Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters











Publication year range
1.
Exp Parasitol ; 259: 108721, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38369179

ABSTRACT

Glutathione S-transferases (GSTs) are a family of multifunctional isoenzymes involved in the neutralization of toxic compounds, drug resistance and several other cellular functions. The glutathione S-transferase enzyme of Schistosoma japonicum (SjGST-26) plays a role in human schistosomiasis and is also a frequently used fusion partner in mammalian and bacterial expression and pull-down systems. GSTs seem not to be naturally associated with metal ions. Exceptionally, in vitro, metal binding sites have been previously described in some schistosome GSTs; however, their possible physiological role is unclear. Molecules of several neurotransmitter transporters also contain a regulatory zinc binding site, which affects their transport cycle. Here we show that among several metals, manganese and zinc are able to induce a specific protein interaction of SjGST-26 with the glycine transporter GlyT1 and the GABA transporter GAT3 in vitro. The results suggest that metal-binding sites on SjGST-26 and neurotransmitter transporters might function in metal-coordinated interactions with other metalloproteins. Our results additionally indicate that the presence of metal ions in SjGST-26-based GST protein pull-down assays may lead to a false-positive interaction if the potential interacting target is the metalloprotein.


Subject(s)
Schistosoma japonicum , Animals , Humans , Manganese , Zinc , Glutathione Transferase/metabolism , Ions , Glutathione , Mammals/metabolism
2.
Front Synaptic Neurosci ; 15: 1198159, 2023.
Article in English | MEDLINE | ID: mdl-37325697

ABSTRACT

The efficiency of neocortical information processing critically depends on the balance between the glutamatergic (excitatory, E) and GABAergic (inhibitory, I) synaptic transmission. A transient imbalance of the E/I-ratio during early development might lead to neuropsychiatric disorders later in life. The transgenic glutamic acid decarboxylase 67-green fluorescent protein (GAD67-GFP) mouse line (KI) was developed to selectively visualize GABAergic interneurons in the CNS. However, haplodeficiency of the GAD67 enzyme, the main GABA synthetizing enzyme in the brain, temporarily leads to a low GABA level in the developing brain of these animals. However, KI mice did not demonstrate any epileptic activity and only few and mild behavioral deficits. In the present study we investigated how the developing somatosensory cortex of KI-mice compensates the reduced GABA level to prevent brain hyperexcitability. Whole-cell patch clamp recordings from layer 2/3 pyramidal neurons at P14 and at P21 revealed a reduced frequency of miniature inhibitory postsynaptic currents (mIPSCs) in KI mice without any change in amplitude or kinetics. Interestingly, mEPSC frequencies were also decreased, while the E/I-ratio was nevertheless shifted toward excitation. Surprisingly, multi-electrode-recordings (MEA) from acute slices revealed a decreased spontaneous neuronal network activity in KI mice compared to wild-type (WT) littermates, pointing to a compensatory mechanism that prevents hyperexcitability. Blockade of GABAB receptors (GABABRs) with CGP55845 strongly increased the frequency of mEPSCs in KI, but failed to affect mIPSCs in any genotype or age. It also induced a membrane depolarization in P14 KI, but not in P21 KI or WT mice. MEA recordings in presence of CGP55845 revealed comparable levels of network activity in both genotypes, indicating that tonically activated GABABRs balance neuronal activity in P14 KI cortex despite the reduced GABA levels. Blockade of GABA transporter 3 (GAT-3) reproduced the CGP55845 effects suggesting that tonic activation of GABABRs is mediated by ambient GABA released via GAT-3 operating in reverse mode. We conclude that GAT-3-mediated GABA release leads to tonic activation of both pre- and postsynaptic GABABRs and restricts neuronal excitability in the developing cortex to compensate for reduced neuronal GABA synthesis. Since GAT-3 is predominantly located in astrocytes, GAD67 haplodeficiency may potentially stimulate astrocytic GABA synthesis through GAD67-independent pathways.

3.
Int J Mol Sci ; 23(24)2022 Dec 13.
Article in English | MEDLINE | ID: mdl-36555501

ABSTRACT

Astrocytes are the most abundant glial cells in the central nervous system (CNS) mediating a variety of homeostatic functions, such as spatial K+ buffering or neurotransmitter reuptake. In addition, astrocytes are capable of releasing several biologically active substances, including glutamate and GABA. Astrocyte-mediated GABA release has been a matter of debate because the expression level of the main GABA synthesizing enzyme glutamate decarboxylase is quite low in astrocytes, suggesting that low intracellular GABA concentration ([GABA]i) might be insufficient to support a non-vesicular GABA release. However, recent studies demonstrated that, at least in some regions of the CNS, [GABA]i in astrocytes might reach several millimoles both under physiological and especially pathophysiological conditions, thereby enabling GABA release from astrocytes via GABA-permeable anion channels and/or via GABA transporters operating in reverse mode. In this review, we summarize experimental data supporting both forms of GABA release from astrocytes in health and disease, paying special attention to possible feedback mechanisms that might govern the fine-tuning of astrocytic GABA release and, in turn, the tonic GABAA receptor-mediated inhibition in the CNS.


Subject(s)
Astrocytes , gamma-Aminobutyric Acid , Astrocytes/metabolism , gamma-Aminobutyric Acid/metabolism , Neuroglia/metabolism , Receptors, GABA-A/metabolism , Glutamic Acid/metabolism
4.
Brain Res Bull ; 183: 104-115, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35278661

ABSTRACT

Traumatic brain injury (TBI) causes neurotransmitter disturbances contributing to neuronal cell death and neurological deficits. In humans, brain injuries impair γ-aminobutyric acid (GABA) uptake and ultimately result in cognitive impairment. GABA transporter 3 (GAT3) is a vital approach of GABA reuptake and catabolism. The contribution of GAT3 in TBI-induced cognitive impairment and its underlying mechanisms remain unknown, which were explored in the present study. Here, we found that expression of GAT3 was downregulated to increase GABA concentration in the mouse brain after TBI. And GAT3 was detected in neurons and astrocytes after TBI unexpectedly, instead of merely expressed on astrocytes in physiological states. Subsequently, activated metabotropic glutamate receptor 5 (mGluR5) reduced GABA content by elevating the expression levels of GAT3. Then, increased mGluR5 activity obviously improved cognitive impairment. Mechanistically, mGluR5 was activated to evidently induce the expression of p-ERK, CREB, and p-CREB after TBI. The inhibition of CREB decreased the expression of CREB, p-CREB, and GAT3 elevated by active mGluR5. However, the CREB inhibitor increased GABA content. Furthermore, Rab11a regulating GAT3 trafficking by endocytosis was elevated after TBI. And Rab11a downregulated by active mGluR5 was reversed by CREB inhibitor. In summary our findings elucidated that activated mGluR5 ameliorated cognitive function by upregulating GAT3 in TBI. And mGluR5 possibly regulated GAT3 by ERK/CREB/Rab11a pathway. GAT3 could serve as a potential target for TBI cognitive treatment.


Subject(s)
Brain Injuries, Traumatic , Cognitive Dysfunction , GATA3 Transcription Factor/metabolism , Animals , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Mice , Receptor, Metabotropic Glutamate 5/metabolism , Up-Regulation , gamma-Aminobutyric Acid/metabolism
5.
BMC Med Genomics ; 15(1): 27, 2022 02 12.
Article in English | MEDLINE | ID: mdl-35151321

ABSTRACT

BACKGROUND: Linkeropathies refers to a series of extremely rare hereditary connective tissue diseases affected by various glycosyltransferases in the biosynthesis of proteoglycans. We report for the first time two heterozygous variants of B3GAT3 in a Chinese infant, in whom Marfan syndrome was suspected at birth. CASE PRESENTATION: A 2-month-old boy from a non-consanguineous Chinese family without a family history presented severe phenotypes of joint dislocation, obvious flexion contractures of the elbow, arachnodactyly with slightly adducted thumbs, cranial dysplasia, foot abnormalities and aortic root dilation; Marfan syndrome was suspected at birth. Our patient was the youngest, at the age of 2 months, to experience aortic root dilation. Two B3GAT3 variants, NM_012200.2, c.752T>C, p.V251A and c.47C>A, p.S16*, with heterozygosity were identified in the patient by whole-exome sequencing; the variants were inherited from his parents. During close follow-up, significant changes in the cranial profile and obvious external hydrocephalus were present at the age of 7 months, which differs from previously reported cases. CONCLUSION: We diagnosed a patient with congenital heart defects at an early age with a B3GAT3-related disorder instead of Marfan syndrome and expanded the spectrum of B3GAT3-related disorders. We also provide a literature review of reported B3GAT3 cases; for at least one of the variants, this is the first report of genotype-phenotype correlations in individuals with cardiovascular defects being related to the acceptor substrate-binding subdomain of B3GAT3.


Subject(s)
Glucuronosyltransferase , Genetic Association Studies , Glucuronosyltransferase/genetics , Heterozygote , Humans , Phenotype , Exome Sequencing
6.
Adv Neurobiol ; 26: 231-254, 2021.
Article in English | MEDLINE | ID: mdl-34888837

ABSTRACT

Astroglia are key regulators of synaptic function, playing central roles in homeostatic ion buffering, energy dynamics, transmitter uptake, maintenance of neurotransmitter pools, and regulation of synaptic plasticity through release of neuroactive chemicals. Given the myriad of crucial homeostatic and signaling functions attributed to astrocytes and the variety of neurotransmitter receptors expressed by astroglia, they serve as prime cellular candidates for establishing maladaptive synaptic plasticity following drug exposure. Initial studies on astroglia and addiction have placed drug-mediated disruptions in the homeostatic regulation of glutamate as a central aspect of relapse vulnerability. However, the generation of sophisticated tools to study and manipulate astroglia have proven that the interaction between addictive substances, astroglia, and relapse-relevant synaptic plasticity extends far beyond the homeostatic regulation of glutamate. Here we present astroglial systems impacted by drug exposure and discuss how changes in astroglial biology contribute to addiction biology.


Subject(s)
Astrocytes , Synapses , Glutamic Acid , Humans , Neuronal Plasticity , Signal Transduction
7.
J Neurophysiol ; 126(4): 1310-1313, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34495776

ABSTRACT

Rett syndrome (RTT) is a neurodevelopmental disorder characterized a spectrum of phenotypes affecting neuronal and glial populations. Recent work by Dong et al. (Dong Q, Kim J, Nguyen L, Bu Q, Chang Q. J Neurosci 40: 6250-6261, 2020) suggests that augmented GABA uptake by astrocytes diminishes tonic inhibition in the hippocampus and contributes to increased seizure propensity in RTT. Here, I will review evidence supporting this possibility and critically evaluate how increased expression of a GABA transporter might contribute to this mechanism.


Subject(s)
Astrocytes , Rett Syndrome , Astrocytes/metabolism , GABA Plasma Membrane Transport Proteins , Humans , Methyl-CpG-Binding Protein 2/metabolism , Pyramidal Cells/metabolism , gamma-Aminobutyric Acid
8.
Eur J Med Genet ; 64(12): 104342, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34537402

ABSTRACT

BACKGROUND: Proteoglycans (PGs) are complex macromolecules consisting of a core protein and glycosaminoglycan (GAG) side chains. PGs are important for the constitution and functioning of the connective tissue. The normal composition of the GAG side chains defines the nature of the PGs and a wide range of biological events. Deficiencies of specific enzymes involved in the linkage of GAGs to the core protein to form functional PGs, lead to a heterogeneous disease group called Linkeropathies. This is a group of multisystem conditions characterized by different phenotypes that include skeletal dysplasia and various extra-skeletal features: developmental delay/intellectual disability, ophthalmological abnormalities including blue sclerae, facial characteristics, cardiac defects, abdominal wall defects (hernias), cutis laxa, hypermobility and hypotonia. The conditions show variable severity and often overlapping phenotypes. The enzyme ß-1,3-glucuronyltransferase 3, encoded by B3GAT3, is involved in the linkage process to form functional PGs. Biallelic pathogenic variants in B3GAT3 hence lead to Linkeropathy due to loss of function or decreased activity of this enzyme. PATIENT PRESENTATION: We describe a 22-year-old female patient, born of consanguineous parents. The disease history includes congenital severe joint malalignment of elbows, hips, knees and feet, hypermobility, severe kyphoscoliosis, osteoporosis with multiple fractures in childhood, congenital diaphragmatic hernia, minor dental anomalies, digital malformations, and characteristic facial features. Whole exome sequencing was performed, and homozygosity for a novel in-frame deletion in B3GAT3, (c.61_63delCTC (p.(Leu21del))) was detected. Both unaffected parents (double second cousins) were shown to be heterozygous carriers. CONCLUSION: This is the first report to describe homozygosity for this specific in-frame deletion in B3GAT3 (p.(Leu21del)). We present a young adult phenotype and a summary of previous reported patients with other biallelic B3GAT3-variants for comparison. Previously described patients of B3GAT3-deficiency were, however, all children with phenotypes ranging from prenatal manifestation and early lethality to less severe. We suggest that this novel homozygous in-frame deletion in B3GAT3 may be the cause of a recessive form of Linkeropathy.


Subject(s)
Eye Abnormalities/genetics , Genetic Predisposition to Disease/genetics , Glucuronosyltransferase/genetics , Osteochondrodysplasias/genetics , Sequence Deletion/genetics , Adult , Female , Homozygote , Humans , Phenotype , Young Adult
9.
Neurochem Res ; 46(10): 2676-2686, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33710537

ABSTRACT

Alterations in neurotransmitter homeostasis, primarily of glutamate and GABA, is strongly implicated in the pathophysiology of Alzheimer's disease (AD). Homeostasis at the synapse is maintained by neurotransmitter recycling between neurons and astrocytes. Astrocytes support neuronal transmission through glutamine synthesis, which can be derived from oxidative metabolism of GABA. However, the precise implications of astrocytic GABA metabolism in AD remains elusive. The aim of this study was to investigate astrocytic GABA metabolism in AD pathology implementing human induced pluripotent stem cells derived astrocytes. Metabolic mapping of GABA was performed with [U-13C]GABA stable isotopic labeling using gas chromatography coupled to mass spectrometry (GC-MS). Neurotransmitter and amino acid content was quantified via high performance liquid chromatography (HPLC) and protein expression was investigated by Western blot assay. Cell lines carrying mutations in either amyloid precursor protein (APP) or presenilin1 (PSEN-1) were used as AD models and were compared to a control cell line of the same genetic background. AD astrocytes displayed a reduced oxidative GABA metabolism mediated by a decreased uptake capacity of GABA, as GABA transporter 3 (GAT3) was downregulated in AD astrocytes compared to the controls. Interestingly, the carbon backbone of GABA in AD astrocytes was utilized to a larger extent to support glutamine synthesis compared to control astrocytes. The results strongly indicate alterations in GABA uptake and metabolism in AD astrocytes linked to reduced GABA transporter expression, hereby contributing further to neurotransmitter disturbances.


Subject(s)
Alzheimer Disease/metabolism , Astrocytes/metabolism , GABA Plasma Membrane Transport Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , gamma-Aminobutyric Acid/metabolism , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Down-Regulation/physiology , Glutamic Acid/metabolism , Glutamine/metabolism , Humans , Mutation , Presenilin-1/genetics
10.
Addict Biol ; 26(4): e13011, 2021 07.
Article in English | MEDLINE | ID: mdl-33527681

ABSTRACT

While most individuals with access to alcohol drink it recreationally, some vulnerable individuals eventually lose control over their intake and progressively develop compulsive alcohol drinking and decreased interest in alternative sources of reinforcement, two key features of addiction. The neural and molecular mechanisms underlying this vulnerability to switch from controlled to compulsive alcohol intake have not been fully elucidated. It has been shown that rats having reduced levels of expression of the gamma-aminobutyric acid (GABA) transporter, GAT-3, in the amygdala tend to persist in seeking and drinking alcohol even when adulterated with quinine, suggesting that pharmacological interventions aimed at restoring GABA homeostasis in these individuals may provide a targeted treatment to limit compulsive alcohol drinking. Here, we tested the hypothesis that the GABAB receptor agonist baclofen, which decreases GABA release, specifically reduces compulsive alcohol drinking in vulnerable individuals. In a large cohort of Sprague-Dawley rats allowed to drink alcohol under an intermittent two-bottle choice procedure, a cluster of individuals was identified that persisted in drinking alcohol despite adulteration with quinine or when an alternative ingestive reinforcer, saccharin, was available. In these rats, which were characterized by decreased GAT-3 mRNA levels in the central amygdala, acute baclofen administration (1.5 mg/kg, intraperitoneal) resulted in a decrease in compulsive drinking. These results indicate that low GAT-3 mRNA levels in the central amygdala may represent an endophenotype of vulnerability to develop a compulsive drinking of alcohol that is shown here to be mitigated by baclofen.


Subject(s)
Alcoholism/metabolism , Baclofen/pharmacology , Polymers/metabolism , Animals , Central Amygdaloid Nucleus/drug effects , Compulsive Behavior/metabolism , Conditioning, Operant/drug effects , Ethanol/pharmacology , Male , Quinine/pharmacology , Rats , Rats, Sprague-Dawley , Reinforcement, Psychology , Self Administration
11.
Curr Med Chem ; 28(4): 750-776, 2021.
Article in English | MEDLINE | ID: mdl-31612818

ABSTRACT

Neurotransmitter γ-aminobutyric acid (GABA) plays a principal role in the regulation of mammalian central nervous system functions. GABA evoked neurotransmission is terminated by a rapid uptake via dependent plasma membrane GABA transporters (GATs) located in the cell membrane. Potent inhibitors of these GATs are of fundamental importance for elucidation of the physiological function of these targets. Over recent years, a wide range of new GAT1-selective and less common non-GAT1-selective inhibitors have been successfully developed. This review highlights development and recent significant achievements in the field of GABA reuptake inhibitors. Special attention is paid to their pharmacological roles, structure and subtype selectivity relationships.


Subject(s)
GABA Uptake Inhibitors , gamma-Aminobutyric Acid , Animals , Biological Transport , GABA Plasma Membrane Transport Proteins/metabolism , Synaptic Transmission , gamma-Aminobutyric Acid/metabolism
12.
Glia ; 68(12): 2601-2612, 2020 12.
Article in English | MEDLINE | ID: mdl-32584476

ABSTRACT

Synaptic transmission is closely linked to brain energy and neurotransmitter metabolism. However, the extent of brain metabolism of the inhibitory neurotransmitter γ-aminobutyric acid (GABA), and the relative metabolic contributions of neurons and astrocytes, are yet unknown. The present study was designed to investigate the functional significance of brain GABA metabolism using isolated mouse cerebral cortical slices and slices of neurosurgically resected neocortical human tissue of the temporal lobe. By using dynamic isotope labeling, with [15 N]GABA and [U-13 C]GABA as metabolic substrates, we show that both mouse and human brain slices exhibit a large capacity for GABA metabolism. Both the nitrogen and the carbon backbone of GABA strongly support glutamine synthesis, particularly in the human cerebral cortex, indicative of active astrocytic GABA metabolism. This was further substantiated by pharmacological inhibition of the primary astrocytic GABA transporter subtype 3 (GAT3), by (S)-SNAP-5114 or 1-benzyl-5-chloro-2,3-dihydro-1H-indole-2,3-dione (compound 34), leading to significant reductions in oxidative GABA carbon metabolism. Interestingly, this was not the case when tiagabine was used to specifically inhibit GAT1, which is predominantly found on neurons. Finally, we show that acute GABA exposure does not directly stimulate glycolytic activity nor oxidative metabolism in cultured astrocytes, but can be used as an additional substrate to enhance uncoupled respiration. These results clearly show that GABA is actively metabolized in astrocytes, particularly for the synthesis of glutamine, and challenge the current view that synaptic GABA homeostasis is maintained primarily by presynaptic recycling.


Subject(s)
Astrocytes , Animals , Carbon , Cerebral Cortex , Glutamic Acid , Glutamine , Mice , Neurotransmitter Agents , gamma-Aminobutyric Acid
13.
Genes (Basel) ; 10(9)2019 08 21.
Article in English | MEDLINE | ID: mdl-31438591

ABSTRACT

The term linkeropathies (LKs) refers to a group of rare heritable connective tissue disorders, characterized by a variable degree of short stature, skeletal dysplasia, joint laxity, cutaneous anomalies, dysmorphism, heart malformation, and developmental delay. The LK genes encode for enzymes that add glycosaminoglycan chains onto proteoglycans via a common tetrasaccharide linker region. Biallelic variants in XYLT1 and XYLT2, encoding xylosyltransferases, are associated with Desbuquois dysplasia type 2 and spondylo-ocular syndrome, respectively. Defects in B4GALT7 and B3GALT6, encoding galactosyltransferases, lead to spondylodysplastic Ehlers-Danlos syndrome (spEDS). Mutations in B3GAT3, encoding a glucuronyltransferase, were described in 25 patients from 12 families with variable phenotypes resembling Larsen, Antley-Bixler, Shprintzen-Goldberg, and Geroderma osteodysplastica syndromes. Herein, we report on a 13-year-old girl with a clinical presentation suggestive of spEDS, according to the 2017 EDS nosology, in whom compound heterozygosity for two B3GAT3 likely pathogenic variants was identified. We review the spectrum of B3GAT3-related disorders and provide a comparison of all LK patients reported up to now, highlighting that LKs are a phenotypic continuum bridging EDS and skeletal disorders, hence offering future nosologic perspectives.


Subject(s)
Antley-Bixler Syndrome Phenotype/genetics , Arachnodactyly/genetics , Bone Diseases/congenital , Craniosynostoses/genetics , Dwarfism/genetics , Glucuronosyltransferase/genetics , Marfan Syndrome/genetics , Mutation , Osteochondrodysplasias/genetics , Phenotype , Skin Diseases, Genetic/genetics , Adolescent , Antley-Bixler Syndrome Phenotype/pathology , Arachnodactyly/pathology , Bone Diseases/genetics , Bone Diseases/pathology , Craniosynostoses/pathology , Dwarfism/pathology , Female , Humans , Marfan Syndrome/pathology , Osteochondrodysplasias/pathology , Skin Diseases, Genetic/pathology
14.
Orphanet J Rare Dis ; 14(1): 138, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31196143

ABSTRACT

BACKGROUND: Proteoglycans are large and structurally complex macromolecules which can be found in abundancy in the extracellular matrix and on the surface of all animal cells. Mutations in the genes encoding the enzymes responsible for the formation of the tetrasaccharide linker region between the proteoglycan core protein and the glycosaminoglycan side chains lead to a spectrum of severe and overlapping autosomal recessive connective tissue disorders, collectively coined the 'glycosaminoglycan linkeropathies'. RESULTS: We report the clinical findings of two novel patients with a complex linkeropathy due to biallelic mutations in B3GAT3, the gene that encodes glucuronosyltransferase I, which catalyzes the addition of the ultimate saccharide to the linker region. We identified a previously reported c.667G > A missense mutation and an unreported homozygous c.416C > T missense mutation. We also performed a genotype and phenotype-oriented literature overview of all hitherto reported patients harbouring B3GAT3 mutations. A total of 23 patients from 10 families harbouring bi-allelic mutations and one patient with a heterozygeous splice-site mutation in B3GAT3 have been reported. They all display a complex phenotype characterized by consistent presence of skeletal dysplasia (including short stature, kyphosis, scoliosis and deformity of the long bones), facial dysmorphology, and spatulate distal phalanges. More variably present are cardiac defects, joint hypermobility, joint dislocations/contractures and fractures. Seven different B3GAT3 mutations have been reported, and although the number of patients is still limited, some phenotype-genotype correlations start to emerge. The more severe phenotypes seem to have mutations located in the substrate acceptor subdomain of the catalytic domain of the glucuronosyltransferase I protein while more mildly affected phenotypes seem to have mutations in the NTP-sugar donor substrate binding subdomain. CONCLUSIONS: Loss-of-function mutations in B3GAT3 are associated with a complex connective tissue phenotype characterized by disproportionate short stature, skeletal dysplasia, facial dysmorphism, spatulate distal phalanges and -to a lesser extent- joint contractures, joint hypermobility with dislocations, cardiac defects and bone fragility. Based on the limited number of reported patients, some genotype-phenotype correlations start to emerge.


Subject(s)
Glucuronosyltransferase/metabolism , Connective Tissue/metabolism , Female , Genetic Association Studies , Genotype , Glucuronosyltransferase/genetics , Homozygote , Humans , Male , Mutation/genetics , Mutation, Missense/genetics , Phenotype
15.
Open Med (Wars) ; 14: 251-258, 2019.
Article in English | MEDLINE | ID: mdl-30847403

ABSTRACT

Liver cancer is one of the most malignant tumors with poor prognosis. Finding molecular markers that can predict prognosis is very important for the treatment of liver cancer. The present research is trying to find a new biomarker for human liver cancer. The analysis of abnormal expression genes and prognosis value on liver cancer by Gene Expression Profiling Interactive Analysis (GEPIA) database, the Pathology Atlas of the Human Protein Atlas (HPA), and Kaplan Meier-plotter (KM plotter), proved that B3GAT3 might be one of the important candidates. Furthermore, we investigated the specific role of B3GAT3 on liver cancer through the transfection of B3GAT3 siRNA in HepG2 cells. The proliferation was detected using CCK8, and migration and invasion were determined using Transwell assay. Our results proved that knockdown of B3GAT3 inhibited the proliferation, migration, and invasion. Moreover, B3GAT3 knockdown inhibited the expression of EMT related proteins, N-cad, Snail, and Twist, while promoting the expression of E-cad, suggesting that B3GAT3 knockdown reversed the EMT process of liver cancer cells. In conclusion, overexpressed B3GAT3 promotes the process of tumor EMT, which is an independent prognostic marker to predict the prognosis of liver cancer and might be a potential new target for liver cancer therapy.

16.
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
17.
Food Chem Toxicol ; 123: 142-150, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30367913

ABSTRACT

Recent experimental and epidemiologic investigations have revealed that the central nervous system is a target for vitamin D3 action and also linked vitamin D3 deficiency to Alzheimer's and Parkinson's disease, autism and dementia. Abnormal homeostasis of glutamate and GABA and signaling disbalance are implicated in the pathogenesis of major neurological diseases. Here, key transport characteristics of glutamate and GABA were analysed in presynaptic nerve terminals (synaptosomes) isolated from the cortex of vitamin D3 deficient (VDD) rats. Puberty rats were kept at the VDD diet up to adulthood. VDD caused: (i) a decrease in the initial rates of L-[14C]glutamate and [3H]GABA uptake by plasma membrane transporters of nerve terminals; (ii) a decrease in exocytotic release of L-[14C]glutamate and [3H]GABA; (iii) changes in expression of glutamate (EAAC-1) and GABA (GAT-3) transporters. Whereas, the synaptosomal ambient levels and Ca2+-independent transporter-mediated release of L-[14C]glutamate and [3H]GABA were not significantly altered in VDD. Vitamin D3 is a potent neurosteroid and its nutritional deficiency can provoke development of neurological consequences changing glutamate/GABA transporter expressions and excitation/inhibition balance. Also, changes in glutamate transport can underlie lower resistance to hypoxia/ischemia, larger infarct volumes and worsened outcomes in ischemic stroke patients with VDD.


Subject(s)
Cholecalciferol/deficiency , Excitatory Amino Acid Transporter 3/metabolism , Glutamic Acid/metabolism , Puberty/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Cholecalciferol/metabolism , Excitatory Amino Acid Transporter 3/genetics , Exocytosis , GABA Plasma Membrane Transport Proteins/genetics , GABA Plasma Membrane Transport Proteins/metabolism , Humans , Male , Protein Transport , Puberty/genetics , Rats , Rats, Wistar , Synaptosomes/metabolism
18.
Mol Brain ; 11(1): 4, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29370841

ABSTRACT

Neuronal inhibition is mediated by glycine and/or GABA. Inferior colliculus (IC) neurons receive glycinergic and GABAergic inputs, whereas inhibition in hippocampus (HC) predominantly relies on GABA. Astrocytes heterogeneously express neurotransmitter transporters and are expected to adapt to the local requirements regarding neurotransmitter homeostasis. Here we analyzed the expression of inhibitory neurotransmitter transporters in IC and HC astrocytes using whole-cell patch-clamp and single-cell reverse transcription-PCR. We show that most astrocytes in both regions expressed functional glycine transporters (GlyTs). Activation of these transporters resulted in an inward current (IGly) that was sensitive to the competitive GlyT1 agonist sarcosine. Astrocytes exhibited transcripts for GlyT1 but not for GlyT2. Glycine did not alter the membrane resistance (RM) arguing for the absence of functional glycine receptors (GlyRs). Thus, IGly was mainly mediated by GlyT1. Similarly, we found expression of functional GABA transporters (GATs) in all IC astrocytes and about half of the HC astrocytes. These transporters mediated an inward current (IGABA) that was sensitive to the competitive GAT-1 and GAT-3 antagonists NO711 and SNAP5114, respectively. Accordingly, transcripts for GAT-1 and GAT-3 were found but not for GAT-2 and BGT-1. Only in hippocampal astrocytes, GABA transiently reduced RM demonstrating the presence of GABAA receptors (GABAARs). However, IGABA was mainly not contaminated by GABAAR-mediated currents as RM changes vanished shortly after GABA application. In both regions, IGABA was stronger than IGly. Furthermore, in HC the IGABA/IGly ratio was larger compared to IC. Taken together, our results demonstrate that astrocytes are heterogeneous across and within distinct brain areas. Furthermore, we could show that the capacity for glycine and GABA uptake varies between both brain regions.


Subject(s)
Astrocytes/metabolism , GABA Plasma Membrane Transport Proteins/metabolism , Glycine Plasma Membrane Transport Proteins/metabolism , Hippocampus/metabolism , Animals , Glycine/pharmacology , Inferior Colliculi , Ion Channel Gating/drug effects , Kinetics , Mice, Inbred C57BL , Single-Cell Analysis , gamma-Aminobutyric Acid/pharmacology
19.
Adv Neurobiol ; 16: 137-167, 2017.
Article in English | MEDLINE | ID: mdl-28828609

ABSTRACT

Imbalances in GABA-mediated tonic inhibition are involved in several pathophysiological conditions. A classical way of controlling tonic inhibition is through pharmacological intervention with extrasynaptic GABAA receptors that sense ambient GABA and mediate a persistent GABAergic conductance. An increase in tonic inhibition may, however, also be obtained indirectly by inhibiting glial GABA transporters (GATs). These are sodium-coupled membrane transport proteins that normally act to terminate GABA neurotransmitter action by taking up GABA into surrounding astrocytes. The aim of the review is to provide an overview of glial GATs in regulating tonic inhibition, especially in epilepsy and stroke. This entails a comprehensive summary of changes known to occur in GAT expression levels and signalling following epileptic and ischemic insults. Further, we discuss the accumulating pharmacological evidence for targeting GATs in these diseases.


Subject(s)
Epilepsy/metabolism , GABA Plasma Membrane Transport Proteins/metabolism , Neuroglia/metabolism , Stroke/metabolism , Animals , Epilepsy/physiopathology , Humans , Stroke/physiopathology
20.
Adv Neurobiol ; 16: 315-332, 2017.
Article in English | MEDLINE | ID: mdl-28828618

ABSTRACT

γ-Aminobutyric acid (GABA) neurotransmission is terminated by the GABA transporters (GATs) via uptake of GABA into neurons and surrounding glial cells. Four different transporters have been identified: GAT1, GAT2, GAT3, and the betaine/GABA transporter 1 (BGT1). The GAT1 subtype is the most explored transporter due to its high abundance in the brain and the existence of selective and potent GAT1 inhibitors. Consequently, less is known about the role and therapeutic potential of the non-GAT1 subtypes. Emerging pharmacological evidence suggests that some of these transporters pose interesting targets in several brain disorders. Pharmacological non-GAT1-selective tool compounds are important to further investigate the involvement of GATs in different pathological conditions. Extensive medicinal chemistry efforts have been put into the development of subtype-selective inhibitors, but truly selective and potent inhibitors of non-GAT1 subtypes are still limited. This review covers the advances within the medicinal chemistry area and the structural basis for obtaining non-GAT1-selective inhibitors.


Subject(s)
GABA Plasma Membrane Transport Proteins/metabolism , GABA Uptake Inhibitors/pharmacology , Animals , Brain/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL