Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Neuroscience ; 2024 May 22.
Article in English | MEDLINE | ID: mdl-38788828

ABSTRACT

Gamma-aminobutyric acid and glycine (GABA/Gly) are predominantly inhibitory neurotransmitters in the mature central nervous system; however, they mediate membrane potential depolarization during development. These differences in actions depend on intracellular Cl- concentrations ([Cl-]i), which are primarily regulated by potassium chloride cotransporter 2 (KCC2). After nerve injury, KCC2 expression markedly decreases and GABA/Gly mediate depolarization. Following nerve regeneration, KCC2 expression recovers and GABA/Gly become inhibitory, suggesting that KCC2 reduction and GABA/Gly excitation may be crucial for axonal regeneration. To directly clarify their involvement in regeneration, we analyzed recovery processes after tibial nerve severance and suturing between heterozygous KCC2 knockout mice (HT), whose KCC2 levels are halved, and their wild-type littermates (WT). Compared with WT mice, the sciatic functional index-indicating lower limb motor function-was significantly higher until 28 days after operation (D28) in HT mice. Furthermore, at D7, many neurofilament-positive fibers were elongated into the distal part of the ligated nerve in HT mice only, and myelinated axonal density was significantly higher at D21 and D28 in HT animals. Electron microscopy and galanin immunohistochemistry indicated a shorter nerve degeneration period in HT mice. Moreover, a less severe decrease in choline acetyltransferase was observed in HT mice. These results suggest that nerve degeneration and regeneration proceed more rapidly in HT mice, resulting in milder motor dysfunction. Via similar microglial activation, nerve surgery may reduce KCC2 levels more rapidly in HT mice, followed by earlier increased [Cl-]i and longer-lasting GABA/Gly excitation. Taken together, reduced KCC2 may accelerate nerve regeneration via GABA/Gly excitation.

2.
Acta Histochem Cytochem ; 55(1): 47-56, 2022 Feb 26.
Article in English | MEDLINE | ID: mdl-35444351

ABSTRACT

Gamma-aminobutyric acid (GABA) is an inhibitory neurotransmitter in the mature brain; however, it acts excitatory during development. This difference in action depends on the intracellular chloride ion concentration, primarily regulated by potassium chloride co-transporter2 (KCC2). Sufficient KCC2 expression results in its inhibitory action. GABA is also abundant in pancreatic islets, where it acts differentially on the islet cells, and is involved in carbohydrate metabolism. However, the mechanisms underlying the differential action remain unknown. We performed immunohistochemistry for glutamic acid decarboxylase (GAD), a synthetic enzyme for GABA, and KCC2 in normal adult islets. GAD was co-localized with insulin in ß cells, whereas KCC2 was expressed in glucagon-positive α cells. These results are in line with previous observations that GABA decreases glucagon release but increases insulin release, and suggest that GABA and insulin may work together in reducing blood glucose levels under hyperglycemia. Next, we examined the streptozotocin-induced type1 diabetes mellitus mouse model. GAD and insulin expression levels were markedly decreased. KCC2 was expressed in glucagon-positive cells, whereas insulin- and somatostatin-positive cells were KCC2-negative. These findings suggest that in diabetes model, reduced GABA release may cause disinhibition of glucagon release, resulting in increased blood sugar levels and the maintenance of hyperglycemic state.

3.
Int J Mol Sci ; 23(2)2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35055019

ABSTRACT

Gamma-aminobutyric acid (GABA) and glycine act as inhibitory neurotransmitters. Three types of inhibitory neurons and terminals, GABAergic, GABA/glycine coreleasing, and glycinergic, are orchestrated in the spinal cord neural circuits and play critical roles in regulating pain, locomotive movement, and respiratory rhythms. In this study, we first describe GABAergic and glycinergic transmission and inhibitory networks, consisting of three types of terminals in the mature mouse spinal cord. Second, we describe the developmental formation of GABAergic and glycinergic networks, with a specific focus on the differentiation of neurons, formation of synapses, maturation of removal systems, and changes in their action. GABAergic and glycinergic neurons are derived from the same domains of the ventricular zone. Initially, GABAergic neurons are differentiated, and their axons form synapses. Some of these neurons remain GABAergic in lamina I and II. Many GABAergic neurons convert to a coreleasing state. The coreleasing neurons and terminals remain in the dorsal horn, whereas many ultimately become glycinergic in the ventral horn. During the development of terminals and the transformation from radial glia to astrocytes, GABA and glycine receptor subunit compositions markedly change, removal systems mature, and GABAergic and glycinergic action shifts from excitatory to inhibitory.


Subject(s)
GABAergic Neurons/metabolism , Glycine/metabolism , Receptors, Glycine/metabolism , Signal Transduction , Spinal Cord/metabolism , Synaptic Transmission , gamma-Aminobutyric Acid/metabolism , Animals , Anterior Horn Cells/metabolism , Astrocytes/metabolism , Axons/metabolism , Biomarkers , Ganglia, Spinal/metabolism , Mice , Spinal Cord/cytology , Synapses/metabolism
4.
Neurosci Res ; 177: 52-63, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34757085

ABSTRACT

Peripheral nerve injury affects motor functions. To reveal the mechanisms underlying motor dysfunction and recovery after nerve compression, which have not been precisely examined, we investigated the temporal relationship among changes in motor function, nerve histopathology, and marker molecule expression in the spinal cord after loose ligation of the mouse sciatic nerve. After ligation, sciatic motor function suddenly declined, and axons gradually degenerated. During degeneration, galanin was localized in motor neuron cell bodies. Then, in the ventral horn, microglia were activated, and expression of choline acetyltransferase (ChAT), a synthetic enzyme of acetylcholine, and potassium chloride co-transporter 2 (KCC2), which shifts the action of γ-amino butyric acid (GABA) and glycine to inhibitory, decreased. Motor function recovery was insufficient although axonal regeneration was complete. ChAT levels gradually recovered during axonal regeneration. When regeneration was nearly complete, microglial activation declined, and KCC2 expression started to increase. The KCC2 level sufficiently recovered when axonal regeneration was complete, suggesting that the excitatory action of GABA/glycine may participate in axonal regeneration. Furthermore, these changes proceeded slower than those after severance, suggesting that loose ligation, compression, may mediate slower progression of degeneration and regeneration than severance, and these changes may cause the motor dysfunction and its recovery.


Subject(s)
Peripheral Nerve Injuries , Symporters , Animals , Choline O-Acetyltransferase/metabolism , Glycine/metabolism , Mice , Microglia/metabolism , Motor Neurons/metabolism , Nerve Degeneration/metabolism , Nerve Regeneration/physiology , Peripheral Nerve Injuries/metabolism , Sciatic Nerve/metabolism , Spinal Cord/metabolism , Spinal Cord Ventral Horn/metabolism , Spinal Cord Ventral Horn/pathology , Symporters/metabolism , gamma-Aminobutyric Acid/metabolism , K Cl- Cotransporters
5.
Brain Res ; 1733: 146718, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32045595

ABSTRACT

Gamma-amino butyric acid (GABA) is an inhibitory neurotransmitter in the mature brain, but is excitatory during development and after motor nerve injury. This difference in GABAergic action depends on the intracellular chloride ion concentration ([Cl-]i), primarily regulated by potassium chloride co-transporter 2 (KCC2). To reveal precise processes of the neuropathic pain through changes in GABAergic action, we prepared tibial nerve ligation and severance models using male mice, and examined temporal relationships amongst changes in (1) the mechanical withdrawal threshold in the sural nerve area, (2) localization of the molecules involved in GABAergic transmission and its upstream signaling in the dorsal horn, and (3) histology of the tibial nerve. In the ligation model, tibial nerve degeneration disappeared by day 56, but mechanical allodynia, reduced KCC2 localization, and increased microglia density remained until day 90. Microglia density was higher in the tibial zone than the sural zone before day 21, but this result was inverted after day 28. In contrast, in the severance model, all above changes were detected until day 28, but were simultaneously and significantly recovered by day 90. These results suggested that in male mice, allodynia may be caused by reduced GABAergic synaptic inhibition, resulting from elevated [Cl-]i after the reduction of KCC2 by activated microglia. Furthermore, our results suggested that factors from degenerating nerve terminals may diffuse into the sural zone, whereby they induced the development of allodynia in the sural nerve area, while other factors in the sural zone may mediate persistent allodynia through the same pathway.


Subject(s)
Microglia/metabolism , Neuralgia/metabolism , Symporters/metabolism , Tibial Nerve/injuries , Tibial Nerve/metabolism , Animals , Male , Mice, Inbred C57BL , Neuralgia/pathology , Pain Threshold , Tibial Nerve/pathology , K Cl- Cotransporters
6.
J Chem Neuroanat ; 88: 13-21, 2018 03.
Article in English | MEDLINE | ID: mdl-29113945

ABSTRACT

After injury, peripheral axons usually re-extend toward their target, and neuronal functions recover. Previous studies have reported that expression of various molecules are transiently altered in motor neurons after nerve injury, but the time course of these changes and their relationship with functional recovery have not been clearly demonstrated. We used the mouse facial nerve transection and suturing model, and examined the changes in expression of five molecules, choline acetyl transferase (ChAT), galanin, calcitonin gene-related protein (CGRP), gephyrin, and potassium chloride co-transporter 2 (KCC2) in the facial motor neurons after surgery until recovery. Number of ChAT-positive neurons was markedly decreased at days 3 and 7, and recovered to the normal level by day 60, when facial motor functions recovered. Localization of two neuropeptides, CGRP and galanin, was increased in the perikarya and axons during regeneration, and returned to the normal levels by days 60 and 28, respectively. Expression of two postsynaptic elements of γ-amino butyric acid synapses, gephyrin and KCC2, was decreased at days 3 and 7, and recovered by day 60. These results suggest that ChAT, CGRP, and KCC2 may be objective indicators of regeneration, and altering their expression may be related to the functional recovery and axonal re-extension.


Subject(s)
Calcitonin Gene-Related Peptide/biosynthesis , Choline O-Acetyltransferase/biosynthesis , Facial Nerve/physiology , Motor Neurons/physiology , Nerve Regeneration/physiology , Symporters/biosynthesis , Animals , Biomarkers/analysis , Carrier Proteins/biosynthesis , Facial Nerve Injuries/metabolism , Galanin/biosynthesis , Male , Membrane Proteins/biosynthesis , Mice , Mice, Inbred C57BL , K Cl- Cotransporters
7.
Neurosci Res ; 134: 39-48, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29174921

ABSTRACT

The inhibitory neurotransmitter gamma-amino butyric acid (GABA) plays important roles in energy balance and feeding behavior in the hypothalamus. To reveal the time course of GABAergic network formation, we examined the immunohistochemical localization of glutamic acid decarboxylase (GAD), a GABAergic neuron marker, vesicular GABA transporter (VGAT), a marker of inhibitory terminals, and K+-Cl--cotransporter2 (KCC2), which shifts GABA action from excitation to inhibition, in the developing mouse hypothalamus. GABAergic terminals, seen as GAD- and VGAT-positive dots, increased in density during embryonic development. Moreover, the onset of KCC2 localization was almost concomitant with GABAergic terminal formation, and KCC2-positive profiles increased in density during development. This suggested that after the formation of GABAergic terminals, GABAergic action may change to inhibition in the hypothalamus. This maturation appears to proceed as follows: the lateral hypothalamus (LH) matures first, followed by the paraventricular nucleus (PVN) by the time of birth, while the ventromedial hypothalamus (VMH) and the arcuate nucleus (Arc) are not fully mature at the time of birth. Our findings suggest that GABAergic networks in the "feeding center" (LH) and the "exit" (PVN) may mature before birth, while those in the "satiety center" (VMH) and "higher control center" (Arc) may mature after birth.


Subject(s)
Feeding Behavior/physiology , GABAergic Neurons/physiology , Hypothalamus/cytology , Hypothalamus/embryology , Age Factors , Animals , Embryo, Mammalian , Female , Glutamate Decarboxylase/metabolism , Mice , Mice, Inbred C57BL , Symporters/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , K Cl- Cotransporters
8.
Diabetologia ; 60(8): 1502-1511, 2017 08.
Article in English | MEDLINE | ID: mdl-28528402

ABSTRACT

AIMS/HYPOTHESIS: Overeating of dietary fats causes obesity in humans and rodents. Recent studies in humans and rodents have demonstrated that addiction to fats shares a common mechanism with addiction to alcohol, nicotine and narcotics in terms of a dysfunction of brain reward systems. It has been highlighted that a high-fat diet (HFD) attenuates dopamine D2 receptor (D2R) signalling in the striatum, a pivotal regulator of the brain reward system, resulting in hedonic overeating. We previously reported that the brown rice-specific bioactive constituent γ-oryzanol attenuated the preference for an HFD via hypothalamic control. We therefore explored the possibility that γ-oryzanol would modulate functioning of the brain reward system in mice. METHODS: Male C57BL/6J mice fed an HFD were orally treated with γ-oryzanol, and striatal levels of molecules involved in D2R signalling were evaluated. The impact of γ-oryzanol on DNA methylation of the D2R promoter and subsequent changes in preferences for dietary fat was examined. In addition, the effects of 5-aza-2'-deoxycytidine, a potent inhibitor of DNA methyltransferases (DNMTs), on food preference, D2R signalling and the levels of DNMTs in the striatum were investigated. The inhibitory effects of γ-oryzanol on the activity of DNMTs were enzymatically evaluated in vitro. RESULTS: In striatum from mice fed an HFD, the production of D2Rs was decreased via an increase in DNA methylation of the promoter region of the D2R. Oral administration of γ-oryzanol decreased the expression and activity of DNMTs, thereby restoring the level of D2Rs in the striatum. Pharmacological inhibition of DNMTs by 5-aza-2'-deoxycytidine also ameliorated the preference for dietary fat. Consistent with these findings, enzymatic in vitro assays demonstrated that γ-oryzanol inhibited the activity of DNMTs. CONCLUSIONS/INTERPRETATION: We demonstrated that γ-oryzanol ameliorates HFD-induced DNA hypermethylation of the promoter region of D2R in the striatum of mice. Our experimental paradigm highlights γ-oryzanol as a promising antiobesity substance with the distinct property of being a novel epigenetic modulator.


Subject(s)
Brain/drug effects , Brain/metabolism , Obesity/drug therapy , Oryza/chemistry , Phenylpropionates/chemistry , Phenylpropionates/therapeutic use , Receptors, Dopamine D2/metabolism , Animals , DNA Methylation/genetics , DNA Methylation/physiology , Diet, High-Fat/adverse effects , Feeding and Eating Disorders/drug therapy , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism
9.
Drug Deliv ; 24(1): 558-568, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28181829

ABSTRACT

Our previous works demonstrated that brown rice-specific bioactive substance, γ-oryzanol acts as a chaperone, attenuates exaggerated endoplasmic reticulum (ER) stress in brain hypothalamus and pancreatic islets, thereby ameliorating metabolic derangement in high fat diet (HFD)-induced obese diabetic mice. However, extremely low absorption efficiency from intestine of γ-oryzanol is a tough obstacle for the clinical application. Therefore, in this study, to overcome extremely low bioavailability of γ-oryzanol with super-high lipophilicity, we encapsulated γ-oryzanol in polymer poly (DL-lactide-co-glycolide) (PLGA) nanoparticles (Nano-Orz), and evaluated its metabolically beneficial impact in genetically obese-diabetic ob/ob mice, the best-known severest diabetic model in mice. To our surprise, Nano-Orz markedly ameliorated fuel metabolism with an unexpected magnitude (∼1000-fold lower dose) compared with regular γ-oryzanol. Furthermore, such a conspicuous impact was achievable by its administration once every 2 weeks. Besides the excellent impact on dysfunction of hypothalamus and pancreatic islets, Nano-Orz markedly decreased ER stress and inflammation in liver and adipose tissue. Collectively, nanotechnology-based developments of functional foods oriented toward γ-oryzanol shed light on the novel approach for the treatment of a variety of metabolic diseases in humans.


Subject(s)
Diabetes Mellitus/drug therapy , Drug Carriers , Energy Metabolism/drug effects , Hypoglycemic Agents/administration & dosage , Hypolipidemic Agents/administration & dosage , Lactic Acid/administration & dosage , Nanoparticles , Obesity/drug therapy , Phenylpropionates/administration & dosage , Polyglycolic Acid/administration & dosage , Administration, Oral , Animals , Behavior, Animal/drug effects , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus/blood , Diabetes Mellitus/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Compounding , Endoplasmic Reticulum Stress/drug effects , Food Preferences/drug effects , Gastrointestinal Microbiome/drug effects , Hypoglycemic Agents/chemistry , Hypolipidemic Agents/chemistry , Insulin Resistance , Intestinal Absorption , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Lactic Acid/chemistry , Lipids/blood , Liver/drug effects , Liver/metabolism , Male , Mice, Obese , Nanomedicine , Obesity/blood , Obesity/genetics , Phenylpropionates/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Solubility , Technology, Pharmaceutical/methods , Time Factors
10.
Neuroscience ; 343: 459-471, 2017 02 20.
Article in English | MEDLINE | ID: mdl-28039040

ABSTRACT

In the spinal cord, glycine and γ-amino butyric acid (GABA) are inhibitory neurotransmitters. However, the ontogeny of the glycinergic network remains unclear. To address this point, we examined the developmental formation of glycinergic terminals by immunohistochemistry for glycine transporter 2 (GlyT2), a marker of glycinergic terminals, in developing mouse cervical spinal cord. Furthermore, the developmental localization of GlyT2 was compared with that of glutamic acid decarboxylase (GAD), a marker of GABAergic terminals, and vesicular GABA transporter (VGAT), a marker of inhibitory terminals, by single and double immunolabeling. GlyT2-positive dots (glycinergic terminals) were first detected in the marginal zone on embryonic day 14 (E14). In the ventral horn, they were detected at E16 and increased in observed density during postnatal development. Until postnatal day 7 (P7), GAD-positive dots (GABAergic terminals) were dominant and GlyT2 immunolabeling was localized at GAD-positive dots. During the second postnatal week, GABAergic terminals markedly decreased and glycinergic terminals became dominant. In the dorsal horn, glycinergic terminals were detected at P0 in lamina IV and P7 in lamina III and developmentally increased. GlyT2 was also localized at GAD-positive dots, and colocalizing dots were dominant at P21. VGAT-positive dots (inhibitory terminals) continued to increase until P21. These results suggest that GABAergic terminals first appear during embryonic development and may often change to colocalizing terminals throughout the gray matter during development. The colocalizing terminals may remain in the dorsal horn, whereas in the ventral horn, colocalizing terminals may give rise to glycinergic terminals.


Subject(s)
Anterior Horn Cells/metabolism , Cervical Cord/growth & development , Cervical Cord/metabolism , Glycine/metabolism , Posterior Horn Cells/metabolism , Synapses/metabolism , Animals , Animals, Newborn , Anterior Horn Cells/cytology , Cervical Cord/cytology , Glycine Plasma Membrane Transport Proteins/metabolism , Immunohistochemistry , Male , Microscopy, Electron , Posterior Horn Cells/cytology , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , gamma-Aminobutyric Acid/metabolism
11.
Biochem Biophys Res Commun ; 482(4): 1327-1333, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27939885

ABSTRACT

Class I histone deacetylase (HDAC) inhibitors are believed to have positive effects on neurite outgrowth, synaptic plasticity, and neurogenesis in adult brain. However, the downstream molecular targets of class I HDAC inhibitors in neurons are not clear. Although class I HDAC inhibitors are thought to broadly promote transcription of many neuronal genes through enhancement of histone acetylation, the affected gene set may include unidentified genes that are essential for neuronal survival and function. To identify novel genes that are targets of class I HDAC inhibitors, we used a microarray to screen transcripts from neuronal cultures and evaluated changes in protein and mRNA expression following treatment with four HDAC inhibitors. We identified tescalcin (Tesc) as the most strongly up-regulated gene following treatment with class I HDAC inhibitors in neurons. Moreover, hippocampal neurons overexpressing TESC showed a greater than 5-fold increase in the total length of neurites and number of branch points compared with controls. These findings highlight a potentially important role for TESC in mediating the neuroprotective effect of class I HDAC inhibitors. TESC may also be involved in the development of brain and neurodegenerative diseases through epigenetic mechanisms.


Subject(s)
Calcium-Binding Proteins/chemistry , Hippocampus/cytology , Histone Deacetylase 1/chemistry , Histone Deacetylase Inhibitors/chemistry , Neurons/metabolism , Animals , Calcineurin/chemistry , Calcium/chemistry , Cluster Analysis , Epigenesis, Genetic , Gene Expression Profiling , Green Fluorescent Proteins/metabolism , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/chemistry , Male , Mice , Mice, Inbred C57BL , Neurites/drug effects , Neurodegenerative Diseases/metabolism , Neurogenesis/drug effects , Neuroprotective Agents/pharmacology , Oligonucleotide Array Sequence Analysis , Plasmids/metabolism , Real-Time Polymerase Chain Reaction , Software , Up-Regulation , Valproic Acid/chemistry , Vorinostat
12.
Br J Pharmacol ; 172(18): 4519-4534, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26140534

ABSTRACT

BACKGROUND AND PURPOSE: γ-Oryzanol, derived from unrefined rice, attenuated the preference for dietary fat in mice, by decreasing hypothalamic endoplasmic reticulum stress. However, no peripheral mechanisms, whereby γ-oryzanol could ameliorate glucose dyshomeostasis were explored. Dopamine D2 receptor signalling locally attenuates insulin secretion in pancreatic islets, presumably via decreased levels of intracellular cAMP. We therefore hypothesized that γ-oryzanol would improve high-fat diet (HFD)-induced dysfunction of islets through the suppression of local D2 receptor signalling. EXPERIMENTAL APPROACH: Glucose metabolism and regulation of molecules involved in D2 receptor signalling in pancreatic islets were investigated in male C57BL/6J mice, fed HFD and treated with γ-oryzanol . In isolated murine islets and the beta cell line, MIN6 , the effects of γ-oryzanol on glucose-stimulated insulin secretion (GSIS) was analysed using siRNA for D2 receptors and a variety of compounds which alter D2 receptor signalling. KEY RESULTS: In islets, γ-oryzanol enhanced GSIS via the activation of the cAMP/PKA pathway. Expression of molecules involved in D2 receptor signalling was increased in islets from HFD-fed mice, which were reciprocally decreased by γ-oryzanol. Experiments with siRNA for D2 receptors and D2 receptor ligands in vitro suggest that γ-oryzanol suppressed D2 receptor signalling and augmented GSIS. CONCLUSIONS AND IMPLICATIONS: γ-Oryzanol exhibited unique anti-diabetic properties. The unexpected effects of γ-oryzanol on D2 receptor signalling in islets may provide a novel; natural food-based, approach to anti-diabetic therapy.

13.
Biochem Biophys Res Commun ; 457(2): 206-12, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25562654

ABSTRACT

Cell-penetrating peptides (CPPs) as a novel biomedical delivery system have been highly anticipated, since they can translocate across biological membranes and are capable of transporting their cargo inside live cells with minimal invasiveness. However, non-selective internalization in various cell types remains a challenge in the clinical application of CPPs, especially in cancer treatment. In this study, we attempted to identify novel cancer-homing CPPs to target glioblastoma multiforme (GBM), which is often refractory and resistant to treatment. We screened for CPPs showing affinity for the human GBM cell line, U87MG, from an mRNA display random peptide library. One of the candidate peptides which amino-acid sequence was obtained from the screening showed selective cell-penetrating activity in U87MG cells. Conjugation of the p16(INK4a) functional peptide to the GBM-selective CPP induced cellular apoptosis and reduced phosphorylated retinoblastoma protein levels. This indicates that the CPP was capable of delivering a therapeutic molecule into U87MG cells inducing apoptosis. These results suggest that the novel CPP identified in this study permeates with high affinity into GBM cells, revealing it to be a promising imaging and therapeutic tool in the treatment of glioblastoma.


Subject(s)
Brain Neoplasms/metabolism , Cell-Penetrating Peptides/pharmacology , Glioblastoma/metabolism , Membrane Transport Proteins/metabolism , Amino Acid Sequence , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/therapeutic use , Humans , Molecular Sequence Data
14.
Endocrinology ; 156(4): 1242-50, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25594697

ABSTRACT

Endoplasmic reticulum (ER) stress is profoundly involved in dysfunction of ß-cells under high-fat diet and hyperglycemia. Our recent study in mice showed that γ-oryzanol, a unique component of brown rice, acts as a chemical chaperone in the hypothalamus and improves feeding behavior and diet-induced dysmetabolism. However, the entire mechanism whereby γ-oryzanol improves glucose metabolism throughout the body still remains unclear. In this context, we tested whether γ-oryzanol reduces ER stress and improves function and survival of pancreatic ß-cells using murine ß-cell line MIN6. In MIN6 cells with augmented ER stress by tunicamycin, γ-oryzanol decreased exaggerated expression of ER stress-related genes and phosphorylation of eukaryotic initiation factor-2α, resulting in restoration of glucose-stimulated insulin secretion and prevention of apoptosis. In islets from high-fat diet-fed diabetic mice, oral administration of γ-oryzanol improved glucose-stimulated insulin secretion on following reduction of exaggerated ER stress and apoptosis. Furthermore, we examined the impact of γ-oryzanol on low-dose streptozotocin-induced diabetic mice, where exaggerated ER stress and resultant apoptosis in ß-cells were observed. Also in this model, γ-oryzanol attenuated mRNA level of genes involved in ER stress and apoptotic signaling in islets, leading to amelioration of glucose dysmetabolism. Taken together, our findings demonstrate that γ-oryzanol directly ameliorates ER stress-induced ß-cell dysfunction and subsequent apoptosis, highlighting usefulness of γ-oryzanol for the treatment of diabetes mellitus.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Endoplasmic Reticulum Stress/drug effects , Insulin-Secreting Cells/drug effects , Phenylpropionates/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Diet, High-Fat , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Male , Mice
15.
Brain Res ; 1601: 31-9, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25596421

ABSTRACT

GABA acts as inhibitory neurotransmitter in the adult central nervous system but as excitatory neurotransmitter during early postnatal development. This shift in GABA's action from excitation to inhibition is caused by a decrease in intracellular chloride concentration ([Cl(-)]i), which in turn is caused by changes in the relative expression levels of the K(+)-Cl(-) co-transporter (KCC2) and the Na(+), K(+)-2Cl(-) co-transporter (NKCC1) proteins. Previous studies have used slices containing the medullary pre-Bötzinger complex (pre-BötC) to record respiration-related rhythmic activity (RRA) from the hypoglossal nucleus (12 N). The role of GABAergic transmission in the regulation of medullary RRA neonatally, however, is yet to be determined. Here, we examined how GABA and chloride co-transporters contribute to RRA during development in the 12 N where inspiratory neurons reside. We recorded extracellular RRA in medullary slices obtained from postnatal day (P) 0-7 mice. RRA was induced by soaking slices in artificial cerebrospinal fluid (aCSF) containing 8mM-K(+). Application of GABA significantly increased the frequency of RRA after P3, whereas application of a KCC2 blocker (R (+)-[(2-n-butyl-6,7-dichloro-2-cyclopentyl-2,3-dihydro-1-oxo-1H-indenyl-5-yl)oxy]acetic acid (DIOA)) significantly decreased the frequency of RRA after P1. In addition, dense KCC2 immunolabeling was seen in the superior longitudinalis (SL) of the 12 N, which is responsible for retraction of the tongue, from P0 and P7. These results indicate that GABA administration can increase RRA frequency during the first week following birth. This in turn suggests that decreasing [Cl(-)]i levels caused by increasing KCC2 levels in the 12 N could play important roles in regulating the frequency of RRA during development.


Subject(s)
Medulla Oblongata/physiology , Respiration , Symporters/physiology , gamma-Aminobutyric Acid/physiology , Animals , Medulla Oblongata/drug effects , Medulla Oblongata/growth & development , Mice , Mice, Inbred C57BL , Respiration/drug effects , Solute Carrier Family 12, Member 2/physiology , Symporters/metabolism , gamma-Aminobutyric Acid/pharmacology , K Cl- Cotransporters
16.
Front Cell Neurosci ; 8: 420, 2014.
Article in English | MEDLINE | ID: mdl-25540610

ABSTRACT

In vitro approaches have suggested that neuropsin (or kallikrein 8/KLK8), which controls gamma-aminobutyric acid (GABA) neurotransmission through neuregulin-1 (NRG-1) and its receptor (ErbB4), is involved in neural plasticity (Tamura et al., 2012, 2013). In the present study, we examined whether parvalbumin (PV)-positive neuronal networks, the majority of which are ErbB4-positive GABAergic interneurons, are controlled by neuropsin in tranquil and stimulated voluntarily behaving mice. Parvalbumin-immunoreactive fibers surrounding hippocampal pyramidal and granular neurons in mice reared in their home cage were decreased in neuropsin-deficient mice, suggesting that neuropsin controls PV immunoreactivity. One- or two-week exposures of wild mice to novel environments, in which they could behave freely and run voluntarily in a wheel resulted in a marked upregulation of both neuropsin mRNA and protein in the hippocampus. To elucidate the functional relevance of the increase in neuropsin during exposure to a rich environment, the intensities of PV-immunoreactive fibers were compared between neuropsin-deficient and wild-type (WT) mice under environmental stimuli. When mice were transferred into novel cages (large cages with toys), the intensity of PV-immunoreactive fibers increased in WT mice and neuropsin-deficient mice. Therefore, behavioral stimuli control a neuropsin-independent form of PV immunoreactivity. However, the neuropsin-dependent part of the change in PV-immunoreactive fibers may occur in the stimulated hippocampus because increased levels of neuropsin continued during these enriched conditions.

17.
Neurosci Lett ; 566: 221-5, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24607929

ABSTRACT

In the mature central nervous system, γ-amino butyric acid (GABA) is an inhibitory neurotransmitter, whereas during development, GABA induces depolarization. To examine the embryonic development of GABAergic transmission in the mouse spinal trigeminal nucleus interpolaris (SpVi), which receives sensory input from the face and is important in survival of rodents, we performed immunohistochemistry for three related molecules: glutamic acid decarboxylase (GAD), a marker of GABAergic neurons; vesicular GABA transporter (VGAT), a marker of GABAergic and glycinergic vesicles; and potassium chloride co-transporter 2 (KCC2), which shifts GABA action from excitatory to inhibitory. GAD-positive longitudinal projection fibers, where VGAT-positive dots were localized, were clearly discernible until embryonic day (E)17, and were markedly decreased in number on postnatal day 0. GAD-positive neurons were detected after E15, and GAD- and VGAT-positive axon varicosities were observed after E17. KCC2 immunolabeling was first localized in the dendrites and cell bodies of several neurons in the lateral part of the SpVi on E13 and throughout the nucleus on E17. These results suggest that the SpVi may first receive GABAergic projection fibers from extra-nuclear area before birth, and GABAergic interneurons may form synapses within the SpVi after E17. In addition, GABA action may gradually shift from excitatory to inhibitory between E13 and E17.


Subject(s)
Embryonic Development , Trigeminal Nucleus, Spinal/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Biomarkers/metabolism , GABAergic Neurons/metabolism , Glutamate Decarboxylase/metabolism , Mice, Inbred C57BL , Signal Transduction , Symporters/metabolism , Synapses/metabolism , Trigeminal Nucleus, Spinal/embryology , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , K Cl- Cotransporters
18.
Epilepsy Res ; 93(2-3): 177-84, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21256718

ABSTRACT

Focal cortical dysplasia (FCD), which is characterized histologically by disorganized cortical lamination and large abnormal cells, is one of the major causes of intractable epilepsies. γ-aminobutyric acid (GABA)(A) receptor-mediated synchronous depolarizing potentials have been observed in FCD tissue. Since alterations in Cl(-) homeostasis might underlie these depolarizing actions of GABA, cation-Cl(-) cotransporters could play critical roles in the generation of these abnormal actions. We examined the expression patterns of NKCC1 and KCC2 by in situ hybridization histochemistry and immunohistochemistry in FCD tissue obtained by surgery from patients with intractable epilepsy. KCC2 mRNA and protein were expressed not only in non-dysplastic neurons in histologically normal portions located in the periphery of the excised cortex, but also in dysplastic cells in FCD tissue. The levels of KCC2 mRNA and protein were significantly decreased in the neurons around large abnormal neurons (giant neurons), but not in giant neurons, compared with non-dysplastic neurons. The neurons localized only around giant neurons significantly smaller than non-dysplastic neurons. However NKCC1 expression did not differ among these cell types. These results suggest that the intracellular Cl(-) concentration ([Cl(-)](i)) of small neurons might increase, so that depolarizing GABA actions could occur in the FCD tissue of epileptic foci.


Subject(s)
Epilepsy/genetics , Epilepsy/metabolism , Malformations of Cortical Development/genetics , Malformations of Cortical Development/metabolism , Neurons/metabolism , Symporters/biosynthesis , Symporters/genetics , Adult , Child , Chlorides/metabolism , Down-Regulation , Drug Resistance , Epilepsy/pathology , Female , Humans , Immunohistochemistry , In Situ Hybridization , Male , Malformations of Cortical Development/pathology , Neurons/classification , Neurons/ultrastructure , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sodium-Potassium-Chloride Symporters/biosynthesis , Sodium-Potassium-Chloride Symporters/genetics , Solute Carrier Family 12, Member 2 , Young Adult , gamma-Aminobutyric Acid/metabolism , K Cl- Cotransporters
19.
Brain Cell Biol ; 36(5-6): 173-89, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19132534

ABSTRACT

Although the binding of synaphin (also called complexin) to the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex is critical for synaptic vesicle exocytosis, the exact role of synaphin remains unclear. Here, we show that synaphin directly binds to synaptotagmin 1, a major Ca(2+) sensor for fast neurotransmitter release, in a 1:1 stoichiometry. Mapping of the synaphin site involved in synaptotagmin 1 binding revealed that the C-terminal region is essential for the interaction between these two proteins. Binding was sensitive to ionic strength, suggesting the involvement of charged residues in the C-terminus region. Mutation of the seven consecutive glutamic acid residues (residues 108-114) at the C-terminal region of synaphin to alanines or glutamines resulted in a dramatic reduction in synaptotagmin 1 binding activity. Furthermore, a peptide from the C-terminus of synaphin (residues 91-124) blocked the binding of synaptotagmin 1 to synaphin, an effect that was abolished by mutating the consecutive glutamic acid residues to alanine. Immunoprecipitation experiments with brain membrane extracts showed the presence of a complex consisting of synaphin, synaptotagmin 1, and SNAREs. We propose that synaphin recruits synaptotagmin 1 to the SNARE-based fusion complex and synergistically functions with synaptotagmin 1 in mediating fast synaptic vesicle exocytosis.


Subject(s)
Exocytosis/physiology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , SNARE Proteins/metabolism , Synaptic Vesicles/metabolism , Synaptotagmin I/metabolism , Adaptor Proteins, Vesicular Transport , Animals , DNA, Complementary , Exocytosis/genetics , Immunoprecipitation , Nerve Tissue Proteins/genetics , Neurons/cytology , Polymerase Chain Reaction , Rats , SNARE Proteins/genetics , Synaptic Vesicles/genetics , Synaptotagmin I/genetics
20.
Neurosci Res ; 59(3): 288-95, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17904674

ABSTRACT

Focal cortical malformations comprise a heterogeneous group of disturbances in brain development, often associated with intractable epilepsy. A focal freeze-lesion of cerebral cortex in newborn rat produces a cortical malformation that resembles human polymicrogyria, clinical conditions that results from abnormal neuronal migration. The change in GABAergic functions that occurs during early brain development is induced by an alteration in Cl(-) homeostasis and plays important roles in neocortical development by modulating such events as laminar organization and synaptogenesis. We therefore investigated the relationship between pathogenesis of polymicrogyria and ontogeny of Cl(-) homeostasis in developing parietal cortex after creation of a freeze-lesion at P0. We demonstrated, by in situ hybridization histochemistry for cation-Cl(-) cotrtansporters, that NKCC1 mRNA expression was upregulated and KCC2 mRNA expression downregulated at P4 in "bridge" structure (formed in lesion site across the gap in intact exofocal cortex) as compared to exfocal cortex. Immunohistochemical investigation revealed a colocalization of NKCC1 and neuron specific enolase (NSE) within this structure, while BrdU-positive cells express GFAP and NKCC1 appeared beneath it. These results suggest that immature cortical plate neurons might produce "bridge" structure during formation of microgyrus, and that altered neuronal Cl(-) homeostasis might be involved in neuronal migration disorder that ultimately results in cortical malformations.


Subject(s)
Cell Movement/physiology , Cerebral Cortex/abnormalities , Cerebral Cortex/metabolism , Chlorides/metabolism , Nervous System Malformations/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Symporters/metabolism , Animals , Biomarkers , Bromodeoxyuridine , Cell Differentiation/physiology , Cerebral Cortex/physiopathology , Chloride Channels/metabolism , Cold Temperature/adverse effects , Denervation , Down-Regulation/physiology , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Male , Nervous System Malformations/physiopathology , Parietal Lobe/abnormalities , Parietal Lobe/metabolism , Parietal Lobe/physiopathology , Phosphopyruvate Hydratase/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Sodium-Potassium-Chloride Symporters/genetics , Solute Carrier Family 12, Member 2 , Symporters/genetics , Up-Regulation/physiology , gamma-Aminobutyric Acid/metabolism , K Cl- Cotransporters
SELECTION OF CITATIONS
SEARCH DETAIL
...