Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Int J Surg Pathol ; 28(7): 728-734, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32403965

ABSTRACT

Papillary renal neoplasm with reverse polarity is a form of recently described tumor. These tumors are defined by GATA3 positivity, negative vimentin staining, and the presence of both papillary structures and a layer of eosinophilic cells with apical nuclei and a granular cytoplasm. In the present report, we review 7 cases of papillary renal neoplasm with reverse polarity that were GATA3+ and vimentin-, consistent with past reports. In all 7 of these cases, we found that these tumors were additionally positive for 34ßE12. All 7 of these tumors were categorized as stage pT1. On histological examination, these tumors exhibited branching papillae with apical nuclei. All 7 of these patients were alive on most recent follow-up, with 6 being disease free and one having developed prostate cancer. Together, this overview of 7 additional cases of papillary renal neoplasm with reverse polarity offers further insight into this rare and poorly understood disease.


Subject(s)
Carcinoma, Papillary/pathology , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Aged , Biomarkers, Tumor , Female , GABA Plasma Membrane Transport Proteins/biosynthesis , Humans , Keratins/biosynthesis , Male , Middle Aged , Vimentin/biosynthesis
2.
J Cereb Blood Flow Metab ; 39(1): 74-88, 2019 01.
Article in English | MEDLINE | ID: mdl-29160736

ABSTRACT

Ischemic stroke triggers an elevation in tonic GABA inhibition that impairs the ability of the brain to form new structural and functional cortical circuits required for recovery. This stroke-induced increase in tonic inhibition is caused by impaired GABA uptake via the glial GABA transporter GAT3, highlighting GAT3 as a novel target in stroke recovery. Using a photothrombotic stroke mouse model, we show that GAT3 protein levels are decreased in peri-infarct tissue from 6 h to 42 days post-stroke. Prior studies have shown that GAT substrates can increase GAT surface expression. Therefore, we aimed to assess whether the GAT3 substrate, L-isoserine, could increase post-stroke functional recovery. L-Isoserine (38 µM or 380 µM) administered directly into the infarct from day 5 to 32 post-stroke, significantly increased motor performance in the grid-walking and cylinder tasks in a concentration-dependent manner, without affecting infarct volumes. Additionally, L-isoserine induced a lasting increase in GAT3 expression in peri-infarct regions accompanied by a small decrease in GFAP expression. This study is the first to show that a GAT3 substrate can increase GAT3 expression and functional recovery after focal ischemic stroke following a delayed long-term treatment. We propose that enhancing GAT3-mediated uptake dampens tonic inhibition and promotes functional recovery after stroke.


Subject(s)
Brain Ischemia/drug therapy , GABA Plasma Membrane Transport Proteins/biosynthesis , Recovery of Function/drug effects , Serine/analogs & derivatives , Stroke/drug therapy , Animals , Brain Ischemia/physiopathology , Dose-Response Relationship, Drug , GABA Plasma Membrane Transport Proteins/genetics , Glial Fibrillary Acidic Protein/biosynthesis , Male , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Psychomotor Performance/drug effects , Serine/pharmacology , Serine/therapeutic use , Stroke/physiopathology , Up-Regulation/drug effects
3.
Pathol Oncol Res ; 24(2): 259-267, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28470571

ABSTRACT

Estrogen and progesterone receptors are possible markers for suggesting a mammary origin of metastatic carcinoma, but are useless in cases of triple negative breast cancers (TNBC). Five other potential markers of breast origin were investigated on tissue microarrays in a series of TNBCs showing keratin 5 expression, consistent with a basal-like phenotype. GATA-3 staining was observed in 82 of 115 triple negative cases (71.3%) including 23 cases with >5% staining. Mammaglobin staining was detected in 30 cases (26.0%) including 12 with >5% staining. GCDFP-15 was seen in 23 cases (20.0%) including 9 with >5% staining. NY-BR-1 positivity was present in 7 cases (6.0%) including 3 patients with >5% staining. BCA-225 staining was observed in 74 cases (64.3%); however this latter marker lacks also specificity owing to the reported widespread staining in other malignancies. GATA-3, mammaglobin and GCDFP-15 coexpression was seen in one case (0.9%), whereas GATA-3 and mammaglobin or mammaglobin and GCDFP-15 coexpression was present in 2 and 2 cases (1.7%), respectively. Using at least 5% staining as cut-off, the expression of any of the last 4 markers was 34.7%. The expression of GATA-3, mammaglobin, GCDFP-15 and NY-BR-1 is lower in TNBC-s than in breast carcinomas in general, and this may be even lower in basal-like carcinomas. Although these markers are not fully specific, by using them, a subset of basal-like TNBC-s can be identified as of mammary origin. However, a substantial proportion will not show any staining with any of these markers.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma/metabolism , Triple Negative Breast Neoplasms/metabolism , Antigens, Neoplasm/biosynthesis , Carrier Proteins/biosynthesis , Female , GABA Plasma Membrane Transport Proteins/biosynthesis , Glycoproteins/biosynthesis , Humans , Immunohistochemistry , Keratin-5/biosynthesis , Mammaglobin A/biosynthesis , Membrane Transport Proteins
4.
J Neuroinflammation ; 13(1): 83, 2016 Apr 18.
Article in English | MEDLINE | ID: mdl-27090509

ABSTRACT

BACKGROUND: Hyperammonemia induces neuroinflammation and increases GABAergic tone in the cerebellum which contributes to cognitive and motor impairment in hepatic encephalopathy (HE). The link between neuroinflammation and GABAergic tone remains unknown. New treatments reducing neuroinflammation and GABAergic tone could improve neurological impairment. The aims were, in hyperammonemic rats, to assess whether: (a) Enhancing endogenous anti-inflammatory mechanisms by sulforaphane treatment reduces neuroinflammation and restores learning and motor coordination. (b) Reduction of neuroinflammation by sulforaphane normalizes extracellular GABA and glutamate-NO-cGMP pathway and identify underlying mechanisms. (c) Identify steps by which hyperammonemia-induced microglial activation impairs cognitive and motor function and how sulforaphane restores them. METHODS: We analyzed in control and hyperammonemic rats, treated or not with sulforaphane, (a) learning in the Y maze; (b) motor coordination in the beam walking; (c) glutamate-NO-cGMP pathway and extracellular GABA by microdialysis; (d) microglial activation, by analyzing by immunohistochemistry or Western blot markers of pro-inflammatory (M1) (IL-1b, Iba-1) and anti-inflammatory (M2) microglia (Iba1, IL-4, IL-10, Arg1, YM-1); and (e) membrane expression of the GABA transporter GAT-3. RESULTS: Hyperammonemia induces activation of astrocytes and microglia in the cerebellum as assessed by immunohistochemistry. Hyperammonemia-induced neuroinflammation is associated with increased membrane expression of the GABA transporter GAT-3, mainly in activated astrocytes. This is also associated with increased extracellular GABA in the cerebellum and with motor in-coordination and impaired learning ability in the Y maze. Sulforaphane promotes polarization of microglia from the M1 to the M2 phenotype, reducing IL-1b and increasing IL-4, IL-10, Arg1, and YM-1 in the cerebellum. This is associated with astrocytes deactivation and normalization of GAT-3 membrane expression, extracellular GABA, glutamate-nitric oxide-cGMP pathway, and learning and motor coordination. CONCLUSIONS: Neuroinflammation increases GABAergic tone in the cerebellum by increasing GAT-3 membrane expression. This impairs motor coordination and learning in the Y maze. Sulforaphane could be a new therapeutic approach to improve cognitive and motor function in hyperammonemia, hepatic encephalopathy, and other pathologies associated with neuroinflammation by promoting microglia differentiation from M1 to M2.


Subject(s)
GABA Plasma Membrane Transport Proteins/biosynthesis , Hepatic Encephalopathy/metabolism , Hyperammonemia/metabolism , Microglia/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Blotting, Western , Cell Membrane/metabolism , Cerebellum , Disease Models, Animal , Hepatic Encephalopathy/complications , Hyperammonemia/etiology , Hyperammonemia/physiopathology , Immunohistochemistry , Inflammation/metabolism , Isothiocyanates/pharmacology , Male , Maze Learning/drug effects , Maze Learning/physiology , Microdialysis , Microglia/drug effects , Motor Activity/drug effects , Motor Activity/physiology , Rats , Rats, Wistar , Sulfoxides , gamma-Aminobutyric Acid/metabolism
5.
Eur J Pharmacol ; 767: 77-81, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26463038

ABSTRACT

Chronic morphine exposure produces morphine tolerance. One of the mechanisms of morphine tolerance involves γ-aminobutric acid (GABA), whose level is regulated by GABA transporter 1 (GAT-1). The aim of this study was to investigate the expression of GAT-1 in the spinal cord during morphine treatment. Morphine was administrated to rats via drinking water for 21 days. On day 21, a single dose of morphine (10mg/kg) was injected, followed by the administration of 5% formalin after 30 min. Expression of GAT-1 in the lumbar spinal cord during morphine treatment was analyzed by Western blotting and immunohistochemistry assay. In another set of experiments, a morphine-tolerant group was treated with a GAT-1 inhibitor, ethyl nipecotate (60 mg/kg), 5 min prior to the formalin test. To assess a possible analgesic effect of the GAT-1 inhibitor, a non-tolerant group was injected only with ethyl nipecotate 5 min prior to the formalin test. Our results indicated that a chronic consumption of morphine led to morphine tolerance. Morphine tolerance was also concomitant with GAT-1 up-regulation in the lumbar spinal cord. The GAT-1 inhibitor ethyl nipecotate improved the antinociceptive effect of morphine in the morphine-tolerant group. Ethyl nipecotate also had an antinociceptive effect on the non-tolerant group. Thus, our data suggest that GAT-1 overexpression in the spinal cord plays an important role in morphine tolerance.


Subject(s)
Drug Tolerance , GABA Plasma Membrane Transport Proteins/biosynthesis , Morphine/pharmacology , Spinal Cord/metabolism , Analgesics/pharmacology , Analgesics, Opioid/pharmacology , Animals , GABA Uptake Inhibitors/pharmacology , Lumbosacral Region , Male , Nipecotic Acids/pharmacology , Pain Measurement/drug effects , Rats , Rats, Wistar , Spinal Cord/drug effects , Up-Regulation
6.
Cell Cycle ; 14(15): 2451-60, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26114472

ABSTRACT

Degeneration or loss of GABAergic neurons frequently may lead to many neuropsychiatric disorders such as epilepsy and autism spectrum disorders. So far no clinically effective therapies can slow and halt the progression of these diseases. Cell-replacement therapy is a promising strategy for treatment of these neuropsychiatric diseases. Although increasing evidence showed that mammalian somatic cells can be directly converted into functional neurons using specific transcription factors or miRNAs via virus delivery, the application of these induced neurons is potentially problematic, due to integration of vectors into the host genome, which results in the disruption or dysfunction of nearby genes. Here, we show that mouse fibroblasts could be efficiently reprogrammed into GABAergic neurons in a combined medium composed of conditioned medium from neurotrophin-3 modified Olfactory Ensheathing Cells (NT3-OECs) plus SB431542, GDNF and RA. Following 3 weeks of induction, these cells derived from fibroblasts acquired the morphological and phenotypical GABAerigic neuronal properties, as demonstrated by the expression of neuronal markers including Tuj1, NeuN, Neurofilament-L, GABA, GABA receptors and GABA transporter 1. More importantly, these converted cells acquired neuronal functional properties such as synapse formation and increasing intracellular free calcium influx when treated with BayK, a specific activator of L-type calcium channel. Therefore, our findings demonstrate for the first time that fibroblasts can be directly converted into GABAergic neurons without ectopic expression of specific transcription factors or miRNA. This study may provide a promising cell source for the application of cell replacement therapy in neuropsychiatric disorders.


Subject(s)
Cellular Reprogramming/physiology , Fibroblasts/cytology , GABAergic Neurons/cytology , Neurogenesis/physiology , Animals , Benzamides/pharmacology , Calcium Channels, L-Type/metabolism , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Culture Media, Conditioned/pharmacology , DNA-Binding Proteins , Dioxoles/pharmacology , GABA Plasma Membrane Transport Proteins/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/biosynthesis , Nervous System Diseases/pathology , Nervous System Diseases/therapy , Neurofilament Proteins/biosynthesis , Nuclear Proteins/biosynthesis , Receptors, GABA/biosynthesis , Tubulin/biosynthesis , gamma-Aminobutyric Acid/biosynthesis
7.
Pharmacol Biochem Behav ; 135: 97-104, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26044967

ABSTRACT

BACKGROUND: Cortical GABA regulates a number of cognitive functions including attention and working memory and is dysregulated in a number of psychiatric conditions. In schizophrenia for example, changes in GABA neurons [reduced expression of glutamic acid decarboxylase (GAD), parvalbumin (PV) and the GABA reuptake transporter (GAT1)] suggest reduced cortical GABA synthesis and release; these changes are hypothesized to cause the cognitive deficits observed in this disorder. The goals of this experiment were to determine whether chronically reducing GAD function within the rat PFC causes attention deficits and alterations in PV and GAT1 expression. METHODS: Male Sprague Dawley rats were trained on the 5-choice serial reaction time task (5CSRTT, a task of attention) until they reached criterion performance and then were implanted with a bilateral cannula aimed at the medial PFC. Cannulae were connected to osmotic minipumps that infused the GAD inhibitor l-allylglycine (LAG, 3.2µg/0.5µl/h) for 13days. Following a 5-day recovery from surgery rats were tested on the standard 5CSRTT for 5 consecutive days and then tested on two modifications of the 5CSRTT. Finally, locomotor activity was assessed and the rats sacrificed. Brains were rapidly extracted and flash frozen and analyzed for the expression of GAD67, PV, GAT1 and the obligatory NMDA receptor subunit NR1. RESULTS: Chronic LAG infusions transiently impaired attention, persistently impaired impulse control and increased locomotor activity. Behavioral changes were associated with an upregulation of GAD67, but no change in PV, GAT1 or NR1 expression. SUMMARY: Chronic inhibition of GABA synthesis within the medial PFC, increased impulsive behavior and locomotion, but did not impair attention; results consistent with previous research following acute inhibition of GABA synthesis. Moreover, our data do not support the hypothesis that decreasing GABA synthesis and release is sufficient to cause changes in other GABA-related proteins.


Subject(s)
Attention/drug effects , GABA Antagonists/pharmacology , Impulsive Behavior , gamma-Aminobutyric Acid/biosynthesis , Animals , Enzyme Inhibitors/pharmacology , GABA Antagonists/therapeutic use , GABA Plasma Membrane Transport Proteins/biosynthesis , Glutamate Decarboxylase/antagonists & inhibitors , Glutamate Decarboxylase/metabolism , Male , Parvalbumins/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/enzymology , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Receptors, N-Methyl-D-Aspartate/biosynthesis , Receptors, N-Methyl-D-Aspartate/genetics
8.
Gene Expr ; 16(3): 145-53, 2015.
Article in English | MEDLINE | ID: mdl-25700370

ABSTRACT

The supraspinal pathophysiology of the painful neuropathy induced by paclitaxel, a chemotherapeutic agent, is not well understood. The γ-aminobutyric acid (GABA) neurotransmitter system has been implicated in the pathogenesis of neuropathic pain. Gene expression of GABAergic system molecules was examined in the anterior cingulate cortex (ACC) of mice brains, by real-time PCR, during paclitaxel-induced neuropathic pain, because this area is involved in pain perception and modulation that might contribute to neuropathic pain. Paclitaxel treatment resulted in thermal hyperalgesia and in increased GABA transporter-1 (GAT-1) mRNA expression, but not that of other GABA transporters or GABA(A) ergic enzymes in the ACC compared to vehicle treatment. Among the 18 GABA(A) receptor subunits analyzed, only ß2, ß3, δ, and γ2 had increased mRNA levels, and for the receptor subunit, only GABA(B2) had increased mRNA levels in the ACC of paclitaxel-treated mice, whereas the rest of the GABA receptor subunits were not altered. The mRNA expression of GABAA receptor subunits α6, θ, π, ρ1, ρ2, and ρ3 were not detected in the ACC. In conclusion, these data show that during paclitaxel-induced neuropathic pain there is significant increase in GAT-1 expression in the ACC. GAT-1 is the main transporter of GABA from the synapse, and thus its increased expression possibly results in less GABA at the synapse and dysregulation of the GABAergic system. GAT-1 is a potential therapeutic target for managing paclitaxel-induced neuropathic pain.


Subject(s)
GABA Plasma Membrane Transport Proteins/biosynthesis , Neuralgia/genetics , Receptors, GABA-A/genetics , Transcriptome , Animals , Disease Models, Animal , Drug-Related Side Effects and Adverse Reactions/genetics , Drug-Related Side Effects and Adverse Reactions/physiopathology , GABA Plasma Membrane Transport Proteins/genetics , Gyrus Cinguli/drug effects , Gyrus Cinguli/metabolism , Gyrus Cinguli/physiopathology , Humans , Mice , Neuralgia/chemically induced , Neuralgia/physiopathology , Paclitaxel/adverse effects , RNA, Messenger/biosynthesis , Receptors, GABA-A/biosynthesis , gamma-Aminobutyric Acid/metabolism
9.
Neurochem Res ; 40(3): 621-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25708016

ABSTRACT

In general, pro-inflammatory cytokines (PICs) contribute to regulation of epilepsy-associated pathophysiological processes in the central nerve system. In this report, we examined the specific activation of PICs, namely IL-1ß, IL-6 and TNF-α in rat brain after kainic acid (KA)-induced status epilepticus (SE). Also, we examined the role played by PICs in regulating expression of GABA transporter type 1 and 3 (GAT-1 and GAT-3, respectively), which are the two important subtypes of GATs responsible for the regulation of extracellular GABA levels in the brain. Our results show that IL-1ß, IL-6 and TNF-α were significantly increased in the parietal cortex, hippocampus and amygdala of KA-rats as compared with sham control animals (P < 0.05, KA rats vs. control rats). KA-induced SE also significantly increased (P < 0.05 vs. controls) the protein expression of GAT-1 and GAT-3 in those brain regions. In addition, central administration of antagonists to IL-1ß and TNF-α receptors significantly attenuated amplified GAT-1 and GAT-3 (P < 0.05 vs. vehicle control for each antagonist group). However, antagonist to IL-6 receptor failed to attenuate enhancement in expression of GAT-1 and GAT-3 induced by KA-induced SE. Overall, our data demonstrate that PIC pathways are activated in the specific brain regions during SE which thereby selectively leads to upregulation of GABA transporters. As a result, it is likely that de-inhibition of GABA system is increased in the brain. This support a role for PICs in engagement of the adaptive mechanisms associated with epileptic activity, and has pharmacological implications to target specific PICs for neuronal dysfunction and vulnerability related to epilepsy.


Subject(s)
Brain/metabolism , Cytokines/physiology , GABA Plasma Membrane Transport Proteins/biosynthesis , Kainic Acid/toxicity , Status Epilepticus/metabolism , Amygdala/drug effects , Amygdala/metabolism , Animals , Brain/drug effects , Gene Expression Regulation , Hippocampus/drug effects , Hippocampus/metabolism , Inflammation Mediators/physiology , Male , Parietal Lobe/drug effects , Parietal Lobe/metabolism , Rats , Rats, Sprague-Dawley , Status Epilepticus/chemically induced
10.
Dev Biol ; 393(1): 171-82, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24929237

ABSTRACT

Motility of sperm is crucial for their directed migration to the egg. The acquisition and modulation of motility are regulated to ensure that sperm move when and where needed, thereby promoting reproductive success. One specific example of this phenomenon occurs during differentiation of the ameboid sperm of Caenorhabditis elegans as they activate from a round spermatid to a mature, crawling spermatozoon. Sperm activation is regulated by redundant pathways to occur at a specific time and place for each sex. Here, we report the identification of the solute carrier 6 (SLC6) transporter protein SNF-10 as a key regulator of C. elegans sperm activation in response to male protease activation signals. We find that SNF-10 is present in sperm and is required for activation by the male but not by the hermaphrodite. Loss of both snf-10 and a hermaphrodite activation factor render sperm completely insensitive to activation. Using in vitro assays, we find that snf-10 mutant sperm show a specific deficit in response to protease treatment but not to other activators. Prior to activation, SNF-10 is present in the plasma membrane, where it represents a strong candidate to receive signals that lead to subcellular morphogenesis. After activation, it shows polarized localization to the cell body region that is dependent on membrane fusions mediated by the dysferlin FER-1. Our discovery of snf-10 offers insight into the mechanisms differentially employed by the two sexes to accomplish the common goal of producing functional sperm, as well as how the physiology of nematode sperm may be regulated to control motility as it is in mammals.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/physiology , GABA Plasma Membrane Transport Proteins/physiology , Sperm Motility/physiology , Spermatozoa/cytology , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/biosynthesis , Caenorhabditis elegans Proteins/metabolism , GABA Plasma Membrane Transport Proteins/biosynthesis , GABA Plasma Membrane Transport Proteins/genetics , Hermaphroditic Organisms , Male , Membrane Proteins/metabolism , Morphogenesis , Mutation , Sperm Motility/genetics , Spermatogenesis
12.
Biomed Pharmacother ; 67(3): 240-5, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23462296

ABSTRACT

Multiple drug resistance of cancer cells is multifactorial. A microarray technique may provide information about new candidate genes playing a role in drug resistance. Drug membrane transporters from ABC and SLC families play a main role in this phenomenon. This study demonstrates alterations in ABC and SLC gene expression levels in methotrexate, cisplatin, doxorubicin, vincristine, topotecan and paclitaxel-resistant variant of W1 ovarian cancer cell line. Resistant W1 cell lines were derived by stepwise selection of cells in increasing concentration of drugs. Affymetrix GeneChip(®) Human Genome U219 Array Strip was used for hybridizations. Statistical significance was determined by independent sample t-test. The genes having altered expression levels in drug-resistant sublines were selected and filtered by scater plot. Genes up/downregulated more than threefolds were selected and listed. Among ABC genes, seven were upregulated and three were downregulated. Three genes: ABCB1, ABCB4 and ABCG2 were upregulated very significantly (over tenfold). One ABCA8 was significantly downregulated. Among 38 SLC genes, 18 were upregulated, 16 were downregulated and four were up- or downregulated dependent on the cell line. Expression of 10 SLC genes was changed very significantly (over tenfold). Four genes were significantly increased: SLC6A1, SLC9A2, SLC12A1, SLC16A6 and six genes were significantly decreased: SLC2A14, SLC7A3, SLC7A8, SLC7A11, SLC16A14, SLC38A9. Based on the expression profiles, our results provide a preliminary insight into the relationship between drug resistance and expression of membrane transporters involved in drug resistance. Correlation of specific drug transporter with drug resistance requires further analysis.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Drug Resistance, Neoplasm/genetics , GABA Plasma Membrane Transport Proteins/genetics , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/metabolism , Protein Array Analysis , ATP-Binding Cassette Transporters/genetics , Animals , Cell Line, Tumor , Female , GABA Plasma Membrane Transport Proteins/biosynthesis , Humans , Ovarian Neoplasms/genetics , Protein Array Analysis/methods
13.
Hum Mol Genet ; 21(13): 2923-35, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22466801

ABSTRACT

Premutation CGG repeat expansions (55-200 CGG repeats; preCGG) within the fragile X mental retardation 1 (FMR1) gene cause fragile X-associated tremor/ataxia syndrome (FXTAS). Defects in neuronal morphology and migration have been described in a preCGG mouse model. Mouse preCGG hippocampal neurons (170 CGG repeats) grown in vitro develop abnormal networks of clustered burst (CB) firing, as assessed by multielectrode array recordings and clustered patterns of spontaneous Ca(2+) oscillations, neither typical of wild-type (WT) neurons. PreCGG neurons have reduced expression of vesicular GABA and glutamate (Glu) transporters (VGAT and VGLUT1, respectively), and preCGG hippocampal astrocytes display a rightward shift on Glu uptake kinetics, compared with WT. These alterations in preCGG astrocytes and neurons are associated with 4- to 8-fold elevated Fmr1 mRNA and occur despite consistent expression of fragile X mental retardation protein levels at ∼50% of WT levels. Abnormal patterns of activity observed in preCGG neurons are pharmacologically mimicked in WT neurons by addition of Glu or the mGluR1/5 agonist, dihydroxyphenylglycine, to the medium, or by inhibition of astrocytic Glu uptake with dl-threo-ß-benzyloxyaspartic acid, but not by the ionotropic Glu receptor agonists, α-2-amino-3-(5-methyl-3-oxo-1,2-oxazol-4-yl) propanoic acid or N-methyl-d-aspartic acid. The mGluR1 (7-(hydroxyimino)cyclopropa [b]chromen-1a-carboxylate ethyl ester) or mGluR5 (2-methyl-6-(phenylethynyl)pyridine hydrochloride) antagonists reversed CB firing. Importantly, the acute addition of the neurosteroid allopregnanolone mitigated functional impairments observed in preCGG neurons in a reversible manner. These results demonstrate abnormal mGluR1/5 signaling in preCGG neurons, which is ameliorated by mGluR1/5 antagonists or augmentation of GABA(A) receptor signaling, and identify allopregnanolone as a candidate therapeutic lead.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Hippocampus/physiology , Neurons/drug effects , Pregnanolone/pharmacology , Amino Acid Transport System X-AG/antagonists & inhibitors , Amino Acid Transport System X-AG/biosynthesis , Animals , Aspartic Acid/pharmacology , Astrocytes/metabolism , Cells, Cultured , Excitatory Amino Acid Antagonists/pharmacology , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , GABA Plasma Membrane Transport Proteins/biosynthesis , Gene Knock-In Techniques , Hippocampus/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Methylaspartate/pharmacology , Neurons/physiology , RNA, Messenger/biosynthesis , Receptor, Metabotropic Glutamate 5 , Receptors, Glutamate , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Trinucleotide Repeat Expansion
14.
Int J Mol Sci ; 13(3): 2578-2589, 2012.
Article in English | MEDLINE | ID: mdl-22489112

ABSTRACT

Betaine/γ-aminobutyric acid (GABA) transporter (BGT1, SLC6A12) is a member of the Na(+)- and Cl(-)-dependent neurotransmitter transporter gene family with a homology to the GABA transporters (GATs), GAT1 (SLC6A1), GAT2 (SLC6A13) and GAT3 (SLC6A11) (HUGO nomenclature). Since antidepressants have been reported to inhibit GABA uptake, we examined those effects on mouse BGT1 (mBGT1) in comparison with other mouse GAT (mGAT) subtypes in the heterologously expressed cell cultures. All antidepressants tested here inhibited the [(3)H]GABA uptake through mBGT1 and mGATs in a rank order of potency with mBGT1 > mGAT1-3. Kinetic analyses for maprotilline, mianserine and trimipramine revealed that they inhibited mBGT1 and mGAT1 noncompetitively, except that mianserine competitively inhibited mBGT1. These results provided a clue to investigate the structure-function relationship of mBGT1 using antidepressants as a tool, leading to the identification of potential candidates for selective and specific inhibitors of mBGT1.


Subject(s)
Antidepressive Agents/pharmacology , GABA Plasma Membrane Transport Proteins/metabolism , Animals , Biological Transport/drug effects , COS Cells , Cells, Cultured , Chlorocebus aethiops , GABA Plasma Membrane Transport Proteins/biosynthesis , Kinetics , Mice , Rats , Serotonin Plasma Membrane Transport Proteins/metabolism , gamma-Aminobutyric Acid/metabolism
15.
Brain Res ; 1388: 1-11, 2011 May 04.
Article in English | MEDLINE | ID: mdl-21382352

ABSTRACT

We used immunohistochemistry to analyze the spatiotemporal patterns of GABA and GABA Transporter-1 (GAT1) immunoreactivities in the developing and adult mouse amygdala. GABA-immunoreactive(ir) neurons were first observed in the mouse amygdala at the embryonic day 15.5 (E15.5), and they became abundant throughout the amygdala perinatally. GAT1 immunoreactivity started to be observed in the mouse amygdala at E18.5. As development proceeds, GAT1 immunoreactivity was more intense, reaching a high density in some amygdalar nuclei at the second postnatal week. In general, GAT1-ir terminals were denser in pallial amygdalar derivatives, such as the basolateral complex than in subpallial derivatives, such as the central nucleus. Distinctive patterns of GABA and GAT1 immunoreactivities distinguish the basolateral complex and the central nucleus during postnatal development and in the adult. GAT1 immunoreactivity appears earlier in the basolateral complex than in the central nucleus. Moreover, the distribution of GAT1-ir fibers and terminals in the basolateral complex parallels the distribution of GABA-ir neurons whereas in the central nucleus the distribution of GAT1-ir terminals was not related with the amount of GABA-ir neurons, especially during development. Another major difference between the basolateral complex and the central nucleus was related to axonal specializations termed here as GAT1 cartridges. Our results indicate that GAT1-ir cartridges were numerous in the basolateral complex but they were completely absent in the central amygdala. Finally, we discuss the patterns of GABA and GAT1 immunoreactivities in relation with the different origin or cellular composition of the basolateral complex and central nucleus.


Subject(s)
Amygdala/embryology , Amygdala/metabolism , GABA Plasma Membrane Transport Proteins/biosynthesis , gamma-Aminobutyric Acid/biosynthesis , Amygdala/growth & development , Animals , Immunohistochemistry , Mice
16.
J Neurochem ; 117(1): 82-90, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21219332

ABSTRACT

Seizure activity can alter GABA transporter and osmoprotective gene expression, which may be involved in the pathogenesis of epilepsy. However, the response of the betaine/GABA transporter (BGT1) is unknown. The goal of the present study was to compare the expression of BGT1 mRNA to that of other osmoprotective genes and GABA transporters following status epilepticus (SE). The possible contributory role of dehydration and inflammation was also investigated because both have been shown to be involved in the regulation of GABA transporter and/or osmoprotective gene expression. BGT1 mRNA was increased 24 h post-SE, as were osmoprotective genes. BGT1 was decreased 72 h and 4 weeks post-SE, as were the GABA transporter mRNAs. The mRNA values for osmoprotective genes following 24-h water withdrawal were significantly lower than the values obtained 24 h post-SE despite similarities in their plasma osmolality values. BGT1 mRNA was not altered by lipopolysaccharide-induced inflammation while the transcription factor tonicity-responsive enhancer binding protein and the GABA transporters 1 and 3 were. These results suggest that neither plasma osmolality nor inflammation fully account for the changes seen in BGT1 mRNA expression post-SE. However, it is evident that BGT1 mRNA expression is altered by SE and displays a temporal pattern with similarities to both GABA and osmolyte transporters. Further investigation of BGT1 regulation in the brain is warranted.


Subject(s)
Betaine/metabolism , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Dehydration/genetics , Gene Expression Regulation , Inflammation Mediators/physiology , RNA, Messenger/biosynthesis , Status Epilepticus/metabolism , Animals , Dehydration/etiology , Dehydration/metabolism , GABA Plasma Membrane Transport Proteins/biosynthesis , Hippocampus/metabolism , Hippocampus/pathology , Male , Mice , Mice, Inbred C57BL , Status Epilepticus/complications , Status Epilepticus/genetics
17.
Glycobiology ; 21(3): 329-39, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21045010

ABSTRACT

The γ-aminobutyric acid (GABA) transporters (GATs) have long been recognized for their key role in the uptake of neurotransmitters. The GAT1 belongs to the family of Na(+)- and Cl(-)-coupled transport proteins, which possess 12 putative transmembrane (TM) domains and three N-glycosylation sites on the extracellular loop between TM domains 3 and 4. Previously, we demonstrated that terminal trimming of N-glycans is important for the GABA uptake activity of GAT1. In this work, we examined the effect of deficiency, removal or oxidation of surface sialic acid residues on GABA uptake activity to investigate their role in the GABA uptake of GAT1. We found that the reduced concentration of sialic acid on N-glycans was paralleled by a decreased GABA uptake activity of GAT1 in Chinese hamster ovary (CHO) Lec3 cells (mutant defective in sialic acid biosynthesis) in comparison to CHO cells. Likewise, either enzymatic removal or chemical oxidation of terminal sialic acids using sialidase or sodium periodate, respectively, resulted in a strong reduction in GAT1 activity. Kinetic analysis revealed that deficiency, removal or oxidation of terminal sialic acids did not affect the K(m) GABA values. However, deficiency and removal of terminal sialic acids of GAT1 reduced the V(max) GABA values with a reduced apparent affinity for extracellular Na(+). Oxidation of cell surface sialic acids also strongly reduced V(max) without affecting both affinities of GAT1 for GABA and Na(+), respectively. These results demonstrated for the first time that the terminal sialic acid of N-linked oligosaccharides of GAT1 plays a crucial role in the GABA transport process.


Subject(s)
GABA Plasma Membrane Transport Proteins/metabolism , Green Fluorescent Proteins/metabolism , N-Acetylneuraminic Acid/metabolism , Recombinant Fusion Proteins/metabolism , Sodium/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , GABA Plasma Membrane Transport Proteins/biosynthesis , GABA Plasma Membrane Transport Proteins/chemistry , GABA Uptake Inhibitors/pharmacology , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/chemistry , HEK293 Cells , Humans , Kinetics , N-Acetylneuraminic Acid/chemistry , Neuraminidase/pharmacology , Oxidation-Reduction , Periodic Acid/pharmacology , Protein Binding , Protein Transport , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry
18.
Neurosci Lett ; 486(3): 150-5, 2010 Dec 17.
Article in English | MEDLINE | ID: mdl-20851161

ABSTRACT

The loss of GABAergic neurotransmission has been closely linked with epileptogenesis. The modulation of the synaptic activity occurs both via the removal of GABA from the synaptic cleft and by GABA transporters (GATs) and by modulation of GABA receptors. The tremor rat (TRM; tm/tm) is the parent strain of the spontaneously epileptic rat (SER; zi/zi, tm/tm), which exhibits absence-like seizure after 8 weeks of age. However, there are no reports that can elucidate the effects of GATs and GABA(A) receptors (GABARs) on TRMs. The present study was conducted to detect GATs and GABAR α1 subunit in TRMs hippocampus at mRNA and protein levels. In this study, total synaptosomal GABA content was significantly decreased in TRMs hippocampus compared with control Wistar rats by high performance liquid chromatography (HPLC); mRNA and protein expressions of GAT-1, GAT-3 and GABAR α1 subunit were all significantly increased in TRMs hippocampus by real time PCR and Western blot, respectively; GAT-1 and GABAR α1 subunit proteins were localized widely in TRMs and control rats hippocampus including CA1, CA3 and dentate gyrus (DG) regions whereas only a wide distribution of GAT-3 was observed in CA1 region by immunohistochemistry. These data demonstrate that excessive expressions of GAT-1 as well as GAT-3 and GABAR α1 subunit in TRMs hippocampus may provide the potential therapeutic targets for genetic epilepsy.


Subject(s)
Epilepsy/metabolism , GABA Plasma Membrane Transport Proteins/biosynthesis , Hippocampus/metabolism , Receptors, GABA-A/biosynthesis , Tremor/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Anticonvulsants/pharmacology , Disease Models, Animal , Epilepsy/genetics , Epilepsy/therapy , GABA Plasma Membrane Transport Proteins/genetics , GABA Plasma Membrane Transport Proteins/metabolism , Genetic Therapy/methods , Hippocampus/drug effects , Kindling, Neurologic , Rats , Rats, Mutant Strains , Rats, Wistar , Receptors, GABA-A/genetics , Tremor/drug therapy , Tremor/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
19.
Neurosci Lett ; 480(2): 132-7, 2010 Aug 16.
Article in English | MEDLINE | ID: mdl-20542084

ABSTRACT

Neuropathic pain is a major health issue and is frequently accompanied by allodynia (painful sensations in response to normally non-painful stimulations), and unpleasant paresthesia/dysesthesia, pointing to alterations in sensory pathways normally dedicated to the processing of non-nociceptive information. Interestingly, mounting evidence indicate that central glial cells are key players in allodynia, partly due to changes in the astrocytic capacity to scavenge extracellular glutamate and gamma-aminobutyric acid (GABA), through changes in their respective transporters (EAAT and GAT). In the present study, we investigated the glial changes occurring in the dorsal column nuclei, the major target of normally innocuous sensory information, in the rat spared nerve injury (SNI) model of neuropathic pain. We report that together with a robust microglial and astrocytic reaction in the ipsilateral gracile nucleus, the GABA transporter GAT-1 is upregulated with no change in GAT-3 or glutamate transporters. Furthermore, [(3)H] GABA reuptake on crude synaptosome preparation shows that transporter activity is functionally increased ipsilaterally in SNI rats. This GAT-1 upregulation appears evenly distributed in the gracile nucleus and colocalizes with astrocytic activation. Neither glial activation nor GAT-1 modulation was detected in the cuneate nucleus. Together, the present results point to GABA transport in the gracile nucleus as a putative therapeutic target against abnormal sensory perceptions related to neuropathic pain.


Subject(s)
GABA Plasma Membrane Transport Proteins/biosynthesis , Medulla Oblongata/metabolism , Pain/metabolism , Peripheral Nervous System Diseases/metabolism , Amino Acid Transport System X-AG/biosynthesis , Animals , Astrocytes/metabolism , Biological Transport , Excitatory Amino Acid Transporter 1/biosynthesis , Excitatory Amino Acid Transporter 2/biosynthesis , Microglia/metabolism , Pain/etiology , Peripheral Nervous System Diseases/etiology , Peroneal Nerve/injuries , Rats , Rats, Sprague-Dawley , Synaptosomes/metabolism , Tibial Nerve/injuries , gamma-Aminobutyric Acid/metabolism
20.
Syst Biol Reprod Med ; 55(5-6): 175-80, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19938951

ABSTRACT

The present study was carried out to identify GABA (gamma-aminobutyric acid) transport protein I (GAT1) in male reproductive organs and to study the effect of GAT1 overexpression on the male reproductive system in GAT1 transgenic mice (TG). Expression and location of GAT1 in testes, epididymis, and sperm of wild-type (WT) mice were identified by immunohistochemistry and western-blot. Histological changes of testes, epididymis, and sperm of transgenic mice overexpressing GAT1 were detected by immunofluorenscent staining and haematoxylin and eosin (HE) staining. GAT1 expression was detected in the testes, epididymis, and sperm of non-transgenic mice. Vacuolization and deformity of spermatogenic cells were observed in the transgenic mice, but the epididymis was unremarkable. Immunofluorenscent staining showed that the number of diastrophic and decapitated sperm increased significantly in transgenic mice to 46.9% from 7.3% in nontransgenic mice. These results suggest that abnormal expression of GAT1 could result in spermiogenesis function injury, sperm paramorphia and dysgenesis.


Subject(s)
Epididymis/metabolism , GABA Plasma Membrane Transport Proteins/biosynthesis , Spermatozoa/metabolism , Testis/metabolism , Animals , Male , Mice , Mice, Transgenic , Reproduction , Testicular Diseases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...