Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
FASEB J ; 35(5): e21420, 2021 05.
Article in English | MEDLINE | ID: mdl-33774855

ABSTRACT

Although an aberrant reduction in pancreatic ß-cell mass contributes to the pathogenesis of diabetes, the mechanism underlying the regulation of ß-cell mass is poorly understood. Here, we show that diacylglycerol kinase δ (DGKδ) is a key enzyme in the regulation of ß-cell mass. DGKδ expression was detected in the nucleus of ß-cells. We developed ß-cell-specific DGKδ knockout (ßDGKδ KO) mice, which showed lower blood glucose, higher plasma insulin levels, and better glucose tolerance compared to control mice. Moreover, an increased number of small islets and Ki-67-positive islet cells, as well as elevated cyclin B1 expression in the islets, were detected in the pancreas of ßDGKδ KO mice. DGKδ knockdown in the ß-cell line MIN6 induced significant increases in bromodeoxyuridine (BrdU) incorporation and cyclin B1 expression. Finally, we confirmed that streptozotocin-induced hyperglycemia and ß-cell loss were alleviated in ßDGKδ KO mice. Thus, suppressing the expression or enzymatic activity of DGKδ that functions as a suppressor of ß-cell proliferation could be a novel therapeutic approach to increase ß-cell mass for the treatment of diabetes.


Subject(s)
Brain/enzymology , Cell Proliferation , Diabetes Mellitus, Experimental/complications , Diacylglycerol Kinase/physiology , Hyperglycemia/prevention & control , Insulin-Secreting Cells/metabolism , Animals , Hyperglycemia/etiology , Hyperglycemia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
2.
Biochimie ; 177: 13-24, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32791090

ABSTRACT

Diacylglycerol kinase (DGK) phosphorylates diacylglycerol to produce phosphatidic acid (PA). The η isozyme of DGK is abundantly expressed in C2C12 myoblasts. However, the role of DGKη in skeletal muscle cells remains unknown. In the present study, we showed that DGKη was downregulated at an early stage of myogenic differentiation. The knockdown of DGKη by siRNAs significantly inhibited C2C12 myoblast proliferation but did not inhibit differentiation. Moreover, the suppression of DGKη expression decreased the expression levels of mammalian target of rapamycin (mTOR), which is a key regulator of cell proliferation, and fatty acid synthase (FASN), which catalyzes the de novo synthesis of fatty acids for cell proliferation and is transcriptionally regulated via mTOR signaling. Furthermore, the knockdown of mTOR or raptor, which is a component of mTOR complex 1 (mTORC1), decreased the amount of FASN. These results indicate that DGKη regulates myoblast proliferation through the mTOR (mTORC1)-FASN pathway. Interestingly, the knockdown of mTOR reduced the expression levels of DGKη, implying mutual regulation between DGKη and mTOR. In DGKη-knockdown myoblasts, C30-C36-PA species, mTOR activators, were decreased, suggesting that the modulation of mTOR activity through these PA species also plays an important role in myoblast proliferation.


Subject(s)
Diacylglycerol Kinase/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Diacylglycerol Kinase/genetics , Diglycerides/metabolism , Down-Regulation , Fatty Acid Synthase, Type I/biosynthesis , Gene Knockdown Techniques , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle Development/genetics , Myoblasts/metabolism , Phosphatidic Acids/chemistry , Phosphatidic Acids/metabolism , Phosphorylation , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Regulatory-Associated Protein of mTOR/genetics , Regulatory-Associated Protein of mTOR/metabolism , Signal Transduction
3.
FEBS Lett ; 594(11): 1787-1796, 2020 06.
Article in English | MEDLINE | ID: mdl-32134507

ABSTRACT

Serotonin transporter (SERT) is involved in serotonergic system regulation and in the pathophysiology/therapeutics of serotonin-/SERT-related diseases such as obsessive-compulsive disorder, depression, autism, and schizophrenia. We recently revealed that diacylglycerol (DG) kinase (DGK) δ induces ubiquitination/degradation of SERT in a DGK activity-dependent manner through Praja-1 E3 ubiquitin-protein ligase. However, it is still unclear how Praja-1 activity is regulated by DGKδ. Here, we reveal that 1-stearoyl-2-docosahexaenoyl (18:0/22:6)-phosphatidic acid (PA) and 18:0/22:6-DG are simultaneously decreased and accumulated, respectively, in the DGKδ-knockout mouse brain, indicating that DGKδ selectively phosphorylates 18:0/22:6-DG to generate 18:0/22:6-PA. Moreover, we find that 18:0/22:6-PA selectively binds to Praja-1 and enhances its activity. These results strongly suggest that 18:0/22:6-PA generated by DGKδ activates Praja-1 to degrade SERT in the brain.


Subject(s)
Brain/metabolism , Phosphatidic Acids/chemistry , Phosphatidic Acids/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , COS Cells , Chlorocebus aethiops , Diacylglycerol Kinase/metabolism , Enzyme Activation , Male , Mice , Substrate Specificity
4.
Biochem Biophys Res Commun ; 497(4): 1031-1037, 2018 03 18.
Article in English | MEDLINE | ID: mdl-29486157

ABSTRACT

We previously reported that brain-specific diacylglycerol kinase (DGK) δ-knockout (KO) mice showed obsessive-compulsive disorder (OCD)-like behaviors, which were alleviated by a serotonin (5-HT) transporter (SERT) inhibitor. However, the molecular mechanisms causing the OCD-like abnormal behaviors remain unclear. In the present study, we found that DGKδ deficiency increased SERT protein levels in the mouse cerebral cortex. Moreover, DGKδ interacted and co-localized with SERT. Furthermore, DGKδ-KO decreased tryptophan hydroxylase-2 expression and increased monoamine oxidase-A expression. Indeed, the amount of 5-HT in the cerebral cortex was significantly decreased in DGKδ-KO mice. These data strongly suggest that OCD-like behaviors in the DGKδ-KO mice are caused by comprehensive and composite serotonergic hypofunction.


Subject(s)
Brain/enzymology , Diacylglycerol Kinase/deficiency , Serotonin/metabolism , Animals , Cerebral Cortex/metabolism , Diacylglycerol Kinase/metabolism , Mice , Mice, Knockout , Obsessive-Compulsive Disorder/etiology , Serotonin Plasma Membrane Transport Proteins/metabolism
5.
Brain Res ; 1648(Pt A): 193-201, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27423518

ABSTRACT

Diacylglycerol kinase (DGK) is a lipid-metabolizing enzyme that phosphorylates diacylglycerol to produce phosphatidic acid. Previously, we reported that the δ isozyme of DGK was abundantly expressed in the mouse brain. However, the functions of DGKδ in the brain are still unclear. Because conventional DGKδ-knockout (KO) mice die within 24h after birth, we have generated brain-specific conditional DGKδ-KO mice to circumvent the lethality. In the novel object recognition test, the number of contacts in the DGKδ-KO mice to novel and familiar objects was greatly increased compared to the control mice, indicating that the DGKδ-KO mice showed irrational contacts with objects such as compulsive checking. In the marble burying test, which is used for analyzing obsessive-compulsive disorder (OCD)-like phenotypes, the DGKδ-KO mice buried more marbles than the control mice. Additionally, these phenotypes were significantly alleviated by the administration of an OCD remedy, fluoxetine. These results indicate that the DGKδ-KO mice showed OCD-like behaviors. Moreover, the number of long axon/neurites increased in both DGKδ-KO primary cortical neurons and DGKδ-knockdown neuroblastoma Neuro-2a cells compared to control cells. Conversely, overexpression of DGKδ decreased the number of long axon/neurites of Neuro-2a cells. Taken together, these results strongly suggest that a deficiency of DGKδ induces OCD-like behavior through enhancing axon/neurite outgrowth.


Subject(s)
Behavior, Animal/physiology , Brain/enzymology , Diacylglycerol Kinase/physiology , Obsessive-Compulsive Disorder/enzymology , Animals , Behavior, Animal/drug effects , Cell Line, Tumor , Diacylglycerol Kinase/genetics , Female , Fluoxetine/administration & dosage , Isoenzymes/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurites/enzymology , Phenotype , Recognition, Psychology/physiology , Selective Serotonin Reuptake Inhibitors/administration & dosage
6.
J Neurochem ; 138(3): 448-56, 2016 08.
Article in English | MEDLINE | ID: mdl-27167678

ABSTRACT

The η isozyme of diacylglycerol kinase (DGK) is highly expressed in the hippocampus and Purkinje cells in the central nervous system. Recently, several genome-wide association studies have implicated DGKη in the etiology of bipolar disorder (BPD). However, it is still unknown whether DGKη is indeed related to BPD. In this study, we generated DGKη-knockout (KO) mice and performed behavioral tests such as the open field test, the elevated plus maze test and tail suspension test using the KO mice to investigate the effects of DGKη deficits on psychomotor behavior. Intriguingly, DGKη-KO mice displayed an overall behavioral profile that is similar to human mania, including hyperactivity, less anxiety and less depression-like behavior. In addition, these phenotypes were significantly attenuated by the administration of a BPD (mania) remedy, namely, lithium. Moreover, DGKη-KO mice showed impairment in glycogen synthase kinase (GSK) 3ß signaling, which is closely related to BPD. These findings clearly support the linkage between BPD and DGKη that is implicated by genome-wide association studies. Moreover, this study provides DGKη-KO mice as a previously unrecognized model that reflects several features of human BPD with manic episodes and revealed an important role for DGKη in regulating behavior and mood through, at least in part, GSK3ß signaling. Several genome-wide association studies have implicated diacylglycerol kinase (DGK) η gene in the etiology of bipolar disorder (BPD). In this study, we revealed that DGKη-knockout (KO) mice displayed an overall behavioral profile that is similar to mania of BPD and is lithium (BPD (mania) remedy)-sensitive. DGKη may regulate behavior and mood through, at least in part, glycogen synthase kinase (GSK) 3ß signaling.


Subject(s)
Behavior/drug effects , Bipolar Disorder/metabolism , Diacylglycerol Kinase/metabolism , Hyperkinesis/metabolism , Lithium/pharmacology , Signal Transduction/drug effects , Animals , Anxiety/genetics , Anxiety/metabolism , Depression/genetics , Depression/metabolism , Diacylglycerol Kinase/deficiency , Genome-Wide Association Study/methods , Male , Mice , Mice, Knockout , Signal Transduction/physiology
7.
J Biol Chem ; 291(15): 8150-61, 2016 Apr 08.
Article in English | MEDLINE | ID: mdl-26887948

ABSTRACT

Type II diacylglycerol kinase (DGK) isozymes (δ, η, and κ) have a pleckstrin homology domain (PH) at their N termini. Here, we investigated the lipid binding properties of the PHs of type II DGK isozymes using protein-lipid overlay and liposome binding assays. The PH of DGKη showed the most pronounced binding activity to phosphatidylinositol (PI) 4,5-bisphosphate (PI(4,5)P2) among the various glycero- and sphingolipids including PI 3,4,5-trisphosphate, PI 3,4-bisphosphate, PI 3-phosphate, PI 4-phosphate, and PI 5-phosphate. Moreover, the PI(4,5)P2binding activity of the DGKη-PH was significantly stronger than that of other type II DGK isozymes. Notably, compared with the PH of phospholipase C (PLC) δ1, which is generally utilized as a cellular PI(4,5)P2- probe, the DGKη-PH is equal to or superior than the PLCδ1-PH in terms of affinity and selectivity for PI(4,5)P2 Furthermore, in COS-7 cells, GFP-fused wild-type DGKη1 and its PH partly translocated from the cytoplasm to the plasma membrane where the PLCδ1-PH was co-localized in response to hyperosmotic stress in an inositol 5-phosphatase-sensitive manner, whereas a PH deletion mutant did not. Moreover, K74A and R85A mutants of DGKη-PH, which lack the conserved basic amino acids thought to ligate PI(4,5)P2, were indeed unable to bind to PI(4,5)P2and co-localize with the PLCδ1-PH even in osmotically shocked cells. Overexpression of wild-type DGKη1 enhanced EGF-dependent phosphorylation of ERK, whereas either K74A or R85A mutant did not. Taken together, these results indicate that the DGKη-PH preferentially interacts with PI(4,5)P2and has crucial roles in regulating the subcellular localization and physiological function of DGKη. Moreover, the DGKη-PH could serve as an excellent cellular sensor for PI(4,5)P2.


Subject(s)
Diacylglycerol Kinase/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Diacylglycerol Kinase/analysis , Humans , MAP Kinase Signaling System , Molecular Sequence Data , Osmotic Pressure , Protein Binding , Protein Structure, Tertiary , Sequence Alignment
8.
BMC Dev Biol ; 15: 6, 2015 Jan 23.
Article in English | MEDLINE | ID: mdl-25613821

ABSTRACT

BACKGROUND: We have revealed that the type II diacylglycerol kinases (DGKs) δ, η and κ were expressed in the testis and ovary. However, these enzymes' functions in the reproductive organs remain unknown. RESULTS: In this study, we first identified the expression sites of type II DGKs in the mouse reproductive organs in detail. Reverse transcription-polymerase chain reaction and Western blotting confirmed that DGKδ2 (splicing variant 2) but not DGKδ1 (splicing variant 1) and DGKκ were expressed in the testis, ovary and uterus. DGKη1 (splicing variant 1) but not DGKη2 (splicing variant 2) was strongly detected in the ovary and uterus. Interestingly, we found that a new alternative splicing product of the DGKη gene, DGKη3, which lacks exon 26 encoding 31 amino acid residues, was expressed only in the testis. Moreover, we investigated the distribution of type II DGKs in the testis, ovary and uterus through in situ hybridization. DGKδ2 was distributed in the primary spermatocytes of the testis and ovarian follicles. DGKη1 was distributed in the oviductal epithelium of the ovary and the luminal epithelium of the uterus. Intriguingly, DGKη3 was strongly expressed in the secondary spermatocytes and round spermatids of the testis. DGKκ was distributed in the primary and secondary spermatocyte of the testis. CONCLUSION: These results indicate that the expression patterns of the type II DGK isoforms δ2, η1, η3 and κ differ from each other, suggesting that these DGK isoforms play specific roles in distinct compartments and developmental stages of the reproductive organs, especially in the processes of spermatogenesis and oocyte maturation.


Subject(s)
Diacylglycerol Kinase/metabolism , Isoenzymes/metabolism , Ovary/enzymology , Uterus/enzymology , Amino Acid Sequence , Animals , Base Sequence , Diacylglycerol Kinase/genetics , Female , Isoenzymes/genetics , Mice , Molecular Sequence Data , RNA, Messenger/genetics
9.
J Histochem Cytochem ; 63(1): 57-68, 2015 01.
Article in English | MEDLINE | ID: mdl-25362140

ABSTRACT

The functions of type II diacylglycerol kinase (DGK) δ and -η in the brain are still unclear. As a first step, we investigated the spatial and temporal expression of DGKδ and -η in the brains of mice. DGKδ2, but not DGKδ1, was highly expressed in layers II-VI of the cerebral cortex; CA-CA3 regions and dentate gyrus of hippocampus; mitral cell, glomerular and granule cell layers of the olfactory bulb; and the granule cell layer in the cerebellum in 1- to 32-week-old mice. DGKδ2 was expressed just after birth, and its expression levels dramatically increased from weeks 1 to 4. A substantial amount of DGKη (η1/η2) was detected in layers II-VI of the cerebral cortex, CA1 and CA2 regions and dentate gyrus of the hippocampus, mitral cell and glomerular layers of the olfactory bulb, and Purkinje cells in the cerebellum of 1- to 32-week-old mice. DGKη2 expression reached maximum levels at P5 and decreased by 4 weeks, whereas DGKη1 increased over the same time frame. These results indicate that the expression patterns of DGK isozymes differ from each other and also from other isozymes, and this suggests that DGKδ and -η play distinct and specific roles in the brain.


Subject(s)
Brain/enzymology , Brain/growth & development , Diacylglycerol Kinase/genetics , Diacylglycerol Kinase/metabolism , Gene Expression Regulation, Developmental , Animals , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spatio-Temporal Analysis
10.
Lipids ; 49(7): 633-40, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24852321

ABSTRACT

Decreased expression of diacylglycerol kinase (DGK) δ in skeletal muscles is closely related to the pathogenesis of type 2 diabetes. However, the regulation of DGKδ expression is not well understood. In this study, we found that myristic acid (14:0) significantly increased DGKδ2 protein expression in a dose-dependent manner (EC(50) = 0.16 mM) in mouse C2C12 myotubes. In contrast, oleic [18:1(n-9)], eicosenoic [20:1(n-9)] and erucic [22:1(n-9)] acids markedly decreased DGKδ2 expression. Myristic acid slowly enhanced DGKδ2 expression at the transcription level. Therefore, DGKδ2 expression is positively regulated by the relatively short-chain saturated fatty acid myristic acid but attenuated by n-9 monounsaturated fatty acids.


Subject(s)
Diacylglycerol Kinase/genetics , Fatty Acids, Nonesterified/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Animals , Cells, Cultured , Diacylglycerol Kinase/metabolism , Mice , Muscle, Skeletal/cytology , Structure-Activity Relationship
11.
Biochim Biophys Acta ; 1823(12): 2210-6, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22974639

ABSTRACT

The type II diacylglycerol kinases (DGKs) contain several functional domains such as a pleckstrin homology (PH) domain, two C1 domains and a sterile α-motif (SAM) domain. It was previously revealed that DGKδ contributes to hyperglycemia-induced peripheral insulin resistance and thereby exacerbate the severity of type 2 diabetes. Moreover, a high extracellular concentration of glucose activated DGKδ in skeletal muscle cells, which was followed by a reduction in the intracellular diacylglycerol levels and the inactivation of protein kinase Cα, the enzyme that phosphorylates and inactivates the insulin receptor. However, the intracellular behavior of DGKδ upon high glucose stimulation remains unclear. In this study, we found that DGKδ1, but not a splice variant DGKδ2 or the other type II DGKη1/2, translocated from the cytoplasm to the plasma membrane in human embryonic kidney HEK293 and mouse myoblast C2C12 cells within 5 min in response to high glucose levels. The translocation was inhibited by phosphatidylinositol 3-kinase inhibitors, LY294002 and GDC-0941, suggesting that the event is regulated via the phosphatidylinositol 3-kinase pathway. Moreover, we revealed that the PH and C1 domains are responsible for the plasma membrane translocation and that the SAM domain negatively regulates the translocation. These results indicate that DGKδ1 is the sole type II DGK isoform that responds rapidly and dynamically to high glucose levels.


Subject(s)
Cell Membrane/drug effects , Diacylglycerol Kinase/metabolism , Glucose/pharmacology , Protein Transport/drug effects , Sweetening Agents/pharmacology , Animals , Blotting, Western , Cell Membrane/metabolism , Cells, Cultured , Cytoplasm/drug effects , Cytoplasm/metabolism , Humans , Hydrogen-Ion Concentration , Isoenzymes , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Mice , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL
...