Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Pflugers Arch ; 475(4): 489-504, 2023 04.
Article in English | MEDLINE | ID: mdl-36749388

ABSTRACT

Uric acid, the end product of purine metabolism in humans, is crucial because of its anti-oxidant activity and a causal relationship with hyperuricemia and gout. Several physiologically important urate transporters regulate this water-soluble metabolite in the human body; however, the existence of latent transporters has been suggested in the literature. We focused on the Escherichia coli urate transporter YgfU, a nucleobase-ascorbate transporter (NAT) family member, to address this issue. Only SLC23A proteins are members of the NAT family in humans. Based on the amino acid sequence similarity to YgfU, we hypothesized that SLC23A1, also known as sodium-dependent vitamin C transporter 1 (SVCT1), might be a urate transporter. First, we identified human SVCT1 and mouse Svct1 as sodium-dependent low-affinity/high-capacity urate transporters using mammalian cell-based transport assays. Next, using the CRISPR-Cas9 system followed by the crossing of mice, we generated Svct1 knockout mice lacking both urate transporter 1 and uricase. In the hyperuricemic mice model, serum urate levels were lower than controls, suggesting that Svct1 disruption could reduce serum urate. Given that Svct1 physiologically functions as a renal vitamin C re-absorber, it could also be involved in urate re-uptake from urine, though additional studies are required to obtain deeper insights into the underlying mechanisms. Our findings regarding the dual-substrate specificity of SVCT1 expand the understanding of urate handling systems and functional evolutionary changes in NAT family proteins.


Subject(s)
Organic Anion Transporters , Uric Acid , Animals , Humans , Mice , Amino Acid Sequence , Ascorbic Acid/metabolism , Biological Transport , Mammals/metabolism , Organic Anion Transporters/metabolism , Sodium-Coupled Vitamin C Transporters/genetics , Sodium-Coupled Vitamin C Transporters/metabolism , Uric Acid/metabolism
2.
Nucleosides Nucleotides Nucleic Acids ; 41(12): 1279-1286, 2022.
Article in English | MEDLINE | ID: mdl-35094660

ABSTRACT

LLC-PK1 renal cells show Na+-dependent and Na+-independent hypoxanthine uptake. While the latter is inhibited by adenine, neither are inhibited by xanthine. In rats, intestinal Na+-dependent hypoxanthine transporter Slc23a4 is not expressed in the kidney, and its action is inhibited by xanthine. This study aimed to clone Slc23a4-paralog SLC23A3 from the human kidney and investigate its hypoxanthine transport activity. We observed Na+-dependent 10 nM [3H]-hypoxanthine uptake in SLC23A3 RNA-injected Xenopus oocytes. Moreover, 100 µM xanthine did not inhibit Na+-independent 300 nM [3H]-hypoxanthine uptake, whereas 100 µM adenine did. These results confirm that SLC23A3 is a hypoxanthine transporter in the human kidney.


Subject(s)
Kidney , Membrane Transport Proteins , Humans , Rats , Animals , Hypoxanthine/metabolism , Kidney/metabolism , Membrane Transport Proteins/metabolism , Biological Transport , Sodium/metabolism , Sodium/pharmacology , Adenine/metabolism , Xanthines/metabolism
3.
J Am Soc Nephrol ; 33(2): 326-341, 2022 02.
Article in English | MEDLINE | ID: mdl-34799437

ABSTRACT

BACKGROUND: Hereditary renal hypouricemia type 1 (RHUC1) is caused by URAT1/SLC22A12 dysfunction, resulting in urolithiasis and exercise-induced AKI (EIAKI). However, because there is no useful experimental RHUC1 animal model, the precise pathophysiologic mechanisms underlying EIAKI have yet to be elucidated. We established a high HPRT activity Urat1-Uox double knockout (DKO) mouse as a novel RHUC1 animal model for investigating the cause of EIAKI and the potential therapeutic effect of xanthine oxidoreductase inhibitors (XOIs). METHODS: The novel Urat1-Uox DKO mice were used in a forced swimming test as loading exercise to explore the onset mechanism of EIAKI and evaluate related purine metabolism and renal injury parameters. RESULTS: Urat1-Uox DKO mice had uricosuric effects and elevated levels of plasma creatinine and BUN as renal injury markers, and decreased creatinine clearance observed in a forced swimming test. In addition, Urat1-Uox DKO mice had increased NLRP3 inflammasome activity and downregulated levels of Na+-K+-ATPase protein in the kidney, as Western blot analysis showed. Finally, we demonstrated that topiroxostat and allopurinol, XOIs, improved renal injury and functional parameters of EIAKI. CONCLUSIONS: Urat1-Uox DKO mice are a useful experimental animal model for human RHUC1. The pathogenic mechanism of EIAKI was found to be due to increased levels of IL-1ß via NLRP3 inflammasome signaling and Na+-K+-ATPase dysfunction associated with excessive urinary urate excretion. In addition, XOIs appear to be a promising therapeutic agent for the treatment of EIAKI.


Subject(s)
Acute Kidney Injury/drug therapy , Hypoxanthine Phosphoribosyltransferase/metabolism , Organic Anion Transporters/deficiency , Urate Oxidase/deficiency , Xanthine Dehydrogenase/antagonists & inhibitors , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Allopurinol/pharmacology , Animals , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Hypoxanthine Phosphoribosyltransferase/genetics , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nitriles/pharmacology , Organic Anion Transporters/genetics , Physical Exertion , Pyridines/pharmacology , Renal Tubular Transport, Inborn Errors/drug therapy , Renal Tubular Transport, Inborn Errors/etiology , Renal Tubular Transport, Inborn Errors/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Urate Oxidase/genetics , Urinary Calculi/drug therapy , Urinary Calculi/etiology , Urinary Calculi/metabolism
4.
Int J Mol Sci ; 21(18)2020 Sep 04.
Article in English | MEDLINE | ID: mdl-32899645

ABSTRACT

The volume, composition, and movement of the cerebrospinal fluid (CSF) are important for brain physiology, pathology, and diagnostics. Nevertheless, few studies have focused on the main structure that produces CSF, the choroid plexus (CP). Due to the presence of monocarboxylate transporters (MCTs) in the CP, changes in blood and brain lactate levels are reflected in the CSF. A lactate receptor, the hydroxycarboxylic acid receptor 1 (HCA1), is present in the brain, but whether it is located in the CP or in other periventricular structures has not been studied. Here, we investigated the distribution of HCA1 in the cerebral ventricular system using monomeric red fluorescent protein (mRFP)-HCA1 reporter mice. The reporter signal was only detected in the dorsal part of the third ventricle, where strong mRFP-HCA1 labeling was present in cells of the CP, the tela choroidea, and the neuroepithelial ventricular lining. Co-labeling experiments identified these cells as fibroblasts (in the CP, the tela choroidea, and the ventricle lining) and ependymal cells (in the tela choroidea and the ventricle lining). Our data suggest that the HCA1-containing fibroblasts and ependymal cells have the ability to respond to alterations in CSF lactate in body-brain signaling, but also as a sign of neuropathology (e.g., stroke and Alzheimer's disease biomarker).


Subject(s)
Choroid Plexus/metabolism , Receptors, G-Protein-Coupled/metabolism , Third Ventricle/metabolism , Animals , Brain/metabolism , Cerebral Ventricles/metabolism , Cerebral Ventricles/physiology , Cerebrospinal Fluid/metabolism , Choroid Plexus/physiology , Fibroblasts/metabolism , Humans , Lactic Acid/metabolism , Mice , Mice, Inbred C57BL , Third Ventricle/physiology
5.
Antioxidants (Basel) ; 9(5)2020 May 06.
Article in English | MEDLINE | ID: mdl-32384764

ABSTRACT

The oxidant/antioxidant imbalance plays a pivotal role in the lung. Uric acid (UA), an endogenous antioxidant, is highly present in lung tissue, however, its impact on lung function under pathophysiological conditions remains unknown. In this work, pharmacological and genetic inhibition of UA metabolism in experimental mouse models of acute and chronic obstructive pulmonary disease (COPD) revealed that increased plasma UA levels improved emphysematous phenotype and lung dysfunction in accordance with reduced oxidative stress specifically in female but not in male mice, despite no impact of plasma UA induction on the pulmonary phenotypes in nondiseased mice. In vitro experiments determined that UA significantly suppressed hydrogen peroxide (H2O2)-induced oxidative stress in female donor-derived primary human bronchial epithelial (NHBE) cells in the absence of estrogen, implying that the benefit of UA is limited to the female airway in postmenopausal conditions. Consistently, our clinical observational analyses confirmed that higher blood UA levels, as well as the SLC2A9/GLUT9 rs11722228 T/T genotype, were associated with higher lung function in elderly human females. Together, our findings provide the first unique evidence that higher blood UA is a protective factor against the pathological decline of lung function in female mice, and possibly against aging-associated physiological decline in human females.

6.
Nucleosides Nucleotides Nucleic Acids ; 39(10-12): 1465-1473, 2020.
Article in English | MEDLINE | ID: mdl-32126884

ABSTRACT

Although xanthinuria is nonfatal in human, xanthine oxidoreductase knockout (Xor-KO) mice have only a short lifespan. Hypoxanthine phosphoribosyltransferase activity (HPRT) in human and wild mice is higher than in laboratory mice. The aim of this study was to investigate the underlying mechanisms that give rise to the longer lifespan of high-HPRT/Xor-KO mice. Before Xor-KO mice die, urinary excretion of hypoxanthine increased with a corresponding decrease in excretion of xanthine. The switch of excretion from xanthine to hypoxanthine might be a cause of death for Xor-KO mice, suggesting inhibition of NAD+-dependent IMP dehydrogenase. Because hypoxanthine inhibits the synthesis of nicotinamide mononucleotide (NMN), a precursor of NAD+, the accumulation of hypoxanthine in Xor-KO mice may cause a depletion in the levels of NAD+. Moreover, urinary excretion of urate in high-HPRT/Uox-KO/Xor-KO mice means urate derived from gut microbiota is absorbed by the intestine. Likewise, over excretion of oxypurine in mice may be caused by intestinal absorption of oxypurine. For NAD+ replenishment, oral supplementation with 1% L-tryptophan, an alternative precursor of NAD+, resulted in a recovery of body weight gain in high-HPRT/Uox-KO/Xor-KO mice. In conclusion, the death of Xor-KO mice by renal failure seems to be caused by a depletion in NAD+ levels due to the intracellular accumulation of hypoxanthine. NAD+ replenishment by oral supplementation of NMN or tryptophan was complicated by the effect of gut microbiota and failed to rescue high-HPRT/Xor-KO mice. The attenuation of intestinal absorption of oxypurines seems to be necessary to avoid hypoxanthine accumulation and over excretion of oxypurine.


Subject(s)
Gene Knockout Techniques , Hypoxanthine Phosphoribosyltransferase/metabolism , Xanthine Dehydrogenase/deficiency , Xanthine Dehydrogenase/genetics , Animals , Longevity , Mice , NAD/pharmacology
7.
Br J Pharmacol ; 177(10): 2274-2285, 2020 05.
Article in English | MEDLINE | ID: mdl-31971609

ABSTRACT

BACKGROUND AND PURPOSE: Purine metabolism in mice and human differ in terms of uricase (Uox) activity as well as hypoxanthine phosphoribosyltransferase (HPRT) activity. The aim of this study was the establishment of high HPRT activity-Uox knockout (KO) mice as a novel hyperuricaemic model. Then to investigate the effects of purine-type xanthine dehydrogenase (XDH) inhibitor, allopurinol, and non-purine-type XDH inhibitor, topiroxostat, on purine metabolism. EXPERIMENTAL APPROACH: A novel hyperuricaemic mouse model was established by mating B6-ChrXCMSM mice with uricase KO mice. The pharmacological effects of allopurinol and topiroxostat were explored by evaluating urate, hypoxanthine, xanthine and creatinine in the plasma and urine of these model mice. Furthermore, we analysed the effect of both drugs on erythrocyte hypoxanthine phosphoribosyltransferase activity. KEY RESULTS: Plasma urate level and urinary urate/creatinine ratio significantly decreased after administration of allopurinol 30 mg·kg-1 or topiroxostat 1 mg·kg-1 for 7 days. The urate-lowering effect was equivalent for allopurinol and topiroxostat. However, the urinary hypoxanthine/creatinine ratio and xanthine/creatinine ratio after treatment with topiroxostat were significantly lower than for allopurinol. In addition, the urinary oxypurine/creatinine ratio was significantly lowered after treatment with topiroxostat, but allopurinol elicited no such effect. Furthermore, allopurinol inhibited mouse erythrocyte hypoxanthine phosphoribosyltransferase, while topiroxostat did not. CONCLUSIONS AND IMPLICATIONS: High hypoxanthine phosphoribosyltransferase activity- uricase KO mice were established as a novel hyperuricaemic animal model. In addition, topiroxostat, a non-purine-type xanthine dehydrogenase inhibitor, elicited a potent plasma urate-lowering effect. However, unlike allopurinol, topiroxostat did not perturb the salvage pathway, resulting in lowered total oxypurine excretion.


Subject(s)
Sexual and Gender Minorities , Urate Oxidase , Allopurinol/pharmacology , Animals , Homosexuality, Male , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Male , Mice , Mice, Knockout , Purines/pharmacology , Xanthine Dehydrogenase/genetics
8.
Shokuhin Eiseigaku Zasshi ; 59(6): 275-281, 2018.
Article in Japanese | MEDLINE | ID: mdl-30626785

ABSTRACT

The aluminium (Al) content of Japanese confectionery and foods containing flour was investigated. Some of these items were investigated in previous studies, which examined foods that made use of baking powder containing aluminium potassium sulfate (Alum). Al was detected in 41 of the 123 samples at levels ranging from 0.01 (limit of quantitation) to 0.40 mg/g. The detection rate of Al in almost all confectionery (except Japanese confectionery) was decreased as compared with previous studies. However, the detection rate of Al in Japanese confectionery and foods containing flour was high. For 4 of the 41 samples tested, consuming one serving once a week would result in an Al intake exceeding the PTWI for young children (body weight=16 kg).


Subject(s)
Alum Compounds , Aluminum/analysis , Calcium Sulfate , Flour/analysis , Food Contamination/analysis , Sodium Bicarbonate , Starch , Child , Humans
9.
Shokuhin Eiseigaku Zasshi ; 58(3): 166-171, 2017.
Article in Japanese | MEDLINE | ID: mdl-28690308

ABSTRACT

Stainless steel kitchenware and tableware on sale in Japan were investigated. Surface elemental composition ratios of 172 samples were analyzed by the fluorescence X-ray method. High levels of manganese (9.59-20.03%)were detected in 17 samples. This finding was confirmed by ICP analysis. Next, we conducted migration tests. Samples conformed to the Italian Specific Migration Limits. Moreover, lead and antimony were not detected in these samples, in accordance with the Japanese Food Sanitation Law.


Subject(s)
Cooking and Eating Utensils , Legislation, Food/standards , Manganese/analysis , Stainless Steel/chemistry , Antimony/analysis , Cadmium/analysis , Chromium/analysis , Iron/analysis , Japan , Lead/analysis , Nickel/analysis , Spectrometry, X-Ray Emission
10.
Nat Commun ; 8: 15800, 2017 06 12.
Article in English | MEDLINE | ID: mdl-28604739

ABSTRACT

Lrfn2/SALM1 is a PSD-95-interacting synapse adhesion molecule, and human LRFN2 is associated with learning disabilities. However its role in higher brain function and underlying mechanisms remain unknown. Here, we show that Lrfn2 knockout mice exhibit autism-like behavioural abnormalities, including social withdrawal, decreased vocal communications, increased stereotyped activities and prepulse inhibition deficits, together with enhanced learning and memory. In the hippocampus, the levels of synaptic PSD-95 and GluA1 are decreased. The synapses are structurally and functionally immature with spindle shaped spines, smaller postsynaptic densities, reduced AMPA/NMDA ratio, and enhanced LTP. In vitro experiments reveal that synaptic surface expression of AMPAR depends on the direct interaction between Lrfn2 and PSD-95. Furthermore, we detect functionally defective LRFN2 missense mutations in autism and schizophrenia patients. Together, these findings indicate that Lrfn2/LRFN2 serve as core components of excitatory synapse maturation and maintenance, and their dysfunction causes immature/silent synapses with pathophysiological state.


Subject(s)
Autistic Disorder/genetics , Membrane Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Neuronal Plasticity/genetics , Animals , Disks Large Homolog 4 Protein/metabolism , Hippocampus/metabolism , Humans , Memory , Mice, Knockout , Mutation, Missense , Receptors, AMPA/metabolism , Schizophrenia/genetics
11.
Fluids Barriers CNS ; 13(1): 22, 2016 Dec 12.
Article in English | MEDLINE | ID: mdl-27955673

ABSTRACT

BACKGROUND: Uric acid (UA) is known to exert neuroprotective effects in the brain. However, the mechanism of UA regulation in the brain is not well characterized. In our previous study, we described that the mouse urate transporter URAT1 is localized to the cilia and apical surface of ventricular ependymal cells. To further strengthen the hypothesis that UA is transported transcellularly at the ependymal cells, we aimed to assess the distribution of other UA transporters in the murine brain. METHODS: Immunostaining and highly-sensitive in situ hybridization was used to assess the distribution of UA transporters: GLUT9/URATv1, ABCG2, and URAT1. RESULTS: Immunostaining for GLUT9 was observed in ependymal cells, neurons, and brain capillaries. Immunostaining for ABCG2 was observed in the choroid plexus epithelium and brain capillaries, but not in ependymal cells. These results were validated by in situ hybridization. CONCLUSIONS: We propose that given their specific expression patterns in ependymal, choroid plexus epithelial, and brain capillary endothelial cells in this study, UA may be transported by these UA transporters in the murine brain. This may provide a novel strategy for targeted neuroprotection.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Brain/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Organic Anion Transporters/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Acetic Acid , Animals , Chloroform , Ependyma/metabolism , Epithelium/metabolism , Fluorescent Antibody Technique , In Situ Hybridization , Male , Methanol , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger , Tissue Fixation
12.
Nucleosides Nucleotides Nucleic Acids ; 35(10-12): 543-549, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27906636

ABSTRACT

Renal hypouricemia (RHUC) is a hereditary disease characterized by a low level of plasma urate but with normal urinary urate excretion. RHUC type 1 is caused by mutations of the urate transporter URAT1 gene (SLC22A12). However, the plasma urate levels of URAT1 knockout mice are no different from those of wild-type mice. In the present study, a double knockout mouse, in which the URAT1 and uricase (Uox) genes were deleted (Urat1-Uox-DKO), were used as an experimental animal model of RHUC type 1 to investigate RHUC and excise-induced acute kidney injury (EIAKI). Mice were given a variable content of allopurinol for one week followed by HPLC measurement of urate and creatinine concentrations in spot urine and blood from the tail. The urinary excretion of urate in Urat1-Uox-DKO mice was approximately 25 times higher than those of humans. With allopurinol, the plasma urate levels of Urat1-Uox-DKO mice were lower than those of Uox-KO mice. There were no differences in the urinary urate excretions between Urat1-Uox-DKO and Uox-KO mice administered with 9 mg allopurinol /100 g feed. In the absence of allopurinol, plasma creatinine levels of some Urat1-Uox-DKO mice were higher than those of Uox-KO mice. Consequently, hypouricemia and normouricosuria may indicate that the Urat1-Uox-DKO mouse administered with allopurinol may represent a suitable animal model of RHUC type 1. Urat1-Uox-DKO mice without allopurinol exhibited acute kidney injury, thus providing additional benefit as a potential animal model for EIAKI. Finally, our data indicate that allopurinol appears to provide prophylactic effects for EIAKI.


Subject(s)
Acute Kidney Injury/genetics , Organic Anion Transporters/genetics , Renal Tubular Transport, Inborn Errors/genetics , Urate Oxidase/genetics , Urinary Calculi/genetics , Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Allopurinol/pharmacology , Allopurinol/therapeutic use , Animals , Creatinine/blood , Disease Models, Animal , Gout Suppressants/pharmacology , Gout Suppressants/therapeutic use , Male , Mice, Knockout , Organic Anion Transporters/metabolism , Physical Conditioning, Animal , Renal Tubular Transport, Inborn Errors/drug therapy , Renal Tubular Transport, Inborn Errors/metabolism , Urate Oxidase/metabolism , Uric Acid/urine , Urinary Calculi/drug therapy , Urinary Calculi/metabolism
13.
Biol Pharm Bull ; 39(7): 1081-4, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27170514

ABSTRACT

Thirty minutes incubation at room temperature elevates the uric acid (UA) level of mouse blood in a test tube, and has previously been reported as "false in vitro elevation of the uric acid level." However the UA level of human blood does not elevate using the same incubation. We clarified the mechanism of the false in vitro UA elevation using mice with highly active hypoxanthine phosphoribosyl transferase (Hprt) of B6-ChrXC(MSM), a consomic mouse strain with the chromosome portion of Mus musculus morocinus in the Hprt gene site, or mice with a targeted deletion of the urate oxidase gene (Uox) (Uox-knockout (KO)). The plasma levels of UA, hypoxanthine, and xanthine, determined by HPLC, were compared with those of C57BL/6J laboratory mice used as controls. The uric acid level of Uox-KO mice was approximately 10 times higher than that of control, did not elevated after incubation in the test tube. With allopurinol, the hypoxanthine levels of B6-ChrXC(MSM) and Uox-KO were significantly lower than that of controls. Without allopurinol, the UA and xanthine levels of B6-ChrXC(MSM) were significantly lower than those of C57BL/6J controls. Even with allopurinol, the UA and xanthine levels were still significantly lower than that of controls. In conclusion, "false in vitro elevation of uric acid level" seems to be caused by low levels of erythrocyte HPRT activity and the low plasma uric acid level of laboratory mice.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/genetics , Urate Oxidase/genetics , Uric Acid/blood , Animals , Hypoxanthine/blood , Male , Mice, Inbred C57BL , Mice, Knockout , Xanthine/blood
14.
Food Addit Contam Part B Surveill ; 9(3): 185-90, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27092423

ABSTRACT

Aluminium (Al) levels of 90 food samples were investigated. Nineteen samples contained Al levels exceeding the tolerable weekly intake (TWI) for young children [body weight (bw): 16 kg] when consuming two servings/week. These samples were purchased multiple times at specific intervals and were evaluated for Al levels. Al was detected in 27 of the 90 samples at levels ranging from 0.01 (limit of quantitation) to 1.06 mg/g. Of these, the Al intake levels in two samples (cookie and scone mix, 1.3 and 2 mg/kg bw/week, respectively) exceeded the TWI as established by European Food Safety Authority, although the level in the scone mix was equivalent to the provisional TWI (PTWI) as established by Joint Food and Agriculture Organization of the United Nations/World Health Organization Expert Committee on Food Additives. The Al levels markedly decreased in 14 of the 19 samples with initially high Al levels. These results indicated reductions in the Al levels to below the PTWI limits in all but two previously identified food samples.


Subject(s)
Aluminum Compounds/chemistry , Aluminum/analysis , Food Additives/chemistry , Food Analysis , Food Contamination , Aluminum/toxicity , Bread/adverse effects , Bread/analysis , Bread/economics , Bread/standards , Child, Preschool , Cooking , Diet/adverse effects , Diet/ethnology , Food Additives/adverse effects , Food Additives/standards , Food Analysis/economics , Food Inspection/methods , Humans , Hydrolysis/radiation effects , Indicators and Reagents/chemistry , Internationality , Limit of Detection , Microwaves , Nitric Acid/chemistry , No-Observed-Adverse-Effect Level , Reproducibility of Results , Snacks , Spectrophotometry, Atomic , Tokyo
15.
Nat Commun ; 5: 4501, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25047565

ABSTRACT

GABAergic interneurons are highly heterogeneous, and much is unknown about the specification and functional roles of their neural circuits. Here we show that a transinteraction of Elfn1 and mGluR7 controls targeted interneuron synapse development and that loss of Elfn1 results in hyperactivity and sensory-triggered epileptic seizures in mice. Elfn1 protein increases during postnatal development and localizes to postsynaptic sites of somatostatin-containing interneurons (SOM-INs) in the hippocampal CA1 stratum oriens and dentate gyrus (DG) hilus. Elfn1 knockout (KO) mice have deficits in mGluR7 recruitment to synaptic sites on SOM-INs, and presynaptic plasticity is impaired at these synapses. In patients with epilepsy and attention deficit hyperactivity disorder (ADHD), we find damaging missense mutations of ELFN1 that are clustered in the carboxy-terminal region required for mGluR7 recruitment. These results reveal a novel mechanism for interneuron subtype-specific neural circuit establishment and define a common basis bridging neurological disorders.


Subject(s)
Epilepsy/genetics , Mutation, Missense , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Metabotropic Glutamate/metabolism , Seizures/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Animals , Attention Deficit Disorder with Hyperactivity/genetics , Autistic Disorder/genetics , Case-Control Studies , Child , Child, Preschool , Female , Humans , Interneurons/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Middle Aged , Molecular Sequence Data , Neuronal Plasticity/genetics , Polymorphism, Single Nucleotide , Rats, Sprague-Dawley , Seizures/genetics , Young Adult
16.
Article in English | MEDLINE | ID: mdl-24940669

ABSTRACT

Uric acid (UA) levels in mouse blood have been reported to range widely from 0.1 µM to 760 µM. The aim of this study was to demonstrate false in vitro and in vivo elevations of UA levels in mouse blood. Male ICR mice were anesthetized with pentobarbital (breathing mice) or sacrificed with overdose ether (non-breathing mice). Collected blood was dispensed into MiniCollect® tubes and incubated in vitro for 0 or 30 min at room temperature. After separation of plasma or serum, the levels of UA and hypoxanthine were determined using HPLC. From the non-incubated plasma of breathing mice, the true value of UA level in vivo was 13.5±1.4 µM. However, UA levels in mouse blood increased by a factor of 3.9 following incubation in vitro. This "false in vitro elevation" of UA levels in mouse blood after blood sampling was inhibited by allopurinol, a xanthine oxidase inhibitor. Xanthine oxidase was converted to UA in mouse serum from hypoxanthine which was released from blood cells during incubation. Plasma UA levels from non-breathing mice were 19 times higher than those from breathing mice. This "false in vivo elevation" of UA levels before blood sampling was inhibited by pre-treatment with phentolamine, an α-antagonist. Over-anesthesia with ether might induce α-vasoconstriction and ischemia and thus degrade intracellular ATP to UA. For the accurate measurement of UA levels in mouse blood, the false in vitro and in vivo elevations of UA level must be avoided by immediate separation of plasma after blood sampling from anesthetized breathing mice.


Subject(s)
Blood Chemical Analysis/methods , Uric Acid/blood , Allopurinol/pharmacology , Animals , Blood Specimen Collection , False Positive Reactions , Male , Mice , Mice, Inbred ICR , Phentolamine/pharmacology , Time Factors
17.
Fluids Barriers CNS ; 10(1): 31, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24156345

ABSTRACT

BACKGROUND: Elevated uric acid (UA) is commonly associated with gout and it is also a known cardiovascular disease risk factor. In contrast to such deleterious effects, UA possesses neuroprotective properties in the brain and elucidating the molecular mechanisms involved may have significant value regarding the therapeutic treatment of neurodegenerative disease. However, it is not yet fully established how UA levels are regulated in the brain. In this study, we investigated the distribution of mouse urate transporter 1 (URAT1) in the brain. URAT1 is a major reabsorptive urate transporter predominantly found in the kidney. METHODS: Immunohistochemistry of wild type and URAT1 knockout mouse brain using paraffin or frozen sections and a rabbit polyclonal anti-mouse URAT1 antibody were employed. RESULTS: Antibody specificity was confirmed by the lack of immunostaining in brain tissue from URAT1 knockout mice. URAT1 was distributed throughout the ventricular walls of the lateral ventricle, dorsal third ventricle, ventral third ventricle, aqueduct, and fourth ventricle, but not in the non-ciliated tanycytes in the lower part of the ventral third ventricle. URAT1 was localized to the apical membrane, including the cilia, of ependymal cells lining the wall of the ventricles that separates cerebrospinal fluid (CSF) and brain tissue. CONCLUSION: In this study, we report that URAT1 is expressed on cilia and the apical surface of ventricular ependymal cells. This is the first report to demonstrate expression of the urate transporter in ventricular ependymal cells and thus raises the possibility of a novel urate transport system involving CSF.

18.
Biol Pharm Bull ; 36(5): 741-7, 2013.
Article in English | MEDLINE | ID: mdl-23649333

ABSTRACT

Preventing the onset of microalbuminuria in diabetic nephropathy is a problem that needs urgent rectification. The use of a mouse model for diabetes is vital in this regard. For example, db/db mice exhibit defects in the leptin receptor Ob-Rb sub-type, while the ob/ob strain exhibits defects in the leptin ligand. These mouse strains demonstrate type 2 diabetes, either with or without microalbuminuria, respectively. The purpose of the present study was to use DNA microarray technology to screen for the gene responsible for the onset of diabetic microalbuminuria. Using Affymetrix Mouse Gene ST 1.0 arrays, microarray analysis was performed using total RNA from the kidneys of ob control, ob/ob, db/m, and db/db mice. Microarray and quantitative reverse transcription-polymerase chain reaction (RT-PCR) indicated that transcription of the macrophage migration inhibitory factor (MIF) gene was significantly enhanced in the kidneys of db/db mice. Western blotting showed that levels of MIF protein was enhanced in the kidneys of both diabetic db/db and ob/ob mice. On the other hand, elevation of urinary MIF excretion detected by enzyme-linked immunosorbent assay (ELISA) was only in db/db mice and preceded the onset of microalbuminuria. Immunofluorescence studies revealed that MIF was expressed in mouse kidney glomeruli. While MIF expression was enhanced in the diabetic kidneys of both mouse strains, the elevated secretion from db/db mouse kidneys may be responsible for initiating the onset of microalbuminuria in diabetic nephropathy.


Subject(s)
Albuminuria/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/metabolism , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Albuminuria/genetics , Animals , Blood Glucose/analysis , Body Weight , Diabetes Mellitus, Type 2/genetics , Diabetic Nephropathies/genetics , Disease Models, Animal , Gene Expression Profiling , Insulin/blood , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Male , Mice , Oligonucleotide Array Sequence Analysis
19.
EMBO J ; 32(9): 1265-79, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23572076

ABSTRACT

Glycerol-3-phosphate acyltransferase (GPAT) is involved in the first step in glycerolipid synthesis and is localized in both the endoplasmic reticulum (ER) and mitochondria. To clarify the functional differences between ER-GPAT and mitochondrial (Mt)-GPAT, we generated both GPAT mutants in C. elegans and demonstrated that Mt-GPAT is essential for mitochondrial fusion. Mutation of Mt-GPAT caused excessive mitochondrial fragmentation. The defect was rescued by injection of lysophosphatidic acid (LPA), a direct product of GPAT, and by inhibition of LPA acyltransferase, both of which lead to accumulation of LPA in the cells. Mitochondrial fragmentation in Mt-GPAT mutants was also rescued by inhibition of mitochondrial fission protein DRP-1 and by overexpression of mitochondrial fusion protein FZO-1/mitofusin, suggesting that the fusion/fission balance is affected by Mt-GPAT depletion. Mitochondrial fragmentation was also observed in Mt-GPAT-depleted HeLa cells. A mitochondrial fusion assay using HeLa cells revealed that Mt-GPAT depletion impaired mitochondrial fusion process. We postulate from these results that LPA produced by Mt-GPAT functions not only as a precursor for glycerolipid synthesis but also as an essential factor of mitochondrial fusion.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Glycerol-3-Phosphate O-Acyltransferase/metabolism , Mitochondria/enzymology , Mitochondrial Dynamics , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/physiology , Female , Gene Deletion , Glycerol-3-Phosphate O-Acyltransferase/genetics , Glycerol-3-Phosphate O-Acyltransferase/physiology , Lysophospholipids/metabolism , Lysophospholipids/pharmacology , Microsomes/metabolism , Mitochondria/drug effects , Mitochondria/genetics , Mitochondrial Size/drug effects , Mitochondrial Size/genetics , Models, Biological , Mutagenesis, Site-Directed , Oogenesis/genetics
20.
Mol Biol Cell ; 21(18): 3114-24, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20668164

ABSTRACT

Phosphatidylinositol (PI), an important constituent of membranes, contains stearic acid as the major fatty acid at the sn-1 position. This fatty acid is thought to be incorporated into PI through fatty acid remodeling by sequential deacylation and reacylation. However, the genes responsible for the reaction are unknown, and consequently, the physiological significance of the sn-1 fatty acid remains to be elucidated. Here, we identified acl-8, -9, and -10, which are closely related to each other, and ipla-1 as strong candidates for genes involved in fatty acid remodeling at the sn-1 position of PI. In both ipla-1 mutants and acl-8 acl-9 acl-10 triple mutants of Caenorhabditis elegans, the stearic acid content of PI is reduced, and asymmetric division of stem cell-like epithelial cells is defective. The defects in asymmetric division of these mutants are suppressed by a mutation of the same genes involved in intracellular retrograde transport, suggesting that ipla-1 and acl genes act in the same pathway. IPLA-1 and ACL-10 have phospholipase A(1) and acyltransferase activity, respectively, both of which recognize the sn-1 position of PI as their substrate. We propose that the sn-1 fatty acid of PI is determined by ipla-1 and acl-8, -9, -10 and crucial for asymmetric divisions.


Subject(s)
Acyltransferases/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Fatty Acids/chemistry , Phosphatidylinositols/chemistry , Phospholipases A1/metabolism , Stem Cells/physiology , Acyltransferases/genetics , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Cell Division/physiology , Fatty Acids/metabolism , HEK293 Cells , Humans , Mice , Mutation , Phosphatidylinositols/metabolism , Phospholipases A1/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Stearic Acids/chemistry , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL