Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
BMC Nephrol ; 25(1): 66, 2024 Feb 23.
Article En | MEDLINE | ID: mdl-38395839

BACKGROUND: IgG4-related disease (IgG4-RD) is a fibroinflammatory disease that affects multiple organs, including the pancreas, lacrimal glands, salivary glands, periaortic/retroperitoneum, and kidney. Interstitial nephritis is a typical renal disorder associated with IgG4-RD, but membranous nephropathy is also seen in some cases. CASE PRESENTATION: Herein we report on the case of a 77-year-old male patient with nephrotic syndrome and IgG4-related lung disease. His serum phospholipase A2 receptor (PLA2R) antibody was positive. His renal biopsy specimen was also positive for PLA2R. The renal biopsy specimen showed membranous nephropathy with equal IgG3 and IgG4 immunofluorescence staining and no interstitial nephritis, suggesting IgG4-RD manifesting as membranous nephropathy. CONCLUSIONS: Nephrotic syndrome caused by membranous nephropathy is sometimes associated with IgG4-RD. In such cases, even if serum PLA2R antibody is positive, it should be considered that the membranous nephropathy may be secondary to IgG4-RD.


Glomerulonephritis, Membranous , Immunoglobulin G4-Related Disease , Nephritis, Interstitial , Nephrotic Syndrome , Male , Humans , Aged , Glomerulonephritis, Membranous/complications , Glomerulonephritis, Membranous/diagnosis , Receptors, Phospholipase A2 , Immunoglobulin G4-Related Disease/complications , Immunoglobulin G4-Related Disease/diagnosis , Nephrotic Syndrome/complications , Nephritis, Interstitial/complications , Nephritis, Interstitial/diagnosis , Immunoglobulin G , Autoantibodies
2.
Ren Fail ; 44(1): 1801-1810, 2022 Dec.
Article En | MEDLINE | ID: mdl-36305202

INTRODUCTION: We examined the combined effect of erythropoietin (EPO) hyporesponsiveness and low handgrip strength (HGS) on the prognosis of patients undergoing hemodialysis (HD). METHODS: We recruited patients with chronic kidney disease (CKD) Stage 5, who were undergoing HD at our dialysis clinic between January 2015 and March 2015 (n = 182). Patients of ≥20 years of age and who had been undergoing HD for ≧3 months at enrollment were eligible for inclusion. Seven patients treated with epoetin-ß pegol were excluded. First, the erythropoietin resistance index (ERI) and HGS were measured. The patients were stratified by the ERI of 9.44 (U/kg/week/g/dL), and by the HGS of 28 kg for men and 18 kg for women. We then observed death and cardiovascular disease (CVD), composite endpoint (deaths or CVD) for a median of 2 years. RESULTS: A total of 175 patients (male, n = 122; female, n = 53; age, 34-92 years) were included in the analysis. During the observation period of 24 months, 57 events (14 deaths and 43 CVD) were observed. High ERI and low HGS were associated with a high incidence of endpoints compared to low ERI and high HGS. Among the four groups classified by ERI and HGS values, the highest risk group was the high ERI/low HGS group (HR: 4.20 95% CI 2.12-8.33). CONCLUSIONS: EPO hyporesponsiveness combined with low HGS were found to be significant predictors of a poor outcome, and the synergistic effects of the two factors had stronger predictive ability than either single factor.


Cardiovascular Diseases , Erythropoietin , Hematinics , Kidney Failure, Chronic , Humans , Female , Male , Adult , Middle Aged , Aged , Aged, 80 and over , Hematinics/pharmacology , Hematinics/therapeutic use , Hand Strength , Erythropoiesis , Prospective Studies , Renal Dialysis/adverse effects , Erythropoietin/therapeutic use , Erythropoietin/pharmacology , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/therapy , Prognosis , Cardiovascular Diseases/etiology
3.
Mol Brain ; 15(1): 23, 2022 03 12.
Article En | MEDLINE | ID: mdl-35279170

Calsyntenins (CLSTNs) are important synaptic molecules whose molecular functions are not fully understood. Although mutations in calsyntenin (CLSTN) genes have been associated with psychiatric disorders in humans, their function is still unclear. One of the reasons why the function of CLSTNs in the nervous system has not been clarified is the functional redundancy among the three paralogs. Therefore, to investigate the functions of mammalian CLSTNs, we generated triple knockout (TKO) mice lacking all CLSTN paralogs and examined their behavior. The mutant mice tended to freeze in novel environments and exhibited hypersensitivity to stress. Consistent with this, glucose levels under stress were significantly higher in the mutant mice than in the wild-type controls. In particular, phenotypes such as decreased motivation, which had not been reported in single Clstn KO mice, were newly discovered. The TKO mice generated in this study represent an important mouse model for clarifying the function of CLSTN in the future.


Interneurons , Membrane Proteins , Animals , Humans , Mammals , Membrane Proteins/genetics , Mice , Mice, Knockout , Phenotype
4.
J Neurosci ; 42(12): 2448-2473, 2022 03 23.
Article En | MEDLINE | ID: mdl-35121636

Signal-induced proliferation-associated 1 (SIPA1)-like 1 (SIPA1L1; also known as SPAR1) has been proposed to regulate synaptic functions that are important in maintaining normal neuronal activities, such as regulating spine growth and synaptic scaling, as a component of the PSD-95/NMDA-R-complex. However, its physiological role remains poorly understood. Here, we performed expression analyses using super-resolution microscopy (SRM) in mouse brain and demonstrated that SIPA1L1 is mainly localized to general submembranous regions in neurons, but surprisingly, not to PSD. Our screening for physiological interactors of SIPA1L1 in mouse brain identified spinophilin and neurabin-1, regulators of G-protein-coupled receptor (GPCR) signaling, but rejected PSD-95/NMDA-R-complex components. Furthermore, Sipa1l1-/- mice showed normal spine size distribution and NMDA-R-dependent synaptic plasticity. Nevertheless, Sipa1l1-/- mice showed aberrant responses to α2-adrenergic receptor (a spinophilin target) or adenosine A1 receptor (a neurabin-1 target) agonist stimulation, and striking behavioral anomalies, such as hyperactivity, enhanced anxiety, learning impairments, social interaction deficits, and enhanced epileptic seizure susceptibility. Male mice were used for all experiments. Our findings revealed unexpected properties of SIPA1L1, suggesting a possible association of SIPA1L1 deficiency with neuropsychiatric disorders related to dysregulated GPCR signaling, such as epilepsy, attention deficit hyperactivity disorder (ADHD), autism, or fragile X syndrome (FXS).SIGNIFICANCE STATEMENT Signal-induced proliferation-associated 1 (SIPA1)-like 1 (SIPA1L1) is thought to regulate essential synaptic functions as a component of the PSD-95/NMDA-R-complex. In our screening for physiological SIPA1L1-interactors, we identified G-protein-coupled receptor (GPCR)-signaling regulators. Moreover, SIPA1L1 knock-out (KO) mice showed striking behavioral anomalies, which may be relevant to GPCR signaling. Our findings revealed an unexpected role of SIPA1L1, which may open new avenues for research on neuropsychiatric disorders that involve dysregulated GPCR signaling. Another important aspect of this paper is that we showed effective methods for checking PSD association and identifying native protein interactors that are difficult to solubilize. These results may serve as a caution for future claims about interacting proteins and PSD proteins, which could eventually save time and resources for researchers and avoid confusion in the field.


GTPase-Activating Proteins/metabolism , N-Methylaspartate , Nerve Tissue Proteins , Animals , Disks Large Homolog 4 Protein , Male , Mice , Mice, Knockout , Microfilament Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptor, Adenosine A1 , Receptors, G-Protein-Coupled/metabolism
5.
Mol Brain ; 13(1): 146, 2020 11 10.
Article En | MEDLINE | ID: mdl-33172474

Protrudin is a protein that resides in the membrane of the endoplasmic reticulum and is highly expressed in the nervous system. Although mutations in the human protrudin gene (ZFYVE27, also known as SPG33) give rise to hereditary spastic paraplegia (HSP), the physiological role of the encoded protein has been largely unclear. We therefore generated mice deficient in protrudin and subjected them to a battery of behavioral tests designed to examine their intermediate phenotypes. The protrudin-deficient mice were found to have a reduced body size and to manifest pleiotropic behavioral abnormalities, including hyperactivity, depression-like behavior, and deficits in attention and fear-conditioning memory. They exhibited no signs of HSP, however, consistent with the notion that HSP-associated mutations of protrudin may elicit neural degeneration, not as a result of a loss of function, but rather as a result of a gain of toxic function. Overall, our results suggest that protrudin might play an indispensable role in normal neuronal development and behavior.


Attention/physiology , Behavior, Animal , Conditioning, Classical , Cues , Depression/physiopathology , Fear/physiology , Vesicular Transport Proteins/deficiency , Animals , Anxiety/complications , Anxiety/physiopathology , Depression/complications , Electrophysiological Phenomena , Gene Targeting , Hippocampus/physiopathology , Memory, Short-Term , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity , Phenotype , Reflex, Startle , Social Interaction , Vesicular Transport Proteins/metabolism
6.
Sci Rep ; 10(1): 15461, 2020 09 22.
Article En | MEDLINE | ID: mdl-32963255

Lemur tail kinase 1 (LMTK1), previously called Apoptosis-Associated Tyrosine Kinase (AATYK), remains an uncharacterized Ser/Thr protein kinase that is predominantly expressed in the brain. It is recently reported that LMTK1A, an isoform of LMTK1, binds to recycling endosomes through its palmitoylation and regulates endosomal trafficking by suppressing the activity of Rab11 small GTPase. In neurons, knockdown or knockout of LMTK1 results in longer axons, greater branching of dendrites and increased number of spines, suggesting that LMTK1 plays a role in neuronal circuit formation. However, its in vivo function remained to be investigated. Here, we examined the brain structures and behaviors of LMTK1 knockout (KO) mice. LMTK1 was expressed in most neurons throughout the brain. The overall brain structure appeared to be normal in LMTK1 KO mice, but the numbers of synapses were increased. LMTK1 KO mice had a slight impairment in memory formation and exhibited distinct psychiatric behaviors such as hyperactivity, impulsiveness and high motor coordination without social interaction deficits. Some of these abnormal behaviors represent core features of attention deficit hyperactive disorder (ADHD), suggesting the possible involvement of LMTK1 in the pathogenesis of ADHD.


Apoptosis Regulatory Proteins/physiology , Attention Deficit Disorder with Hyperactivity/pathology , Behavior, Animal , Brain/physiopathology , Impulsive Behavior , Neurons/pathology , Protein-Tyrosine Kinases/physiology , Animals , Attention Deficit Disorder with Hyperactivity/etiology , Attention Deficit Disorder with Hyperactivity/psychology , Female , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Neurons/metabolism
7.
Sci Rep ; 10(1): 14187, 2020 08 25.
Article En | MEDLINE | ID: mdl-32843708

Down syndrome is a complex genetic disorder caused by the presence of three copies of the chromosome 21 in humans. The most common models, carrying extra-copies of overlapping fragments of mouse chromosome 16 that is syntenic to human chromosome 21, are Ts2Cje, Ts1Cje and Ts1Rhr mice. In electrophysiological analyses using hippocampal slices, we found that the later phase of the depolarization during tetanic stimulation, which was regulated by GABAB receptors, was significantly smaller in Ts1Cje and Ts2Cje mice than that in WT controls but not in Ts1Rhr mice. Furthermore, isolated GABAB receptor-mediated inhibitory synaptic responses were larger in Ts1Cje mice. To our knowledge, this is the first report that directly shows the enhancement of GABAB receptor-mediated synaptic currents in Ts1Cje mice. These results suggest that GABAB receptor-mediated synaptic inhibition was enhanced in Ts1Cje and Ts2Cje mice but not in Ts1Rhr mice. The Cbr1 gene, which is present in three copies in Ts1Cje and Ts2Cje but not in Ts1Rhr, encodes carbonyl reductase that may facilitate GABAB-receptor activity through a reduction of prostaglandin E2 (PGE2). Interestingly, we found that a reduction of PGE2 and an memory impairment in Ts1Cje mice were alleviated when only Cbr1 was set back to two copies (Ts1Cje;Cbr1+/+/-). However, the GABAB receptor-dependent enhancement of synaptic inhibition in Ts1Cje was unaltered in Ts1Cje;Cbr1+/+/- mice. These results indicate that Cbr1 is one of the genes responsible for DS cognitive impairments and the gene(s) other than Cbr1, which is included in Ts1Cje but not in Ts1Rhr, is responsible for the GABAB receptor-dependent over-inhibition.


Alcohol Oxidoreductases/genetics , Down Syndrome/genetics , Down Syndrome/metabolism , Receptors, GABA-B/genetics , Spatial Memory/physiology , Alcohol Oxidoreductases/metabolism , Animals , Brain/metabolism , Brain/pathology , DNA Copy Number Variations , Disease Models, Animal , Down Syndrome/pathology , Down Syndrome/psychology , Female , Hippocampus/metabolism , Hippocampus/pathology , Inhibition, Psychological , Male , Mice , Mice, Inbred C57BL , Receptors, GABA-B/metabolism , Synapses/genetics , Synapses/metabolism , gamma-Aminobutyric Acid/genetics , gamma-Aminobutyric Acid/metabolism
8.
Sci Rep ; 9(1): 15562, 2019 10 29.
Article En | MEDLINE | ID: mdl-31664108

The long-chain fatty acid receptor GPR40 plays an important role in potentiation of glucose-induced insulin secretion (GIIS) from pancreatic ß-cells. Previous studies demonstrated that GPR40 activation enhances Ca2+ release from the endoplasmic reticulum (ER) by activating inositol 1,4,5-triphosphate (IP3) receptors. However, it remains unknown how ER Ca2+ release via the IP3 receptor is linked to GIIS potentiation. Recently, stromal interaction molecule (STIM) 1 was identified as a key regulator of store-operated Ca2+ entry (SOCE), but little is known about its contribution in GPR40 signaling. We show that GPR40-mediated potentiation of GIIS is abolished by knockdown of IP3 receptor 1 (IP3R1), STIM1 or Ca2+-channel Orai1 in insulin-secreting MIN6 cells. STIM1 and Orai1 knockdown significantly impaired SOCE and the increase of intracellular Ca2+ by the GPR40 agonist, fasiglifam. Furthermore, ß-cell-specific STIM1 knockout mice showed impaired fasiglifam-mediated GIIS potentiation not only in isolated islets but also in vivo. These results indicate that the IP3R1/STIM1/Orai1 pathway plays an important role in GPR40-mediated SOCE initiation and GIIS potentiation in pancreatic ß-cells.


Inositol 1,4,5-Trisphosphate Receptors/genetics , ORAI1 Protein/genetics , Receptors, G-Protein-Coupled/genetics , Stromal Interaction Molecule 1/genetics , Animals , Calcium/metabolism , Calcium Signaling/genetics , Endoplasmic Reticulum/genetics , Glucose/genetics , Glucose/metabolism , Humans , Insulin/biosynthesis , Insulin/genetics , Insulin-Secreting Cells/metabolism , Mice , Mice, Knockout
9.
Neuroscience ; 414: 154-167, 2019 08 21.
Article En | MEDLINE | ID: mdl-31310731

Accumulating evidence suggests that glutamatergic signaling and synaptic plasticity underlie one of a number of ways psychiatric disorders appear. The present study reveals a possible mechanism by which this occurs, through highlighting the importance of LMTK3, in the brain. Behavioral analysis of Lmtk3-KO mice revealed a number of abnormalities that have been linked to psychiatric disease such as hyper-sociability, PPI deficits and cognitive dysfunction. Treatment with clozapine suppressed these behavioral changes in Lmtk3-KO mice. As synaptic dysfunction is implicated in human psychiatric disease, we analyzed the LTP of Lmtk3-KO mice and found that induction is severely impaired. Further investigation revealed abnormalities in GluA1 trafficking after AMPA stimulation in Lmtk3-KO neurons, along with a reduction in GluA1 expression in the post-synaptic density. Therefore, we hypothesize that LMTK3 is an important factor involved in the trafficking of GluA1 during LTP, and that disruption of this pathway contributes to the appearance of behavior associated with human psychiatric disease in mice.


Behavior, Animal/physiology , Membrane Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, AMPA/metabolism , Animals , Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Clozapine/pharmacology , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Maze Learning/drug effects , Maze Learning/physiology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Prepulse Inhibition/drug effects , Prepulse Inhibition/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Transport/genetics , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Reflex, Startle/drug effects , Reflex, Startle/genetics , Social Behavior , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
10.
Biol Psychiatry ; 84(8): 591-600, 2018 10 15.
Article En | MEDLINE | ID: mdl-29945718

BACKGROUND: Ketamine, an N-methyl-D-aspartate receptor antagonist, exerts robust antidepressant effects in patients with treatment-resistant depression. The precise mechanisms underlying ketamine's antidepressant actions remain unclear, although previous research suggests that alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) activation plays a role. We investigated whether (S)-norketamine and (R)-norketamine, the two main metabolites of (R,S)-ketamine, also play a significant role in ketamine's antidepressant effects and whether the effects are mediated by AMPAR. METHODS: Cellular mechanisms of antidepressant action of norketamine enantiomers were examined in mice. RESULTS: (S)-Norketamine had more potent antidepressant effects than (R)-norketamine in inflammation and chronic social defeat stress models. Furthermore, (S)-norketamine induced more beneficial effects on decreased dendritic spine density and synaptogenesis in the prefrontal cortex and hippocampus compared with (R)-norketamine. Unexpectedly, AMPAR antagonists did not block the antidepressant effects of (S)-norketamine. The electrophysiological data showed that, although (S)-norketamine inhibited N-methyl-D-aspartate receptor-mediated synaptic currents, (S)-norketamine did not enhance AMPAR-mediated neurotransmission in hippocampal neurons. Furthermore, (S)-norketamine improved reductions in brain-derived neurotrophic factor-tropomyosin receptor kinase B signaling in the prefrontal cortex of mice susceptible to chronic social defeat stress, whereas the tropomyosin receptor kinase B antagonist and a mechanistic target of rapamycin inhibitor blocked the antidepressant effects of (S)-norketamine. In contrast to (S)-ketamine, (S)-norketamine did not cause behavioral abnormalities, such as prepulse inhibition deficits, reward effects, loss of parvalbumin immunoreactivity in the medial prefrontal cortex, or baseline gamma-band oscillation increase. CONCLUSIONS: Our data identified a novel AMPAR activation-independent mechanism underlying the antidepressant effects of (S)-norketamine. (S)-Norketamine and its prodrugs could be novel antidepressants without the detrimental side effects of (S)-ketamine.


Depression/drug therapy , Hippocampus/metabolism , Ketamine/analogs & derivatives , Animals , Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Dendritic Spines/metabolism , Ketamine/pharmacology , Mice , Mice, Inbred C57BL , Prefrontal Cortex/metabolism , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Stress, Psychological/drug therapy
11.
Sci Rep ; 7(1): 7996, 2017 08 11.
Article En | MEDLINE | ID: mdl-28801590

Neurotransmitter release is mediated by the SNARE complex, but the role of its phosphorylation has scarcely been elucidated. Although PKC activators are known to facilitate synaptic transmission, there has been a heated debate on whether PKC mediates facilitation of neurotransmitter release through phosphorylation. One of the SNARE proteins, SNAP-25, is phosphorylated at the residue serine-187 by PKC, but its physiological significance has been unclear. To examine these issues, we analyzed mutant mice lacking the phosphorylation of SNAP-25 serine-187 and found that they exhibited reduced release probability and enhanced presynaptic short-term plasticity, suggesting that not only the release process, but also the dynamics of synaptic vesicles was regulated by the phosphorylation. Furthermore, it has been known that the release probability changes with development, but the precise mechanism has been unclear, and we found that developmental changes in release probability of neurotransmitters were regulated by the phosphorylation. These results indicate that SNAP-25 phosphorylation developmentally facilitates neurotransmitter release but strongly inhibits presynaptic short-term plasticity via modification of the dynamics of synaptic vesicles in presynaptic terminals.


Neuronal Plasticity , Protein Processing, Post-Translational , Synaptosomal-Associated Protein 25/genetics , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mutation , Phosphorylation , Serine/genetics , Synaptic Potentials , Synaptosomal-Associated Protein 25/chemistry , Synaptosomal-Associated Protein 25/metabolism
12.
Neurobiol Dis ; 106: 158-170, 2017 Oct.
Article En | MEDLINE | ID: mdl-28688852

Mutations in the Cyclin-dependent kinase-like 5 (CDKL5) gene cause severe neurodevelopmental disorders accompanied by intractable epilepsies, i.e. West syndrome or atypical Rett syndrome. Here we report generation of the Cdkl5 knockout mouse and show that CDKL5 controls postsynaptic localization of GluN2B-containing N-methyl-d-aspartate (NMDA) receptors in the hippocampus and regulates seizure susceptibility. Cdkl5 -/Y mice showed normal sensitivity to kainic acid; however, they displayed significant hyperexcitability to NMDA. In concordance with this result, electrophysiological analysis in the hippocampal CA1 region disclosed an increased ratio of NMDA/α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor-mediated excitatory postsynaptic currents (EPSCs) and a significantly larger decay time constant of NMDA receptor-mediated EPSCs (NMDA-EPSCs) as well as a stronger inhibition of the NMDA-EPSCs by the GluN2B-selective antagonist ifenprodil in Cdkl5 -/Y mice. Subcellular fractionation of the hippocampus from Cdkl5 -/Y mice revealed a significant increase of GluN2B and SAP102 in the PSD (postsynaptic density)-1T fraction, without changes in the S1 (post-nuclear) fraction or mRNA transcripts, indicating an intracellular distribution shift of these proteins to the PSD. Immunoelectron microscopic analysis of the hippocampal CA1 region further confirmed postsynaptic overaccumulation of GluN2B and SAP102 in Cdkl5 -/Y mice. Furthermore, ifenprodil abrogated the NMDA-induced hyperexcitability in Cdkl5 -/Y mice, suggesting that upregulation of GluN2B accounts for the enhanced seizure susceptibility. These data indicate that CDKL5 plays an important role in controlling postsynaptic localization of the GluN2B-SAP102 complex in the hippocampus and thereby regulates seizure susceptibility, and that aberrant NMDA receptor-mediated synaptic transmission underlies the pathological mechanisms of the CDKL5 loss-of-function.


Hippocampus/metabolism , Post-Synaptic Density/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Seizures/metabolism , Animals , Disease Models, Animal , Disease Susceptibility/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Guanylate Kinases/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Kainic Acid , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , N-Methylaspartate , Piperidines/pharmacology , Post-Synaptic Density/drug effects , Post-Synaptic Density/pathology , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Seizures/pathology , Tissue Culture Techniques
13.
Eur J Neurosci ; 44(5): 2272-84, 2016 09.
Article En | MEDLINE | ID: mdl-27422015

Synaptic efficacy is determined by various factors, including the quantal size, which is dependent on the amount of neurotransmitters in synaptic vesicles at the presynaptic terminal. It is essential for stable synaptic transmission that the quantal size is kept within a constant range and that synaptic efficacy during and after repetitive synaptic activation is maintained by replenishing release sites with synaptic vesicles. However, the mechanisms for these fundamental properties have still been undetermined. We found that the active zone protein CAST (cytomatrix at the active zone structural protein) played pivotal roles in both presynaptic regulation of quantal size and recycling of endocytosed synaptic vesicles. In the CA1 region of hippocampal slices of the CAST knockout mice, miniature excitatory synaptic responses were increased in size, and synaptic depression after prolonged synaptic activation was larger, which was attributable to selective impairment of synaptic vesicle trafficking via the endosome in the presynaptic terminal likely mediated by Rab6. Therefore, CAST serves as a key molecule that regulates dynamics and neurotransmitter contents of synaptic vesicles in the excitatory presynaptic terminal in the central nervous system.


CA1 Region, Hippocampal/metabolism , Cytoskeletal Proteins/metabolism , Endocytosis , Synaptic Vesicles/metabolism , Animals , CA1 Region, Hippocampal/cytology , Cytoskeletal Proteins/genetics , Excitatory Postsynaptic Potentials , Exocytosis , Mice , Mice, Inbred C57BL , Miniature Postsynaptic Potentials , Presynaptic Terminals/metabolism , rab GTP-Binding Proteins/metabolism
14.
Case Rep Nephrol Urol ; 4(2): 88-94, 2014 May.
Article En | MEDLINE | ID: mdl-24926309

Juxtaglomerular cell tumor is a rare renal neoplasm. Secondary hypertension with juxtaglomerular cell tumor can be seen in females in their 20s and 30s. We present a case of juxtaglomerular cell tumor during pregnancy. A 32-year-old female was hospitalized for refractory hypertension and nephrotic syndrome in the 23rd gestational week. One year before admission, she had been diagnosed with hypertension; plasma renin activity at that time had been 2.3 ng/ml/h. Her blood pressure was uncontrolled during pregnancy, and proteinuria was detected in the 12th gestational week despite the administration of antihypertensive medications. Laboratory data showed proteinuria, hypokalemia, and hypoalbuminemia. In the 25th gestational week, she underwent surgical termination of the pregnancy because of congestive heart failure and acute renal injury. After the termination of the pregnancy and the delivery of a viable fetus, her hypertension and nephrotic syndrome were found to persist with a high plasma renin activity (13 ng/ml/h). Ultrasonography showed a 5.5-cm left renal cystic mass with a partially solid component at the lower renal pole. The left kidney with the renal mass was excised by laparoscopic nephrectomy. Plasma renin activity normalized the next day, with a decrease in blood pressure to 120-130/80-90 mm Hg; however, proteinuria remained at ≥3.5 g/day. On the basis of histopathological findings, the patient was diagnosed with a juxtaglomerular cell tumor and focal segmental glomerulosclerosis. Juxtaglomerular cell tumor is a rare renin-secreting tumor associated with refractory hypertension in young females and is a possible cause of hypertension during pregnancy.

15.
J Neurosci ; 34(17): 5927-37, 2014 Apr 23.
Article En | MEDLINE | ID: mdl-24760852

LMTK3 belongs to the LMTK family of protein kinases that are predominantly expressed in the brain. Physiological functions of LMTK3 and other members of the LMTK family in the CNS remain unknown. In this study, we performed a battery of behavioral analyses using Lmtk3(-/-) mice and showed that these mice exhibit abnormal behaviors, including pronounced locomotor hyperactivity, reduced anxiety behavior, and decreased depression-like behavior. Concurrently, the dopamine metabolite levels and dopamine turnover rate are increased in the striata of Lmtk3(-/-) mice compared with wild-type controls. In addition, using cultured primary neurons from Lmtk3(-/-) mice, we found that LMTK3 is involved in the endocytic trafficking of N-methyl-d-aspartate receptors, a type of ionotropic glutamate receptor. Altered membrane traffic of the receptor in Lmtk3(-/-) neurons may underlie behavioral abnormalities in the mutant animals. Together, our data suggest that LMTK3 plays an important role in regulating locomotor behavior in mice.


Behavior, Animal/physiology , Endocytosis/genetics , Hyperkinesis/genetics , Membrane Proteins/genetics , Motor Activity/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Anxiety/genetics , Anxiety/metabolism , Cells, Cultured , Corpus Striatum/metabolism , Depression/genetics , Depression/metabolism , Dopamine/metabolism , Hyperkinesis/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
16.
Mol Brain ; 5: 8, 2012 Feb 08.
Article En | MEDLINE | ID: mdl-22316320

BACKGROUND: Retinoid signaling pathways mediated by retinoic acid receptor (RAR)/retinoid × receptor (RXR)-mediated transcription play critical roles in hippocampal synaptic plasticity. Furthermore, recent studies have shown that treatment with retinoic acid alleviates age-related deficits in hippocampal long-term potentiation (LTP) and memory performance and, furthermore, memory deficits in a transgenic mouse model of Alzheimer's disease. However, the roles of the RAR/RXR signaling pathway in learning and memory at the behavioral level have still not been well characterized in the adult brain. We here show essential roles for RAR/RXR in hippocampus-dependent learning and memory. In the current study, we generated transgenic mice in which the expression of dominant-negative RAR (dnRAR) could be induced in the mature brain using a tetracycline-dependent transcription factor and examined the effects of RAR/RXR loss. RESULTS: The expression of dnRAR in the forebrain down-regulated the expression of RARß, a target gene of RAR/RXR, indicating that dnRAR mice exhibit dysfunction of the RAR/RXR signaling pathway. Similar with previous findings, dnRAR mice displayed impaired LTP and AMPA-mediated synaptic transmission in the hippocampus. More importantly, these mutant mice displayed impaired hippocampus-dependent social recognition and spatial memory. However, these deficits of LTP and memory performance were rescued by stronger conditioning stimulation and spaced training, respectively. Finally, we found that pharmacological blockade of RARα in the hippocampus impairs social recognition memory. CONCLUSIONS: From these observations, we concluded that the RAR/RXR signaling pathway greatly contributes to learning and memory, and LTP in the hippocampus in the adult brain.


Hippocampus/physiopathology , Memory/physiology , Neuronal Plasticity/physiology , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors/metabolism , Signal Transduction , Synapses/physiology , Animals , Anxiety/physiopathology , Down-Regulation , Doxycycline/pharmacology , Genes, Dominant/genetics , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Memory/drug effects , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neuronal Plasticity/drug effects , Signal Transduction/drug effects , Social Behavior , Synapses/drug effects , Synaptic Transmission/drug effects
17.
J Neurosci ; 29(23): 7607-18, 2009 Jun 10.
Article En | MEDLINE | ID: mdl-19515929

Ca2+/calmodulin-dependent protein kinase IIalpha (CaMKIIalpha) is an essential mediator of activity-dependent synaptic plasticity that possesses multiple protein functions. So far, the autophosphorylation site-mutant mice targeted at T286 and at T305/306 have demonstrated the importance of the autonomous activity and Ca2+/calmodulin-binding capacity of CaMKIIalpha, respectively, in the induction of long-term potentiation (LTP) and hippocampus-dependent learning. However, kinase activity of CaMKIIalpha, the most essential enzymatic function, has not been genetically dissected yet. Here, we generated a novel CaMKIIalpha knock-in mouse that completely lacks its kinase activity by introducing K42R mutation and examined the effects on hippocampal synaptic plasticity and behavioral learning. In homozygous CaMKIIalpha (K42R) mice, kinase activity was reduced to the same level as in CaMKIIalpha-null mice, whereas CaMKII protein expression was well preserved. Tetanic stimulation failed to induce not only LTP but also sustained dendritic spine enlargement, a structural basis for LTP, at the Schaffer collateral-CA1 synapse, whereas activity-dependent postsynaptic translocation of CaMKIIalpha was preserved. In addition, CaMKIIalpha (K42R) mice showed a severe impairment in inhibitory avoidance learning, a form of memory that is dependent on the hippocampus. These results demonstrate that kinase activity of CaMKIIalpha is a common critical gate controlling structural, functional, and behavioral expression of synaptic memory.


Avoidance Learning/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dendritic Spines/enzymology , Hippocampus/enzymology , Long-Term Potentiation/physiology , Neurons/enzymology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cells, Cultured , Dendritic Spines/physiology , Dendritic Spines/ultrastructure , Excitatory Postsynaptic Potentials/physiology , Female , Gene Knock-In Techniques , Hippocampus/physiology , In Vitro Techniques , Male , Mice , Mice, Knockout , Mice, Transgenic , Mutation, Missense , Neuronal Plasticity/physiology , Neurons/physiology , Synapses/enzymology , Synapses/physiology
18.
Brain Dev ; 30(5): 313-20, 2008 May.
Article En | MEDLINE | ID: mdl-18358657

Synaptic release of the excitatory amino acid glutamate is considered as an important mechanism in the pathogenesis of ischemic brain damage in neonates. Synaptotagmin I is one of exocytosis-related proteins at nerve terminals and considered to accelerate the exocytosis of synaptic vesicles by promoting fusion between the vesicles and plasma membrane. To test the possibility that antisense in vivo knockdown of synaptotagmin I modulates the exocytotic release of glutamate, thus suppressing the excitotoxic intracellular processes leading to neuronal death following ischemia in the neonatal brain, we injected antisense oligodeoxynucleotides (ODNs) targeting synaptotagmin I (0.3 (AS), 0.15 (0.5 AS), or 0.03 microg (0.1 AS), or vehicle) into the lateral ventricles of 7-day-old rats by using a hemagglutinating virus of Japan (HVJ)-liposome mediated gene transfer technique. At 10 days of age, these rats were subjected to an electrical coagulation of the right external and internal carotid arteries, then the insertion of a solid nylon thread into the right common carotid artery toward the ascending aorta up to 10-12 mm from the upper edge of the sternocleidomastoid muscle. Cerebral ischemia was induced by clamping the left external and internal carotid arteries with a clip, and ended by removing the clip 2h later. Twenty-four hours after the end of ischemia, the extent of ischemic brain damage was neuropathologically and quantitatively evaluated in the neocortex and striatum. While the relative volume of damage in the cerebral cortex and striatum of the vehicle group was extended to 40% and 13.7%, respectively, that in the AS group was significantly reduced to 4.8% and 0.6%. In the 0.5 AS group, the relative volume of ischemic damage in the cerebral cortex and striatum was reduced to 20.5% and 15.4%, respectively, and the difference between the 0.5 AS group and vehicle group was statistically significant in the neocortex, but not in the striatum. These results indicated that antisense in vivo knockdown of synaptotagmin I successfully attenuated ischemic brain damage in neonatal rats and that the effect was dose-dependent. It was also suggested that this treatment was more effective in the neocortex than in the striatum in neonatal rats.


Brain Injuries/therapy , Gene Transfer Techniques , Oligodeoxyribonucleotides, Antisense/therapeutic use , Sendai virus/physiology , Synaptotagmin I/metabolism , Animals , Animals, Newborn , Brain Injuries/pathology , Brain Ischemia/complications , Corpus Striatum/drug effects , Corpus Striatum/pathology , Corpus Striatum/virology , Dose-Response Relationship, Drug , Liposomes/therapeutic use , Neocortex/drug effects , Neocortex/pathology , Neocortex/virology , Rats , Rats, Wistar , Synaptotagmin I/genetics
19.
Mol Cell Biol ; 25(10): 4166-75, 2005 May.
Article En | MEDLINE | ID: mdl-15870286

Neuronal leucine-rich repeat proteins (NLRRs) are type I transmembrane proteins and expressed in neuronal tissues, but their function remains unknown. Here, we describe the identification and characterization of a new member of the NLRR family, NLRR4, and its potential role in long-lasting memory. We generated NLRR4-deficient (NLRR4(-/-)) mice and found that they showed impaired memory retention. In hippocampus-dependent learning tasks, NLRR4(-/-) mice were able to learn and maintain the memories for one day but unable to retain the memories for four days after learning. In contrast, in a hippocampus-independent task, NLRR4(-/-) mice were able to retain the memory normally for at least seven days. These results suggest that NLRR4 plays a key role in hippocampus-dependent long-lasting memory.


Hippocampus/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Memory/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Brain/physiopathology , Cloning, Molecular , Cues , Fear/physiology , Gene Deletion , Hippocampus/metabolism , Hippocampus/physiopathology , Long-Term Potentiation/physiology , Maze Learning/physiology , Membrane Proteins/chemistry , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/deficiency , RNA, Messenger/genetics , RNA, Messenger/metabolism , Synaptic Transmission/physiology
20.
Neurosci Res ; 45(3): 285-96, 2003 Mar.
Article En | MEDLINE | ID: mdl-12631464

Neurotransmitter release during and after ischemic event is thought to be involved in excitotoxicity as a pathogenesis for the ischemic brain damage, which is mediated by excessive activation of glutamate receptors and attendant calcium overload. To ascertain the role of transmitter release from nerve terminals in promoting the ischemic neurodegeneration, we delivered antisense oligodeoxynucleotides (ODNs) to synaptotagmin I or synapsin I into the rat brain by using HVJ-liposome gene transfer technique. The antisense ODNs were injected into the lateralventricle in rats 4 days prior to transient forebrain ischemia of 20 min. With a single antisense treatment, long-lasting downregulation of the transmitter release relating protein levels at overall synaptic terminals was achieved. The antisense in vivo knockdown of synaptotagmin I prevented almost completely the ischemic damage of hippocampal CA1 neurons, while the in vivo knockdown of synapsin I markedly promoted the ischemic damage of CA1 pyramidal neurons and extended the injury to relatively resistant CA2/CA3 region. The modulation of ischemic hippocampal damage by the in vivo knockdown of synaptotagmin I or synapsin I suggests that transmitter release from terminals plays an important role in the evolution of ischemic brain damage and therefore the transmitter release strategy by the use of antisense ODNs-HVJ-liposome complex is reliable for neuroprotective therapies.


Brain Ischemia/prevention & control , Calcium-Binding Proteins , Gene Transfer Techniques , Hippocampus/metabolism , Membrane Glycoproteins/deficiency , Nerve Tissue Proteins/deficiency , Oligonucleotides, Antisense/administration & dosage , Synapsins/deficiency , Animals , Brain Ischemia/genetics , Brain Ischemia/pathology , Down-Regulation/genetics , Down-Regulation/physiology , Hippocampus/pathology , Liposomes/administration & dosage , Membrane Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Oligonucleotides, Antisense/genetics , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Rats , Synapsins/genetics , Synaptotagmin I , Synaptotagmins
...