Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Physiol Rep ; 12(11): e16091, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38862270

ABSTRACT

Sildenafil, a phosphodiesterase-5 (PDE5) inhibitor, has been shown to improve insulin sensitivity in animal models and prediabetic patients. However, its other metabolic effects remain poorly investigated. This study examines the impact of sildenafil on insulin secretion in MIN6-K8 mouse clonal ß cells. Sildenafil amplified insulin secretion by enhancing Ca2+ influx. These effects required other depolarizing stimuli in MIN6-K8 cells but not in KATP channel-deficient ß cells, which were already depolarized, indicating that sildenafil-amplified insulin secretion is depolarization-dependent and KATP channel-independent. Interestingly, sildenafil-amplified insulin secretion was inhibited by pharmacological inhibition of R-type channels, but not of other types of voltage-dependent Ca2+ channels (VDCCs). Furthermore, sildenafil-amplified insulin secretion was barely affected when its effect on cyclic GMP was inhibited by PDE5 knockdown. Thus, sildenafil stimulates insulin secretion and Ca2+ influx through R-type VDCCs independently of the PDE5/cGMP pathway, a mechanism that differs from the known pharmacology of sildenafil and conventional insulin secretory pathways. Our results reposition sildenafil as an insulinotropic agent that can be used as a potential antidiabetic medicine and a tool to elucidate the novel mechanism of insulin secretion.


Subject(s)
Calcium , Insulin Secretion , Insulin-Secreting Cells , Insulin , Phosphodiesterase 5 Inhibitors , Sildenafil Citrate , Sildenafil Citrate/pharmacology , Animals , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Mice , Insulin Secretion/drug effects , Phosphodiesterase 5 Inhibitors/pharmacology , Calcium/metabolism , Insulin/metabolism , Cell Line
2.
Peptides ; 179: 171267, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38908517

ABSTRACT

Signs and symptoms of hypernatremia largely indicate central nervous system dysfunction. Acute hypernatremia can cause demyelinating lesions similar to that observed in osmotic demyelination syndrome (ODS). We have previously demonstrated that microglia accumulate in ODS lesions and minocycline protects against ODS by inhibiting microglial activation. However, the direct effect of rapid rise in the sodium concentrations on microglia is largely unknown. In addition, the effect of chronic hypernatremia on microglia also remains elusive. Here, we investigated the effects of acute (6 or 24 h) and chronic (the extracellular sodium concentration was increased gradually for at least 7 days) high sodium concentrations on microglia using the microglial cell line, BV-2. We found that both acute and chronic high sodium concentrations increase NOS2 expression and nitric oxide (NO) production. We also demonstrated that the expression of nuclear factor of activated T-cells-5 (NFAT5) is increased by high sodium concentrations. Furthermore, NFAT5 knockdown suppressed NOS2 expression and NO production. We also demonstrated that high sodium concentrations decreased intracellular Ca2+ concentration and an inhibitor of Na+/Ca2+ exchanger, NCX, suppressed a decrease in intracellular Ca2+ concentrations and NOS2 expression and NO production induced by high sodium concentrations. Furthermore, minocycline inhibited NOS2 expression and NO production induced by high sodium concentrations. These in vitro data suggest that microglial activity in response to high sodium concentrations is regulated by NFAT5 and Ca2+ efflux through NCX and is suppressed by minocycline.

3.
BMC Res Notes ; 16(1): 202, 2023 Sep 11.
Article in English | MEDLINE | ID: mdl-37697384

ABSTRACT

OBJECTIVE: Insulin secretion is regulated by ATP-sensitive potassium (KATP) channels in pancreatic beta-cells. Peroxisome proliferator-activated receptors (PPAR) α ligands are clinically used to treat dyslipidemia. A PPARα ligand, fenofibrate, and PPARγ ligands troglitazone and 15-deoxy-∆12,14-prostaglandin J2 are known to close KATP channels and induce insulin secretion. The recently developed PPARα ligand, pemafibrate, became a new entry for treating dyslipidemia. Because pemafibrate is reported to improve glucose intolerance in mice treated with a high fat diet and a novel selective PPARα modulator, it may affect KATP channels or insulin secretion. RESULTS: The effect of fenofibrate (100 µM) and pemafibrate (100 µM) on insulin secretion from MIN6 cells was measured by using batch incubation for 10 and 60 min in low (2 mM) and high (10 mM) glucose conditions. The application of fenofibrate for 10 min significantly increased insulin secretion in low glucose conditions. Pemafibrate failed to increase insulin secretion in all of the conditions experimented in this study. The KATP channel activity was measured by using whole-cell patch clamp technique. Although fenofibrate (100 µM) reduced the KATP channel current, the same concentration of pemafibrate had no effect. Both fenofibrate and pemafibrate had no effect on insulin mRNA expression.


Subject(s)
Fenofibrate , Animals , Mice , PPAR alpha , Ligands , Insulin Secretion , Glucose , KATP Channels , Adenosine Triphosphate
4.
J Diabetes Investig ; 14(9): 1045-1055, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37300240

ABSTRACT

AIMS/INTRODUCTION: Glucagon is secreted from pancreatic α-cells and plays an important role in amino acid metabolism in liver. Various animal models deficient in glucagon action show hyper-amino acidemia and α-cell hyperplasia, indicating that glucagon contributes to feedback regulation between the liver and the α-cells. In addition, both insulin and various amino acids, including branched-chain amino acids and alanine, participate in protein synthesis in skeletal muscle. However, the effect of hyperaminoacidemia on skeletal muscle has not been investigated. In the present study, we examined the effect of blockade of glucagon action on skeletal muscle using mice deficient in proglucagon-derived peptides (GCGKO mice). MATERIALS AND METHODS: Muscles isolated from GCGKO and control mice were analyzed for their morphology, gene expression and metabolites. RESULTS: GCGKO mice showed muscle fiber hypertrophy, and a decreased ratio of type IIA and an increased ratio of type IIB fibers in the tibialis anterior. The expression levels of myosin heavy chain (Myh) 7, 2, 1 and myoglobin messenger ribonucleic acid were significantly lower in GCGKO mice than those in control mice in the tibialis anterior. GCGKO mice showed a significantly higher concentration of arginine, asparagine, serine and threonine in the quadriceps femoris muscles, and also alanine, aspartic acid, cysteine, glutamine, glycine and lysine, as well as four amino acids in gastrocnemius muscles. CONCLUSIONS: These results show that hyperaminoacidemia induced by blockade of glucagon action in mice increases skeletal muscle weight and stimulates slow-to-fast transition in type II fibers of skeletal muscle, mimicking the phenotype of a high-protein diet.


Subject(s)
Glucagon , Muscle, Skeletal , Proglucagon , Animals , Mice , Amino Acids , Glucagon/metabolism , Muscle, Skeletal/metabolism , Proglucagon/genetics , Proglucagon/metabolism
5.
Sci Rep ; 12(1): 21840, 2022 12 17.
Article in English | MEDLINE | ID: mdl-36528738

ABSTRACT

There are no available therapies targeting the underlying molecular mechanisms of neurodegenerative diseases. Although chaperone therapies that alleviate endoplasmic reticulum (ER) stress recently showed promise in the treatment of neurodegenerative diseases, the detailed mechanisms remain unclear. We previously reported that mice with central nervous system-specific deletion of Derlin-1, which encodes an essential component for ER quality control, are useful as models of neurodegenerative diseases such as spinocerebellar degeneration. Cholesterol biosynthesis is essential for brain development, and its disruption inhibits neurite outgrowth, causing brain atrophy. In this study, we report a novel mechanism by which chemical chaperones ameliorate brain atrophy and motor dysfunction. ER stress was induced in the cerebella of Derlin-1 deficiency mice, whereas the administration of a chemical chaperone did not alleviate ER stress. However, chemical chaperone treatment ameliorated cholesterol biosynthesis impairment through SREBP-2 activation and simultaneously relieved brain atrophy and motor dysfunction. Altogether, these findings demonstrate that ER stress may not be the target of action of chaperone therapies and that chemical chaperone-mediated improvement of brain cholesterol biosynthesis is a promising novel therapeutic strategy for neurodegenerative diseases.


Subject(s)
Membrane Proteins , Neurodegenerative Diseases , Animals , Mice , Atrophy/drug therapy , Cholesterol , Endoplasmic Reticulum Stress , Membrane Proteins/chemistry , Molecular Chaperones , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/genetics
6.
Mol Metab ; 55: 101414, 2022 01.
Article in English | MEDLINE | ID: mdl-34871777

ABSTRACT

OBJECTIVE: Age is a risk factor for type 2 diabetes (T2D). We aimed to elucidate whether ß-cell glucose metabolism is altered with aging and contributes to T2D. METHODS: We used senescence-accelerated mice (SAM), C57BL/6J (B6) mice, and ob/ob mice as aging models. As a diabetes model, we used db/db mice. The glucose responsiveness of insulin secretion and the [U-13C]-glucose metabolic flux were examined in isolated islets. We analyzed the expression of ß-cell-specific genes in isolated islets and pancreatic sections as molecular signatures of ß-cell identity. ß cells defective in the malate-aspartate (MA) shuttle were previously generated from MIN6-K8 cells by the knockout of Got1, a component of the shuttle. We analyzed Got1 KO ß cells as a model of increased glycolysis. RESULTS: We identified hyperresponsiveness to glucose and compromised cellular identity as dysfunctional phenotypes shared in common between aged and diabetic mouse ß cells. We also observed a metabolic commonality between aged and diabetic ß cells: hyperactive glycolysis through the increased expression of nicotinamide mononucleotide adenylyl transferase 2 (Nmnat2), a cytosolic nicotinamide adenine dinucleotide (NAD)-synthesizing enzyme. Got1 KO ß cells showed increased glycolysis, ß-cell dysfunction, and impaired cellular identity, phenocopying aging and diabetes. Using Got1 KO ß cells, we show that attenuation of glycolysis or Nmnat2 activity can restore ß-cell function and identity. CONCLUSIONS: Our study demonstrates that hyperactive glycolysis is a metabolic signature of aged and diabetic ß cells, which may underlie age-related ß-cell dysfunction and loss of cellular identity. We suggest Nmnat2 suppression as an approach to counteract age-related T2D.


Subject(s)
Aging/physiology , Glycolysis/physiology , Insulin-Secreting Cells/physiology , Animals , Cell Line , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Glycemic Control/methods , Insulin/metabolism , Insulin Secretion/physiology , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Obesity/metabolism
7.
iScience ; 24(7): 102758, 2021 Jul 23.
Article in English | MEDLINE | ID: mdl-34355142

ABSTRACT

Derlin family members (Derlins) are primarily known as components of the endoplasmic reticulum-associated degradation pathway that eliminates misfolded proteins. Here we report a function of Derlins in the brain development. Deletion of Derlin-1 or Derlin-2 in the central nervous system of mice impaired postnatal brain development, particularly of the cerebellum and striatum, and induced motor control deficits. Derlin-1 or Derlin-2 deficiency reduced neurite outgrowth in vitro and in vivo and surprisingly also inhibited sterol regulatory element binding protein 2 (SREBP-2)-mediated brain cholesterol biosynthesis. In addition, reduced neurite outgrowth due to Derlin-1 deficiency was rescued by SREBP-2 pathway activation. Overall, our findings demonstrate that Derlins sustain brain cholesterol biosynthesis, which is essential for appropriate postnatal brain development and function.

8.
J Diabetes Investig ; 12(6): 920-930, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33417747

ABSTRACT

AIMS/INTRODUCTION: Glutamine is the most abundant amino acid in the circulation. In this study, we investigated cell signaling in the amplification of insulin secretion by glutamine. MATERIALS AND METHODS: Clonal pancreatic ß-cells MIN6-K8, wild-type B6 mouse islets, glutamate dehydrogenase (GDH) knockout clonal ß-cells (Glud1KOßCL), and glutamate-oxaloacetate transaminase 1 (GOT1) knockout clonal ß-cells (Got1KOßCL) were studied. Insulin secretion from these cells and islets was examined under various conditions, and intracellular glutamine metabolism was assessed by metabolic flux analysis. Intracellular Ca2+ concentration ([Ca2+ ]i ) was also measured. RESULTS: Glutamine dose-dependently amplified insulin secretion in the presence of high glucose in both MIN6-K8 cells and Glud1KOßCL. Inhibition of glutaminases, the enzymes that convert glutamine to glutamate, dramatically reduced the glutamine-amplifying effect on insulin secretion. A substantial amount of glutamate was produced from glutamine through direct conversion by glutaminases. Glutamine also increased [Ca2+ ]i at high glucose, which was abolished by inhibition of glutaminases. Glutamic acid dimethylester (dm-Glu), a membrane permeable glutamate precursor that is converted to glutamate in cells, increased [Ca2+ ]i as well as induced insulin secretion at high glucose. These effects of glutamine and dm-Glu were dependent on calcium influx. Glutamine also induced insulin secretion in clonal ß-cells MIN6-m14, which otherwise exhibit no insulin secretory response to glucose. CONCLUSIONS: Glutamate converted from glutamine is an essential mediator that enhances calcium signaling in the glutamine-amplifying effect on insulin secretion. Our data also suggest that glutamine exerts a permissive effect on glucose-induced insulin secretion.


Subject(s)
Glutamic Acid/metabolism , Glutamine/metabolism , Insulin Secretion/physiology , Insulin-Secreting Cells/metabolism , Animals , Aspartate Aminotransferase, Cytoplasmic , Cells, Cultured , Glucose/metabolism , Glutamate Dehydrogenase , Insulin/metabolism , Islets of Langerhans/cytology , Mice , Signal Transduction
9.
J Clin Invest ; 130(12): 6639-6655, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33196462

ABSTRACT

By restoring glucose-regulated insulin secretion, glucagon-like peptide-1-based (GLP-1-based) therapies are becoming increasingly important in diabetes care. Normally, the incretins GLP-1 and glucose-dependent insulinotropic polypeptide (GIP) jointly maintain normal blood glucose levels by stimulation of insulin secretion in pancreatic ß cells. However, the reason why only GLP-1-based drugs are effective in improving insulin secretion after presentation of diabetes has not been resolved. ATP-sensitive K+ (KATP) channels play a crucial role in coupling the systemic metabolic status to ß cell electrical activity for insulin secretion. Here, we have shown that persistent membrane depolarization of ß cells due to genetic (ß cell-specific Kcnj11-/- mice) or pharmacological (long-term exposure to sulfonylureas) inhibition of the KATP channel led to a switch from Gs to Gq in a major amplifying pathway of insulin secretion. The switch determined the relative insulinotropic effectiveness of GLP-1 and GIP, as GLP-1 can activate both Gq and Gs, while GIP only activates Gs. The findings were corroborated in other models of persistent depolarization: a spontaneous diabetic KK-Ay mouse and nondiabetic human and mouse ß cells of pancreatic islets chronically treated with high glucose. Thus, a Gs/Gq signaling switch in ß cells exposed to chronic hyperglycemia underlies the differential insulinotropic potential of incretins in diabetes.


Subject(s)
Chromogranins/metabolism , Diabetes Mellitus, Experimental/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Incretins/pharmacology , Insulin-Secreting Cells/metabolism , Signal Transduction , Animals , Chromogranins/genetics , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Humans , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/pathology , Mice , Mice, Knockout , Potassium Channels, Inwardly Rectifying/deficiency , Potassium Channels, Inwardly Rectifying/metabolism
10.
J Diabetes Investig ; 11(6): 1434-1447, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32279428

ABSTRACT

AIMS/INTRODUCTION: Pancreatic islets are heterogenous. To clarify the relationship between islet heterogeneity and incretin action in the islets, we studied gene expression and metabolic profiles of non-large and enlarged islets of the Zucker fatty diabetes mellitus rat, an obese diabetes model, as well as incretin-induced insulin secretion (IIIS) in these islets. MATERIALS AND METHODS: Pancreatic islets of control (fa/+) and fatty (fa/fa) rats at 8 and 12 weeks-of-age were isolated. The islets of fa/fa rats at 12 weeks-of-age were separated into non-large islets (≤200 µm in diameter) and enlarged islets (>300 µm in diameter). Morphological analyses, insulin secretion experiments, transcriptome analysis, metabolome analysis and oxygen consumption analysis were carried out on these islets. RESULTS: The number of enlarged islets was increased with age in fatty rats, and IIIS was significantly reduced in the enlarged islets. Markers for ß-cell differentiation were markedly decreased in the enlarged islets, but those for cell proliferation were increased. Glycolysis was enhanced in the enlarged islets, whereas the tricarboxylic acid cycle was suppressed. The oxygen consumption rate under glucose stimulation was reduced in the enlarged islets. Production of glutamate, a key signal for IIIS, was decreased in the enlarged islets. CONCLUSIONS: The enlarged islets of Zucker fatty diabetes mellitus rats, which are defective for IIIS, show tumor cell-like metabolic features, including a dedifferentiated state, accelerated aerobic glycolysis and impaired mitochondrial function. The age-dependent increase in such islets could contribute to the pathophysiology of obese diabetes.


Subject(s)
Gene Expression Regulation/drug effects , Incretins/toxicity , Insulin Secretion/drug effects , Islets of Langerhans/pathology , Metabolome/drug effects , Obesity/physiopathology , Pancreatic Neoplasms/pathology , Animals , Gene Expression Profiling , Islets of Langerhans/drug effects , Male , Pancreatic Neoplasms/chemically induced , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Rats , Rats, Zucker
11.
Neurosci Res ; 143: 31-43, 2019 Jun.
Article in English | MEDLINE | ID: mdl-29859850

ABSTRACT

Adult neurogenesis is a process of generating new neurons from neural stem/precursor cells (NS/PCs) in restricted adult brain regions throughout life. It is now generally known that adult neurogenesis in the hippocampal dentate gyrus (DG) and subventricular zone participates in various higher brain functions, such as learning and memory formation, olfactory discrimination and repair after brain injury. However, the mechanisms underlying adult neurogenesis remain to be fully understood. Here, we show that Nuclear protein 95 KDa (Np95, also known as UHRF1 or ICBP90), which is an essential protein for maintaining DNA methylation during cell division, is involved in multiple processes of adult neurogenesis. Specific ablation of Np95 in adult NS/PCs (aNS/PCs) led to a decrease in their proliferation and an impairment of neuronal differentiation and to suppression of neuronal maturation associated with the impairment of dendritic formation in the hippocampal DG. We also found that deficiency of Np95 in NS/PCs increased the expression of tumor suppressor genes p16 and p53, and confirmed that expression of these genes in NS/PCs recapitulates the phenotype of Np95-deficient NS/PCs. Taken together, our findings suggest that Np95 plays an essential role in proliferation and differentiation of aNS/PCs through the regulation of tumor suppressor gene expression in adult neurogenesis.


Subject(s)
Adult Stem Cells/physiology , Gene Expression Regulation , Genes, Tumor Suppressor , Neural Stem Cells/physiology , Nuclear Proteins/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , CCAAT-Enhancer-Binding Proteins , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Dentate Gyrus/metabolism , Hippocampus/metabolism , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/physiology , Nuclear Proteins/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases
12.
Diabetes ; 67(9): 1795-1806, 2018 09.
Article in English | MEDLINE | ID: mdl-29954738

ABSTRACT

ß-Cell-ß-cell interactions are required for normal regulation of insulin secretion. We previously found that formation of spheroid clusters (called K20-SC) from MIN6-K20 clonal ß-cells lacking incretin-induced insulin secretion (IIIS) under monolayer culture (called K20-MC) drastically induced incretin responsiveness. Here we investigated the mechanism by which an incretin-unresponsive state transforms to an incretin-responsive state using K20-SC as a model. Glutamate production by glucose through the malate-aspartate shuttle and cAMP signaling, both of which are critical for IIIS, were enhanced in K20-SC. SC formed from ß-cells deficient for aspartate aminotransferase 1, a critical enzyme in the malate-aspartate shuttle, exhibited reduced IIIS. Expression of the sodium-coupled neutral amino acid transporter 5 (SNAT5), which is involved in glutamine transport, was downregulated in K20-SC and pancreatic islets of normal mice but was upregulated in K20-MC and islets of rodent models of obesity and diabetes, both of which exhibit impaired IIIS. Inhibition of SNAT5 significantly increased cellular glutamate content and improved IIIS in islets of these models and in K20-MC. These results suggest that suppression of SNAT5 activity, which results in increased glutamate production, and enhancement of cAMP signaling endows incretin-unresponsive ß-cells with incretin responsiveness.


Subject(s)
Amino Acid Transport Systems, Neutral/antagonists & inhibitors , Diabetes Mellitus, Type 2/drug therapy , Incretins/pharmacology , Insulin-Secreting Cells/drug effects , Membrane Transport Modulators/pharmacology , Models, Biological , Obesity/drug therapy , Amino Acid Transport Systems, Neutral/agonists , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Anti-Obesity Agents/pharmacology , Cell Communication/drug effects , Cell Line , Cells, Cultured , Clone Cells , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Drug Resistance/drug effects , Gene Expression Regulation/drug effects , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/ultrastructure , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans/ultrastructure , Male , Mice, Inbred Strains , Microscopy, Electron, Transmission , Obesity/metabolism , Obesity/pathology , RNA Interference , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Spheroids, Cellular/ultrastructure , Tissue Culture Techniques
13.
Dev Dyn ; 247(1): 229-238, 2018 01.
Article in English | MEDLINE | ID: mdl-28771884

ABSTRACT

BACKGROUND: Although quiescent neural stem cells (NSCs) in the adult hippocampus proliferate in response to neurogenic stimuli and subsequently give rise to new neurons continuously throughout life, misregulation of NSCs in pathological conditions, including aging, leads to the impairment of learning and memory. High mobility group B family 1 (HMGB1) and HMGB2, HMG family proteins that function as transcriptional activators through the modulation of chromatin structure, have been assumed to play some role in the regulation of adult NSCs; however, their precise functions and even expression patterns in the adult hippocampus remain elusive. RESULTS: Here we show that expression of HMGB2 but not HMGB1 is restricted to the subset of NSCs and their progenitors. Furthermore, running, a well-known positive neurogenic stimulus, increased the proliferation of HMGB2-expressing cells, whereas aging was accompanied by a marked decrease in these cells. Intriguingly, HMGB2-expressing quiescent NSCs, which were shifted toward the proliferative state, were decreased as aging progressed. CONCLUSIONS: HMGB2 expression is strongly associated with transition from the quiescent to the proliferative state of NSCs, supporting the possibility that HMGB2 is involved in the regulation of adult neurogenesis and can be used as a novel marker to identify NSCs primed for activation in the adult hippocampus. Developmental Dynamics 247:229-238, 2018. © 2017 Wiley Periodicals, Inc.


Subject(s)
Adult Stem Cells/metabolism , HMGB2 Protein/metabolism , Hippocampus/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Adult Stem Cells/cytology , Animals , HMGB2 Protein/genetics , Hippocampus/cytology , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , Mice , Neural Stem Cells/cytology , Neurons/cytology , Neurons/metabolism
14.
PLoS One ; 12(11): e0187213, 2017.
Article in English | MEDLINE | ID: mdl-29091932

ABSTRACT

Incretins (GLP-1 and GIP) potentiate insulin secretion through cAMP signaling in pancreatic ß-cells in a glucose-dependent manner. We recently proposed a mechanistic model of incretin-induced insulin secretion (IIIS) that requires two critical processes: 1) generation of cytosolic glutamate through the malate-aspartate (MA) shuttle in glucose metabolism and 2) glutamate transport into insulin granules by cAMP signaling to promote insulin granule exocytosis. To directly prove the model, we have established and characterized CRISPR/Cas9-engineered clonal mouse ß-cell lines deficient for the genes critical in these two processes: aspartate aminotransferase 1 (AST1, gene symbol Got1), a key enzyme in the MA shuttle, which generates cytosolic glutamate, and the vesicular glutamate transporters (VGLUT1, VGLUT2, and VGLUT3, gene symbol Slc17a7, Slc17a6, and Slc17a8, respectively), which participate in glutamate transport into secretory vesicles. Got1 knockout (KO) ß-cell lines were defective in cytosolic glutamate production from glucose and showed impaired IIIS. Unexpectedly, different from the previous finding that global Slc17a7 KO mice exhibited impaired IIIS from pancreatic islets, ß-cell specific Slc17a7 KO mice showed no significant impairment in IIIS, as assessed by pancreas perfusion experiment. Single Slc17a7 KO ß-cell lines also retained IIIS, probably due to compensatory upregulation of Slc17a6. Interestingly, triple KO of Slc17a7, Slc17a6, and Slc17a8 diminished IIIS, which was rescued by exogenously introduced wild-type Slc17a7 or Slc17a6 genes. The present study provides direct evidence for the essential roles of AST1 and VGLUTs in ß-cell glutamate signaling for IIIS and also shows the usefulness of the CRISPR/Cas9 system for studying ß-cells by simultaneous disruption of multiple genes.


Subject(s)
Aspartate Aminotransferases/metabolism , Glutamic Acid/metabolism , Incretins/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Signal Transduction , Vesicular Glutamate Transport Proteins/metabolism , Animals , Aspartate Aminotransferases/genetics , Cell Line , Insulin Secretion , Mice , Mice, Knockout , Mutation , Vesicular Glutamate Transport Proteins/genetics
15.
J Biochem ; 162(3): 155-162, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28903548

ABSTRACT

The unfolded protein response (UPR) is an intracellular homeostatic signalling pathway that is induced by accumulated misfolded/unfolded proteins in the endoplasmic reticulum (ER). The UPR is closely associated with the development of disease in several tissues, including the central nervous system (CNS), in response to ER stress. More recently, the unique features and importance of the UPR have been revealed in neural stem cells (NSCs) and differentiated CNS cells [neurons and glial cells (astrocytes and oligodendrocytes)]. Although several UPR signalling pathways dynamically change in each CNS cell during brain development, the role of UPR signalling in CNS cells (especially NSCs and glial cells) under pathological or physiological conditions is poorly understood. Here, we discuss and summarize the recent progress in understanding how the UPR regulates the proliferation, differentiation, maturation and viability of CNS cells.


Subject(s)
Central Nervous System/metabolism , Unfolded Protein Response , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , Central Nervous System/cytology , Endoplasmic Reticulum/metabolism , Humans
16.
Neurogenesis (Austin) ; 3(1): e1232679, 2016.
Article in English | MEDLINE | ID: mdl-27844025

ABSTRACT

Despite recent advances in our understanding of epigenetic regulation of central nervous system development, little is known regarding the effects of epigenetic dysregulation on neurogenesis and brain function in adulthood. In the present study, we show that prenatal deletion of DNA methyltransferase 1 (Dnmt1) in neural stem cells results in impaired neurogenesis as well as increases in inflammatory features (e.g., elevated glial fibrillary acidic protein [GFAP] expression in astrocytes and increased numbers of microglia) in the adult mouse brain. Moreover, these mice exhibited anxiety-like behavior during an open-field test. These findings suggest that Dnmt1 plays a critical role in regulating neurogenesis and behavior in the developing brain and into adulthood.

17.
J Neurosci ; 36(22): 6050-68, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27251626

ABSTRACT

UNLABELLED: Development of the hippocampal dentate gyrus (DG) in the mammalian brain is achieved through multiple processes during late embryonic and postnatal stages, with each developmental step being strictly governed by extracellular cues and intracellular mechanisms. Here, we show that the maintenance DNA methyltransferase 1 (Dnmt1) is critical for development of the DG in the mouse. Deletion of Dnmt1 in neural stem cells (NSCs) at the beginning of DG development led to a smaller size of the granule cell layer in the DG. NSCs lacking Dnmt1 failed to establish proper radial processes or to migrate into the subgranular zone, resulting in aberrant neuronal production in the molecular layer of the DG and a reduction of integrated neurons in the granule cell layer. Interestingly, prenatal deletion of Dnmt1 in NSCs affected not only the developmental progression of the DG but also the properties of NSCs maintained into adulthood: Dnmt1-deficient NSCs displayed impaired neurogenic ability and proliferation. We also found that Dnmt1 deficiency in NSCs decreased the expression of Reelin signaling components in the developing DG and increased that of the cell cycle inhibitors p21 and p57 in the adult DG. Together, these findings led us to propose that Dnmt1 functions as a key regulator to ensure the proper development of the DG, as well as the proper status of NSCs maintained into adulthood, by modulating extracellular signaling and intracellular mechanisms. SIGNIFICANCE STATEMENT: Here, we provide evidence that Dnmt1 is required for the proper development of the hippocampal dentate gyrus (DG). Deletion of Dnmt1 in neural stem cells (NSCs) at an early stage of DG development impaired the ability of NSCs to establish secondary radial glial scaffolds and to migrate into the subgranular zone of the DG, leading to aberrant neuronal production in the molecular layer, increased cell death, and decreased granule neuron production. Prenatal deletion of Dnmt1 in NSCs also induced defects in the proliferation and neurogenic ability of adult NSCs. Furthermore, we found that Dnmt1 regulates the expression of key extracellular signaling components during developmental stages while modulating intracellular mechanisms for proliferation and neuronal production of NSCs in the adult.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Dentate Gyrus , Gene Expression Regulation, Developmental , Neurons/physiology , Animals , Animals, Newborn , Cell Differentiation/genetics , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferase 1 , Dentate Gyrus/cytology , Dentate Gyrus/embryology , Dentate Gyrus/growth & development , Doublecortin Domain Proteins , Embryo, Mammalian , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred ICR , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin/genetics , Nestin/metabolism , Neural Stem Cells/physiology , Neurogenesis/genetics , Neuropeptides/genetics , Neuropeptides/metabolism , Reelin Protein
18.
Nat Commun ; 6: 6514, 2015 Mar 09.
Article in English | MEDLINE | ID: mdl-25751136

ABSTRACT

Pathological conditions such as epilepsy cause misregulation of adult neural stem/progenitor populations in the adult hippocampus in mice, and the resulting abnormal neurogenesis leads to impairment in learning and memory. However, how animals cope with abnormal neurogenesis remains unknown. Here we show that microglia in the mouse hippocampus attenuate convulsive seizure-mediated aberrant neurogenesis through the activation of Toll-like receptor 9 (TLR9), an innate immune sensor known to recognize microbial DNA and trigger inflammatory responses. We found that microglia sense self-DNA from degenerating neurons following seizure, and secrete tumour necrosis factor-α, resulting in attenuation of aberrant neurogenesis. Furthermore, TLR9 deficiency exacerbated seizure-induced cognitive decline and recurrent seizure severity. Our findings thus suggest the existence of bidirectional communication between the innate immune and nervous systems for the maintenance of adult brain integrity.


Subject(s)
Cognitive Dysfunction/immunology , Hippocampus/immunology , Microglia/immunology , Neurogenesis/immunology , Seizures/immunology , Toll-Like Receptor 9/genetics , Animals , Cognition , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Gene Expression Regulation , Hippocampus/growth & development , Hippocampus/metabolism , Hippocampus/pathology , Immunity, Innate/genetics , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Neurogenesis/genetics , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Seizures/genetics , Seizures/metabolism , Seizures/pathology , Severity of Illness Index , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 7/deficiency , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
19.
Neurosci Res ; 95: 1-11, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25659757

ABSTRACT

Adult neurogenesis persists throughout life in the dentate gyrus (DG) of the hippocampus, and its importance has been highlighted in hippocampus-dependent learning and memory. Adult neurogenesis consists of multiple processes: maintenance and neuronal differentiation of neural stem/precursor cells (NS/PCs), followed by survival and maturation of newborn neurons and their integration into existing neuronal circuitry. However, the mechanisms that govern these processes remain largely unclear. Here we show that DNA methyltransferase 1 (DNMT1), an enzyme responsible for the maintenance of DNA methylation, is highly expressed in proliferative cells in the adult DG and plays an important role in the survival of newly generated neurons. Deletion of Dnmt1 in adult NS/PCs (aNS/PCs) did not affect the proliferation and differentiation of aNS/PCs per se. However, it resulted in a decrease of newly generated mature neurons, probably due to gradual cell death after aNS/PCs differentiated into neurons in the hippocampus. Interestingly, loss of DNMT1 in post-mitotic neurons did not influence their survival. Taken together, these findings suggest that the presence of DNMT1 in aNS/PCs is crucial for the survival of newly generated neurons, but is dispensable once they accomplish neuronal differentiation in the adult hippocampus.


Subject(s)
Adult Stem Cells/enzymology , DNA (Cytosine-5-)-Methyltransferases/metabolism , Dentate Gyrus/enzymology , Neural Stem Cells/enzymology , Neurons/enzymology , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , Dentate Gyrus/cytology , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Neurons/cytology
20.
Neurogenesis (Austin) ; 1(1): e976026, 2014.
Article in English | MEDLINE | ID: mdl-27504471

ABSTRACT

Nuclear protein 95 KDa (Np95, also known as UHRF1 or ICBP90) plays an important role in maintaining DNA methylation of newly synthesized DNA strands by recruiting DNA methyltransferase 1 (DNMT1) during cell division. In addition, Np95 participates in chromatin remodeling by interacting with histone modification enzymes such as histone deacetylases. However, its expression pattern and function in the brain have not been analyzed extensively. We here investigated the expression pattern of Np95 in the mouse brain, from developmental to adult stages. In the fetal brain, Np95 is abundantly expressed at the midgestational stage, when a large number of neural stem/precursor cells (NS/PCs) exist. Interestingly, Np95 is expressed specifically in NS/PCs but not in differentiated cells such as neurons or glial cells. Furthermore, we demonstrate that Np95 is preferentially expressed in type 2a cells, which are highly proliferative NS/PCs in the dentate gyrus of the adult hippocampus. Moreover, the number of Np95-expressing cells increases in response to kainic acid administration or to voluntary running, which are known to enhance the proliferation of adult NS/PCs. These results suggest that Np95 participates in the process of proliferation and differentiation of NS/PCs, and that it should be a useful novel marker for proliferating NS/PCs, facilitating the analysis of the complex behavior of NS/PCs in the brain.

SELECTION OF CITATIONS
SEARCH DETAIL