Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Sci Rep ; 14(1): 3187, 2024 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-38326353

RESUMO

Global cerebral ischemia (GCI) caused by clinical conditions such as cardiac arrest leads to delayed neuronal death in the hippocampus, resulting in physical and mental disability. However, the mechanism of delayed neuronal death following GCI remains unclear. To elucidate the mechanism, we performed a metabolome analysis using a mouse model in which hypothermia (HT) during GCI, which was induced by the transient occlusion of the bilateral common carotid arteries, markedly suppressed the development of delayed neuronal death in the hippocampus after reperfusion. Fifteen metabolites whose levels were significantly changed by GCI and 12 metabolites whose levels were significantly changed by HT were identified. Furthermore, the metabolites common for both changes were narrowed down to two, adenosine monophosphate (AMP) and xanthosine monophosphate (XMP). The levels of both AMP and XMP were found to be decreased by GCI, but increased by HT, thereby preventing their decrease. In contrast, the levels of adenosine, inosine, hypoxanthine, xanthine, and guanosine, the downstream metabolites of AMP and XMP, were increased by GCI, but were not affected by HT. Our results may provide a clue to understanding the mechanism by which HT during GCI suppresses the development of delayed neuronal death in the hippocampus.


Assuntos
Isquemia Encefálica , Hipotermia , Ribonucleotídeos , Humanos , Hipotermia/metabolismo , Isquemia Encefálica/metabolismo , Xantina/metabolismo , Infarto Cerebral/metabolismo , Hipocampo/metabolismo , Monofosfato de Adenosina/metabolismo
2.
Pflugers Arch ; 475(4): 489-504, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36749388

RESUMO

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.


Assuntos
Transportadores de Ânions Orgânicos , Ácido Úrico , Animais , Humanos , Camundongos , Sequência de Aminoácidos , Ácido Ascórbico/metabolismo , Transporte Biológico , Mamíferos/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Transportadores de Sódio Acoplados à Vitamina C/genética , Transportadores de Sódio Acoplados à Vitamina C/metabolismo , Ácido Úrico/metabolismo
3.
Plast Reconstr Surg Glob Open ; 10(10): e4601, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36320623

RESUMO

Bovine-derived collagen gel has been used in the medical field as an injection formulation, but there are concerns about cross-infection such as bovine spongiform encephalopathy. In this study, we attempted to use fish as a safe alternative to bovine collagen. Objective: Fish collagen has not been used in clinical settings, so we examined its potential by comparing its properties with those of bovine-derived collagen. Methods: Collagen was extracted from the ventral skin of flatfish. It was cross-linked with 1%, 3%, or 5% of 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDC) and treated with 1%, 5%, or 10% of lactose. Hydroxyproline contents and Young's modulus (elasticity) were measured. In addition, these were injected under the back of BALB/c nude mice and the amount of hydroxyproline was observed. Histological examination of the samples was also conducted. Results: The amount of hydroxyproline in fish collagen was 3.3 ± 0.3 µg/mg. The 3% collagen gel treated with 5% EDC and 5% lactose had the highest Young's modulus and was closest to the bovine-derived collagen injection formulation. When injected into mice, it was retained in vivo for about 90 days. Conclusions: Fish collagen has a low denaturation temperature and is unstable and easily biodegrades in mammalian organisms. However, it is possible to approach the properties of conventional mammalian collagen by cross-linking and lactose treatment, suggesting that fish collagen can be used as a scaffold for cells in regenerative medicine.

4.
J Am Soc Nephrol ; 33(2): 326-341, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34799437

RESUMO

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.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Hipoxantina Fosforribosiltransferase/metabolismo , Transportadores de Ânions Orgânicos/deficiência , Urato Oxidase/deficiência , Xantina Desidrogenase/antagonistas & inibidores , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/metabolismo , Alopurinol/farmacologia , Animais , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Hipoxantina Fosforribosiltransferase/genética , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Nitrilas/farmacologia , Transportadores de Ânions Orgânicos/genética , Esforço Físico , Piridinas/farmacologia , Erros Inatos do Transporte Tubular Renal/tratamento farmacológico , Erros Inatos do Transporte Tubular Renal/etiologia , Erros Inatos do Transporte Tubular Renal/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Urato Oxidase/genética , Cálculos Urinários/tratamento farmacológico , Cálculos Urinários/etiologia , Cálculos Urinários/metabolismo
5.
Br J Pharmacol ; 177(10): 2274-2285, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31971609

RESUMO

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.


Assuntos
Minorias Sexuais e de Gênero , Urato Oxidase , Alopurinol/farmacologia , Animais , Homossexualidade Masculina , Humanos , Hipoxantina Fosforribosiltransferase/genética , Masculino , Camundongos , Camundongos Knockout , Purinas/farmacologia , Xantina Desidrogenase/genética
6.
Ann Rheum Dis ; 76(5): 869-877, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27899376

RESUMO

OBJECTIVE: A genome-wide association study (GWAS) of gout and its subtypes was performed to identify novel gout loci, including those that are subtype-specific. METHODS: Putative causal association signals from a GWAS of 945 clinically defined gout cases and 1213 controls from Japanese males were replicated with 1396 cases and 1268 controls using a custom chip of 1961 single nucleotide polymorphisms (SNPs). We also first conducted GWASs of gout subtypes. Replication with Caucasian and New Zealand Polynesian samples was done to further validate the loci identified in this study. RESULTS: In addition to the five loci we reported previously, further susceptibility loci were identified at a genome-wide significance level (p<5.0×10-8): urate transporter genes (SLC22A12 and SLC17A1) and HIST1H2BF-HIST1H4E for all gout cases, and NIPAL1 and FAM35A for the renal underexcretion gout subtype. While NIPAL1 encodes a magnesium transporter, functional analysis did not detect urate transport via NIPAL1, suggesting an indirect association with urate handling. Localisation analysis in the human kidney revealed expression of NIPAL1 and FAM35A mainly in the distal tubules, which suggests the involvement of the distal nephron in urate handling in humans. Clinically ascertained male patients with gout and controls of Caucasian and Polynesian ancestries were also genotyped, and FAM35A was associated with gout in all cases. A meta-analysis of the three populations revealed FAM35A to be associated with gout at a genome-wide level of significance (p meta =3.58×10-8). CONCLUSIONS: Our findings including novel gout risk loci provide further understanding of the molecular pathogenesis of gout and lead to a novel concept for the therapeutic target of gout/hyperuricaemia.


Assuntos
Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Gota/genética , Adulto , Idoso , Povo Asiático/genética , Estudos de Casos e Controles , Proteínas de Transporte de Cátions/genética , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Loci Gênicos , Genótipo , Gota/classificação , Histonas/genética , Humanos , Japão , Masculino , Pessoa de Meia-Idade , Havaiano Nativo ou Outro Ilhéu do Pacífico/genética , Transportadores de Ânions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/genética , Polimorfismo de Nucleotídeo Único , Proteínas/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo I/genética , População Branca/genética
7.
Fluids Barriers CNS ; 13(1): 22, 2016 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-27955673

RESUMO

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.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Encéfalo/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Ácido Acético , Animais , Clorofórmio , Epêndima/metabolismo , Epitélio/metabolismo , Imunofluorescência , Hibridização In Situ , Masculino , Metanol , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro , Fixação de Tecidos
8.
Nucleosides Nucleotides Nucleic Acids ; 35(10-12): 550-558, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27906625

RESUMO

Uric acid (UA) is a potential risk factor of the progression of chronic kidney disease (CKD). Recently, we reported that intestinal UA excretion might be enhanced via upregulation of the ATP-binding cassette transporter G2 (Abcg2) in a 5/6 nephrectomy (Nx) rat model. In the present study, we examined the mRNA and protein expressions of UA transporters, URAT1, GLUT9/URATv1, ABCG2 and NPT4 in the kidney and ileum in the same rat model. Additionally, we investigated the Abcg2 mRNA expression of ileum in hyperuricemic rat model by orally administering oxonic acid. Male Wistar rats were randomly assigned to three groups consisting of Nx group, oxonic acid-treated (Ox) group and sham-operated control group, and sacrificed at 8 weeks. Creatinine and UA were measured and the mRNA expressions of UA transporters in the kidney and intestine were evaluated by a real time PCR. UA transporters in the kidney sections were also examined by immunohistochemistry. Serum creatinine elevated in the Nx group whereas serum UA increased in the Ox group. Both the mRNA expression and the immunohistochemistry of the UA transporters were decreased in the Nx group, suggesting a marginal role in UA elevation in decreased kidney function. In contrast, the mRNA expression of Abcg2 in the ileum significantly increased in the Ox group. These results suggest that the upregulation of Abcg2 mRNA in the ileum triggered by an elevation of serum UA may play a compensatory role in increasing intestinal UA excretion.


Assuntos
Rim/metabolismo , Insuficiência Renal Crônica/sangue , Ácido Úrico/sangue , Animais , Colo/metabolismo , Creatinina/sangue , Duodeno/metabolismo , Expressão Gênica , Íleo/metabolismo , Masculino , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar
9.
Nucleosides Nucleotides Nucleic Acids ; 35(10-12): 543-549, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27906636

RESUMO

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.


Assuntos
Injúria Renal Aguda/genética , Transportadores de Ânions Orgânicos/genética , Erros Inatos do Transporte Tubular Renal/genética , Urato Oxidase/genética , Cálculos Urinários/genética , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/metabolismo , Alopurinol/farmacologia , Alopurinol/uso terapêutico , Animais , Creatinina/sangue , Modelos Animais de Doenças , Supressores da Gota/farmacologia , Supressores da Gota/uso terapêutico , Masculino , Camundongos Knockout , Transportadores de Ânions Orgânicos/metabolismo , Condicionamento Físico Animal , Erros Inatos do Transporte Tubular Renal/tratamento farmacológico , Erros Inatos do Transporte Tubular Renal/metabolismo , Urato Oxidase/metabolismo , Ácido Úrico/urina , Cálculos Urinários/tratamento farmacológico , Cálculos Urinários/metabolismo
10.
Artigo em Inglês | MEDLINE | ID: mdl-24940669

RESUMO

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.


Assuntos
Análise Química do Sangue/métodos , Ácido Úrico/sangue , Alopurinol/farmacologia , Animais , Coleta de Amostras Sanguíneas , Reações Falso-Positivas , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fentolamina/farmacologia , Fatores de Tempo
11.
Sci Rep ; 3: 2014, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23774753

RESUMO

Gout is a common disease which mostly occurs after middle age, but more people nowadays develop it before the age of thirty. We investigated whether common dysfunction of ABCG2, a high-capacity urate transporter which regulates serum uric acid levels, causes early-onset gout. 705 Japanese male gout cases with onset age data and 1,887 male controls were genotyped, and the ABCG2 functions which are estimated by its genotype combination were determined. The onset age was 6.5 years earlier with severe ABCG2 dysfunction than with normal ABCG2 function (P = 6.14 × 10(-3)). Patients with mild to severe ABCG2 dysfunction accounted for 88.2% of early-onset cases (twenties or younger). Severe ABCG2 dysfunction particularly increased the risk of early-onset gout (odds ratio 22.2, P = 4.66 × 10(-6)). Our finding that common dysfunction of ABCG2 is a major cause of early-onset gout will serve to improve earlier prevention and therapy for high-risk individuals.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Gota/genética , Proteínas de Neoplasias/genética , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Adolescente , Adulto , Idade de Início , Estudos de Casos e Controles , Humanos , Masculino , Adulto Jovem
12.
Nat Commun ; 3: 764, 2012 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-22473008

RESUMO

ABCG2, also known as BCRP, is a high-capacity urate exporter, the dysfunction of which raises gout/hyperuricemia risk. Generally, hyperuricemia has been classified into urate 'overproduction type' and/or 'underexcretion type' based solely on renal urate excretion, without considering an extra-renal pathway. Here we show that decreased extra-renal urate excretion caused by ABCG2 dysfunction is a common mechanism of hyperuricemia. Clinical parameters, including urinary urate excretion, are examined in 644 male outpatients with hyperuricemia. Paradoxically, ABCG2 export dysfunction significantly increases urinary urate excretion and risk ratio of urate overproduction. Abcg2-knockout mice show increased serum uric acid levels and renal urate excretion, and decreased intestinal urate excretion. Together with high ABCG2 expression in extra-renal tissues, our data suggest that the 'overproduction type' in the current concept of hyperuricemia be renamed 'renal overload type', which consists of two subtypes-'extra-renal urate underexcretion' and genuine 'urate overproduction'-providing a new concept valuable for the treatment of hyperuricemia and gout.


Assuntos
Hiperuricemia/metabolismo , Rim/metabolismo , Ácido Úrico/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Adulto , Idoso , Animais , Transporte Biológico , Regulação para Baixo , Humanos , Hiperuricemia/genética , Hiperuricemia/fisiopatologia , Rim/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
13.
Nucleosides Nucleotides Nucleic Acids ; 30(12): 1112-6, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22132965

RESUMO

Primary renal hypouricemia is a genetic disorder characterized by defective renal uric acid (UA) reabsorption with complications such as nephrolithiasis and exercise-induced acute renal failure. The known causes are: defects in the SLC22A12 gene, encoding the human urate transporter 1 (hURAT1), and also impairment of voltage urate transporter (URATv1), encoded by SLC2A9 (GLUT9) gene. Diagnosis is based on hypouricemia (<119 µmol/L) and increased fractional excretion of UA (>10%). To date, the cases with mutations in hURAT1 gene have been reported in East Asia only. More than 100 Japanese patients have been described. Hypouricemia is sometimes overlooked; therefore, we have set up the flowchart for this disorder. The patients were selected for molecular analysis from 620 Czech hypouricemic patients. Secondary causes of hyperuricosuric hypouricemia were excluded. The estimations of (1) serum UA, (2) excretion fraction of UA, and (3) analysis of hURAT1 and URATv1 genes follow. Three transitions and one deletion (four times) in SLC22A12 gene and one nucleotide insertion in SLC2A9 gene in seven Czech patients were found. Three patients had acute renal failure and urate nephrolithiasis. In addition, five nonsynonymous sequence variants and three nonsynonymous sequence variants in SLC2A9 gene were found in two UK patients suffering from acute renal failure. Our finding of the defects in SLC22A12 and SLC2A9 genes gives further evidence of the causative genes of primary renal hypouricemia and supports their important role in regulation of serum urate levels in humans.


Assuntos
Técnicas e Procedimentos Diagnósticos , Erros Inatos do Transporte Tubular Renal/diagnóstico , Cálculos Urinários/diagnóstico , Adolescente , Adulto , Idoso , Proteínas Facilitadoras de Transporte de Glucose/genética , Humanos , Pessoa de Meia-Idade , Mutação/genética , Transportadores de Ânions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/genética , Erros Inatos do Transporte Tubular Renal/genética , Cálculos Urinários/genética , Adulto Jovem
14.
Nucleosides Nucleotides Nucleic Acids ; 30(12): 1290-4, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22132989

RESUMO

To elucidate the mechanism of obesity/metabolic syndrome-related hyperuricemia, this study aimed to determine the expression levels of transport systems for urate absorption (Urat1, Smct1, Glut9) and urate secretion (Abcg2). The kidneys of two obesity models in mice were used: 1) leptin-deficient mice (ob/ob mice) and 2) Quick fat diet model. 1) 8-week-old male ob/ob mice demonstrated the increased protein levels of Slc22a12 (Urat1), Slc2a9 (Glut9), and Abcg2 (Abcg2) and a decreased protein level of Slc5a8 (Smct1). However, no significant changes in the mRNA levels of these genes were observed. 2) C57BL/6 mice were fed with a Quick fat diet (crude fat content: 13.6%) from the age of 24 to 28 weeks (Quick fat diet group). The average body weights of the Quick fat diet group were heavier than those of the control group fed with a normal diet (crude fat content: 4.8%). The mRNA levels of Slc22a12, Slc2a9, Abcg2, or Slc5a8 did not change significantly in both groups. The protein levels of Slc22a12 (Urat1) and Abcg2 (Abcg2) increased significantly in the Quick fat diet group. Those of Slc2a9 (Glut9) and Slc5a8 (Smct1) were not changed significantly in the Quick fat diet group. In conclusion, the Quick fat diet enhanced the protein levels of Urat1 and Abcg2 without any changes in their mRNA transcription levels. The cause of obesity/metabolic syndrome-associated hyperuricemia appears to be associated with the urate reabsorption transporter Urat1 protein enhanced by fat.


Assuntos
Síndrome Metabólica/metabolismo , Obesidade/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Animais , Glicemia/metabolismo , Peso Corporal , Modelos Animais de Doenças , Regulação da Expressão Gênica , Insulina/sangue , Masculino , Síndrome Metabólica/sangue , Síndrome Metabólica/genética , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/genética , Transportadores de Ânions Orgânicos/sangue , Transportadores de Ânions Orgânicos/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
15.
Nucleosides Nucleotides Nucleic Acids ; 30(2): 113-9, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21360409

RESUMO

Increased levels of serum urate in postmenopausal women are thought to be caused by a change in renal urate elimination associated with the loss of female hormones. In this study, we investigated the regulation of renal urate transporter expression by female hormones using ovariectomized mice with or without hormone replacement. Estradiol suppressed the protein levels of urate reabsorptive transporters urate transporter 1 and glucose transporter 9 (Urat1 and Glut9), and that of urate efflux transporter ATP-binding cassette sub-family G member 2 (Abcg2). Progesterone suppressed protein levels of sodium-coupled monocarboxylate transporter 1 (Smct1). However, neither estradiol nor progesterone influenced the respective levels of mRNA.


Assuntos
Estradiol/farmacologia , Rim/efeitos dos fármacos , Rim/metabolismo , Progesterona/farmacologia , Ácido Úrico/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Proteínas de Transporte de Cátions/genética , Feminino , Proteínas Facilitadoras de Transporte de Glucose/genética , Camundongos , Camundongos Endogâmicos ICR , Transportadores de Ácidos Monocarboxílicos , Transportadores de Ânions Orgânicos/genética , Ovariectomia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
16.
Arzneimittelforschung ; 60(4): 186-8, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20486468

RESUMO

Both an angiotensin II receptor blocker, losartan (CAS 124750-99-8) and a serum urate lowering agent, benzbromarone (CAS 3562-84-3) exert a uricosuric action by inhibiting urate transporter 1 (URAT1). A recent clinical trial indicated that losartan could reduce the level of serum urate in hypertensive patients treated with urate lowering agents, suggesting the different mode of action of losartan from benzbromarone. In the present study, the effect of losartan and benzbromarone on the level of URAT1 mRNA was determined in transfected HEK293 cells. Losartan caused a significant reduction of its mRNA level, whereas it was not affected by benzbromarone. These results indicate that losartan decreases the level of human URAT1 mRNA, which may underlie the uricosuric action of losartan in hypertensive patients treated with serum urate lowering agents.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Benzobromarona/farmacologia , Losartan/farmacologia , Transportadores de Ânions Orgânicos/biossíntese , Proteínas de Transporte de Cátions Orgânicos/biossíntese , RNA Mensageiro/biossíntese , Uricosúricos/farmacologia , Linhagem Celular , Humanos , Rim/metabolismo , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ácido Úrico/metabolismo
17.
Biol Pharm Bull ; 31(12): 2250-4, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19043208

RESUMO

The effects of fluvoxamine, a selective serotonin reuptake inhibitor (SSRI), were studied in normophagic and food-restriction-induced hyperphagic middle-aged Wistar rats. Normophagic intact Wistar rats were given fluvoxamine (100 mg/kg/d, per os (p.o.)) or vehicle for 10 d. Hyperphagic middle-aged Wistar rats were subjected to 10 d of food restriction; they were allowed to refeed for 10 d, with ad libitum food access and administered fluvoxamine (100 mg/kg/d, p.o.) or vehicle during the 10-d refeeding period. Fluvoxamine administration to normophagic middle-aged Wistar rats affected neither their weight nor food intake. However, administration to food-restricted rats showed inhibitory effects of weight gain and food intake during 10 d of refeeding. Fluvoxamine-treated rats showed significantly lower neuropeptide Y (NPY) immunostaining levels in the paraventricular nucleus (PVN) and dorsomedial hypothalamic nucleus (DMH) than untreated controls. Hypophagic and weight-inhibiting effects of fluvoxamine might be mediated via decreased NPY in PVN and DMH. These results suggest that the appetite-controlling effect of fluvoxamine might be responsive to the rats' appetite condition.


Assuntos
Antidepressivos de Segunda Geração/farmacologia , Ingestão de Alimentos/efeitos dos fármacos , Fluvoxamina/farmacologia , Hiperfagia/psicologia , Aumento de Peso/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Hipotálamo/fisiologia , Masculino , Neuropeptídeo Y/fisiologia , Ratos , Ratos Wistar
18.
Am J Hypertens ; 21(10): 1157-62, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18670416

RESUMO

BACKGROUND: The angiotensin receptor blocker losartan inhibited urate transporter 1 (URAT1) according to in vitro experiments. However, it is still unknown whether the inhibitory effect of losartan on URAT1 contributes to its uricosuric action in humans. METHODS: Thirty-two patients with hypertension and nine patients with idiopathic renal hypouricemia (five with and four without hypertension) were enrolled for this study. Hypertensive patients were prescribed oral losartan (50 mg/day, n = 16) or candesartan (8 mg/day, n = 16). Before and after 1-month treatment, the serum concentration of urate (Sur) and creatinine (Scr), and the clearance value of urate (Cur) and creatinine (Ccr) were determined. Clearance studies using the URAT1 inhibitor benzbromarone (100 mg/day) or losartan (50 mg/day) loading test were also performed in these patients. RESULTS: Blood pressure (BP) significantly decreased in the patients treated with either losartan or candesartan. Losartan significantly reduced Sur, which was associated with a concomitant increase in the Cur/Ccr ratio, whereas candesartan did not alter these parameters. In hypertensive patients with loss-of-function mutation of URAT1, losartan did not alter either Sur or Cur/Ccr, nor did benzbromarone. The lack of effect of URAT1 inhibitors on renal excretion of urate was independent of the renal function of hypouricemic patients. On the other hand, both losartan and benzbromarone increased Cur/Ccr ratio in hypertensive patients harboring the wild URAT1 gene, regardless of the presence of hypouricemia. CONCLUSIONS: These findings suggested that losartan inhibited URAT1 and thereby it lowered Sur levels in hypertensive patients.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Hipertensão/tratamento farmacológico , Hiperuricemia/metabolismo , Losartan/uso terapêutico , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Administração Oral , Idoso , Bloqueadores do Receptor Tipo 1 de Angiotensina II/administração & dosagem , Benzimidazóis/administração & dosagem , Compostos de Bifenilo , Pressão Sanguínea/fisiologia , DNA/genética , Relação Dose-Resposta a Droga , Feminino , Seguimentos , Humanos , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Hiperuricemia/etiologia , Hiperuricemia/genética , Losartan/administração & dosagem , Masculino , Mutação , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , Proteínas de Transporte de Cátions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Reação em Cadeia da Polimerase , Tetrazóis/administração & dosagem , Resultado do Tratamento , Ácido Úrico/metabolismo
19.
Biol Pharm Bull ; 31(3): 405-11, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18310901

RESUMO

Familial juvenile hyperuricemic nephropathy (FJHN) and medullary cystic kidney disease type 2 (MCKD2) are autosomal dominant disorders characterized by juvenile hyperuricemia of the underexcretion type, juvenile gout and chronic renal failure in the adult. FJHN/MCKD2 constitute diseases caused by mutations of the human uromodulin (UMOD) gene that encodes uromodulin, the most abundant glycoprotein in normal human urine. The mutations affect the transport of uromodulin, resulting in the accumulation of uromodulin in the kidneys of FJHN/MCKD2 patients. The purpose of this study was to confirm the accumulation of uromodulin in the kidneys of transgenic mice harboring the mutant human UMOD gene with mouse UMOD gene promoter, and to determine the relationship between its accumulation and the effect on uromodulin transport. The mutant human UMOD mRNA and its protein were expressed in the kidneys of transgenic mice. Moreover, the staining of human uromodulin was colocalized with that of mouse uromodulin. Although the human UMOD mRNA levels increased, the protein levels did not change and the accumulation of human uromodulin was not observed. However, the mouse uromodulin consists of two forms, 103 and 117 kDa, and the 103 kDa protein was gradually increased in the kidneys of transgenic mice. Human and mouse uromodulins in the kidneys of transgenic mice were mainly detected in the Triton X-100 insoluble microsomal fraction. Therefore, the progressive accumulation of uromodulin was observed in the plasma membrane of the kidneys of transgenic mice but the accumulated uromodulin protein was not that encoded by the transgene.


Assuntos
Rim/metabolismo , Mucoproteínas/genética , Mucoproteínas/metabolismo , Mutação , Transgenes , Animais , Western Blotting , Membrana Celular/metabolismo , DNA/genética , Imunofluorescência , Humanos , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Transporte Proteico , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Uromodulina
20.
Toxicol Appl Pharmacol ; 227(3): 462-7, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18076961

RESUMO

Divalent metal transporter 1 (DMT1) is a mammalian iron (Fe) transporter and also transports Cadmium (Cd) in vitro. This study compared Cd absorption in DMT1-dysfunctional MK/Rej-(mk)/(mk) mice (mk/mk mice) and in DMT1-functional, Fe-deficient wild-type (WT) mice, to clarify the role of DMT1 in intestinal Cd absorption in vivo. Mice were given 1 ppm CdCl2 aq in drinking water for 2 weeks, and the concentrations of Cd and Fe in liver, kidney, and intestinal epithelium were subsequently determined. The Fe concentration in intestinal epithelia of WT mice was decreased in proportion to the level of dietary Fe limitation, while Cd accumulation under the same conditions was increased. DMT1 mRNA expression in the small intestine of Fe-deficient WT mice was clearly increased compared to that in Fe-sufficient WT mice. Iron deficiency resulted in up-regulation of Cd uptake in the intestine of Fe-deficient WT mice. The mk/mk mice have a mutation in DMT1 and loss of its function led to decreased intestinal Fe concentration. However, intestinal Cd accumulation was the same as in WT mice and it was also increased in Fe-deficient situation. There is the possibility that an unknown Cd pathway has taken a role on Cd intestinal absorption in vivo and that this pathway is regulated by food Fe concentrations. Therefore, DMT1 is not the sole transporter of intestinal cadmium absorption in vivo.


Assuntos
Cádmio/metabolismo , Proteínas de Transporte de Cátions/fisiologia , Absorção Intestinal , Mucosa Intestinal/metabolismo , Alelos , Anemia Ferropriva/genética , Animais , Cádmio/análise , Cloreto de Cálcio/administração & dosagem , Proteínas de Transporte de Cátions/genética , Absorção Intestinal/genética , Mucosa Intestinal/química , Ferro/análise , Deficiências de Ferro , Camundongos , Camundongos Mutantes , Mutação , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA