Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
J Nat Med ; 77(3): 584-595, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37148454

ABSTRACT

Reductions in brain-derived neurotrophic factor (BDNF) expression levels have been reported in the brains of patients with neurological disorders such as Alzheimer's disease. Therefore, upregulating BDNF and preventing its decline in the diseased brain could help ameliorate neurological dysfunctions. Accordingly, we sought to discover agents that increase Bdnf expression in neurons. Here, we screened a library of 42 Kampo extracts to identify those with the ability to induce Bdnf expression in cultured cortical neurons. Among the active extracts identified in the screen, we focused on the extract based on the Kampo formula daikenchuto. The extract of daikenchuto in the library used in this study was prepared using the mixture of Zingiberis Rhizoma Processum (ZIN), Zanthoxyli Piperiti Pericarpium (ZAN), and Ginseng Radix (GIN) without Koi. In this study, we defined DKT as the mixture of ZIN, ZAN, and GIN without Koi (DKT extract means the extract prepared from the mixture of ZIN, ZAN, and GIN without Koi). DKT extract significantly increased endogenous Bdnf expression by mediated, at least in part, via Ca2+ signaling involving L-type voltage-dependent Ca2+ channels in cultured cortical neurons. Furthermore, DKT extract significantly improved the survival of cultured cortical neurons and increased neurite complexity in immature neurons. Taken together, our findings suggest that DKT extract induces Bdnf expression and has a neurotrophic effect in neurons. Because BDNF inducers are expected to have therapeutic potential for neurological disorders, re-positioning of Kampo formulations such as daikenchuto may lead to clinical application in diseases associated with reduced BDNF in the brain.


Subject(s)
Brain-Derived Neurotrophic Factor , Medicine, Kampo , Humans , Brain-Derived Neurotrophic Factor/metabolism , Plant Extracts/pharmacology , Plant Extracts/metabolism , Neurons , Cells, Cultured
2.
Biol Pharm Bull ; 46(4): 636-639, 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-36801840

ABSTRACT

Serum response factor (SRF) is a transcription factor that plays essential roles in multiple brain functions in concert with SRF cofactors such as ternary complex factor (TCF) and megakaryoblastic leukemia (MKL)/myocardin-related transcription factor (MRTF), which comprises MKL1/MRTFA and MKL2/MRTFB. Here, we stimulated primary cultured rat cortical neurons with brain-derived neurotrophic factor (BDNF) and investigated the levels of SRF and SRF cofactor mRNA expression. We found that SRF mRNA was transiently induced by BDNF, whereas the levels of SRF cofactors were differentially regulated: mRNA expression of Elk1, a TCF family member, and MKL1/MRTFA were unchanged, while in contrast, mRNA expression of MKL2/MRTFB was transiently decreased. Inhibitor experiments revealed that BDNF-mediated alteration in mRNA levels detected in this study was mainly due to the extracellular signal-regulated protein kinase (ERK)/mitogen-activated protein kinase (MAPK) pathway. Collectively, BDNF mediates the reciprocal regulation of SRF and MKL2/MRTFB at the mRNA expression level through ERK/MAPK, which may fine-tune the transcription of SRF target genes in cortical neurons. Accumulating evidence regarding the alteration of SRF and SRF cofactor levels detected in several neurological disorders suggests that the findings of this study might also provide novel insights into valuable therapeutic strategies for the treatment of brain diseases.


Subject(s)
Brain-Derived Neurotrophic Factor , Serum Response Factor , Rats , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Serum Response Factor/genetics , Serum Response Factor/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Regulation , Neurons/metabolism
3.
J Neurochem ; 159(4): 762-777, 2021 11.
Article in English | MEDLINE | ID: mdl-32639614

ABSTRACT

Megakaryoblastic leukemia 2 (MKL2)/myocardin-related transcription factor-B (MRTFB), a serum response factor (SRF) coactivator, is an important regulator of gene expression and neuronal morphology. Here, we show that different mouse MRTFB splice isoforms, including a novel fourth MRTFB isoform named spliced neuronal long isoform of SRF transcriptional coactivator (SOLOIST)/MRTFB isoform 4 (MRTFB i4), play distinct roles in this process. SOLOIST/MRTFB i4 has a short exon that encodes 21 amino acid residues ahead of the first RPXXXEL (RPEL) motif in MRTFB isoform 3. Quantitative PCR revealed that SOLOIST/MRTFB i4 and isoform 1 were enriched in the forebrain and neurons, and up-regulated during brain development. Conversely, isoform 3 was detected in various tissues, including both neurons and astrocytes, and was down-regulated in the developing brain. Reporter assays supported the SRF-coactivator function of SOLOIST/MRTFB i4 as well as isoform 1. Acute expression of MRTFB isoform 1, but not isoform 3 or SOLOIST/MRTFB i4, in neuronal cells within 24 hr drastically increased endogenous immediate early gene [c-fos, egr1, and activity-regulated cytoskeleton-associated protein] expression, but not endogenous actinin α1, ß-actin, gelsolin, or srf gene expression measured by qPCR. Over-expression of SOLOIST/MRTFB i4 reduced the dendritic complexity of cortical neurons, whereas over-expression of isoform 1 increased this complexity. Co-expression of isoform 1 and SOLOIST/MRTFB i4 in cortical neurons revealed that isoform 1 competitively counteracted down-regulation by SOLOIST/MRTFB i4. Our findings indicate that MRTFB isoforms have unique expression patterns and differential effects on gene expression and dendritic complexity, which contribute to shaping neuronal circuits, at least in part.


Subject(s)
Neurons/metabolism , Transcription Factors/genetics , Animals , Astrocytes/metabolism , Dendrites/ultrastructure , Down-Regulation/genetics , Female , Gene Expression , Genes, Immediate-Early , Male , Mice , Mice, Inbred C57BL , Nerve Net/ultrastructure , Neurons/ultrastructure , Pregnancy , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Tissue Distribution
4.
Biochem Biophys Res Commun ; 529(3): 615-621, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32736682

ABSTRACT

Suppressor of cancer cell invasion (SCAI) is a suppressor of myocardin-related transcription factor (MRTF)-mediated transcription and cancer cell invasion. However, roles of SCAI in the brain and neuronal cells are not fully resolved. In this study, we initially investigated the distribution of Scai mRNA in the developing rat brain and in neurons. We found that, although Scai mRNA levels decreased during brain development, it was highly expressed in several brain regions and in neurons but not astrocytes. Subsequently, in addition to Scai variant 1, we identified novel rat Scai variants 2 and 3 and characterized their functions in Neuro-2a cells. The novel Scai variants 2 and 3 contain unique exons that possess stop codons and therefore encode shorter proteins compared with the full-length Scai variant 1. SCAI variants 2 and 3 possess a nuclear localization signal, but do not have an MRTF-binding site. Immunostaining of green fluorescent protein (GFP)-tagged SCAI variants revealed a nuclear localization of variant 1, whereas localization of variants 2 and 3 was throughout the cytoplasm and nucleus, suggesting that other nuclear localization signals, which act in Neuro-2a cells, exist in SCAI. All three SCAI variants suppressed the neuron-like morphological change of Neuro-2a cells induced by a Rho effector, constitutively active mDia; however, the suppressive effects of variants 2 and 3 were weaker than that of full-length SCAI variant 1, indicating that the SCAI-mediated change toward a neuronal morphology appeared to be consistent with their nuclear localization. These findings indicate that generation of multiple SCAI splice variants fines-tune neuronal morphology.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Neurons/metabolism , RNA Splicing , Transcription Factors/genetics , Animals , Animals, Newborn , Brain/cytology , Brain/growth & development , Cell Line, Tumor , Cells, Cultured , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Male , Mice , NIH 3T3 Cells , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats, Sprague-Dawley , Transcription Factors/metabolism
5.
Biochem Biophys Res Commun ; 528(2): 322-329, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32423795

ABSTRACT

Phosphatase and actin regulator 3/nuclear scaffold-associated protein phosphatase 1-inhibiting protein (Phactr3/Scapinin) is an actin- and protein phosphatase 1 (PP1)-binding protein known to negatively regulate axon elongation. In this study, we examined the expression pattern of Phactr3/Scapinin in several tissues and investigated the effect of Phactr3/Scapinin on dendritic morphology of cortical neurons. Results showed that Phactr3/Scapinin expression was up-regulated in the developing brain and enriched in neurons and in the postsynaptic density fraction, but not in astrocytes. Overexpression of wild type or mutant Phactr3/Scapinin, which lacked actin-binding activity, resulted in increased dendritic complexity and percentage of spines with a mushroom or stubby shape, as well as a decrease in spine density. However, overexpression of mutant Phactr3/Scapinin that lacked PP1-binding activity did not. Taken together, these findings suggest that Phactr3/Scapinin expression is neuronal and might contribute to synaptic formation via distinct actin- and PP1-binding domains involved in dendritic and axonal morphology, respectively.


Subject(s)
Dendrites/metabolism , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Nuclear Matrix-Associated Proteins/chemistry , Nuclear Matrix-Associated Proteins/metabolism , Nuclear Matrix/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Phosphatase 1/metabolism , Animals , Cerebral Cortex/cytology , Female , Male , Mice, Inbred C57BL , Mutation/genetics , Protein Binding , Protein Domains , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Subcellular Fractions/metabolism , Up-Regulation/genetics
6.
Sci Rep ; 9(1): 11833, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31413298

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is a key player in synaptic plasticity, and consequently, learning and memory. Because of its fundamental role in numerous neurological functions in the central nervous system, BDNF has utility as a biomarker and drug target for neurodegenerative and neuropsychiatric disorders. Here, we generated a screening assay to mine inducers of Bdnf transcription in neuronal cells, using primary cultures of cortical cells prepared from a transgenic mouse strain, specifically, Bdnf-Luciferase transgenic (Bdnf-Luc) mice. We identified several active extracts from a library consisting of 120 herbal extracts. In particular, we focused on an active extract prepared from Ginseng Radix (GIN), and found that GIN activated endogenous Bdnf expression via cAMP-response element-binding protein-dependent transcription. Taken together, our current screening assay can be used for validating herbal extracts, food-derived agents, and chemical compounds for their ability to induce Bdnf expression in neurons. This method will be beneficial for screening of candidate drugs for ameliorating symptoms of neurological diseases associated with reduced Bdnf expression in the brain, as well as candidate inhibitors of aging-related cognitive decline.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Cerebral Cortex/cytology , Luciferases/metabolism , Mass Screening , Neurons/metabolism , Transcription, Genetic , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Dopamine/metabolism , Ginsenosides/pharmacology , Mice, Transgenic , Neurons/drug effects , Plant Extracts/pharmacology , Rats, Sprague-Dawley , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction , Transcription, Genetic/drug effects
7.
Eur J Pharmacol ; 851: 69-79, 2019 May 15.
Article in English | MEDLINE | ID: mdl-30753865

ABSTRACT

Brain-derived neurotrophic factor (BDNF), an essential factor for maintaining brain functions, has been reported to be reduced in various neurological diseases, including Alzheimer's disease and major depression. Therefore, new drugs to increase the BDNF expression need to be developed. Since phosphatidylinositol (3,4,5)-trisphosphate, a membrane signaling molecule produced by phosphoinositide 3 (PI3)-kinase in the BDNF signaling, is a candidate target of SH2 domain-containing inositol 5' phosphatase 2 (SHIP2, a 5'-lipid phosphatase), the present study examined the effect of a SHIP2 inhibitor AS1949490 on Bdnf expression in cultured cortical neurons. BDNF increased its own mRNA levels, and AS1949490 enhanced this positive feedback regulation. The effects of BDNF in combination with AS1949490 on the Bdnf mRNA levels were blocked by inhibitors of mitogen-activated protein kinase kinase (U0126), PI3-kinase (LY294002), phospholipase Cγ (U73122), and protein kinase C (bisindolylmaleimide I), whereas the effect of BDNF alone was inhibited only by U0126. The mRNA stability assay using actinomycin D demonstrated that AS1949490 reduced degradation of the self-amplified Bdnf mRNA levels, and this effect was disappeared in the presence of bisindolylmaleimide I. These results suggest that BDNF promoted the Bdnf mRNA stabilization in a protein kinase C-dependent manner only in the presence of AS1949490, thereby enhancing Bdnf expression. Furthermore, behavioral analyses indicated that central administration of AS1949490 caused memory-improving and anti-depressant effects in passive avoidance test and forced swim test, respectively. Therefore, inhibition of SHIP2 appears to be valuable therapeutic strategy against neurological disorders associated with insufficient BDNF functions.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Cerebral Cortex/cytology , Neurons/drug effects , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/antagonists & inhibitors , Protein Kinase C/metabolism , RNA Stability/drug effects , Thiophenes/pharmacology , Amyloid beta-Peptides/toxicity , Animals , Cells, Cultured , Cytoprotection/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Memory/drug effects , Mice , Neurons/metabolism , Peptide Fragments/toxicity , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , Rats
8.
J Neurochem ; 148(2): 204-218, 2019 01.
Article in English | MEDLINE | ID: mdl-30244496

ABSTRACT

The expression of immediate early genes (IEGs) is thought to be an essential molecular basis of neuronal plasticity for higher brain function. Many IEGs contain serum response element in their transcriptional regulatory regions and their expression is controlled by serum response factor (SRF). SRF is known to play a role in concert with transcriptional cofactors. However, little is known about how SRF cofactors regulate IEG expression during the process of neuronal plasticity. We hypothesized that one of the SRF-regulated neuronal IEGs, activity-regulated cytoskeleton-associated protein (Arc; also termed Arg3.1), is regulated by an SRF coactivator, megakaryoblastic leukemia (MKL). To test this hypothesis, we initially investigated which binding site of the transcription factor or SRF cofactor contributes to brain-derived neurotrophic factor (BDNF)-induced Arc gene transcription in cultured cortical neurons using transfection and reporter assays. We found that BDNF caused robust induction of Arc gene transcription through a cAMP response element, binding site of myocyte enhancer factor 2, and binding site of SRF in an Arc enhancer, the synaptic activity-responsive element (SARE). Regardless of the requirement for the SRF-binding site, the binding site of a ternary complex factor, another SRF cofactor, did not affect BDNF-mediated Arc gene transcription. In contrast, chromatin immunoprecipitation revealed occupation of MKL at the SARE. Furthermore, knockdown of MKL2, but not MKL1, significantly decreased BDNF-mediated activation of the SARE. Taken together, these findings suggest a novel mechanism by which MKL2 controls the Arc SARE in response to BDNF stimulation.


Subject(s)
Cytoskeletal Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Neurons/physiology , Transcription Factors/metabolism , Transcriptional Activation/physiology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Cytoskeletal Proteins/genetics , Female , Nerve Tissue Proteins/genetics , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Serum Response Factor/genetics , Serum Response Factor/metabolism , Transcriptional Activation/drug effects
9.
Cell Struct Funct ; 42(2): 141-148, 2017 Oct 28.
Article in English | MEDLINE | ID: mdl-28943602

ABSTRACT

Deltamethrin (DM), a type II pyrethroid, robustly increases brain-derived neurotrophic factor (Bdnf) expression and has a neurotrophic effect in primary cultures of rat cortical neurons. In this study, we investigated the effect of DM on neurite morphology in cultured rat cortical neurons. DM significantly increased neurite outgrowth, but this increase was abolished when the BDNF scavenger tropomyosin receptor kinase B (TrkB)-Fc was added 10 min before the DM treatment. In contrast, the addition of TrkB-Fc 1 h after the treatment did not affect DM-induced neurite outgrowth. Our previous research has indicated that type II, but not type I, pyrethroids have the ability to induce Bdnf mRNA expression, but neither permethrin nor cypermethrin, which are type I and type II pyrethroids, respectively, affected neurite outgrowth in the current study. These results suggest that this effect is not due to increased Bdnf expression, and the effect is unique to DM. We previously demonstrated that calcineurin plays a role in the DM-mediated induction of Bdnf expression. However, the calcineurin inhibitor FK506 did not significantly affect DM-induced neurite outgrowth. DM-induced neurite outgrowth was abolished by U0126 and rapamycin, indicating the involvement of the mitogen-activated protein kinase (MAPK) and mammalian target of rapamycin (mTOR) pathways. Taken together, these findings suggest that DM activates endogenous BDNF/TrkB-mediated MAPK and mTOR pathways, thereby increasing neurite outgrowth.Key words: BDNF, Deltamethrin, MAPK, mTOR, Neurite outgrowth.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/cytology , Insecticides/pharmacology , Neuronal Outgrowth/drug effects , Neurons/drug effects , Nitriles/pharmacology , Pyrethrins/pharmacology , Receptor, trkB/metabolism , Animals , Cells, Cultured , Neurons/cytology , Neurons/metabolism , Rats , Rats, Sprague-Dawley
10.
Biochem Biophys Res Commun ; 492(3): 474-479, 2017 10 21.
Article in English | MEDLINE | ID: mdl-28830810

ABSTRACT

CCG-1423, a chemical inhibitor of Rho signaling, blocks serum response factor (SRF)/megakaryoblastic leukemia 1 (MKL1)-mediated gene expression by inhibiting the nuclear accumulation of MKL1. Several studies have suggested that CCG-1423 interacts not only with MKL1, which has a critical role in the regulation of neuronal morphology, but also with phosphatase and actin regulator 1 (Phactr1), which is localized at synapses. However, the effect of CCG-1423 on neuronal cells, especially on neuronal morphology, remains to be determined. In this study, we focused on the effect of CCG-1423 on axonal elongation, dendritic length, dendritic complexity and dendritic spine morphology. Incubation of cortical neuron cultures with up to 10 µM CCG-1423 for 72 h did not significantly affect cell viability. CCG-1423 inhibited axonal elongation and blocked the increase of dendritic length and complexity, but did not affect dendritic spine morphology. Here, we demonstrated for the first time that CCG-1423 affects neurite elongation, except for dendritic spines, without affecting neuronal cell viability. This study provides a better understanding of the effects of CCG-1423 on neurons, which may be useful for the assessment of the potential clinical application of CCG-1423 and its derivatives.


Subject(s)
Anilides/pharmacology , Axons/drug effects , Benzamides/pharmacology , Cerebral Cortex/cytology , Dendrites/drug effects , Neurons/drug effects , Animals , Cell Survival/drug effects , Dose-Response Relationship, Drug , Female , Neurons/cytology , Pregnancy , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
11.
Sci Rep ; 7(1): 4949, 2017 07 10.
Article in English | MEDLINE | ID: mdl-28694523

ABSTRACT

Brain-derived neurotrophic factor (BDNF) plays a fundamental role in expressing various neural functions including memory consolidation. Alterations of BDNF levels in the brain are associated with neurodegenerative and neuropsychiatric disorders. Therefore, it is important to understand how levels of BDNF are controlled. Recently we generated a novel transgenic mouse strain, termed the Bdnf-Luciferase transgenic (Bdnf-Luc Tg) mouse, to monitor changes in Bdnf expression. In the present study, we detected the bioluminescence signal from living Bdnf-Luc Tg mice after intraperitoneal administration of d-luciferin. Despite high levels of Bdnf expression in the brain, it was difficult to detect a signal from the brain region, probably because of its poorly penetrable (short-wavelength) bioluminescence. However, we could detect the changes in the bioluminescence signal in the brain region using a luciferin analogue generating a near-infrared wavelength of bioluminescence. We also found a strong correlation between increases in body weight and bioluminescence signal in the abdominal region of Tg mice fed a high-fat diet. These results show that changes in Bdnf expression can be visualized using living mice, and that the Tg mouse could be a powerful tool for clarification of the role of Bdnf expression in pathophysiological and physiological conditions.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Gene Expression , Luminescent Measurements , Molecular Imaging , Animals , Body Weight , Brain-Derived Neurotrophic Factor/metabolism , Diet, High-Fat , Genes, Reporter , Luminescent Measurements/methods , Mice , Mice, Transgenic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
12.
Biochem Biophys Res Commun ; 490(3): 682-687, 2017 08 26.
Article in English | MEDLINE | ID: mdl-28634074

ABSTRACT

The activity-regulated expression of immediate early genes (IEGs) contributes to long-lasting neuronal functions underlying long-term memory. However, their response properties following neuronal activity are unique and remain poorly understood. To address this knowledge gap, here we further investigated the response properties of two representative IEGs, c-fos and brain-derived neurotrophic factor (Bdnf). Treatment of cultured cortical cells with KCl produces a depolarization process that results in the increase of intracellular calcium concentration in a KCl concentration-dependent manner. Consistent with this increase, c-fos expression was induced in a KCl concentration-dependent manner. In contrast, however, Bdnf expression was optimally activated by both 25 and 50 mM concentration of KCl. Similar results were observed when the cells were treated with okadaic acid, which inhibits protein phosphatases and elicits the hyper-phosphorylation of signaling molecules. Thus, Bdnf expression is strictly regulated by a neuronal activity threshold in an all or nothing manner, whereas c-fos expression is activated in a neuronal activity-dependent manner. Our findings also suggest that these differential responses might be due to the presence or absence of a TATA box.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Genes, Immediate-Early , Neurons/metabolism , Proto-Oncogene Proteins c-fos/genetics , Transcriptional Activation , Animals , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Memory, Long-Term , Neurons/cytology , Okadaic Acid/metabolism , Phosphorylation , Potassium Chloride/metabolism , Rats , Rats, Sprague-Dawley
13.
Diabetes Res Clin Pract ; 129: 62-72, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28511140

ABSTRACT

AIMS: Centrally administered insulin improves cognitive functions in patients with Alzheimer's disease; however, it remains unknown whether long-acting insulin analogs exert more pronounced effects than insulin. In the present study, we directly compared the effects of insulin and its analogs on neural functions in vitro and in vivo. METHODS: Cultured rat cerebral cortical neurons were treated with insulin, insulin glargine U100 (Gla), insulin detemir (Det), or insulin degludec (Deg). Moreover, these drugs were intracerebroventricularly administered to mice. Their efficacies were evaluated by biochemical and behavioral analyses. RESULTS: In cultured neurons, insulin, Gla, and Det increased phosphorylation of Akt and enhanced gene expression of brain-derived neurotrophic factor to a similar extent, although Deg was less effective. The effects of Det and Deg, but not insulin and Gla were suppressed by addition of albumin. When the drug was centrally administered, the increasing effects of insulin on the Akt phosphorylation were comparable to those of Gla but greater than those of Det in hippocampus and cerebral cortex of diabetic db/db and non-diabetic db/m+ mice. Moreover, insulin and Gla enhanced memory functions in Y-maze test and suppressed depression-like behavior in forced swim test in normal mice to a similar extent, and these effects were more potent than those of Det. CONCLUSIONS: Insulin and Gla have greater impacts on central nervous system than insulin analogs with high albumin sensitivity, such as Det and Deg. These pharmacological profiles should be taken into account for developing an insulin-based therapy to treat Alzheimer's disease.


Subject(s)
Cerebral Cortex/drug effects , Hypoglycemic Agents/pharmacology , Insulin Detemir/pharmacology , Insulin Glargine/pharmacology , Insulin, Long-Acting/pharmacology , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley
14.
Front Biosci (Landmark Ed) ; 22(7): 1052-1072, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28199192

ABSTRACT

There is no doubt that synaptic activity-regulated expression of immediate early genes (IEGs) contributes to long-lasting changes in neural functions, including learning and memory. Consequently, dysregulation of IEG expression has been involved in the conditions of neural and psychiatric disorders and cognitive dysfunction. This has mainly been demonstrated using genetically modified animal models and neuropharmacological analyses. The regulatory mechanisms of IEG expression have been investigated recently and have re-emphasized the role of IEG expression in plasticity-related processes as well as elucidating molecular mechanisms and drug targets for neurological and psychiatric disorders. This review summarizes recent studies of IEG regulation, including our findings of IEG expression regulated by excitatory and modulatory neurotransmissions. In addition, we propose possible roles for IEG regulation in neurological and psychiatric disorders and long-lasting neural functions. This review improves our understanding of the association between IEG regulation and neural function and diseases, and may promote the discovery of novel drug targets for psychiatric disorders.


Subject(s)
Genes, Immediate-Early , Synaptic Transmission/genetics , Animals , Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/physiology , Cell Nucleus/genetics , Cell Nucleus/physiology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/physiology , Dopamine/physiology , Gene Expression Regulation , Humans , Learning/physiology , Memory, Long-Term/physiology , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Neurotransmitter Agents/physiology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Reward , Schizophrenia/physiopathology , Substance-Related Disorders/genetics , Substance-Related Disorders/physiopathology , Synaptic Transmission/physiology
15.
Genes Cells ; 21(8): 921-9, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27383213

ABSTRACT

Mice lacking the gene encoding pituitary adenylate cyclase-activating polypeptide (PACAP) or its specific receptor, PAC1, show abnormal behaviors related to schizophrenia. However, the regulation of PACAP expression in neurons remains unclear. Here, we report that Pacap mRNA levels are regulated transcriptionally and post-transcriptionally by cAMP and Ca(2+) signals in cultured rat cortical cells. Pacap mRNA levels decreased proportionately with the intensity of cAMP signaling, and this decrease was accelerated by N-methyl-D-aspartate (NMDA) receptor blockade, suggesting that cAMP signaling enhances the degradation of Pacap mRNA, whereas NMDA receptor-mediated signals inhibit its degradation. However, depolarization (which produced a robust increase in Ca(2+) signals) together with cAMP signaling resulted in a synergistic induction of Pacap mRNA through calcineurin and its substrate, cAMP-response element-binding protein (CREB)-regulated transcription coactivator 1. These results strongly support the concept that while cAMP signaling can accelerate the degradation of Pacap mRNA, it can also synergistically enhance Ca(2+) signaling-induced transcriptional activation of Pacap. Taken together, our findings suggest that a balance between Ca(2+) and cAMP signals regulates PACAP levels in neurons and that a perturbation of this balance may result in psychiatric disorders, such as schizophrenia.


Subject(s)
Cyclic AMP/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Schizophrenia/genetics , Transcription Factors/genetics , Animals , Calcineurin/genetics , Calcineurin/metabolism , Calcium Signaling/genetics , Cells, Cultured , Cyclic AMP/metabolism , Gene Expression Regulation , Humans , Mice , Neurons/metabolism , Neurons/pathology , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Proteolysis , RNA, Messenger/genetics , Rats , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/biosynthesis , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Schizophrenia/metabolism , Schizophrenia/pathology , Transcription Factors/metabolism
16.
Biochem Biophys Res Commun ; 466(3): 572-7, 2015 Oct 23.
Article in English | MEDLINE | ID: mdl-26386156

ABSTRACT

How the expression of immediate early genes (IEGs) is controlled in response to neurotransmissions is unknown. Using cultured rat cortical cells, we investigated the expression of IEGs regulated by Ca(2+) and/or cAMP signals. The expression of c-fos was transiently induced by treatment of cells with high potassium (high K(+)), which evoked depolarization, or forskolin, an adenylate cyclase activator. c-fos expression was persistently and synergistically induced by simultaneous treatment with high K(+) and forskolin via cAMP-response element (CRE). Microarray analysis indicated the expression profiles of IEGs caused by depolarization in the presence or absence of forskolin. When a novel index was included to investigate the profile of IEGs, we found that high K(+)-induced expression of IEGs was stimulatory or negatively changed in the presence of forskolin, suggesting distinct convergent effects of Ca(2+) and cAMP signals on the expression of IEGs.


Subject(s)
Calcium Signaling , Cyclic AMP/metabolism , Genes, Immediate-Early , Neurons/metabolism , Animals , Cells, Cultured , Colforsin/pharmacology , Gene Expression/drug effects , Genes, Immediate-Early/drug effects , Genes, fos/drug effects , Neurons/drug effects , Potassium/pharmacology , Rats , Response Elements
17.
Yakugaku Zasshi ; 135(4): 597-617, 2015.
Article in Japanese | MEDLINE | ID: mdl-25832840

ABSTRACT

My former research focused on silk fibroin gene transcription. The in vivo transcription initiation site of the fibroin gene, which is similar to the site corresponding to the 5'-terminal of mature fibroin mRNA, was determined. By developing a cell-free transcription system prepared from silk glands, it was found that the upstream region of the fibroin gene is responsible for efficient transcription initiation, which has enhancer-like features. More recent research has switched my focus to cellular neurobiology to understand the molecular mechanisms of long-term memory at the level of gene expression in terms of cell differentiation. I first developed an experimental system to analyze promoter activity in primary cultured neuronal cells. Particularly focusing on the transcription regulation of the brain-derived neurotrophic factor (BDNF) gene (Bdnf), I found that the interaction of the cAMP response element-binding protein (CREB) with the CRE sequence is important for the activity-dependent activation of the Bdnf promoter. In addition, this activity-dependent transcriptional regulation occurs in cultured neurons stimulated with excitatory GABAergic inputs, which plays a critical role in promoting the step of neuronal differentiation. Finally, I found that stimulation of the G-protein coupled receptor (GPCR) effectively activates Bdnf promoter IV through selective activation of the calcineurin pathway, irrespective of the type of GPCR if the protein kinase A or C pathway is activated. This induction mechanism appears important to understand intracellular mechanisms evoked via simultaneous neurotransmission of excitatory and modulatory inputs into neurons of the brain.


Subject(s)
Gene Expression Regulation , Memory, Long-Term , Animals , Brain , Genomics , Humans , Promoter Regions, Genetic , Transcription, Genetic
18.
J Neurosci ; 35(14): 5606-24, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25855176

ABSTRACT

Although coordinated molecular signaling through excitatory and modulatory neurotransmissions is critical for the induction of immediate early genes (IEGs), which lead to effective changes in synaptic plasticity, the intracellular mechanisms responsible remain obscure. Here we measured the expression of IEGs and used bioluminescence imaging to visualize the expression of Bdnf when GPCRs, major neuromodulator receptors, were stimulated. Stimulation of pituitary adenylate cyclase-activating polypeptide (PACAP)-specific receptor (PAC1), a Gαs/q-protein-coupled GPCR, with PACAP selectively activated the calcineurin (CN) pathway that is controlled by calcium signals evoked via NMDAR. This signaling pathway then induced the expression of Bdnf and CN-dependent IEGs through the nuclear translocation of CREB-regulated transcriptional coactivator 1 (CRTC1). Intracerebroventricular injection of PACAP and intraperitoneal administration of MK801 in mice demonstrated that functional interactions between PAC1 and NMDAR induced the expression of Bdnf in the brain. Coactivation of NMDAR and PAC1 synergistically induced the expression of Bdnf attributable to selective activation of the CN pathway. This CN pathway-controlled expression of Bdnf was also induced by stimulating other Gαs- or Gαq-coupled GPCRs, such as dopamine D1, adrenaline ß, CRF, and neurotensin receptors, either with their cognate agonists or by direct stimulation of the protein kinase A (PKA)/PKC pathway with chemical activators. Thus, the GPCR-induced expression of IEGs in coordination with NMDAR might occur via the selective activation of the CN/CRTC1/CREB pathway under simultaneous excitatory and modulatory synaptic transmissions in neurons if either the Gαs/adenylate cyclase/PKA or Gαq/PLC/PKC-mediated pathway is activated.


Subject(s)
Calcineurin/metabolism , Calcium/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Animals , Calcineurin/genetics , Calcineurin Inhibitors/pharmacology , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Embryo, Mammalian , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Signal Transduction/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism
19.
J Biol Chem ; 290(11): 6825-36, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25623071

ABSTRACT

We examined the transcriptional regulation of the activity-regulated cytoskeleton-associated protein gene (Arc), focusing on BDNF-induced Arc expression in cultured rat cortical cells. Although the synaptic activity-responsive element (SARE), located -7 kbp upstream of the Arc transcription start site, responded to NMDA, BDNF, or FGF2, the proximal region of the promoter (Arc/-1679) was activated by BDNF or FGF2, but not by NMDA, suggesting the presence of at least two distinct Arc promoter regions, distal and proximal, that respond to extracellular stimuli. Specificity protein 4 (SP4) and early growth response 1 (EGR1) controlled Arc/-1679 transcriptional activity via the region encompassing -169 to -37 of the Arc promoter. We found that trichostatin A (TSA), a histone deacetylase (HDAC) inhibitor, significantly enhanced the inductive effects of BDNF or FGF2, but not those of NMDA on Arc expression. Inhibitors of class I/IIb HDACs, SAHA, and class I HDACs, MS-275, but not of class II HDACs, MC1568, enhanced BDNF-induced Arc expression. The enhancing effect of TSA was mediated by the region from -1027 to -1000 bp, to which serum response factor (SRF) and HDAC1 bound. The binding of HDAC1 to this region was reduced by TSA. Thus, Arc expression was suppressed by class I HDAC-mediated mechanisms via chromatin modification of the proximal promoter whereas the inhibition of HDAC allowed Arc expression to be markedly enhanced in response to BDNF or FGF2. These results contribute to our understanding of the physiological role of Arc expression in neuronal functions such as memory consolidation.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cytoskeletal Proteins/genetics , Histone Deacetylases/metabolism , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic , Transcriptional Activation , Animals , Base Sequence , Cells, Cultured , Early Growth Response Protein 1/metabolism , Fibroblast Growth Factor 2/metabolism , Molecular Sequence Data , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Response Elements , Sp4 Transcription Factor/metabolism , Zinc Fingers
20.
Sci Rep ; 4: 5224, 2014 Jun 09.
Article in English | MEDLINE | ID: mdl-24909411

ABSTRACT

The megakaryoblastic leukemia 1 (MKL1) protein functions as a transcriptional coactivator of the serum response factor. MKL1 has three RPEL motifs (RPEL1, RPEL2, and RPEL3) in its N-terminal region. MKL1 binds to monomeric G-actin through RPEL motifs, and the dissociation of MKL1 from G-actin promotes the translocation of MKL1 to the nucleus. Although structural data are available for RPEL motifs of MKL1 in complex with G-actin, the structural characteristics of RPEL motifs in the free state have been poorly defined. Here we characterized the structures of free RPEL motifs using NMR and CD spectroscopy. NMR and CD measurements showed that free RPEL motifs are largely unstructured in solution. However, NMR analysis identified transient α-helices in the regions where helices α1 and α2 are induced upon binding to G-actin. Proline mutagenesis showed that the transient α-helices are locally formed without helix-helix interactions. The helix content is higher in the order of RPEL1, RPEL2, and RPEL3. The amount of preformed structure may correlate with the binding affinity between the intrinsically disordered protein and its target molecule.


Subject(s)
Protein Structure, Secondary/physiology , Serum Response Factor/chemistry , Trans-Activators/chemistry , Actins/chemistry , Actins/metabolism , Amino Acid Motifs/physiology , Cell Nucleus/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Magnetic Resonance Spectroscopy/methods , Protein Binding/physiology , Serum Response Factor/metabolism , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...