Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 759
1.
Biomolecules ; 13(4)2023 03 26.
Article En | MEDLINE | ID: mdl-37189346

BACKGROUND: Translocator protein (TSPO) is a neuroinflammation hallmark. Different TSPO affinity compounds have been produced and over time, the techniques of radiolabeling have been refined. The aim of this systematic review is to summarize the development of new radiotracers for dementia and neuroinflammation imaging. METHODS: An online search of the literature was conducted in the PubMed, Scopus, Medline, Cochrane Library, and Web of Science databases, selecting published studies from January 2004 to December 2022. The accepted studies considered the synthesis of TSPO tracers for nuclear medicine imaging in dementia and neuroinflammation. RESULTS: A total of 50 articles was identified. Twelve papers were selected from the included studies' bibliographies and 34 were excluded. Thus, 28 articles were ultimately selected for quality assessment. CONCLUSION: Huge efforts in developing specific and stable tracers for PET/SPECT imaging have been made. The long half-life of 18F makes this isotope a preferable choice to 11C. An emerging limitation to this however is that neuroinflammation involves all of the brain which inhibits the possibility of detecting a slight inflammation status change in patients. A partial solution to this is using the cerebellum as a reference region and developing higher TSPO affinity tracers. Moreover, it is necessary to consider the presence of distomers and racemic compounds interfering with pharmacological tracers' effects and increasing the noise ratio in images.


Dementia , Molecular Imaging , Neuroinflammatory Diseases , Positron-Emission Tomography , Radiopharmaceuticals , Receptors, GABA-A , Tomography, Emission-Computed, Single-Photon , Humans , Dementia/diagnostic imaging , Neuroinflammatory Diseases/diagnostic imaging , Nuclear Medicine , Fluorine Radioisotopes/chemistry , Receptors, GABA-A/analysis , Radiopharmaceuticals/chemistry , Cerebellum/diagnostic imaging , Animals , Molecular Imaging/methods
2.
Laryngoscope ; 131(10): 2332-2340, 2021 10.
Article En | MEDLINE | ID: mdl-34156095

OBJECTIVES/HYPOTHESIS: Tinnitus can develop due to, or be aggravated by, stress in a rat model. To investigate stress as a possible causal factor in the development of tinnitus, we designed an animal study that included tinnitus behavior and excitatory/inhibitory neurotransmitter expression after noise exposure as well as restraint stress. STUDY DESIGN: An experimental animal study. METHODS: Wistar rats were grouped according to single or double exposure to noise and restraint stress. The noise exposure (NE) group was subjected to 110 dB sound pressure level (SPL) of 16 kHz narrow-band noise (NBN) for 1 hour, and the restraint stress (RS) group was restrained for 1 hour with or without noise exposure. Gap prepulse inhibition of the acoustic startle (GPIAS) reflex was measured at an NBN of 16 kHz to investigate tinnitus development. Various immunohistopathologic and molecular biologic studies were undertaken to evaluate possible mechanisms of tinnitus development after noise and/or restraint stress. RESULTS: The RS-only group showed a reduced GPIAS response, which is a reliable sign of tinnitus development. In the double-stimulus groups, more tinnitus-development signs of reduced GPIAS responses were observed. The expression of γ-aminobutyric acid A receptor α1 (GABAAR α1) in the hippocampus decreased in the NE│RS group. Increased N-methyl-d-aspartate receptor1 intensities in the NE│RS group and decreased GABAAR α1 intensities in the RS and NE│RS groups were observed in the CA3 region of the hippocampus. CONCLUSIONS: Tinnitus appeared to develop after stress alone in this animal study. An imbalance in excitatory and inhibitory neurotransmitters in the hippocampus may be related to the development of tinnitus after acute NE and/or stress. LEVEL OF EVIDENCE: NA Laryngoscope, 131:2332-2340, 2021.


CA3 Region, Hippocampal/pathology , Noise/adverse effects , Stress, Psychological/complications , Tinnitus/etiology , Acoustic Stimulation/adverse effects , Acoustic Stimulation/methods , Animals , Disease Models, Animal , Humans , Male , Rats , Receptors, GABA-A/analysis , Receptors, GABA-A/metabolism , Receptors, N-Methyl-D-Aspartate/analysis , Receptors, N-Methyl-D-Aspartate/metabolism , Reflex, Startle , Stress, Psychological/psychology , Tinnitus/diagnosis , Tinnitus/pathology , Tinnitus/psychology
3.
J Clin Lab Anal ; 35(1): e23584, 2021 Jan.
Article En | MEDLINE | ID: mdl-32951260

BACKGROUND: The GABA-A receptor signaling pathway regulates proliferation, differentiation, apoptosis, and responses to overt DNA damage during embryonic development. METHODS: To analyze the gene expression after intracytoplasmic sperm injection (ICSI) and in in vivo mouse embryos, the global pattern of gene expression dataset, GSE23009, was obtained from the Gene Expression Omnibus database. Genes with differential expression were identified using the R software package, and RT-qPCR was performed to confirm the microarray results. RESULTS: Mouse blastocysts derived from ICSI fertilization had decreased expression of GABA-A receptor signaling pathway genes. However, the mechanisms underlying these changes were not elucidated. The gene expression of the GABA-A pathway was not significantly different between blastocysts obtained from IVF and in vivo fertilization. However, microinjection after IVF significantly reduced the expression of the GABA-A pathway gene to levels similar to those in the ICSI group. CONCLUSION: Based on our results, decreased gene expression is a result of the microinjection manipulation performed during ICSI.


Embryonic Development/genetics , Microinjections/adverse effects , Receptors, GABA-A , Signal Transduction/genetics , Sperm Injections, Intracytoplasmic/adverse effects , Animals , Embryo, Mammalian/chemistry , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental/genetics , Mice , Receptors, GABA-A/analysis , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Transcriptome/genetics
4.
Int J Radiat Oncol Biol Phys ; 109(4): 1040-1053, 2021 03 15.
Article En | MEDLINE | ID: mdl-33289666

PURPOSE: Most patients with metastatic melanoma show variable responses to radiation therapy and do not benefit from immune checkpoint inhibitors. Improved strategies for combination therapy that leverage potential benefits from radiation therapy and immune checkpoint inhibitors are critical. METHODS AND MATERIALS: We analyzed metastatic melanoma tumors in the TCGA cohort for expression of genes coding for subunits of type A γ-aminobutyric acid (GABA) receptor (GABAAR), a chloride ion channel and major inhibitory neurotransmitter receptor. Electrophysiology was used to determine whether melanoma cells possess intrinsic GABAAR activity. Melanoma cell viability studies were conducted to test whether enhancing GABAAR mediated chloride transport using benzodiazepine-impaired viability. A syngeneic melanoma mouse model was used to assay the effect of benzodiazepine on tumor volume and its ability to potentiate radiation therapy or immunotherapy. Treated tumors were analyzed for changes in gene expression by RNA sequencing and presence of tumor-infiltrating lymphocytes by flow cytometry. RESULTS: Genes coding for subunits of GABAARs express functional GABAARs in melanoma cells. By enhancing GABAAR-mediated anion transport, benzodiazepines depolarize melanoma cells and impair their viability. In vivo, benzodiazepine alone reduces tumor growth and potentiates radiation therapy and α-PD-L1 antitumor activity. The combination of benzodiazepine, radiation therapy, and α-PD-L1 results in near complete regression of treated tumors and a potent abscopal effect, mediated by increased infiltration of polyfunctional CD8+ T cells. Treated tumors show expression of cytokine-cytokine receptor interactions and overrepresentation of p53 signaling. CONCLUSIONS: This study identifies an antitumor strategy combining radiation and/or an immune checkpoint inhibitor with modulation of GABAARs in melanoma using benzodiazepine.


Immune Checkpoint Inhibitors/therapeutic use , Melanoma/therapy , Receptors, GABA-A/physiology , T-Lymphocytes/immunology , Animals , Benzodiazepines/pharmacology , Benzodiazepines/therapeutic use , Cell Proliferation/drug effects , Combined Modality Therapy , Female , Humans , Melanoma/pathology , Membrane Proteins/analysis , Mice , Mice, Inbred C57BL , Radiation-Sensitizing Agents/pharmacology , Receptors, GABA-A/analysis
5.
J Endocrinol ; 247(1): 1-10, 2020 10.
Article En | MEDLINE | ID: mdl-32698131

Despite being a highly conserved protein, the precise role of the mitochondrial translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor (PBR), remains elusive. The void created by studies that overturned a presumptive model that described TSPO/PBR as a mitochondrial cholesterol transporter for steroidogenesis has been filled with evidence that it can affect mitochondrial metabolic functions across different model systems. We previously reported that TSPO/PBR deficient steroidogenic cells upregulate mitochondrial fatty acid oxidation and presented a strong positive correlation between TSPO/PBR expression and tissues active in triglyceride metabolism or lipid storage. Nevertheless, the highlighting of inconsistencies in prior work has provoked reprisals that threaten to stifle progress. One frequent factoid presented as being supportive of a cholesterol import function is that there are no steroid-synthesizing cell types without high TSPO/PBR expression. In this study, we examine the hamster adrenal gland that is devoid of lipid droplets in the cortex and largely relies on de novo cholesterol biosynthesis and uptake for steroidogenesis. We find that Tspo expression in the hamster adrenal is imperceptible compared to the mouse. This observation is consistent with a substantially low expression of Cpt1a in the hamster adrenal, indicating minimal mitochondrial fatty acid oxidation capacity compared to the mouse. These findings provide further reinforcement that the much sought-after mechanism of TSPO/PBR function remains correlated with the extent of cellular triglyceride metabolism. Thus, TSPO/PBR could have a homeostatic function relevant only to steroidogenic systems that manage triglycerides associated with lipid droplets.


Adrenal Glands/metabolism , Gene Expression , Mesocricetus/metabolism , Receptors, GABA-A/genetics , Triglycerides/metabolism , Adrenal Cortex/chemistry , Adrenal Glands/ultrastructure , Animals , Cholesterol/metabolism , Fatty Acids/metabolism , Female , Lipids/analysis , Mice , Mice, Inbred C57BL , Mitochondria/chemistry , Mitochondria/metabolism , Ovary/metabolism , Receptors, GABA-A/analysis , Receptors, GABA-A/physiology , Species Specificity , Steroids/biosynthesis
6.
J Comp Neurol ; 528(15): 2551-2568, 2020 10 15.
Article En | MEDLINE | ID: mdl-32220012

GABAA receptors are composed of five subunits arranged around a central chloride channel. Their subunits originate from different genes or gene families. The majority of GABAA receptors in the mammalian brain consist of two α-, two ß- and one γ- or δ-subunit. This subunit organization crucially determines the physiological and pharmacological properties of the GABAA receptors. Using immunohistochemistry, we investigated the distribution of 10 GABAA receptor subunits (α1, α2, α3, α4, α5, ß1, ß2, ß3, γ2, and δ) in the fore brain of three female rhesus monkeys (Macaca mulatta). Within the cerebral cortex, subunits α1, α5, ß2, ß3, and γ2 were found in all layers, α2, α3, and ß1 were more concentrated in the inner and outer layers. The caudate/putamen was rich in α1, α2, α5, all three ß-subunits, γ2, and δ. Subunits α3 and α5 were more concentrated in the caudate than in the putamen. In contrast, α1, α2, ß1, ß2, γ2, and δ were highest in the pallidum. Most dorsal thalamic nuclei contained subunits α1, α2, α4, ß2, ß3, and γ2, whereas α1, α3, ß1, and γ2 were most abundant in the reticular nucleus. Within the amygdala, subunits α1, α2, α5, ß1, ß3, γ2, and δ were concentrated in the cortical nucleus, whereas in the lateral and basolateral amygdala α1, α2, α5, ß1, ß3, and δ, and in the central amygdala α1, α2, ß3, and γ2 were most abundant. Interestingly, subunit α3-IR outlined the intercalated nuclei of the amygdala. In the hippocampus, subunits α1, α2, α5, ß2, ß3, γ2, and δ were highly expressed in the dentate molecular layer, whereas α1, α2, α3, α5, ß1, ß2, ß3, and γ2 were concentrated in sector CA1 and the subiculum. The distribution of GABAA receptor subunits in the rhesus monkey was highly heterogeneous indicating a high number of differently assembled receptors. In most areas investigated, notably in the striatum/pallidum, amygdaloid nuclei and in the hippocampus it was more diverse than in the rat and mouse indicating a more heterogeneous and less defined receptor assembly in the monkey than in rodent brain.


Prosencephalon/chemistry , Prosencephalon/metabolism , Protein Subunits/biosynthesis , Receptors, GABA-A/biosynthesis , Age Factors , Amino Acid Sequence , Animals , Female , Immunohistochemistry , Macaca mulatta , Protein Subunits/analysis , Protein Subunits/genetics , Receptors, GABA-A/analysis , Receptors, GABA-A/genetics
7.
Neuropharmacology ; 169: 107643, 2020 06 01.
Article En | MEDLINE | ID: mdl-31108109

The flexibility of neuronal networks is believed to rely mainly on the plasticity of excitatory synapses. However, like their excitatory counterparts, inhibitory synapses also undergo several forms of synaptic plasticity. This review examines recent advances in the understanding of the molecular mechanisms leading to postsynaptic GABAergic plasticity. Specifically, modulation of GABAA receptor (GABAAR) number at postsynaptic sites plays a key role, with the interaction of GABAARs with the scaffold protein gephyrin and other postsynaptic scaffold/regulatory proteins having particular importance. Our understanding of these molecular interactions are progressing, based on recent insights into the processes of GABAAR lateral diffusion, gephyrin dynamics, and gephyrin nanoscale organization. This article is part of the special issue entitled 'Mobility and trafficking of neuronal membrane proteins'.


Neuronal Plasticity/physiology , Receptors, GABA-A/metabolism , Synapses/metabolism , Synaptic Potentials/physiology , Animals , GABA-A Receptor Agonists/metabolism , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Antagonists/metabolism , GABA-A Receptor Antagonists/pharmacology , Humans , Membrane Proteins/analysis , Membrane Proteins/metabolism , Neuronal Plasticity/drug effects , Receptors, GABA-A/analysis , Synapses/chemistry , Synapses/drug effects , Synaptic Potentials/drug effects
8.
ACS Chem Neurosci ; 10(11): 4669-4684, 2019 11 20.
Article En | MEDLINE | ID: mdl-31589403

The critical roles played by GABAA receptors as inhibitory regulators of excitation in the central nervous system has been known for many years. Aberrant GABAA receptor function and trafficking deficits have also been associated with several diseases including anxiety, depression, epilepsy, and insomnia. As a consequence, important drug groups such as the benzodiazepines, barbiturates, and many general anesthetics have become established as modulators of GABAA receptor activity. Nevertheless, there is much we do not understand about the roles and mechanisms of GABAA receptors at neural network and systems levels. It is therefore crucial to develop novel technologies and especially chemical entities that can interrogate GABAA receptor function in the nervous system. Here, we describe the chemistry and characterization of a novel set of 4-PIOL and 4-PHP analogues synthesized with the aim of developing a toolkit of drugs that can photoinactivate GABAA receptors. Most of these new analogues show higher affinities/potencies compared with the respective lead compounds. This is indicative of cavernous areas being present near their binding sites that can be potentially associated with novel receptor interactions. The 4-PHP azide-analogue, 2d, possesses particularly impressive nanomolar affinity/potency and is an effective UV-inducible photoinhibitor of GABAA receptors with considerable potential for photocontrol of GABAA receptor function in situ.


Isoxazoles/metabolism , Photoaffinity Labels/metabolism , Piperidines/metabolism , Receptors, GABA-A/metabolism , HEK293 Cells , Humans , Isoxazoles/analysis , Photoaffinity Labels/analysis , Piperidines/analysis , Protein Structure, Secondary , Receptors, GABA-A/analysis , Receptors, GABA-A/chemistry
9.
JCI Insight ; 4(20)2019 10 17.
Article En | MEDLINE | ID: mdl-31619586

Intrathecal (IT) delivery and pharmacology of antisense oligonucleotides (ASOs) for the CNS have been successfully developed to treat spinal muscular atrophy. However, ASO pharmacokinetic (PK) and pharmacodynamic (PD) properties remain poorly understood in the IT compartment. We applied multimodal imaging techniques to elucidate the IT PK and PD of unlabeled, radioactively labeled, or fluorescently labeled ASOs targeting ubiquitously expressed or neuron-specific RNAs. Following lumbar IT bolus injection in rats, all ASOs spread rostrally along the neuraxis, adhered to meninges, and were partially cleared to peripheral lymph nodes and kidneys. Rapid association with the pia and arterial walls preceded passage of ASOs across the glia limitans, along arterial intramural basement membranes, and along white-matter axonal bundles. Several neuronal and glial cell types accumulated ASOs over time, with evidence of probable glial accumulation preceding neuronal uptake. IT doses of anti-GluR1 and anti-Gabra1 ASOs markedly reduced the mRNA and protein levels of their respective neurotransmitter receptor protein targets by 2 weeks and anti-Gabra1 ASOs also reduced binding of the GABAA receptor PET ligand 18F-flumazenil in the brain over 4 weeks. Our multimodal imaging approaches elucidate multiple transport routes underlying the CNS distribution, clearance, and efficacy of IT-dosed ASOs.


Brain/metabolism , GABA-A Receptor Antagonists/pharmacokinetics , Muscular Atrophy, Spinal/drug therapy , Oligonucleotides, Antisense/pharmacokinetics , Animals , Arteries/diagnostic imaging , Arteries/metabolism , Brain/blood supply , Brain/cytology , Brain/diagnostic imaging , Flumazenil/administration & dosage , Flumazenil/analogs & derivatives , GABA-A Receptor Antagonists/administration & dosage , Gene Knockdown Techniques , Humans , Injections, Spinal , Intravital Microscopy , Male , Molecular Targeted Therapy/methods , Neuroglia/metabolism , Neurons/metabolism , Oligonucleotides, Antisense/administration & dosage , Pia Mater/diagnostic imaging , Pia Mater/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Receptors, AMPA/analysis , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/genetics , Receptors, GABA-A/analysis , Receptors, GABA-A/genetics , Single Photon Emission Computed Tomography Computed Tomography , Spatio-Temporal Analysis , Thionucleotides/administration & dosage , Thionucleotides/pharmacokinetics , Tissue Distribution
10.
J Comp Neurol ; 527(12): 2030-2046, 2019 08 15.
Article En | MEDLINE | ID: mdl-30773633

The striatum is the main input nucleus of the basal ganglia, mediating motor and cognitive functions. Striatal projection neurons are GABAergic medium spiny neurons (MSN), expressing either the dopamine receptor type 1 (D1 -R MSN) and forming the direct, movement-promoting pathway, or dopamine receptor type 2 (D2 -R MSN), forming the indirect movement-suppressing pathway. Locally, activity and synchronization of MSN are modulated by several subtypes of GABAergic and cholinergic interneurons. Overall, GABAergic circuits in the striatum remain poorly characterized, and little is known about the intrastriatal connectivity of interneurons and the distribution of GABAA receptor (GABAA R) subtypes, distinguished by their subunit composition, in striatal synapses. Here, by using immunofluorescence in mouse tissue, we investigated the distribution of GABAA Rs containing the α1 , α2 , or α3 subunit in perisomatic synapses of striatal MSN and interneurons, as well as the innervation pattern of D1 R- and D2 R-MSN soma and axonal initial segment (AIS) by GABAergic and cholinergic interneurons. Our results show that perisomatic GABAergic synapses of D1 R- and D2 R-MSN contain the GABAA R α1 and/or α2 subunits, but not the α3 subunit; D2 R-MSN have significantly more α1 -GABAA Rs on their soma than D1 R-MSN. Further, interneurons have few perisomatic synapses containing α2 -GABAA Rs, whereas α3 -GABAA Rs (along with the α1 -GABAA Rs) are abundant in perisomatic synapses of CCK+ , NPY+ /SOM+ , and vAChT+ interneurons. Each MSN and interneuron population analyzed received a distinct pattern of GABAergic and cholinergic innervation, complementing this postsynaptic heterogeneity. In conclusion, intra-striatal GABAergic circuits are distinguished by cell-type specific innervation patterns, differential expression and postsynaptic targeting of GABAA R subtypes.


Corpus Striatum/cytology , Corpus Striatum/metabolism , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Neural Pathways/cytology , Neural Pathways/metabolism , Receptors, GABA-A/analysis , Receptors, GABA-A/metabolism
11.
J Neurosci ; 39(14): 2581-2605, 2019 04 03.
Article En | MEDLINE | ID: mdl-30683685

Presynaptic α2δ subunits of voltage-gated calcium channels regulate channel abundance and are involved in glutamatergic synapse formation. However, little is known about the specific functions of the individual α2δ isoforms and their role in GABAergic synapses. Using primary neuronal cultures of embryonic mice of both sexes, we here report that presynaptic overexpression of α2δ-2 in GABAergic synapses strongly increases clustering of postsynaptic GABAARs. Strikingly, presynaptic α2δ-2 exerts the same effect in glutamatergic synapses, leading to a mismatched localization of GABAARs. This mismatching is caused by an aberrant wiring of glutamatergic presynaptic boutons with GABAergic postsynaptic positions. The trans-synaptic effect of α2δ-2 is independent of the prototypical cell-adhesion molecules α-neurexins (α-Nrxns); however, α-Nrxns together with α2δ-2 can modulate postsynaptic GABAAR abundance. Finally, exclusion of the alternatively spliced exon 23 of α2δ-2 is essential for the trans-synaptic mechanism. The novel function of α2δ-2 identified here may explain how abnormal α2δ subunit expression can cause excitatory-inhibitory imbalance often associated with neuropsychiatric disorders.SIGNIFICANCE STATEMENT Voltage-gated calcium channels regulate important neuronal functions such as synaptic transmission. α2δ subunits modulate calcium channels and are emerging as regulators of brain connectivity. However, little is known about how individual α2δ subunits contribute to synapse specificity. Here, we show that presynaptic expression of a single α2δ variant can modulate synaptic connectivity and the localization of inhibitory postsynaptic receptors. Our findings provide basic insights into the development of specific synaptic connections between nerve cells and contribute to our understanding of normal nerve cell functions. Furthermore, the identified mechanism may explain how an altered expression of calcium channel subunits can result in aberrant neuronal wiring often associated with neuropsychiatric disorders such as autism or schizophrenia.


Axons/metabolism , Calcium Channels/biosynthesis , Presynaptic Terminals/metabolism , Receptors, GABA-A/metabolism , Synaptic Potentials/physiology , Animals , Axons/chemistry , Brain/cytology , Brain/physiology , Calcium Channels/analysis , Cells, Cultured , Coculture Techniques , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Presynaptic Terminals/chemistry , Protein Subunits/analysis , Protein Subunits/biosynthesis , Receptors, GABA-A/analysis
12.
Biochem Biophys Res Commun ; 506(1): 216-222, 2018 11 17.
Article En | MEDLINE | ID: mdl-30343885

Rheumatoid arthritis (RA) is a chronic disease with systemic inflammation resulting in destruction of multiple articular cartilages and bones. Activated macrophage plays a pivotal role during the disease course and has been one of main targets to inhibit inflammatory reaction of RA by using biological disease-modifying anti-rheumatic drugs (bDMARDs). 18F-FEDAC is one of PET imaging agents targeting TSPO, which is overexpressed in activated macrophages. The aim of this study was to evaluate the roles of 18F-FEDAC PET as an in vivo imaging of activated macrophages on etanercept (ETN), a TNF-antagonist as one of bDMARDs in collagen induced arthritis mice. In RAW 264.7 cells, the expressions of TSPO as well as iNOS and infiltrated nucleus of NF-κB were induced by activation with lipopolysaccharide and interferon-gamma. TSPO expression was slightly attenuated by ETN treatment, not by methotrexate (MTX) as a cytotoxic agent. However, cell uptake of 18F-FEDAC did not show significant changes according to both of the treatments. Similarly in CIA mice, 18F-FEDAC uptake in inflamed paws on PET imaging did not show significant changes during both of the treatments, contrary to the uptake decrease of 18F-FDG, a glucose analog to reflect metabolic or active inflammatory activity. Interestingly, when we divided joints according to the degree of 18F-FEDAC uptake before ETN treatment, the joints of high 18F-FEDAC uptake showed better response to ETN than the joints with low 18F-FEDAC uptakes. In case of 18F-FDG, there was no such kinds of patterns. We can speculate that 18F-FEDAC PET imaging may identify activated macrophage-induced arthritis because that 18F-FEDAC can reflect activated macrophages, which is the therapeutic target of ETN by TNF antagonistic effect. Thus, in vivo imaging using 18F-FEDAC may be used as a predictor of therapeutic effects among those kinds of bDMARDs having anti-inflammatory actions to inhibit activated macrophage.


Acetamides/therapeutic use , Antirheumatic Agents/therapeutic use , Macrophages/metabolism , Positron-Emission Tomography/methods , Purines/therapeutic use , Acetamides/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Antirheumatic Agents/analysis , Arthritis, Experimental/diagnostic imaging , Arthritis, Experimental/drug therapy , Carrier Proteins/analysis , Carrier Proteins/metabolism , Diagnostic Imaging/methods , Drug Monitoring/methods , Etanercept/pharmacology , Fluorodeoxyglucose F18 , Humans , Ligands , Macrophages/chemistry , Mice , Purines/metabolism , RAW 264.7 Cells , Radiopharmaceuticals , Receptors, GABA-A/analysis , Receptors, GABA-A/metabolism
13.
J Biol Chem ; 293(35): 13427-13439, 2018 08 31.
Article En | MEDLINE | ID: mdl-29986886

Cell surface expression of type A GABA receptors (GABAARs) is a critical determinant of the efficacy of inhibitory neurotransmission. Pentameric GABAARs are assembled from a large pool of subunits according to precise co-assembly rules that limit the extent of receptor structural diversity. These rules ensure that particular subunits, such as ρ1 and ß3, form functional cell surface ion channels when expressed alone in heterologous systems, whereas other brain-abundant subunits, such as α and γ, are retained within intracellular compartments. Why some of the most abundant GABAAR subunits fail to form homomeric ion channels is unknown. Normally, surface expression of α and γ subunits requires co-assembly with ß subunits via interactions between their N-terminal sequences in the endoplasmic reticulum. Here, using molecular biology, imaging, and electrophysiology with GABAAR chimeras, we have identified two critical residues in the transmembrane domains of α and γ subunits, which, when substituted for their ρ1 counterparts, permit cell surface expression as homomers. Consistent with this, substitution of the ρ1 transmembrane residues for the α subunit equivalents reduced surface expression and altered channel gating, highlighting their importance for GABAAR trafficking and signaling. Although not ligand-gated, the formation of α and γ homomeric ion channels at the cell surface was revealed by incorporating a mutation that imparts the functional signature of spontaneous channel activity. Our study identifies two single transmembrane residues that enable homomeric GABAAR subunit cell surface trafficking and demonstrates that α and γ subunits can form functional ion channels.


Cell Membrane/metabolism , Receptors, GABA-A/metabolism , Amino Acid Sequence , Animals , Cell Membrane/chemistry , Conserved Sequence , HEK293 Cells , Humans , Mice , Models, Molecular , Protein Domains , Protein Multimerization , Protein Subunits/analysis , Protein Subunits/metabolism , Protein Transport , Receptors, GABA-A/analysis , Signal Transduction , gamma-Aminobutyric Acid/metabolism
14.
Int Psychogeriatr ; 30(9): 1385-1391, 2018 09.
Article En | MEDLINE | ID: mdl-29559018

ABSTRACTBackground:Although recent studies have suggested that the γ-aminobutyric acid type A (GABAA) receptor binding affinity can be a more sensitive marker of age-related neuronal loss than regional gray matter (GM) volume, knowledge about the relationship between decreased GABAA receptor binding affinity and cognitive decline during normal aging is still limited. METHODS: Thirty-seven healthy elderly individuals (aged 50-77 years (mean, 64.5 ± 7.3 years); 15 males and 22 females) were enrolled in this study. We investigated the association of the performance of the healthy elderly in the attentional function test with regional GM volume, regional cerebral bold flow (rCBF), and GABAA receptor binding affinity in the resting state by structural magnetic resonance imaging (MRI), arterial spin labeling (ASL), and 123I-iomazenil (IMZ) SPECT, with the analysis focusing on the bilateral inferior frontal gyri. RESULTS: The score of the rapid visual information processing (RVP) test, which is used to assess visual sustained attention, showed a positive correlation with GABAA receptor binding affinity in the right inferior frontal gyrus. No significant correlation was found between RVP test score and regional GM volume or rCBF. CONCLUSION: The findings of 123I-IMZ SPECT, but not those of structural MRI or ASL, suggest that a decreased GABAA receptor binding affinity can be a sensitive marker of cognitive impairment.


Attention , Brain Mapping/methods , Prefrontal Cortex/diagnostic imaging , Receptors, GABA-A/analysis , Aged , Cerebrovascular Circulation , Female , Healthy Volunteers , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Middle Aged , Prefrontal Cortex/physiology , Rest , Spin Labels , Tomography, Emission-Computed, Single-Photon
15.
Mol Med Rep ; 17(5): 6961-6968, 2018 05.
Article En | MEDLINE | ID: mdl-29568893

Potassium-chloride cotransporter 2 (KCC2) has been indicated to serve a crucial role during chronic neuropathic pain (NP). Following the emergence of NP, γ­aminobutyric acid (GABA) A receptor­mediated signaling may be further impaired by the changes of KCC2 chloride anion gradient. In the present study, the authors investigate the effect of electro-acupuncture (EA) on the behavior and the expression of KCC2 and GABAA receptor γ2 subunit in the spinal cord of chronic constriction injury (CCI) model rats. A total of 60 adult male Sprague­Dawley rats were divided into four groups: Normal group, sham­CCI group, CCI group and CCI+EA group. The effect of EA was assessed via the values of mechanical withdrawal threshold and thermal withdrawal latency, which were significantly improved upon stimulation of the ST­36 and GB­34 acupoints. In addition, a marked reduction in both the mRNA and protein levels of KCC2 and GABAA receptor γ2 subunit was observed in the spinal cord following loose ligation of the sciatic nerve. The reductions in KCC2 and GABAA receptor γ2 subunit expression were reversed by EA treatment. These results support the notion that KCC2 and GABAA receptor γ2 subunit contribute to NP following peripheral nerve injury and extend the understanding of the analgesic effects of EA on NP.


Electroacupuncture , Hyperalgesia/therapy , Receptors, GABA-A/genetics , Signal Transduction , Spinal Cord Injuries/therapy , Symporters/genetics , Animals , Down-Regulation , Electroacupuncture/methods , Hyperalgesia/etiology , Hyperalgesia/genetics , Hyperalgesia/metabolism , Male , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/analysis , Receptors, GABA-A/metabolism , Spinal Cord/metabolism , Spinal Cord Injuries/complications , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism , Symporters/analysis , Symporters/metabolism , Up-Regulation , K Cl- Cotransporters
16.
Sci Rep ; 7(1): 14897, 2017 11 02.
Article En | MEDLINE | ID: mdl-29097694

Numerous studies have reported age-dependent degradation of neuronal function in the visual cortex and have attributed this functional decline to weakened intracortical inhibition, especially GABAergic inhibition. However, whether this type of functional decline is linked to compromised GABAergic inhibition has not been fully confirmed. Here, we compared the neuronal response properties and markers of GABAergic inhibition in the primary visual cortex (V1) of young adult and senescent rats. Compared with those of young adult rats, old rats' V1 neurons exhibited significantly increased visually evoked responses and spontaneous activity, a decreased signal-to-noise ratio and reduced response selectivity for the stimulus orientation and motion direction. Additionally, the ratio of GABA-positive neurons to total cortical neurons in old rats was significantly decreased compared with that in young rats. Expression of the key GABA-synthesizing enzyme GAD67 was significantly lower in old rats than in young rats, although GAD65 expression showed a marginal difference between the two age groups. Further, expression of an important GABAA receptor subunit, GABAAR α1, was significantly attenuated in old rats relative to young ones. These results demonstrate that ageing may result in decreased GABAergic inhibition in the visual cortex and that this decrease in GABAergic inhibition accompanies neuronal function degradation.


Aging , GABAergic Neurons/physiology , Receptors, GABA-A/metabolism , Visual Cortex/physiology , Animals , GABAergic Neurons/cytology , GABAergic Neurons/ultrastructure , Glutamate Decarboxylase/analysis , Glutamate Decarboxylase/metabolism , Male , Neural Inhibition , Orientation , Rats , Receptors, GABA-A/analysis , Visual Cortex/cytology , Visual Cortex/ultrastructure
17.
J Chem Neuroanat ; 82: 39-55, 2017 Jul.
Article En | MEDLINE | ID: mdl-28445782

The GABAA receptor is the most abundant inhibitory receptor in the human brain and is assembled from a variety of different subunit subtypes which determines their pharmacology and physiology. To determine which GABAA receptor subunit proteins are found in the human thalamus we investigated the distribution of five major GABAA receptor subunits α1, α2, α3, ß2,3 and γ2 using immunohistochemical techniques. The α1-, ß2,3- and γ2- subunits which combine to form a benzodiazepine sensitive GABAA receptor showed the most intense levels of staining and were the most common subunits found throughout the human thalamus especially in the ventral and posterior nuclear groups. The next most intense staining was for the α3-subunit followed by the α2-subunit. The intralaminar nuclear group, the mediodorsal nucleus and the thalamic reticular nucleus contained α1-, ß2,3- and γ2- subunits staining as well as the highest levels of the α2- and α3- subunits. The sensory dorsal lateral geniculate nucleus contained very high levels of α1- and ß2,3- and γ2-subunits. The highest densities of GABAA receptors found throughout the thalamus which contained the subunits α1, ß2,3, and γ2 included nuclei which are especially involved in the control or the modulation of the cortico-basal ganglia-thalamo-cortical motor circuits and are thus important in disorders such as Huntington's disease where the GABAergic projections of the basal ganglia are compromised. In addition the majority of receptors in the thalamic reticular nucleus contain α3 and γ2 subunits whilst the intralaminar nuclei contain high levels of α2 and α3 subunits.


Receptors, GABA-A/analysis , Thalamus/chemistry , Adult , Aged , Aged, 80 and over , Humans , Male , Middle Aged , Receptors, GABA-A/metabolism , Thalamus/metabolism
18.
AJNR Am J Neuroradiol ; 38(5): 935-941, 2017 May.
Article En | MEDLINE | ID: mdl-28255031

BACKGROUND AND PURPOSE: Misery perfusion may cause selective neuronal damage in atherosclerotic ICA or MCA disease. Bypass surgery can improve misery perfusion and may prevent neuronal damage. On the other hand, surgery conveys a risk for neuronal damage. The purpose of this retrospective study was to determine whether progression of cortical neuronal damage in surgically treated patients with misery perfusion is larger than that in surgically treated patients without misery perfusion or medically treated patients with misery perfusion. MATERIALS AND METHODS: We evaluated the distribution of benzodiazepine receptors twice by using PET and 11C-labeled flumazenil in 18 surgically treated patients with atherosclerotic ICA or MCA disease (9 with misery perfusion and 9 without) and no perioperative stroke before and after bypass surgery; in 8 medically treated patients with misery perfusion and no intervening ischemic event; and in 7 healthy controls. We quantified abnormal decreases in the benzodiazepine receptors of the cerebral cortex within the MCA distribution and compared changes in the benzodiazepine receptor index among the 3 groups. RESULTS: The change in the benzodiazepine receptor index in surgically treated patients with misery perfusion (27.5 ± 15.6) during 7 ± 5 months was significantly larger than that in surgically treated patients without misery perfusion (-5.2 ± 9.4) during 6 ± 4 months (P < .001) and in medically treated patients with misery perfusion (3.2 ± 15.4) during 16 ± 6 months (P < .01). CONCLUSIONS: Progression of cortical neuronal damage in surgically treated patients with misery perfusion and no perioperative stroke may occur and may be larger than that in medically treated patients with misery perfusion and no intervening ischemic event.


Carotid Artery Diseases/surgery , Cerebral Cortex/blood supply , Cerebral Cortex/pathology , Cerebral Revascularization/adverse effects , Receptors, GABA-A/analysis , Aged , Cerebral Cortex/metabolism , Female , Humans , Male , Middle Aged , Positron-Emission Tomography , Receptors, GABA-A/metabolism , Retrospective Studies
19.
Methods Enzymol ; 589: 253-280, 2017.
Article En | MEDLINE | ID: mdl-28336066

Protein-based fluorescent biosensors are powerful tools for quantitative detection of biomolecules or drugs with high sensitivity under physiological conditions. However, conventional methods for construction of biosensors require structural data with high resolution or amino acid sequence information in most cases, which hampers applicability of this method to structurally complicated receptor proteins. To sidestep such limitations, we recently developed a new method that employs ligand-directed chemistry coupled with a bimolecular fluorescence quenching and recovery system, which enabled the conversion of various kinds of membrane-bound receptors to "turn-on" type fluorescent sensors. Here, we describe a protocol for construction of "turn-on" type fluorescent biosensors based on the GABAA receptor which permits quantitative analysis of the ligand affinity.


Biosensing Techniques/methods , Drug Evaluation, Preclinical/methods , Receptors, GABA-A/metabolism , Animals , Fluorescent Dyes/analysis , Fluorescent Dyes/metabolism , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Antagonists/pharmacology , HEK293 Cells , Humans , Ligands , Microscopy, Fluorescence/methods , Models, Molecular , Optical Imaging/methods , Receptors, GABA-A/analysis , Spectrometry, Fluorescence/methods
20.
Neuroimage ; 150: 279-291, 2017 04 15.
Article En | MEDLINE | ID: mdl-28192273

Classical benzodiazepines, which are widely used as sedatives, anxiolytics and anticonvulsants, exert their therapeutic effects through interactions with heteropentameric GABAA receptors composed of two α, two ß and one γ2 subunit. Their high affinity binding site is located at the interface between the γ2 and the adjacent α subunit. The α-subunit gene family consists of six members and receptors can be homomeric or mixed with respect to the α-subunits. Previous work has suggested that benzodiazepine binding site ligands with selectivity for individual GABAA receptor subtypes, as defined by the benzodiazepine-binding α subunit, may have fewer side effects and may even be effective in diseases, such as schizophrenia, autism or chronic pain, that do not respond well to classical benzodiazepines. The distributions of the individual α subunits across the CNS have been extensively characterized. However, as GABAA receptors may contain two different α subunits, the distribution of the subunits does not necessarily reflect the distribution of receptor subtypes with respect to benzodiazepine pharmacology. In the present study, we have used in vivo [18F]flumazenil PET and in vitro [3H]flumazenil autoradiography in combination with GABAA receptor point-mutated mice to characterize the distribution of the two most prevalent GABAA receptor subtypes (α1 and α2) throughout the mouse brain. The results were in agreement with published in vitro data. High levels of α2-containing receptors were found in brain regions of the neuronal network of anxiety. The α1/α2 subunit combinations were predictable from the individual subunit levels. In additional experiments, we explored in vivo [18F]flumazenil PET to determine the degree of receptor occupancy at GABAA receptor subtypes following oral administration of diazepam. The dose to occupy 50% of sensitive receptors, independent of the receptor subtype(s), was 1-2mg/kg, in agreement with published data from ex vivo studies with wild type mice. In conclusion, we have resolved the quantitative distribution of α1- and α2-containing homomeric and mixed GABAA receptors in vivo at the millimeter scale and demonstrate that the regional drug receptor occupancy in vivo at these GABAA receptor subtypes can be determined by [18F]flumazenil PET. Such information should be valuable for drug development programs aiming for subtype-selective benzodiazepine site ligands for new therapeutic indications.


Brain/metabolism , Neuroimaging/methods , Positron-Emission Tomography/methods , Receptors, GABA-A/biosynthesis , Animals , Autoradiography , Diazepam/pharmacology , Flumazenil , Fluorine Radioisotopes , GABA Modulators/pharmacology , Mice , Mice, Mutant Strains , Radiopharmaceuticals , Receptors, GABA-A/analysis
...