrocyte lactate supply alone rescued stress-impaired synaptic plasticity, which was blocked by inhibiting neural lactate uptake. This gating of synaptic plasticity in stress by astrocytic metabolic networks indicates a broader role of astrocyte bioenergetics in determining how experience-dependent information is controlled.


Subject(s)
Astrocytes/metabolism , Energy Metabolism/physiology , Long-Term Potentiation/physiology , Neurons/physiology , Stress, Psychological/metabolism , Adaptation, Psychological/physiology , Animals , Disease Models, Animal , Female , Glucose/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Humans , Lactic Acid/metabolism , Male , Metabolic Networks and Pathways/physiology , Mice , Neocortex/cytology , Neocortex/metabolism , Patch-Clamp Techniques
11.
J Neurosci ; 40(4): 729-742, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31818976

ABSTRACT

The impact of pannexin-1 (Panx1) channels on synaptic transmission is poorly understood. Here, we show that selective block of Panx1 in single postsynaptic hippocampal CA1 neurons from male rat or mouse brain slices causes intermittent, seconds long increases in the frequency of sEPSC following Schaffer collateral stimulation. The increase in sEPSC frequency occurred without an effect on evoked neurotransmission. Consistent with a presynaptic origin of the augmented glutamate release, the increased sEPSC frequency was prevented by bath-applied EGTA-AM or TTX. Manipulation of a previously described metabotropic NMDAR pathway (i.e., by preventing ligand binding to NMDARs with competitive antagonists or blocking downstream Src kinase) also increased sEPSC frequency similar to that seen when Panx1 was blocked. This facilitated glutamate release was absent in transient receptor potential vanilloid 1 (TRPV1) KO mice and prevented by the TRPV1 antagonist, capsazepine, suggesting it required presynaptic TRPV1. We show presynaptic expression of TRPV1 by immunoelectron microscopy and link TRPV1 to Panx1 because Panx1 block increases tissue levels of the endovanilloid, anandamide. Together, these findings demonstrate an unexpected role for metabotropic NMDARs and postsynaptic Panx1 in suppression of facilitated glutamate neurotransmission.SIGNIFICANCE STATEMENT The postsynaptic ion and metabolite channel, pannexin-1, is regulated by metabotropic NMDAR signaling through Src kinase. This pathway suppresses facilitated release of presynaptic glutamate during synaptic activity by regulating tissue levels of the transient receptor potential vanilloid 1 agonist anandamide.


Subject(s)
Connexins/metabolism , Glutamic Acid/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Presynaptic Terminals/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Calcium Chelating Agents/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Mice, Knockout , Neurons/drug effects , Presynaptic Terminals/drug effects , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , Sodium Channel Blockers/pharmacology , Synapses/drug effects , Synapses/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Tetrodotoxin/pharmacology , src-Family Kinases/metabolism
12.
J Physiol ; 598(2): 361-379, 2020 01.
Article in English | MEDLINE | ID: mdl-31698505

ABSTRACT

KEY POINTS: The large-pore channel pannexin 1 (Panx1) is expressed in many cell types and can open upon different, yet not fully established, stimuli. Panx1 permeability is often inferred from channel permeability to fluorescent dyes, but it is currently unknown whether dye permeability translates to permeability to other molecules. Cell shrinkage and C-terminal cleavage led to a Panx1 open-state with increased permeability to atomic ions (current), but did not alter ethidium uptake. Panx1 inhibitors affected Panx1-mediated ion conduction differently from ethidium permeability, and inhibitor efficiency towards a given molecule therefore cannot be extrapolated to its effects on the permeability of another. We conclude that ethidium permeability does not reflect equal permeation of other molecules and thus is no measure of general Panx1 activity. ABSTRACT: Pannexin 1 (Panx1) is a large-pore membrane channel connecting the extracellular milieu with the cell interior. While several activation regimes activate Panx1 in a variety of cell types, the selective permeability of an open Panx1 channel remains unresolved: does a given activation paradigm increase Panx1's permeability towards all permeants equally and does fluorescent dye flux serve as a proxy for biological permeation through an open channel? To explore permeant-selectivity of Panx1 activation and inhibition, we employed Panx1-expressing Xenopus laevis oocytes and HEK293T cells. We report that different mechanisms of activation of Panx1 differentially affected ethidium and atomic ion permeation. Most notably, C-terminal truncation or cell shrinkage elevated Panx1-mediated ion conductance, but had no effect on ethidium permeability. In contrast, extracellular pH changes predominantly affected ethidium permeability but not ionic conductance. High [K+ ]o did not increase the flux of either of the two permeants. Once open, Panx1 demonstrated preference for anionic permeants, such as Cl- , lactate and glutamate, while not supporting osmotic water flow. Panx1 inhibitors displayed enhanced potency towards Panx1-mediated currents compared to that of ethidium uptake. We conclude that activation or inhibition of Panx1 display permeant-selectivity and that permeation of ethidium does not necessarily reflect an equal permeation of smaller biological molecules and atomic ions.


Subject(s)
Connexins/physiology , Ion Channels/physiology , Nerve Tissue Proteins/physiology , Animals , Fluorescent Dyes , Glutamic Acid , HEK293 Cells , Humans , Lactic Acid , Oocytes , Xenopus laevis
13.
Sci Rep ; 9(1): 13903, 2019 09 25.
Article in English | MEDLINE | ID: mdl-31554905

ABSTRACT

Pannexins (Panxs), glycoproteins that oligomerize to form hemichannels on the cell membrane, are topologically similar to connexins, but do not form cell-to-cell gap junction channels. There are 3 members of the family, 1-3, with Panx1 being the most abundant. All Panxs are expressed in bone, but their role in bone cell biology is not completely understood. We now report that osteocytic Panx1 deletion (Panx1Δot) alters bone mass and strength in female mice. Bone mineral density after reaching skeletal maturity is higher in female Panx1Δot mice than in control Panx1fl/fl mice. Further, osteocytic Panx1 deletion partially prevented aging effects on cortical bone structure and mechanical properties. Young 4-month-old female Panx1Δot mice exhibited increased lean body mass, even though pannexin levels in skeletal muscle were not affected; whereas no difference in lean body mass was detected in male mice. Furthermore, female Panx1-deficient mice exhibited increased muscle mass without changes in strength, whereas Panx1Δot males showed unchanged muscle mass and decreased in vivo maximum plantarflexion torque, indicating reduced muscle strength. Our results suggest that osteocytic Panx1 deletion increases bone mass in young and old female mice and muscle mass in young female mice, but has deleterious effects on muscle strength only in males.


Subject(s)
Bone and Bones/metabolism , Connexins/metabolism , Muscle Strength/physiology , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/metabolism , Osteocytes/metabolism , Animals , Body Mass Index , Female , Male , Mice , Muscular Diseases/metabolism
14.
J Biol Chem ; 294(17): 6940-6956, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30814251

ABSTRACT

Pannexin 1 (PANX1)-mediated ATP release in vascular smooth muscle coordinates α1-adrenergic receptor (α1-AR) vasoconstriction and blood pressure homeostasis. We recently identified amino acids 198-200 (YLK) on the PANX1 intracellular loop that are critical for α1-AR-mediated vasoconstriction and PANX1 channel function. We report herein that the YLK motif is contained within an SRC homology 2 domain and is directly phosphorylated by SRC proto-oncogene, nonreceptor tyrosine kinase (SRC) at Tyr198 We demonstrate that PANX1-mediated ATP release occurs independently of intracellular calcium but is sensitive to SRC family kinase (SFK) inhibition, suggestive of channel regulation by tyrosine phosphorylation. Using a PANX1 Tyr198-specific antibody, SFK inhibitors, SRC knockdown, temperature-dependent SRC cells, and kinase assays, we found that PANX1-mediated ATP release and vasoconstriction involves constitutive phosphorylation of PANX1 Tyr198 by SRC. We specifically detected SRC-mediated Tyr198 phosphorylation at the plasma membrane and observed that it is not enhanced or induced by α1-AR activation. Last, we show that PANX1 immunostaining is enriched in the smooth muscle layer of arteries from hypertensive humans and that Tyr198 phosphorylation is detectable in these samples, indicative of a role for membrane-associated PANX1 in small arteries of hypertensive humans. Our discovery adds insight into the regulation of PANX1 by post-translational modifications and connects a significant purinergic vasoconstriction pathway with a previously identified, yet unexplored, tyrosine kinase-based α1-AR constriction mechanism. This work implicates SRC-mediated PANX1 function in normal vascular hemodynamics and suggests that Tyr198-phosphorylated PANX1 is involved in hypertensive vascular pathology.


Subject(s)
Tyrosine/metabolism , src-Family Kinases/metabolism , Animals , Calcium/metabolism , Cell Membrane/metabolism , Cells, Cultured , Connexins/drug effects , Connexins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/metabolism , Phenylephrine/pharmacology , Phosphorylation , Proto-Oncogene Mas , src-Family Kinases/chemistry
15.
Neurosci Lett ; 695: 65-70, 2019 03 16.
Article in English | MEDLINE | ID: mdl-28911820

ABSTRACT

Pannexins form single membrane channels that regulate the passage of ions, small molecules and metabolites between the intra- and extracellular compartments. In the central nervous system, these channels are integrated into numerous signaling cascades that shape brain physiology and pathology. Post-translational modification of pannexins is complex, with phosphorylation emerging as a prominent form of functional regulation. While much is still not known regarding the specific kinases and modified amino acids, recent reports support a role for Src family tyrosine kinases (SFK) in regulating pannexin channel activity. This review outlines the current evidence supporting SFK-dependent pannexin phosphorylation in the CNS and examines the importance of these modifications in the healthy and diseased brain.


Subject(s)
Central Nervous System/metabolism , Connexins/metabolism , src-Family Kinases/metabolism , Animals , Central Nervous System/enzymology , Humans , Phosphorylation , Protein Processing, Post-Translational , Signal Transduction
17.
J Neurosci ; 37(42): 10154-10172, 2017 10 18.
Article in English | MEDLINE | ID: mdl-28924009

ABSTRACT

Tolerance to the analgesic effects of opioids is a major problem in chronic pain management. Microglia are implicated in opioid tolerance, but the core mechanisms regulating their response to opioids remain obscure. By selectively ablating microglia in the spinal cord using a saporin-conjugated antibody to Mac1, we demonstrate a causal role for microglia in the development, but not maintenance, of morphine tolerance in male rats. Increased P2X7 receptor (P2X7R) activity is a cardinal feature of microglial activation, and in this study we found that morphine potentiates P2X7R-mediated Ca2+ responses in resident spinal microglia acutely isolated from morphine tolerant rats. The increased P2X7R function was blocked in cultured microglia by PP2, a Src family protein tyrosine kinase inhibitor. We identified Src family kinase activation mediated by µ-receptors as a key mechanistic step required for morphine potentiation of P2X7R function. Furthermore, we show by site-directed mutagenesis that tyrosine (Y382-384) within the P2X7R C-terminus is differentially modulated by repeated morphine treatment and has no bearing on normal P2X7R function. Intrathecal administration of a palmitoylated peptide corresponding to the Y382-384 site suppressed morphine-induced microglial reactivity and preserved the antinociceptive effects of morphine in male rats. Thus, site-specific regulation of P2X7R function mediated by Y382-384 is a novel cellular determinant of the microglial response to morphine that critically underlies the development of morphine analgesic tolerance.SIGNIFICANCE STATEMENT Controlling pain is one of the most difficult challenges in medicine and its management is a requirement of a large diversity of illnesses. Although morphine and other opioids offer dramatic and impressive relief of pain, their impact is truncated by loss of efficacy (analgesic tolerance). Understanding why this occurs and how to prevent it are of critical importance in improving pain therapies. We uncovered a novel site (Y382-384) within the P2X7 receptor that can be targeted to blunt the development of morphine analgesic tolerance, without affecting normal P2X7 receptor function. Our findings provide a critical missing mechanistic piece, site-specific modulation by Y382-384, that unifies P2X7R function to the activation of spinal microglia and the development of morphine tolerance.


Subject(s)
Analgesics, Opioid/administration & dosage , Microglia/physiology , Morphine/administration & dosage , Pain Measurement/drug effects , Receptors, Purinergic P2X7/physiology , Amino Acid Sequence , Analgesics, Opioid/metabolism , Animals , Animals, Newborn , Binding Sites/drug effects , Binding Sites/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Drug Tolerance/physiology , Injections, Intraperitoneal , Injections, Spinal , Male , Mice , Microglia/drug effects , Morphine/metabolism , Pain Measurement/methods , Rats , Rats, Sprague-Dawley
18.
Nat Methods ; 14(4): 391-394, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28288123

ABSTRACT

To expand the range of experiments that are accessible with optogenetics, we developed a photocleavable protein (PhoCl) that spontaneously dissociates into two fragments after violet-light-induced cleavage of a specific bond in the protein backbone. We demonstrated that PhoCl can be used to engineer light-activatable Cre recombinase, Gal4 transcription factor, and a viral protease that in turn was used to activate opening of the large-pore ion channel Pannexin-1.


Subject(s)
Optogenetics/methods , Protein Engineering/methods , Recombinant Proteins/metabolism , Connexins/genetics , Connexins/metabolism , Directed Molecular Evolution , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Localization Signals/genetics , Patch-Clamp Techniques , Photochemistry/methods , Recombinant Proteins/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Red Fluorescent Protein
19.
J Cereb Blood Flow Metab ; 37(5): 1626-1633, 2017 May.
Article in English | MEDLINE | ID: mdl-26994044

ABSTRACT

Spreading depolarization-induced focal dendritic swelling (beading) is an early hallmark of neuronal cytotoxic edema. Pyramidal neurons lack membrane-bound aquaporins posing a question of how water enters neurons during spreading depolarization. Recently, we have identified chloride-coupled transport mechanisms that can, at least in part, participate in dendritic beading. Yet transporter-mediated ion and water fluxes could be paralleled by water entry through additional pathways such as large-pore pannexin-1 channels opened by spreading depolarization. Using real-time in vivo two-photon imaging in mice with pharmacological inhibition or conditional genetic deletion of pannexin-1, we showed that pannexin-1 channels are not required for spreading depolarization-induced focal dendritic swelling.


Subject(s)
Brain/physiopathology , Connexins/physiology , Cortical Spreading Depression/physiology , Dendrites/pathology , Nerve Tissue Proteins/physiology , Water/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Biological Transport , Brain/drug effects , Brain/metabolism , Brain/pathology , Connexins/antagonists & inhibitors , Connexins/genetics , Cortical Spreading Depression/drug effects , Dendrites/drug effects , Dendrites/metabolism , Mefloquine/pharmacology , Mice, Transgenic , Microscopy, Confocal , Microscopy, Fluorescence, Multiphoton , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics
20.
Nat Neurosci ; 19(3): 432-42, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26854804

ABSTRACT

Overactivation of neuronal N-methyl-D-aspartate receptors (NMDARs) causes excitotoxicity and is necessary for neuronal death. In the classical view, these ligand-gated Ca(2+)-permeable ionotropic receptors require co-agonists and membrane depolarization for activation. We report that NMDARs signal during ligand binding without activation of their ion conduction pore. Pharmacological pore block with MK-801, physiological pore block with Mg(2+) or a Ca(2+)-impermeable NMDAR variant prevented NMDAR currents, but did not block excitotoxic dendritic blebbing and secondary currents induced by exogenous NMDA. NMDARs, Src kinase and Panx1 form a signaling complex, and activation of Panx1 required phosphorylation at Y308. Disruption of this NMDAR-Src-Panx1 signaling complex in vitro or in vivo by administration of an interfering peptide either before or 2 h after ischemia or stroke was neuroprotective. Our observations provide insights into a new signaling modality of NMDARs that has broad-reaching implications for brain physiology and pathology.


Subject(s)
Connexins/physiology , Nerve Tissue Proteins/physiology , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Signal Transduction/physiology , src-Family Kinases/physiology , Animals , Calcium/metabolism , Cell Death/physiology , Connexins/metabolism , Dizocilpine Maleate/pharmacology , Magnesium/pharmacology , Membrane Potentials/physiology , N-Methylaspartate/pharmacology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neuroprotective Agents/pharmacology , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Stroke/metabolism , Stroke/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...