Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pflugers Arch ; 475(4): 489-504, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36749388

RESUMEN

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.


Asunto(s)
Transportadores de Anión Orgánico , Ácido Úrico , Animales , Humanos , Ratones , Secuencia de Aminoácidos , Ácido Ascórbico/metabolismo , Transporte Biológico , Mamíferos/metabolismo , Transportadores de Anión Orgánico/metabolismo , Transportadores de Sodio Acoplados a la Vitamina C/genética , Transportadores de Sodio Acoplados a la Vitamina C/metabolismo , Ácido Úrico/metabolismo
2.
Proc Natl Acad Sci U S A ; 117(31): 18175-18177, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32690690

RESUMEN

Recent genome-wide association studies have revealed some genetic loci associated with serum uric acid levels and susceptibility to gout/hyperuricemia which contain potential candidates of physiologically important urate transporters. One of these novel loci is located upstream of SGK1 and SLC2A12, suggesting that variations in these genes increase the risks of hyperuricemia and gout. We herein focused on SLC2A12 encoding a transporter, GLUT12, the physiological function of which remains unclear. As GLUT12 belongs to the same protein family as a well-recognized urate transporter GLUT9, we hypothesized that GLUT12 mediates membrane transport of urate. Therefore, we conducted functional assays and analyzed Glut12 knockout hyperuricemia model mice, generated using the CRISPR-Cas9 system. Our results revealed that GLUT12 acts as a physiological urate transporter and its dysfunction elevates the blood urate concentration. This study provides insights into the deeper understanding of the urate regulatory system in the body, which is also important for pathophysiology of gout/hyperuricemia.


Asunto(s)
Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Hiperuricemia/sangre , Ácido Úrico/sangre , Animales , Regulación de la Expresión Génica , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Ratones , Ratones Noqueados , Ácido Úrico/metabolismo
3.
J Am Soc Nephrol ; 33(2): 326-341, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34799437

RESUMEN

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.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Hipoxantina Fosforribosiltransferasa/metabolismo , Transportadores de Anión Orgánico/deficiencia , Urato Oxidasa/deficiencia , Xantina Deshidrogenasa/antagonistas & inhibidores , Lesión Renal Aguda/etiología , Lesión Renal Aguda/metabolismo , Alopurinol/farmacología , Animales , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Hipoxantina Fosforribosiltransferasa/genética , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Nitrilos/farmacología , Transportadores de Anión Orgánico/genética , Esfuerzo Físico , Piridinas/farmacología , Defectos Congénitos del Transporte Tubular Renal/tratamiento farmacológico , Defectos Congénitos del Transporte Tubular Renal/etiología , Defectos Congénitos del Transporte Tubular Renal/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Urato Oxidasa/genética , Cálculos Urinarios/tratamiento farmacológico , Cálculos Urinarios/etiología , Cálculos Urinarios/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 40(3): 570-582, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31996020

RESUMEN

OBJECTIVE: Uric acid is supposed but not yet determined to be associated with atherosclerosis. Uric acid is released from damaged cells to form urate crystal, which is recognized by the immune system to produce IL (interleukin)-1. Danger signals and IL-1 have been shown to play an important role in atherosclerosis. We determined whether the physiological level of soluble uric acid promotes inflammation and develops atherosclerosis. Approach and Results: The secretion of IL-1ß from human peripheral blood mononuclear cells mediated by NLRP3 (NACHT, LRR, and PYD domain-containing protein 3) inflammasome was promoted by physiological levels in serum uric acid. This augmentation of inflammation was mediated by the regulation of the AMPK (AMP-activated protein kinase)-mTOR (mammalian target of rapamycin) mitochondrial reactive oxygen species and HIF-1α (hypoxia-inducible factor-1α) pathway. In both of uricase transgenic and xanthine oxidase inhibitor-treated mice, decreased levels of uric acid resulted in the activation of AMPK and attenuation of the development of atherosclerotic plaques. Further, acute uric acid reduction by the administration of benzbromarone in healthy humans for 2 weeks significantly decreased plasma IL-18-an inflammasome-dependent cytokine. CONCLUSIONS: The data indicate that the development of atherosclerosis and inflammation is promoted by uric acid in vivo. Moreover, the lowering of uric acid levels attenuated inflammation via the activation of the AMPK pathway. This study provides mechanistic evidence of uric acid-lowering therapies for atherosclerosis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Aterosclerosis/enzimología , Inflamación/enzimología , Leucocitos Mononucleares/enzimología , Ácido Úrico/sangre , Adulto , Animales , Aterosclerosis/sangre , Aterosclerosis/patología , Aterosclerosis/prevención & control , Benzbromarona/administración & dosificación , Biomarcadores/sangre , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Células HEK293 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamasomas/genética , Inflamasomas/metabolismo , Inflamación/sangre , Inflamación/patología , Inflamación/prevención & control , Mediadores de Inflamación/sangre , Interleucina-18/sangre , Interleucina-1beta/sangre , Interleucina-1beta/genética , Leucocitos Mononucleares/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Persona de Mediana Edad , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína del Factor Nuclear 45/sangre , Placa Aterosclerótica , Especies Reactivas de Oxígeno/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Urato Oxidasa/genética , Urato Oxidasa/metabolismo , Uricosúricos/administración & dosificación , Xantina Oxidasa/antagonistas & inhibidores , Xantina Oxidasa/metabolismo , Adulto Joven
5.
Biol Pharm Bull ; 44(12): 1824-1831, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34853265

RESUMEN

Thyroid hormones (THs) have been suggested to play an important role in both physiological and pathological events in the central nervous system. Hypothyroidism, which is characterized by low levels of serum THs, has been associated with aggravation of ischemic neuronal injuries in stroke patients. We hypothesized that administration of T3, the main active form of THs, may attenuate the ischemic neuronal injuries. In mice, global cerebral ischemia (GCI), which is induced by transient occlusion of the bilateral common carotid artery, causes neuronal injuries by inducing neuronal death and activating inflammatory responses after reperfusion in the hippocampus. In this study, we examined the effect of T3 administration on DNA fragmentation induced by neuronal death and the activation of inflammatory cells such as astrocytes and microglia in the hippocampus following GCI. The content of nucleosomes generated by DNA fragmentation in the hippocampus was increased by GCI and further increased by T3 administration. The protein expression levels of glial fibrillary acidic protein (GFAP), an astrocytic marker, and Ionized calcium binding adaptor protein 1 (Iba1), a microglial marker, in the hippocampus were also increased by GCI and further increased by T3 administration. The levels of T3 in both the serum and hippocampus were elevated by T3 administration. Our results indicate that T3 administration aggravates GCI-reperfusion injury in mice. There may be an increased risk of aggravation of ischemic stroke by the excessive elevation of T3 levels during the drug treatment of hypothyroidism.


Asunto(s)
Isquemia Encefálica , Hipocampo/efectos de los fármacos , Daño por Reperfusión , Índice de Severidad de la Enfermedad , Triyodotironina/efectos adversos , Animales , Astrocitos , Muerte Celular , Infarto Cerebral , Fragmentación del ADN , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía , Hipotiroidismo/complicaciones , Hipotiroidismo/tratamiento farmacológico , Inflamación , Masculino , Ratones Endogámicos C57BL , Microglía , Neuronas , Nucleosomas , Reperfusión , Triyodotironina/sangre
6.
Int J Mol Sci ; 21(18)2020 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-32899645

RESUMEN

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).


Asunto(s)
Plexo Coroideo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Tercer Ventrículo/metabolismo , Animales , Encéfalo/metabolismo , Ventrículos Cerebrales/metabolismo , Ventrículos Cerebrales/fisiología , Líquido Cefalorraquídeo/metabolismo , Plexo Coroideo/fisiología , Fibroblastos/metabolismo , Humanos , Ácido Láctico/metabolismo , Ratones , Ratones Endogámicos C57BL , Tercer Ventrículo/fisiología
7.
Am J Physiol Renal Physiol ; 313(3): F826-F834, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28679589

RESUMEN

Accumulating data indicate that renal uric acid (UA) handling is altered in diabetes and by hypoglycemic agents. In addition, hyperinsulinemia is associated with hyperuricemia and hypouricosuria. However, the underlying mechanisms remain unclear. In this study, we aimed to investigate how diabetes and hypoglycemic agents alter the levels of renal urate transporters. In insulin-depleted diabetic rats with streptozotocin treatment, both UA excretion and fractional excretion of UA were increased, suggesting that tubular handling of UA is altered in this model. In the membrane fraction of the kidney, the expression of urate transporter 1 (URAT1) was significantly decreased, whereas that of ATP-binding cassette subfamily G member 2 (ABCG2) was increased, consistent with the increased renal UA clearance. Administration of insulin to the diabetic rats decreased UA excretion and alleviated UA transporter-level changes, while sodium glucose cotransporter 2 inhibitor (SGLT2i) ipragliflozin did not change renal UA handling in this model. To confirm the contribution of insulin in the regulation of urate transporters, normal rats received insulin and separately, ipragliflozin. Insulin significantly increased URAT1 and decreased ABCG2 levels, resulting in increased UA reabsorption. In contrast, the SGLT2i did not alter URAT1 or ABCG2 levels, although blood glucose levels were similarly reduced. Furthermore, we found that insulin significantly increased endogenous URAT1 levels in the membrane fraction of NRK-52E cells, the kidney epithelial cell line, demonstrating the direct effects of insulin on renal UA transport mechanisms. These results suggest a previously unrecognized mechanism for the anti-uricosuric effects of insulin and provide novel insights into the renal UA handling in the diabetic state.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Proteínas de Transporte de Anión/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Hipoglucemiantes/farmacología , Insulina/farmacología , Túbulos Renales/efectos de los fármacos , Reabsorción Renal/efectos de los fármacos , Ácido Úrico/metabolismo , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Línea Celular , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/fisiopatología , Glucósidos/farmacología , Túbulos Renales/metabolismo , Túbulos Renales/fisiopatología , Masculino , Ratas Sprague-Dawley , Eliminación Renal/efectos de los fármacos , Transportador 2 de Sodio-Glucosa/metabolismo , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Estreptozocina , Tiofenos/farmacología , Factores de Tiempo , Ácido Úrico/orina
8.
Ann Rheum Dis ; 76(5): 869-877, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27899376

RESUMEN

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.


Asunto(s)
Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Gota/genética , Adulto , Anciano , Pueblo Asiatico/genética , Estudios de Casos y Controles , Proteínas de Transporte de Catión/genética , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Sitios Genéticos , Genotipo , Gota/clasificación , Histonas/genética , Humanos , Japón , Masculino , Persona de Mediana Edad , Nativos de Hawái y Otras Islas del Pacífico/genética , Transportadores de Anión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/genética , Polimorfismo de Nucleótido Simple , Proteínas/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/genética , Población Blanca/genética
9.
Biol Pharm Bull ; 39(7): 1081-4, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27170514

RESUMEN

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.


Asunto(s)
Hipoxantina Fosforribosiltransferasa/genética , Urato Oxidasa/genética , Ácido Úrico/sangre , Animales , Hipoxantina/sangre , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Xantina/sangre
10.
Clin Exp Nephrol ; 19(4): 576-84, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25239792

RESUMEN

BACKGROUND: Familial juvenile hyperuricemic nephropathy (FJHN) is an autosomal dominant disorder caused by mutations in UMOD that encodes uromodulin. Topiroxostat, a novel non-purine analog, selectively inhibits xanthine oxidase and reduces the serum uric acid levels and the urinary albuminuria. METHODS: Genomic DNA of a patient was extracted from peripheral white blood. Exons and flanking sequences of UMOD were amplified by PCR with primers. Mutation analysis was performed by direct sequencing of the PCR products. The wild-type and mutant uromodulin were expressed in HEK293 cells and analyzed by western blotting, immunoprecipitation, immunofluorescence, and flow cytometry. RESULTS: We identified an FJHN patient who carried a novel UMOD mutation G335A (C112Y). The levels of both cytosolic and secreted C112Y protein were significantly decreased compared with the wild-type, whereas the level of ubiquitination was higher in C112Y than that in the wild type. The half-life of C112Y was shortened and it was restored by a proteasome inhibitor MG132. Immunofluorescence revealed decreased levels of C112Y in the Golgi apparatus and on the plasma membrane. Expression of C112Y induced cellular apoptosis as revealed by flow cytometry. Apoptosis induced by C112Y was suppressed by topiroxostat. CONCLUSION: C112Y causes its protein instability resulting cellular apoptosis which could be suppressed with topiroxostat.


Asunto(s)
Apoptosis/efectos de los fármacos , Gota/genética , Hiperuricemia/genética , Enfermedades Renales/genética , Nitrilos/uso terapéutico , Piridinas/uso terapéutico , Uromodulina/genética , Adulto , Gota/tratamiento farmacológico , Células HEK293 , Humanos , Hiperuricemia/tratamiento farmacológico , Enfermedades Renales/tratamiento farmacológico , Masculino , Mutación , Nitrilos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Piridinas/farmacología
11.
Sci Rep ; 14(1): 3187, 2024 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-38326353

RESUMEN

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.


Asunto(s)
Isquemia Encefálica , Hipotermia , Ribonucleótidos , Humanos , Hipotermia/metabolismo , Isquemia Encefálica/metabolismo , Xantina/metabolismo , Infarto Cerebral/metabolismo , Hipocampo/metabolismo , Adenosina Monofosfato/metabolismo
12.
Biol Pharm Bull ; 36(5): 741-7, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23649333

RESUMEN

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.


Asunto(s)
Albuminuria/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Nefropatías Diabéticas/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Albuminuria/genética , Animales , Glucemia/análisis , Peso Corporal , Diabetes Mellitus Tipo 2/genética , Nefropatías Diabéticas/genética , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Insulina/sangre , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Masculino , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos
13.
Nucleosides Nucleotides Nucleic Acids ; 41(12): 1279-1286, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35094660

RESUMEN

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.


Asunto(s)
Riñón , Proteínas de Transporte de Membrana , Humanos , Ratas , Animales , Hipoxantina/metabolismo , Riñón/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Transporte Biológico , Sodio/metabolismo , Sodio/farmacología , Adenina/metabolismo , Xantinas/metabolismo
14.
Plast Reconstr Surg Glob Open ; 10(10): e4601, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36320623

RESUMEN

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.

15.
J Pharmacol Sci ; 115(3): 383-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21358122

RESUMEN

Uromodulin storage diseases are characterized by hyperuricemia of underexcretion type and renal insufficiency. Although these diseases are caused by mutations in the UMOD gene that encodes the kidney-specific glycoprotein uromodulin, the effect of uromodulin mutation on the kidney has not been clearly established. In this study, we investigated the effect by comparing transgenic mice expressing human uromodulin with and without mutation. Change in the intracellular localization of human uromodulin protein was shown in the kidney of transgenic mice expressing mutant human uromodulin by a deglycosylation experiment. Then, we determined by microarray technology and quantitative real-time PCR that the strongly induced gene in the kidney of these mice was 5-α-reductase 2, an enzyme that converts testosterone into the more potent androgen. Moreover, the expressions of androgen-induced genes ß-glucuronidase, ornithine decarboxylase structural 1, and cytochrome P450 4a12a were increased. The increase in mRNA levels of urate reabsorptive transport system urate transporter 1 could be investigated, but the changes in its protein level and renal urate handling could not be demonstrated. Therefore, it is suggested that a uromodulin mutation may be responsible for the enhancement of renal androgen action.


Asunto(s)
Andrógenos/metabolismo , Riñón/metabolismo , Testosterona/metabolismo , Uromodulina/genética , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/genética , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/metabolismo , Alantoína/sangre , Animales , Perfilación de la Expresión Génica , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Mutación , Ácido Úrico/sangre , Ácido Úrico/orina , Uromodulina/metabolismo
16.
Biomedicines ; 9(12)2021 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-34944661

RESUMEN

Renal hypouricemia (RHUC) is a hereditary disease that presents with increased renal urate clearance and hypouricemia due to genetic mutations in the urate transporter URAT1 or GLUT9 that reabsorbs urates in the renal proximal tubule. Exercise-induced acute kidney injury (EIAKI) is known to be a complication of renal hypouricemia. In the skeletal muscle of RHUC patients during exhaustive exercise, the decreased release of endothelial-derived hyperpolarization factor (EDHF) due to hypouricemia might cause the disturbance of exercise hyperemia, which might increase post-exercise urinary urate excretion. In the kidneys of RHUC patients after exhaustive exercise, an intraluminal high concentration of urates in the proximal straight tubule and/or thick ascending limb of Henle's loop might stimulate the luminal Toll-like receptor 4-myeloid differentiation factor 88-phosphoinositide 3-kinase-mammalian target of rapamycin (luminal TLR4-MyD88-PI3K-mTOR) pathway to activate the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome and may release interleukin-1ß (IL-1ß), which might cause the symptoms of EIAKI.

17.
Psychogeriatrics ; 10(1): 34-8, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20594285

RESUMEN

BACKGROUND: Elderly patients with Alzheimer's disease (AD) take more medicines, other than those for anti-dementia agents, than healthy people and are sensitive to anticholinergic medications. There are only a few reports, however, on the relationship between cognitive function and anticholinergic activity in AD patients, which is caused by taking prescribed medication. METHODS: We measured serum anticholinergic activity (SAA) in 76 AD patients referred to a Psychogeriatric Unit and separated them into SAA positive group (n= 26, SAA (+) group) and SAA negative group (n= 50, SAA (-) group). The difference in demographic data and cognitive functions were compared between the two groups. RESULTS AND CONCLUSIONS: The total scores of the Mini-Mental State Examination (MMSE), the score of MMSE domain of registration and recall were significantly lower (P < 0.05) and the Functional Assessment Staging (FAST) score, the number of different kinds of prescribed psychotropic medications (the number of prescribed psychotropic medications) were significantly higher (P < 0.05) in the SAA (+) group than in the SAA (-). These results suggest that a higher number of psychotropic medications prescribed leads to a tendency for SAA to be positive and that anticholinergic activity accelerates Alzheimer's pathology and decreases cognitive function, especially memory in AD patients. We should more prudently prescribe psychotropic medications to AD patients, because the prescribed psychotropic medications are one of the important causes of decline in cognitive function of AD patients by way of anticholinergic activity.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/fisiopatología , Antagonistas Colinérgicos/efectos adversos , Trastornos del Conocimiento/inducido químicamente , Psicotrópicos/efectos adversos , Anciano , Anciano de 80 o más Años , Atropina , Femenino , Humanos , Masculino , Escala del Estado Mental , Receptores Colinérgicos/efectos de los fármacos , Receptores Colinérgicos/metabolismo
18.
Nucleosides Nucleotides Nucleic Acids ; 39(10-12): 1465-1473, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32126884

RESUMEN

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.


Asunto(s)
Técnicas de Inactivación de Genes , Hipoxantina Fosforribosiltransferasa/metabolismo , Xantina Deshidrogenasa/deficiencia , Xantina Deshidrogenasa/genética , Animales , Longevidad , Ratones , NAD/farmacología
19.
Br J Pharmacol ; 177(10): 2274-2285, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31971609

RESUMEN

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.


Asunto(s)
Minorías Sexuales y de Género , Urato Oxidasa , Alopurinol/farmacología , Animales , Homosexualidad Masculina , Humanos , Hipoxantina Fosforribosiltransferasa/genética , Masculino , Ratones , Ratones Noqueados , Purinas/farmacología , Xantina Deshidrogenasa/genética
20.
Artículo en Inglés | MEDLINE | ID: mdl-31983315

RESUMEN

To elucidate roles of the intestine in uric acid (UA) metabolism, we examined ABCG2 expression, tissue UA content and xanthine oxidoreductase (XOR) activity in different intestinal segments. Male SD rats were assigned to control group or oxonic acid-induced hyperuricemia (HUA) group. In control rats, ABCG2 was present both in villi and crypts in each segment. Tissue UA content and XOR activity were relatively high in duodenum and jejunum. However, in HUA rats, tissue UA content was significantly elevated in the ileum, whereas it remained unaltered in other segments. Moreover, ABCG2 expression in the HUA group was upregulated both in the villi and crypts of the ileum. These data indicate that the ileum may play an important role in the extra-renal UA excretion.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Hiperuricemia/metabolismo , Ácido Úrico/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Animales , Hiperuricemia/inducido químicamente , Intestinos , Masculino , Ácido Oxónico , Ratas , Ratas Sprague-Dawley , Xantina Deshidrogenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA