Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
World J Urol ; 42(1): 28, 2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-38214752

RESUMEN

PURPOSE: Oxalate is an excellent calcium ion attractor with great abundance in the human body, and the liver is the major source of oxalate. The Glycolate oxidase-1 (GOX1) gene is solely responsible for the glycolate and glyoxylate metabolism and produces oxalate. This study has been designed to comprehend the association of genetic variants of the GOX1 gene with the risk of hyperoxaluria and renal stone disease in the Indian population. METHOD: The present study is a candidate gene approach prospective case-control study carried out on 300 participants (150 cases and 150 controls) at Muljibhai Patel Urological Hospital, Gujarat, India. Biochemical parameters, including serum levels of calcium, creatinine, parathyroid hormone, and 24-h urine metabolites, were performed. The genotyping of GOX1 gene variants rs6086287, rs2235250, rs2255183, and rs2294303 was performed using a customized TaqMan assay probe by RT-PCR. RESULT: Parathyroid hormone, serum creatinine, and urine metabolites were significantly elevated in nephrolithiasis compared to healthy individuals. All mutated homozygous genotypes GG (rs6086287), TT (rs2235250), GG (rs2255183), and CC (rs2294303) were significantly associated with a high risk of renal stone disease. Individuals diagnosed with hyperoxaluria and carrying TG (rs6086287), AG (rs2255183), and TT (rs2294303) genotypes have a significantly high risk of renal stone disease. Moreover, haplotype analysis and correlation analysis also confirmed the strong association between genetic variants and nephrolithiasis. CONCLUSION: Genetic variants of the GOX1 genes were associated with renal stone disease. In the presence of risk genotype and hyperoxaluria, the susceptibility to develop renal stone disease risk gets modulated.


Asunto(s)
Oxidorreductasas de Alcohol , Hiperoxaluria , Cálculos Renales , Humanos , Calcio , Estudios de Casos y Controles , Cálculos Renales/complicaciones , Hiperoxaluria/genética , Oxalatos/orina , Hormona Paratiroidea , Creatinina
2.
Biomed Res Int ; 2023: 2883623, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38027040

RESUMEN

Purpose: To investigate whether ADSC-derived miR-23-enriched exosomes could protect against calcium oxalate stone formation in a hyperoxaluria rat model. Methods: An ethylene glycol- (EG-) induced hyperoxaluria rat model and an in vitro model of COM-induced HK-2 cells coculturing with RAW264.7 cells were established to explore the protective mechanisms of ADSC-derived miR-23-enriched exosomes. Results: The results showed that treatment with miR-23-enriched exosomes from ADSCs protected EG-induced hyperoxaluria rats, and cell experiments confirmed that coculturing with miR-23-enriched exosomes alleviated COM-induced cell autophagy. Overexpressed miR-23 suppressed M1 macrophage polarization by inhibiting IRF1 expression. Furthermore, the predicted binding site between the IRF1 messenger RNA 3'-untranslated region (3'-UTR) and miR-23 was confirmed by the dual-luciferase reporter assay. Conclusion: In conclusion, our research gave the first evidence that ADSC-derived miR-23-enriched exosomes affected the polarization of M1 macrophages by directly inhibiting IRF1 and protecting against calcium oxalate stone formation in a hyperoxaluria rat model.


Asunto(s)
Calcinosis , Exosomas , Hiperoxaluria , MicroARNs , Ratas , Animales , Oxalatos , Oxalato de Calcio/metabolismo , Exosomas/genética , Exosomas/metabolismo , Hiperoxaluria/genética , Hiperoxaluria/metabolismo , Macrófagos/metabolismo , Células del Estroma/metabolismo , Calcinosis/metabolismo , MicroARNs/metabolismo
3.
Sci Rep ; 13(1): 9029, 2023 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-37270618

RESUMEN

The risk of enteric hyperoxaluria is significantly increased after malabsorptive bariatric surgery (MBS). However, its underlying determinants are only poorly characterized. In this case-control study, we aimed at identifying clinical and genetic factors to dissect their individual contributions to the development of post-surgical hyperoxaluria. We determined the prevalence of hyperoxaluria and nephrolithiasis after MBS by 24-h urine samples and clinical questionnaires at our obesity center. Both hyperoxaluric and non-hyperoxaluric patients were screened for sequence variations in known and candidate genes implicated in hyperoxaluria (AGXT, GRHPR, HOGA1, SLC26A1, SLC26A6, SLC26A7) by targeted next generation sequencing (tNGS). The cohort comprised 67 patients, 49 females (73%) and 18 males (27%). While hyperoxaluria was found in 29 patients (43%), only one patient reported postprocedural nephrolithiasis within 41 months of follow-up. Upon tNGS, we did not find a difference regarding the burden of (rare) variants between hyperoxaluric and non-hyperoxaluric patients. However, patients with hyperoxaluria showed significantly greater weight loss accompanied by markers of intestinal malabsorption compared to non-hyperoxaluric controls. While enteric hyperoxaluria is very common after MBS, genetic variation of known hyperoxaluria genes contributes little to its pathogenesis. In contrast, the degree of postsurgical weight loss and levels of malabsorption parameters may allow for predicting the risk of enteric hyperoxaluria and consecutive kidney stone formation.


Asunto(s)
Cirugía Bariátrica , Hiperoxaluria , Cálculos Renales , Masculino , Femenino , Humanos , Estudios de Casos y Controles , Hiperoxaluria/genética , Hiperoxaluria/complicaciones , Cirugía Bariátrica/efectos adversos , Cálculos Renales/complicaciones , Pérdida de Peso , Variación Genética
4.
Commun Biol ; 6(1): 270, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36922584

RESUMEN

Hyperoxaluria is well known to cause renal injury and end-stage kidney disease. Previous studies suggested that acetate treatment may improve the renal function in hyperoxaluria rat model. However, its underlying mechanisms remain largely unknown. Using an ethylene glycol (EG)-induced hyperoxaluria rat model, we find the oral administration of 5% acetate reduced the elevated serum creatinine, urea, and protected against hyperoxaluria-induced renal injury and fibrosis with less infiltrated macrophages in the kidney. Treatment of acetate in renal tubular epithelial cells in vitro decrease the macrophages recruitment which might have reduced the oxalate-induced renal tubular cells injury. Mechanism dissection suggests that acetate enhanced acetylation of Histone H3 in renal tubular cells and promoted expression of miR-493-3p by increasing H3K9 and H3K27 acetylation at its promoter region. The miR-493-3p can suppress the expression of macrophage migration inhibitory factor (MIF), thus inhibiting the macrophages recruitment and reduced oxalate-induced renal tubular cells injury. Importantly, results from the in vivo rat model also demonstrate that the effects of acetate against renal injury were weakened after blocking the miR-493-3p by antagomir treatment. Together, these results suggest that acetate treatment ameliorates the hyperoxaluria-induced renal injury via inhibiting macrophages infiltration with change of the miR-493-3p/MIF signals. Acetate could be a new therapeutic approach for the treatment of oxalate nephropathy.


Asunto(s)
Acetatos , Hiperoxaluria , Factores Inhibidores de la Migración de Macrófagos , MicroARNs , Animales , Ratas , Acetatos/farmacología , Hiperoxaluria/complicaciones , Hiperoxaluria/tratamiento farmacológico , Hiperoxaluria/genética , Oxidorreductasas Intramoleculares/metabolismo , Riñón/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Oxalatos/efectos adversos
5.
J Med Genet ; 59(11): 1035-1043, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35115415

RESUMEN

BACKGROUND: Nephrolithiasis (NL) is a complex multifactorial disease affecting up to 10%-20% of the human population and causing a significant burden on public health systems worldwide. It results from a combination of environmental and genetic factors. Hyperoxaluria is a major risk factor for NL. METHODS: We used a whole exome-based approach in a patient with calcium oxalate NL. The effects of the mutation were characterised using cell culture and in silico analyses. RESULTS: We identified a rare heterozygous missense mutation (c.1519C>T/p.R507W) in the SLC26A6 gene that encodes a secretory oxalate transporter. This mutation cosegregated with hyperoxaluria in the family. In vitro characterisation of mutant SLC26A6 demonstrated that Cl--dependent oxalate transport was dramatically reduced because the mutation affects both SLC26A6 transport activity and membrane surface expression. Cotransfection studies demonstrated strong dominant-negative effects of the mutant on the wild-type protein indicating that the phenotype of patients heterozygous for this mutation may be more severe than predicted by haploinsufficiency alone. CONCLUSION: Our study is in line with previous observations made in the mouse showing that SLC26A6 inactivation can cause inherited enteric hyperoxaluria with calcium oxalate NL. Consistent with an enteric form of hyperoxaluria, we observed a beneficial effect of increasing calcium in the patient's diet to reduce urinary oxalate excretion.


Asunto(s)
Antiportadores , Hiperoxaluria , Nefrolitiasis , Transportadores de Sulfato , Humanos , Antiportadores/genética , Calcio/metabolismo , Oxalato de Calcio/metabolismo , Hiperoxaluria/complicaciones , Hiperoxaluria/genética , Mutación , Nefrolitiasis/genética , Nefrolitiasis/complicaciones , Nefrolitiasis/metabolismo , Oxalatos/metabolismo , Transportadores de Sulfato/genética
6.
Am J Kidney Dis ; 79(1): 125-128, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34245816

RESUMEN

Primary hyperoxaluria (PH) is a group of genetic disorders that result in an increased hepatic production of oxalate. PH type 3 (PH3) is the most recently identified subtype and results from mutations in the mitochondrial 4-hydroxy-2-oxoglutarate aldolase gene (HOGA1). To date, there have been 2 cases of kidney failure reported in PH3 patients. We present a case of a young man with a history of recurrent urinary tract infections and voiding dysfunction who developed kidney failure at 33 years of age. He developed a bladder stone and bilateral staghorn calculi at 12 years of age. Initial metabolic evaluation revealed hyperoxaluria with very low urinary citrate excretion on multiple measurements for which he was placed on oral citrate supplements. Further investigation of the hyperoxaluria was not completed as the patient was lost to follow-up observation until he presented at 29 years of age with chronic kidney disease stage 4 (estimated glomerular filtration rate 24mL/min/1.73m2). Hemodialysis 3 times a week was started at 33 years of age, and subsequent genetic testing revealed a homozygous HOGA1 mutation (C.973G>A p.Gly325Ser) diagnostic of PH3. The patient is currently being evaluated for all treatment options including possible liver/kidney transplantation. All cases of a childhood history of recurrent urinary stone disease with marked hyperoxaluria should prompt genetic testing for the 3 known PH types. Hyperhydration and crystallization inhibitors (citrate) are standard of care, but the role of RNA interference agents for all 3 forms of PH is also under active study.


Asunto(s)
Hiperoxaluria Primaria , Hiperoxaluria , Oxo-Ácido-Liasas , Insuficiencia Renal , Humanos , Hiperoxaluria/complicaciones , Hiperoxaluria/diagnóstico , Hiperoxaluria/genética , Hiperoxaluria Primaria/complicaciones , Hiperoxaluria Primaria/diagnóstico , Hiperoxaluria Primaria/genética , Masculino , Oxalatos
7.
Cell Rep ; 36(8): 109526, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34433051

RESUMEN

Epigenetic modifications (e.g. DNA methylation) in NAFLD and their contribution to disease progression and extrahepatic complications are poorly explored. Here, we use an integrated epigenome and transcriptome analysis of mouse NAFLD hepatocytes and identify alterations in glyoxylate metabolism, a pathway relevant in kidney damage via oxalate release-a harmful waste product and kidney stone-promoting factor. Downregulation and hypermethylation of alanine-glyoxylate aminotransferase (Agxt), which detoxifies glyoxylate, preventing excessive oxalate accumulation, is accompanied by increased oxalate formation after metabolism of the precursor hydroxyproline. Viral-mediated Agxt transfer or inhibiting hydroxyproline catabolism rescues excessive oxalate release. In human steatotic hepatocytes, AGXT is also downregulated and hypermethylated, and in NAFLD adolescents, steatosis severity correlates with urinary oxalate excretion. Thus, this work identifies a reduced capacity of the steatotic liver to detoxify glyoxylate, triggering elevated oxalate, and provides a mechanistic explanation for the increased risk of kidney stones and chronic kidney disease in NAFLD patients.


Asunto(s)
Epigenoma , Glioxilatos/metabolismo , Hepatocitos/metabolismo , Hiperoxaluria/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Transcriptoma , Animales , Epigenómica , Perfilación de la Expresión Génica , Humanos , Hiperoxaluria/genética , Masculino , Ratones , Ratones Obesos , Enfermedad del Hígado Graso no Alcohólico/genética , Factores de Riesgo
8.
Nephrol Ther ; 17S: S23-S26, 2021 Apr.
Artículo en Francés | MEDLINE | ID: mdl-33910694

RESUMEN

Primary hyperoxalurias are rare disease with autosomal recessive inheritance; they often lead to kidney failure and can lead to life-threatening conditions, especially in early onset forms. There are three types, responding to distinct enzyme deficits. Type 1 represents 85% of cases and results from an enzyme deficiency (alanine-glyoxylate aminotransferase) in the peroxisomes of the liver, causing hyperoxaluria leading to urolithiasis with or without nephrocalcinosis. As glomerular filtration decreases, a systemic overload appears and spares no organ. Treatment has hitherto been based on combined liver and kidney transplantation, with significant mortality and morbidity. The recent introduction of interfering RNA treatments opens up new perspectives. By blocking an enzymatic synthesis (glycolate oxidase or lacticodehydrogenase a) upstream of the deficit that causes the disease, oxaluria normalizes and the tolerance of the drug (administered by injection every 1 to 3 months) is good. This strategy will help prevent kidney failure in patients treated early and avoid liver transplantation in those who are diagnosed at an advanced stage of kidney failure.


Asunto(s)
Hiperoxaluria Primaria , Hiperoxaluria , Trasplante de Riñón , Trasplante de Hígado , Humanos , Hiperoxaluria/genética , Hiperoxaluria/terapia , Hiperoxaluria Primaria/genética , Hiperoxaluria Primaria/terapia , ARN
9.
Am J Physiol Renal Physiol ; 320(3): F475-F484, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33491567

RESUMEN

Primary hyperoxaluria type 1 (PH1) is a severe inherited disorder caused by a genetic defect in alanine-glyoxylate aminotransferase (AGXT), which results in recurrent urolithiasis and renal failure. Animal models that precisely reflect human PH1 phenotypes are lacking. We aimed to develop a novel PH1 rat model and study the mechanisms involved in PH1 deterioration. One cell stage Sprague-Dawley embryos were injected with the CRISPR/Cas9 system to introduce a Q84X mutation in Agxt. Liver tissues were harvested to determine Agxt expression. Urine oxalate, crystals, and electrolyte levels in AgxtQ84X and wild-type (WT) littermates were evaluated. Kidney tissues were used for Pizzolato staining and kidney injury evaluation. Data showed that Agxt mRNA and protein were absent in AgxtQ84X rats. At 4 and 24 wk, AgxtQ84X rats displayed 2.1- and 2.9-fold higher urinary oxalate levels, respectively, compared with WT littermates. As a result, calcium oxalate (CaOx) crystals in urine were revealed in all AgxtQ84X rats but in none of the WT rats. We also observed bladder stones in 36.4% of AgxtQ84X rats, of which 44.4% had renal CaOx deposition. Moreover, the elevated serum urea and creatinine levels indicated the impaired renal function in AgxtQ84X rats. Further investigation revealed significantly increased expression of inflammation-, necroptosis-, and fibrosis-related genes in the kidneys of AgxtQ84X rats with spontaneous renal CaOx deposition, indicating that these pathways are involved in PH1 deterioration. Collectively, these results suggest that this rat model has broad applicability in mechanistic studies and innovative therapeutics development for PH1 and other kidney stone diseases.NEW & NOTEWORTHY Primary hyperoxaluria type 1 is a severe inherited disorder that results in recurrent urolithiasis and renal failure. We generated an alanine-glyoxylate aminotransferase (Agxt)Q84X nonsense mutant rat model that displayed an early onset of hyperoxaluria, spontaneous renal CaOx precipitation, bladder stone, and kidney injuries. Our results suggest an interaction of renal CaOx crystals with the activation of inflammation-, fibrosis-, and necroptosis-related pathways. In all, the AgxtQ84X rat strain has broad applicability in mechanistic studies and the development of innovative therapeutics.


Asunto(s)
Hiperoxaluria/metabolismo , Riñón/metabolismo , Nefrocalcinosis/metabolismo , Transaminasas/genética , Animales , Oxalato de Calcio/metabolismo , Hiperoxaluria/genética , Cálculos Renales/sangre , Mutación/genética , Nefrocalcinosis/genética , Oxalatos/metabolismo , Ratas , Insuficiencia Renal/genética , Transaminasas/metabolismo
10.
Nat Prod Res ; 35(6): 1029-1034, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31135219

RESUMEN

Oxidative stress has been identified as an underlying factor in the development of insulin resistance, ß-cell dysfunction, impaired glucose tolerance and type 2 diabetes mellitus and it also play major role in kidney stone formation. The present study is aimed to elucidate the in vitro nephroprotective activity of two isoflavonoid glycosides, tectorigenin 7-O-ß-D-glucosyl-(1→6)-ß-D-glucoside (1) and tectorigenin 7-O-ß-D-glucosyl-4'-O-ß-D-glucoside (2) isolated from the n-BuOH fraction of Iris spuria L. (Zeal) rhizome MeOH extract against oxalate and high glucose-induced oxidative stress in NRK-49F cells. The results revealed that compounds 1 and 2 significantly increased the antioxidant enzyme activities and decreased MDA levels in both oxalate and high glucose stress. Treatment with these phytochemicals effectively down-regulated expression of crystal modulator genes and pro-fibrotic genes in oxalate and high glucose-mediated stress respectively. This study indicates cytoprotective, antioxidant, anti-urolithic and anti-diabetic effects of compounds 1 and 2 against oxalate and high glucose stress.[Figure: see text].


Asunto(s)
Glicósidos/aislamiento & purificación , Glicósidos/uso terapéutico , Hiperglucemia/tratamiento farmacológico , Hiperoxaluria/tratamiento farmacológico , Género Iris/química , Isoflavonas/uso terapéutico , Riñón/patología , Sustancias Protectoras/uso terapéutico , Animales , Antioxidantes/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citoprotección/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/toxicidad , Glicósidos/química , Glicósidos/farmacología , Hiperglucemia/genética , Hiperoxaluria/genética , Isoflavonas/química , Isoflavonas/farmacología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Sustancias Protectoras/química , Sustancias Protectoras/farmacología , Ratas , Especies Reactivas de Oxígeno/metabolismo
11.
J Urol ; 205(5): 1394-1399, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33350326

RESUMEN

PURPOSE: The etiology of calcium-oxalate kidney stone formation remains elusive. Biallelic mutations in HOGA1 are responsible for primary hyperoxaluria type 3 and result in oxalate overproduction and kidney stone disease. Our previous study showed that carriers of HOGA1 mutations have elevated urinary levels of oxalate precursors. In this study we explored the possibility that mutations in HOGA1 confer a dominant phenotype in the form of kidney stone disease or hyperoxaluria. MATERIALS AND METHODS: An observational analytic case control study was designed to determine the prevalence of pathogenic HOGA1 mutations among adults with calcium-oxalate kidney stone disease. Given the high prevalence of HOGA1 mutations among Ashkenazi Jews, this group was evaluated separately. Carrier frequency of any of the 52 reported pathogenic mutations was compared to data derived from gnomAD for the corresponding ethnic group. Sanger sequencing of HOGA1 gene was performed on DNA samples from the following groups: 60 Ashkenazi Jews and 86 nonAshkenazi calcium-oxalate stone formers, 150 subjects with low and 150 with high urinary oxalate levels. RESULTS: The carrier prevalence of pathogenic mutations among the Ashkenazi Jews was 1.7% compared to 2.8% in the corresponding control group (p=0.9 OR=0.6 95% CI 0.01-3.51). We did not detect any mutation among the nonAshkenazi study group. No correlation was detected between hyperoxaluria and HOGA1 variants. CONCLUSIONS: This study shows that mutations in HOGA1 do not confer a dominant phenotype in the form of calcium-oxalate kidney stone disease or hyperoxaluria.


Asunto(s)
Oxalato de Calcio , Hiperoxaluria/genética , Cálculos Renales/genética , Mutación , Oxo-Ácido-Liasas/genética , Fenotipo , Adulto , Anciano , Oxalato de Calcio/análisis , Estudios de Casos y Controles , Estudios de Cohortes , Femenino , Humanos , Cálculos Renales/química , Masculino , Persona de Mediana Edad
13.
J Cell Mol Med ; 23(11): 7268-7278, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31489770

RESUMEN

Hyperoxaluria-induced calcium oxalate (CaOx) deposition is the key factor in kidney stone formation, for which adipose-derived stromal cells (ADSCs) have been used as a therapeutic treatment. Studies revealed that miR-20b-3p is down-regulated in hypercalciuric stone-forming rat kidney. To investigate whether ADSC-derived miR-20b-3p-enriched exosomes protect against kidney stones, an ethylene glycol (EG)-induced hyperoxaluria rat model and an in vitro model of oxalate-induced NRK-52E cells were established to explore the protective mechanism of miR-20b-3p. The results showed that miR-20b-3p levels were decreased following hyperoxaluria in the urine of patients and in kidney tissues from animal models. Furthermore, treatment with miR-20b-3p-enriched exosomes from ADSCs protected EG-induced hyperoxaluria rats, and cell experiments confirmed that co-culture with miR-20b-3p-enriched exosomes alleviated oxalate-induced cell autophagy and the inflammatory response by inhibiting ATG7 and TLR4. In conclusion, ADSC-derived miR-20b-3p-enriched exosomes protected against kidney stones by suppressing autophagy and inflammatory responses.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Oxalato de Calcio/toxicidad , Exosomas/genética , Hiperoxaluria/prevención & control , MicroARNs/administración & dosificación , Células del Estroma/efectos de los fármacos , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Apoptosis , Autofagia , Adhesión Celular , Proliferación Celular , Células Cultivadas , Humanos , Hiperoxaluria/inducido químicamente , Hiperoxaluria/genética , Hiperoxaluria/patología , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Ratas , Ratas Sprague-Dawley , Células del Estroma/metabolismo , Células del Estroma/patología
14.
Am J Physiol Renal Physiol ; 317(1): F137-F151, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31091119

RESUMEN

Interaction of pioglitazone (PGZ) and macrophages (Mps) in renal crystal formation remains unclear. In the present study, we investigated the possible mechanisms involved with Mps of PGZ in suppressing renal crystal formation. Crystal formation in the mouse kidney was detected using polarized light optical microscopy and Pizzolato staining. Gene expression was detected by Western blot analysis, quantitative RT-PCR, immunohistochemistry, immunofluorescence, and ELISA. Mp phenotypes were identified by flow cytometric analysis. Cell apoptosis was detected with TUNEL assay, and tubular injury was detected with periodic acid-Schiff staining. Interaction of peroxisome proliferator-activated receptor (PPAR)-γ and promoter was determined by chromatin immunoprecipitation assay. Luciferase reporter assay was performed to authenticate target genes of miRNA-23 (miR-23). Recombinant adenovirus was used to elucidate the role of miR-23 in vivo. Renal crystal formation, inflammation, tubular injury, and cell apoptosis were significantly marked in glyoxylic acid-treated groups and significantly decreased in PGZ-treated groups. PGZ significantly reduced Mp infiltration and M1 Mp polarization in the kidney. In vitro, PGZ shifted crystal-stimulated M1-predominant Mps to M2-predominant Mps, which were anti-inflammatory. PPAR-γ could directly bind to one PPAR-γ regulatory element in the promoter of pre-miR-23 to promote expression of miR-23 in Mps. We identified two downstream target genes of miR-23, interferon regulatory factor 1 and Pknox1. Moreover, miR-23 decreased crystal deposition, M1 Mp polarization, and injury in the kidney. This study has proven that PGZ decreased renal calcium oxalate crystal formation and renal inflammatory injury by suppressing M1 Mp polarization through a PPAR-γ-miR-23-interferon regulatory factor 1/Pknox1 axis. PGZ is liable to be a potential therapeutic medicine for treating urolithiasis.


Asunto(s)
Antiinflamatorios/farmacología , Oxalato de Calcio/metabolismo , Hiperoxaluria/prevención & control , Riñón/efectos de los fármacos , Macrófagos/efectos de los fármacos , MicroARNs/metabolismo , PPAR gamma/agonistas , Pioglitazona/farmacología , Urolitiasis/prevención & control , Animales , Apoptosis/efectos de los fármacos , Sitios de Unión , Cristalización , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Hiperoxaluria/genética , Hiperoxaluria/metabolismo , Hiperoxaluria/patología , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/metabolismo , Riñón/metabolismo , Riñón/patología , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , MicroARNs/genética , PPAR gamma/genética , PPAR gamma/metabolismo , Fenotipo , Regiones Promotoras Genéticas , Transducción de Señal , Urolitiasis/genética , Urolitiasis/metabolismo , Urolitiasis/patología
15.
Am J Physiol Cell Physiol ; 315(5): C687-C698, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30020825

RESUMEN

Most kidney stones (KS) are composed of calcium oxalate, and small increases in urine oxalate affect the stone risk. Intestinal oxalate secretion mediated by anion exchanger SLC26A6 (PAT1) plays a crucial role in limiting net absorption of ingested oxalate, thereby preventing hyperoxaluria and related KS, reflecting the importance of understanding regulation of intestinal oxalate transport. We previously showed that ATP and UTP inhibit oxalate transport by human intestinal Caco2-BBE cells (C2). Since ATP is rapidly degraded to adenosine (ADO), we examined whether intestinal oxalate transport is regulated by ADO. We measured [14C]oxalate uptake in the presence of an outward Cl gradient as an assay of Cl-oxalate exchange activity, ≥49% of which is PAT1-mediated in C2 cells. We found that ADO significantly inhibited oxalate transport by C2 cells, an effect completely blocked by the nonselective ADO receptor antagonist 8- p-sulfophenyltheophylline. ADO also significantly inhibited oxalate efflux by C2 cells, which is important since PAT1 mediates oxalate efflux in vivo. Using pharmacological antagonists and A2B adenosine receptor (A2B AR) siRNA knockdown studies, we observed that ADO inhibits oxalate transport through the A2B AR, phospholipase C, and PKC. ADO inhibits oxalate transport by reducing PAT1 surface expression as shown by biotinylation studies. We conclude that ADO inhibits oxalate transport by lowering PAT1 surface expression in C2 cells through signaling pathways including the A2B AR, PKC, and phospholipase C. Given higher ADO levels and overexpression of the A2B AR in inflammatory bowel disease (IBD), our findings have potential relevance to pathophysiology of IBD-associated hyperoxaluria and related KS.


Asunto(s)
Adenosina/metabolismo , Sistemas de Transporte de Aminoácidos/genética , Enfermedades Inflamatorias del Intestino/genética , Receptor de Adenosina A2B/genética , Simportadores/genética , Adenosina/administración & dosificación , Antagonistas del Receptor de Adenosina A2/administración & dosificación , Adenosina Trifosfato/metabolismo , Transporte Biológico/genética , Células CACO-2 , Humanos , Hiperoxaluria/genética , Hiperoxaluria/metabolismo , Hiperoxaluria/patología , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Cálculos Renales/genética , Cálculos Renales/metabolismo , Cálculos Renales/patología , Oxalatos/metabolismo , Receptor de Adenosina A2B/metabolismo , Factores de Riesgo , Transducción de Señal/efectos de los fármacos , Teofilina/administración & dosificación , Fosfolipasas de Tipo C/genética
16.
J Mol Med (Berl) ; 96(7): 621-630, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29777253

RESUMEN

Primary hyperoxaluria type 1 is a severe kidney stone disease caused by abnormalities of the peroxisomal alanine-glyoxylate aminotransferase (AGT). The most frequent mutation G170R results in aberrant mitochondrial localization of the active enzyme. To evaluate the population of peroxisome-localized AGT, we developed a quantitative Glow-AGT assay based on the self-assembly split-GFP approach and used it to identify drugs that can correct mislocalization of the mutant protein. In line with previous reports, the Glow-AGT assay showed that mitochondrial transport inhibitors DECA and monensin increased peroxisomal localization of the mutant. Here, we demonstrate that prolonged treatment with the translation elongation inhibitor emetine, a medicinal alkaloid used in treatment of amoebiasis, corrected G170R-AGT mislocalization. Furthermore, emetine reduced the augmented oxalate level in culture media of patient-derived hepatocytes bearing the G170R mutation. A distinct translation inhibitor GC7 had a similar effect on the mutant Glow-AGT relocalization indicating that mild translation inhibition is a promising therapeutic approach for primary hyperoxaluria type 1 caused by AGT misfolding/mistargeting. KEY MESSAGES: • There is no effective conservative treatment to decrease oxalate production in PH1 patients. • Chemical chaperones rescue mislocalization of mutant AGT and reduce oxalate levels. • We have developed an assay for precise monitoring of the peroxisomal AGT. • Inhibition of translation by emetine reroutes the mutant protein to peroxisome. • Mild translation inhibition is a promising cure for conformational disorders.


Asunto(s)
Mutación , Biosíntesis de Proteínas , Transaminasas/genética , Transaminasas/metabolismo , Animales , Biomarcadores , Células CHO , Supervivencia Celular , Cricetulus , Hepatocitos/metabolismo , Hiperoxaluria/tratamiento farmacológico , Hiperoxaluria/genética , Hiperoxaluria/metabolismo , Espacio Intracelular/metabolismo , Mitocondrias/metabolismo , Terapia Molecular Dirigida , Oxalatos/metabolismo , Peroxisomas/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Transporte de Proteínas
17.
SLAS Discov ; 22(7): 887-896, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28346094

RESUMEN

Primary hyperoxaluria is the underlying cause of oxalosis and is a life-threatening autosomal recessive disease, for which treatment may require dialysis or dual liver-kidney transplantation. The most common primary hyperoxaluria type 1 (PH1) is caused by genetic mutations of a liver-specific enzyme alanine:glyoxylate aminotransferase (AGT), which results in the misrouting of AGT from the peroxisomes to the mitochondria. Pharmacoperones are small molecules with the ability to modify misfolded proteins and route them correctly within the cells, which may present an effective strategy to treat AGT misrouting in PH1 disorders. We miniaturized a cell-based high-content assay into 1536-well plate format and screened ~4200 pharmacologically relevant compounds including Food and Drug Administration, European Union, and Japanese-approved drugs. This assay employs CHO cells stably expressing AGT-170, a mutant that predominantly resides in the mitochondria, where we monitor for its relocation to the peroxisomes through automated image acquisition and analysis. The miniaturized 1536-well assay yielded a Z' averaging 0.70 ± 0.07. Three drugs were identified as potential pharmacoperones from this pilot screen, demonstrating the applicability of this assay for large-scale high-throughput screening.


Asunto(s)
Hiperoxaluria/tratamiento farmacológico , Ionóforos/farmacología , Enfermedades Renales/tratamiento farmacológico , Animales , Células CHO , Cricetulus , Evaluación Preclínica de Medicamentos/métodos , Hiperoxaluria/genética , Hiperoxaluria/metabolismo , Hiperoxaluria Primaria/tratamiento farmacológico , Hiperoxaluria Primaria/genética , Hiperoxaluria Primaria/metabolismo , Enfermedades Renales/genética , Enfermedades Renales/metabolismo , Trasplante de Riñón/métodos , Hígado/efectos de los fármacos , Hígado/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Mutación/genética , Peroxisomas/efectos de los fármacos , Peroxisomas/genética , Peroxisomas/metabolismo , Diálisis Renal/métodos , Transaminasas/genética , Transaminasas/metabolismo
18.
World J Urol ; 34(1): 89-95, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25981400

RESUMEN

INTRODUCTION AND OBJECTIVES: Reactive oxygen species (ROS) are produced during the interaction between oxalate/calcium oxalate monohydrate (COM) crystals and renal epithelial cells and are responsible for the various cellular responses through the activation of NADPH oxidase (Nox). Ox and COM also activate the renin-angiotensin-aldosterone system (RAAS). Aldosterone stimulates ROS production through activation of Nox with the involvement of mineralocorticoid receptor (MR), Rac1 and mitogen-activated protein kinases (MAPK). We investigated RAAS pathways in vivo in an animal model of hyperoxaluria and in vitro by exposing renal epithelial cells to COM crystals. METHODS: Hyperoxaluria was induced in male SD rats by administering ethylene glycol. One group of rats was additionally given spironolactone. Total RNA was extracted and subjected to genomic microarrays to obtain global transcriptome data. Normal rat kidney cell line (NRK-52E) was incubated with aldosterone(10(-7) M) and COM(67 µg/cm(2)) with or without spironolactone(10(-5) M), a selective inhibitor of SRC family of kinases; protein phosphatase 2(pp2) (10(-5) M) and Nox inhibitor; diphenylene iodonium (DPI) (10(-5) M). RESULTS: Relative expression of genes encoding for AGT, angiotensin receptors 1b and 2, Renin 1, Cyp11b, HSD11B2, Nr3c2, NOx4 and Rac1 was upregulated in the kidneys of rats with hyperoxaluria. Treatment with spironolactone reversed the effect of hyperoxaluria. Both aldosterone and COM crystals activated Nox and Rac1 expression in NRK52E, while spironolactone inhibited Nox and Rac1 expression. Increased Rac1 expression was significantly attenuated by treatment with PP2 and spironolactone. CONCLUSIONS: Results indicate that hyperoxaluria-induced production of ROS, injury and inflammation are in part associated with the activation of Nox through renin-angiotensin-aldosterone pathway.


Asunto(s)
Oxalato de Calcio/metabolismo , Hiperoxaluria/genética , NADPH Oxidasas/metabolismo , ARN Mensajero/metabolismo , Sistema Renina-Angiotensina/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/efectos de los fármacos , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Angiotensinógeno/efectos de los fármacos , Angiotensinógeno/genética , Angiotensinógeno/metabolismo , Animales , Línea Celular , Citocromo P-450 CYP11B2/efectos de los fármacos , Citocromo P-450 CYP11B2/genética , Citocromo P-450 CYP11B2/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Glicol de Etileno/toxicidad , Perfilación de la Expresión Génica , Hiperoxaluria/inducido químicamente , Hiperoxaluria/metabolismo , Masculino , Antagonistas de Receptores de Mineralocorticoides/farmacología , NADPH Oxidasa 4 , NADPH Oxidasas/efectos de los fármacos , NADPH Oxidasas/genética , Compuestos Onio/farmacología , Proteína Fosfatasa 2/farmacología , ARN Mensajero/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Angiotensina/efectos de los fármacos , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Receptores de Mineralocorticoides/efectos de los fármacos , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo , Renina/efectos de los fármacos , Renina/genética , Renina/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Espironolactona/farmacología , Esteroide 11-beta-Hidroxilasa/efectos de los fármacos , Esteroide 11-beta-Hidroxilasa/genética , Esteroide 11-beta-Hidroxilasa/metabolismo , Proteína de Unión al GTP rac1/efectos de los fármacos , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
19.
PLoS One ; 10(8): e0134764, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26241473

RESUMEN

Enhanced sodium excretion is associated with intrarenal oxidative stress. The present study evaluated whether oxidative stress caused by high sodium (HS) may be involved in calcium oxalate crystal formation. Male rats were fed a sodium-depleted diet. Normal-sodium and HS diets were achieved by providing drinking water containing 0.3% and 3% NaCl, respectively. Rats were fed a sodium-depleted diet with 5% hydroxyl-L-proline (HP) for 7 and 42 days to induce hyperoxaluria and/or calcium oxalate deposition. Compared to normal sodium, HS slightly increased calcium excretion despite diuresis; however, the result did not reach statistical significance. HS did not affect the hyperoxaluria, hypocalciuria or supersaturation caused by HP; however, it increased calcium oxalate crystal deposition soon after 7 days of co-treatment. Massive calcium oxalate formation and calcium crystal excretion in HS+HP rats were seen after 42 days of treatment. HP-mediated hypocitraturia was further exacerbated by HS. Moreover, HS aggravated HP-induced renal injury and tubular damage via increased apoptosis and oxidative stress. Increased urinary malondialdehyde excretion, in situ superoxide production, NAD(P)H oxidase and xanthine oxidase expression and activity, and decreased antioxidant enzyme expression or activity in the HS+HP kidney indicated exaggerated oxidative stress. Interestingly, this redox imbalance was associated with reduced renal osteopontin and Tamm-Horsfall protein expression (via increased excretion) and sodium-dependent dicarboxylate cotransporter NaDC-1 upregulation. Collectively, our results demonstrate that a HS diet induces massive crystal formation in the hyperoxaluric kidney; this is not due to increased urinary calcium excretion but is related to oxidative injury and loss of anticrystallization defense.


Asunto(s)
Oxalato de Calcio/química , Hiperoxaluria/metabolismo , Cálculos Renales/etiología , Túbulos Renales/metabolismo , Natriuresis/fisiología , Estrés Oxidativo/efectos de los fármacos , Sodio en la Dieta/farmacología , Animales , Antioxidantes/metabolismo , Biomarcadores , Citratos/orina , Creatinina/orina , Cristalización , Transportadores de Ácidos Dicarboxílicos/genética , Transportadores de Ácidos Dicarboxílicos/fisiología , Dieta Hiposódica , Diuresis/efectos de los fármacos , Inducción Enzimática , Regulación de la Expresión Génica , Hidroxiprolina/toxicidad , Hiperoxaluria/inducido químicamente , Hiperoxaluria/genética , Cálculos Renales/metabolismo , Cálculos Renales/orina , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Masculino , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Transportadores de Anión Orgánico Sodio-Dependiente/fisiología , Osteopontina/genética , Osteopontina/fisiología , Ratas , Ratas Wistar , Sodio en la Dieta/administración & dosificación , Sodio en la Dieta/toxicidad , Superóxidos/metabolismo , Simportadores/genética , Simportadores/fisiología , Uromodulina/genética , Uromodulina/fisiología
20.
J Oral Maxillofac Surg ; 73(11): 2142-8, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26071361

RESUMEN

Chronic renal failure can give rise to a wide spectrum of oral manifestations, owing mainly to secondary hyperparathyroidism complicating this disease. However, any systemic disease responsible for kidney failure can produce oral manifestations, which can be misdiagnosed. This report describes the case of a 40-year-old male patient referred for oral assessment before kidney and liver transplantation. He had primary hyperoxaluria complicated by end-stage renal failure and secondary hyperparathyroidism. Panoramic radiography indicated not only external root resorption, but also maxillary and mandibular radiolucencies consistent with brown tumors. Unexpectedly, histologic study of the bone biopsy specimen led to the diagnosis of jaws oxalosis. Primary hyperoxaluria is a systemic genetic disease. The affected genes are involved in glyoxylate metabolism and their deficiency results in overproduction of oxalates. Inability of the kidney to excrete oxalates leads to deposition of these crystals in almost all tissues (oxalosis) and to multiple-organ failure. Several oral findings have been described in patients with oxalosis, such as periodontal disease and root resorptions, but radiolucencies in the jaws have rarely been described. This case report is of particular interest because of the unusual location of oxalate crystal deposition in the jaws, which could be misdiagnosed in a patient with renal failure and secondary hyperparathyroidism.


Asunto(s)
Hiperoxaluria/complicaciones , Fallo Renal Crónico/complicaciones , Enfermedades Estomatognáticas/complicaciones , Adulto , Humanos , Hiperoxaluria/genética , Masculino , Radiografía Panorámica , Enfermedades Estomatognáticas/diagnóstico por imagen
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...