Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
J Clin Endocrinol Metab ; 107(3): 813-824, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-34636899

RESUMO

PURPOSE: In X-linked hypophosphatemia (XLH), excess fibroblast growth factor-23 causes hypophosphatemia and low calcitriol, leading to musculoskeletal disease with clinical consequences. XLH treatment options include conventional oral phosphate with active vitamin D, or monotherapy with burosumab, a monoclonal antibody approved to treat children and adults with XLH. We have previously reported outcomes up to 64 weeks, and here we report safety and efficacy follow-up results up to 160 weeks from an open-label, multicenter, randomized, dose-finding trial of burosumab for 5- to 12-year-old children with XLH. METHODS: After 1 week of conventional therapy washout, patients were randomized 1:1 to burosumab every 2 weeks (Q2W) or every 4 weeks (Q4W) for 64 weeks, with dosing titrated based on fasting serum phosphorus levels between baseline and week 16. From week 66 to week 160, all patients received Q2W burosumab. RESULTS: Twenty-six children were randomized initially into each Q2W and Q4W group and all completed treatment to week 160. In 41 children with open distal femoral and proximal tibial growth plates (from both treatment groups), total Rickets Severity Score significantly decreased by 0.9 ±â€…0.1 (least squares mean ±â€…SE; P < 0.0001) from baseline to week 160. Fasting serum phosphorus increases were sustained by burosumab therapy throughout the study, with an overall population mean (SD) of 3.35 (0.39) mg/dL, within the pediatric normal range (3.2-6.1 mg/dL) at week 160 (mean change from baseline P < 0.0001). Most adverse events were mild to moderate in severity. MAIN CONCLUSIONS: In children with XLH, burosumab administration for 160 weeks improved phosphate homeostasis and rickets and was well-tolerated. Long-term safety was consistent with the reported safety profile of burosumab. CLINICALTRIALS.GOV: NCT02163577.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Raquitismo Hipofosfatêmico Familiar/tratamento farmacológico , Fator de Crescimento de Fibroblastos 23/antagonistas & inibidores , Anticorpos Monoclonais Humanizados/efeitos adversos , Criança , Pré-Escolar , Relação Dose-Resposta a Droga , Raquitismo Hipofosfatêmico Familiar/sangue , Raquitismo Hipofosfatêmico Familiar/diagnóstico , Feminino , Fator de Crescimento de Fibroblastos 23/metabolismo , Humanos , Masculino , Fosfatos/sangue , Fosfatos/metabolismo , Reabsorção Renal/efeitos dos fármacos , Índice de Gravidade de Doença
2.
Am J Physiol Renal Physiol ; 321(5): F645-F655, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34605273

RESUMO

Fine tuning of Na+ reabsorption takes place along the aldosterone-sensitive distal nephron, which includes the collecting duct (CD), where it is mainly regulated by aldosterone. In the CD, Na+ reabsorption is mediated by the epithelial Na+ channel and Na+ pump (Na+-K+-ATPase). Paracellular ion permeability is mainly dependent on tight junction permeability. Claudin-8 is one of the main tight junction proteins expressed along the aldosterone-sensitive distal nephron. We have previously shown a coupling between transcellular Na+ reabsorption and paracellular Na+ barrier. We hypothesized that aldosterone controls the expression levels of both transcellular Na+ transporters and paracellular claudin-8 in a coordinated manner. Here, we show that aldosterone increased mRNA and protein levels as well as lateral membrane localization of claudin-8 in cultured CD principal cells. The increase in claudin-8 mRNA levels in response to aldosterone was prevented by preincubation with 17-hydroxyprogesterone, a mineralocorticoid receptor antagonist, and by inhibition of transcription with actinomycin D. We also showed that a low-salt diet, which stimulated aldosterone secretion, was associated with increased claudin-8 abundance in the mouse kidney. Reciprocally, mice subjected to a high-salt diet, which inhibits aldosterone secretion, or treated with spironolactone, a mineralocorticoid receptor antagonist, displayed decreased claudin-8 expression. Inhibition of glycogen synthase kinase-3, Lyn, and Abl signaling pathways prevented the effect of aldosterone on claudin-8 mRNA and protein abundance, suggesting that signaling of protein kinases plays a permissive role on the transcriptional activity of the mineralocorticoid receptor. This study shows that signaling via multiple protein kinases working in concert mediates aldosterone-induced claudin-8 expression in the CD.NEW & NOTEWORTHY In this study, we showed that aldosterone modulates claudin-8 expression in cultured collecting duct principal cells and in the mouse kidney. The upregulation of claudin-8 expression in response to aldosterone is dependent on at least glycogen synthase kinase-3, Lyn, and Abl signaling pathways, indicating the participation of multiple protein kinases to the effect of aldosterone.


Assuntos
Aldosterona/farmacologia , Claudinas/metabolismo , Túbulos Renais Coletores/efeitos dos fármacos , Reabsorção Renal/efeitos dos fármacos , Sódio/metabolismo , Animais , Linhagem Celular , Claudinas/genética , Dieta Hipossódica , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/metabolismo , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/metabolismo , Camundongos , Antagonistas de Receptores de Mineralocorticoides/farmacologia , Inibidores da Síntese de Ácido Nucleico/farmacologia , Proteínas Proto-Oncogênicas c-abl/genética , Proteínas Proto-Oncogênicas c-abl/metabolismo , Sódio na Dieta/toxicidade , Transcrição Gênica , Regulação para Cima , Quinases da Família src/genética , Quinases da Família src/metabolismo
3.
Sci Rep ; 11(1): 9191, 2021 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-33911165

RESUMO

Anticholinergics, therapeutic agents for overactive bladder, are clinically suggested to reduce urine output. We investigated whether this effect is due to bladder or kidney urine reabsorption. Various solutions were injected into the bladder of urethane-anesthetized SD rats. The absorption rate for 2 h was examined following the intravenous administration of the anticholinergics imidafenacin (IM), atropine (AT), and tolterodine (TO). The bilateral ureter was then canulated and saline was administered to obtain a diuretic state. Anticholinergics or 1-deamino-[8-D-arginine]-vasopressin (dDAVP) were intravenously administered. After the IM and dDAVP administrations, the rat kidneys were immunostained with AQP2 antibody, and intracellular cAMP was measured. The absorption rate was ~ 10% of the saline injected into the bladder and constant even when anticholinergics were administered. The renal urine among peaked 2 h after the saline administration. Each of the anticholinergics significantly suppressed the urine production in a dose-dependent manner, as did dDAVP. IM and dDAVP increased the intracellular cAMP levels and caused the AQP2 molecule to localize to the collecting duct cells' luminal side. The urinary reabsorption mechanism through the bladder epithelium was not activated by anticholinergic administration. Thus, anticholinergics suppress urine production via an increase in urine reabsorption in the kidneys' collecting duct cells via AQP2.


Assuntos
Antagonistas Colinérgicos/farmacologia , Rim/efeitos dos fármacos , Reabsorção Renal/efeitos dos fármacos , Animais , Antidiuréticos/efeitos adversos , Antidiuréticos/farmacologia , Aquaporina 2/metabolismo , AMP Cíclico/metabolismo , Desamino Arginina Vasopressina/efeitos adversos , Desamino Arginina Vasopressina/farmacologia , Eletrólitos/metabolismo , Feminino , Rim/metabolismo , Concentração Osmolar , Ratos Sprague-Dawley , Reabsorção Renal/fisiologia , Sódio/urina , Bexiga Urinária/efeitos dos fármacos , Micção/efeitos dos fármacos
4.
Mar Drugs ; 18(12)2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-33271765

RESUMO

This study was designed to investigate the effects and underlying mechanisms of Astaxanthin (AST) on high-fructose-induced hyperuricemia (HUA) from the perspectives of the uric acid (UA) synthesis and excretion in rat models. Following six weeks of a 10% fructose diet, the level of serum UA effectively decreased in the AST groups as compared to the model group. The enzymatic activities of xanthine oxidase (XOD) and adenosine deaminase (ADA) were significantly inhibited, and the mRNA expression levels of XOD and ADA significantly decreased after the AST administration. These results suggested that the AST reduced UA synthesis by inhibiting the mRNA expressions and enzyme activities of XOD and ADA, thereby contributing to HUA improvement. On the hand, the relative expressions of the mRNA and protein of kidney reabsorption transport proteins (GLUT9 and URAT1) were significantly down-regulated by AST, while that of the kidney secretion proteins (OAT1, OAT3 and ABCG2) were significantly up-regulated by AST. These results indicated that the AST promoted UA excretion by regulating the urate transport proteins, and thus alleviated HUA. This study suggested that the AST could serve as an effective alternative to traditional medicinal drugs for the prevention of fructose-induced HUA.


Assuntos
Inibidores de Adenosina Desaminase/farmacologia , Adenosina Desaminase/metabolismo , Hiperuricemia/prevenção & controle , Proteínas de Membrana Transportadoras/efeitos dos fármacos , Ácido Úrico/sangue , Xantina Oxidase/antagonistas & inibidores , Adenosina Desaminase/genética , Animais , Biomarcadores/sangue , Biomarcadores/urina , Modelos Animais de Doenças , Frutose , Hiperuricemia/induzido quimicamente , Hiperuricemia/enzimologia , Rim/efeitos dos fármacos , Rim/metabolismo , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Ratos Sprague-Dawley , Reabsorção Renal/efeitos dos fármacos , Ácido Úrico/urina , Xantina Oxidase/genética , Xantina Oxidase/metabolismo , Xantofilas/farmacologia
5.
Nutrients ; 12(6)2020 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-32486008

RESUMO

The beneficial effects of fatty acids (FAs) on human health have attracted widespread interest. However, little is known about the impact of FAs on the handling of urate, the end-product of human purine metabolism, in the body. Increased serum urate levels occur in hyperuricemia, a disease that can lead to gout. In humans, urate filtered by the glomerulus of the kidney is majorly re-absorbed from primary urine into the blood via the urate transporter 1 (URAT1)-mediated pathway. URAT1 inhibition, thus, contributes to decreasing serum urate concentration by increasing net renal urate excretion. Here, we investigated the URAT1-inhibitory effects of 25 FAs that are commonly contained in foods or produced in the body. For this purpose, we conducted an in vitro transport assay using cells transiently expressing URAT1. Our results showed that unsaturated FAs, especially long-chain unsaturated FAs, inhibited URAT1 more strongly than saturated FAs. Among the tested unsaturated FAs, eicosapentaenoic acid, α-linolenic acid, and docosahexaenoic acid exhibited substantial URAT1-inhibitory activities, with half maximal inhibitory concentration values of 6.0, 14.2, and 15.2 µM, respectively. Although further studies are required to investigate whether the ω-3 polyunsaturated FAs can be employed as uricosuric agents, our findings further confirm FAs as nutritionally important substances influencing human health.


Assuntos
Ácidos Graxos Ômega-3/farmacologia , Glomérulos Renais/metabolismo , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Transportadores de Ânions Orgânicos/fisiologia , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Proteínas de Transporte de Cátions Orgânicos/fisiologia , Reabsorção Renal/efeitos dos fármacos , Ácido Úrico/metabolismo , Células Cultivadas , Ácidos Docosa-Hexaenoicos/farmacologia , Relação Dose-Resposta a Droga , Ácido Eicosapentaenoico/farmacologia , Humanos , Hiperuricemia/sangue , Eliminação Renal/efeitos dos fármacos , Ácido Úrico/sangue , Ácido alfa-Linolênico/farmacologia
6.
Am J Physiol Renal Physiol ; 318(4): F870-F877, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31984792

RESUMO

Adenosine plays an important role in various aspects of kidney physiology, but the specific targets and mechanisms of actions are not completely understood. The collecting duct has the highest expression of adenosine receptors, particularly adenosine A1 receptors (A1Rs). Interstitial adenosine levels are greatly increased up to a micromolar range in response to dietary salt loading. We have previously shown that the basolateral membrane of principal cells has primarily K+ conductance mediated by Kir4.1/5.1 channels to mediate K+ recycling and to set up a favorable driving force for Na+/K+ exchange (47). Intercalated cells express the Cl- ClC-K2/b channel mediating transcellular Cl- reabsorption. Using patch-clamp electrophysiology in freshly isolated mouse collecting ducts, we found that acute application of adenosine reversely inhibits ClC-K2/b open probability from 0.31 ± 0.04 to 0.17 ± 0.06 and to 0.10 ± 0.05 for 1 and 10 µM, respectively. In contrast, adenosine (10 µM) had no measureable effect on Kir4.1/5.1 channel activity in principal cells. The inhibitory effect of adenosine on ClC-K2/b was abolished in the presence of the A1R blocker 8-cyclopentyl-1,3-dipropylxanthine (10 µM). Consistently, application of the A1R agonist N6-cyclohexyladenosine (1 µM) recapitulated the inhibitory action of adenosine on ClC-K2/b open probability. The effects of adenosine signaling in the collecting duct were independent from its purinergic counterpartner, ATP, having no measurable actions on ClC-K2/b and Kir4.1/5.1. Overall, we demonstrated that adenosine selectively inhibits ClC-K2/b activity in intercalated cells by targeting A1Rs. We propose that inhibition of transcellular Cl- reabsorption in the collecting duct by adenosine would aid in augmenting NaCl excretion during high salt intake.


Assuntos
Agonistas do Receptor A1 de Adenosina/farmacologia , Adenosina/farmacologia , Proteínas de Transporte de Ânions/antagonistas & inibidores , Canais de Cloreto/antagonistas & inibidores , Cloretos/metabolismo , Túbulos Renais Coletores/efeitos dos fármacos , Receptor A1 de Adenosina/efeitos dos fármacos , Reabsorção Renal/efeitos dos fármacos , Animais , Proteínas de Transporte de Ânions/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Canais de Cloreto/metabolismo , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Receptor A1 de Adenosina/metabolismo
7.
Pediatr Nephrol ; 35(10): 1843-1854, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31392510

RESUMO

Hypophosphatemic rickets is caused by renal phosphate wasting that is most commonly due to X-linked dominant mutations in PHEX. PHEX mutations cause hypophosphatemia indirectly, through the increased expression of fibroblast growth factor 23 (FGF23) by osteocytes. FGF23 decreases renal phosphate reabsorption and thereby increases phosphate excretion. The lack of phosphate leads to a mineralization defect at the level of growth plates (rickets), bone tissue (osteomalacia), and teeth, where the defect facilitates the formation of abscesses. The bone tissue immediately adjacent to osteocytes often remains unmineralized ("periosteocytic lesions"), highlighting the osteocyte defect in this disorder. Common clinical features of XLH include deformities of the lower extremities, short stature, enthesopathies, dental abscesses, as well as skull abnormalities such as craniosynostosis and Chiari I malformation. For the past four decades, XLH has been treated by oral phosphate supplementation and calcitriol, which improves rickets and osteomalacia and the dental manifestations, but often does not resolve all aspects of the mineralization defects. A newer treatment approach using inactivating FGF23 antibodies leads to more stable control of serum inorganic phosphorus levels and seems to heal rickets more reliably. However, the long-term benefits of FGF23 antibody treatment remain to be elucidated.


Assuntos
Raquitismo Hipofosfatêmico Familiar/patologia , Fatores de Crescimento de Fibroblastos/metabolismo , Osteomalacia/patologia , Endopeptidase Neutra Reguladora de Fosfato PHEX/genética , Fosfatos/metabolismo , Absorciometria de Fóton , Desenvolvimento Ósseo/efeitos dos fármacos , Desenvolvimento Ósseo/genética , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/patologia , Calcificação Fisiológica/efeitos dos fármacos , Calcificação Fisiológica/genética , Calcitriol/administração & dosagem , Raquitismo Hipofosfatêmico Familiar/diagnóstico , Raquitismo Hipofosfatêmico Familiar/tratamento farmacológico , Raquitismo Hipofosfatêmico Familiar/genética , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Humanos , Osteócitos/metabolismo , Osteomalacia/diagnóstico , Osteomalacia/tratamento farmacológico , Osteomalacia/genética , Endopeptidase Neutra Reguladora de Fosfato PHEX/metabolismo , Comunicação Parácrina/genética , Fosfatos/administração & dosagem , Fosfatos/sangue , Reabsorção Renal/efeitos dos fármacos , Reabsorção Renal/genética , Dente/crescimento & desenvolvimento , Dente/patologia , Resultado do Tratamento
8.
Biochem Biophys Res Commun ; 517(3): 413-420, 2019 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-31378369

RESUMO

GLUT9 is generally considered to be associated with the uric acid transport, which plays an important role in the regulation of serum uric acid level. In this study, the expression level of miR-143-3p was significantly decreased in hyperuricemia mice model group compared with the normal control by miRNA microarray, the same results were confirmed in the hyperuricemia patients and the healthy control group. It is predicted that GLUT9 may be the target gene of miR-143-3p by target scan and other net-software. GLUT9 as the downstream target gene of miR-143-3p was determinated by fluorescence enzyme activity assay. Western blotting and qRT-PCR indicated that the expression of GLUT9 in human renal tubular epithelial cells transfected with miR-143-3p mimics was significantly reduced. Meanwhile inflammatory factors IL-1ß and MCP-1 significantly decreased. In conclusion, miR-143-3p can reduce uric acid reabsorption by inhibiting its downstream target gene GLUT9.


Assuntos
Proteínas Facilitadoras de Transporte de Glucose/genética , Hiperuricemia/genética , Córtex Renal/metabolismo , MicroRNAs/genética , Ácido Úrico/sangue , Animais , Sequência de Bases , Estudos de Casos e Controles , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Humanos , Hiperuricemia/sangue , Hiperuricemia/induzido quimicamente , Hiperuricemia/fisiopatologia , Hipoxantina/administração & dosagem , Inflamação , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Córtex Renal/efeitos dos fármacos , Córtex Renal/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , Proteínas de Transporte de Cátions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Ácido Oxônico/administração & dosagem , Reabsorção Renal/efeitos dos fármacos , Reabsorção Renal/fisiologia , Transdução de Sinais
9.
Toxicol Lett ; 311: 27-36, 2019 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-31039415

RESUMO

Methotrexate (MTX) is a derivate of folic acid, commonly used as an anchor drug for the treatment and management of malignant diseases and autoimmune disorders. However, nephrotoxicity is an important drawback of MTX therapy. Unfortunately, there are not enough studies reporting the nature of the renal failure induced by MTX. Thus, the aim of this study was to evaluate the time course of renal handling of water and electrolytes in male Wistar rats, after the exposure to a unique dose of MTX (80 mg/kg b.w.). Experiments were carried out at day 2, day 4, day 8 and day 14 after MTX administration. Several parameters of kidney function related to water and electrolytes handling were evaluated. Renal expression and urinary excretion of aquaporin-2 (AQP2) and Na-K-2Cl-cotransporter (NKCC2) were determined by Western blotting. MTX produced alterations on water handling on the second day after treatment, showing a significant increase in solute free water reabsorption which might be mediated by the increased expression of AQP2 in apical membranes. On the other hand, MTX produced alterations on electrolytes handling on the fourth day after treatment, showing a significant decrease of sodium chloride excretion, mediated at least in part, by the increase renal expression of NKCC2. These results provide valuable information to clinical practice in order to be able to find therapeutic targets that diminish adverse effects and health deterioration. Moreover, MTX treatment altered AQP2 and NKCC2 urinary excretion allowing postulating these transporters as potential biomarkers of MTX induced nephrotoxicity.


Assuntos
Aquaporina 2/metabolismo , Eletrólitos/metabolismo , Nefropatias/induzido quimicamente , Túbulos Renais/efeitos dos fármacos , Metotrexato/toxicidade , Reabsorção Renal/efeitos dos fármacos , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Água/metabolismo , Animais , Biomarcadores/metabolismo , Cloretos/metabolismo , Nefropatias/metabolismo , Túbulos Renais/metabolismo , Masculino , Potássio/metabolismo , Ratos Wistar , Sódio/metabolismo , Fatores de Tempo , Urodinâmica/efeitos dos fármacos
10.
Toxicol Appl Pharmacol ; 363: 47-56, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30465787

RESUMO

Clozapine, an atypical antipsychotic drug, is widely utilized for the treatment of schizophrenia; however, clozapine-induced metabolic disorders, such as fatty liver and weight gain, warrant increased attention. Considering the crucial role of l-carnitine (L-Car) in fatty acid oxidation and carnitine/organic cation transporter (OCTN) 2 in renal reabsorption of L-Car, we aimed to study whether clozapine-induced liver lipid metabolic disorder is associated with L-Car dysregulation via inhibition/down-regulation of renal OCTN2. Our results reveal that clozapine inhibits L-Car uptake in MDCK-hOCTN2 cells with an IC50 value of 1.78 µM. Additionally, clozapine significantly reduces the uptake of L-Car in HK-2 cells, mouse primary cultured proximal tubular (mPCPT) cells and HepG2 cells. Acute (intraperitoneal injection) and 21-day successive oral administration of clozapine at 12.5, 25, and 50 mg/kg to mice resulted in 2-3-fold greater renal excretion of L-Car than in the vehicle group, and the concentration of L-Car in plasma and liver was significantly decreased. Concomitantly, mRNA and protein levels of mOctn2 in the kidney were markedly down regulated. Additionally, 28-day oral administration of clozapine induced increased triglyceride (TG) and total cholesterol (TCHO) levels in mouse livers, while L-Car (40 mg/kg - 1 g/kg) attenuated clozapine-induced liver TG and TCHO increase in a dose-dependent manner. These results indicate that clozapine-induced reduction of L-Car reabsorption via inhibition/down-regulation of renal OCTN2 contributes to liver lipid metabolic disorder. L-Car supplementation is probably an effective strategy to attenuate clozapine-induced abnormal lipid metabolism.


Assuntos
Antipsicóticos/efeitos adversos , Carnitina/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Clozapina/efeitos adversos , Membro 5 da Família 22 de Carreadores de Soluto/metabolismo , Animais , Linhagem Celular , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Modelos Animais de Doenças , Cães , Regulação para Baixo/efeitos dos fármacos , Células Hep G2 , Humanos , Concentração Inibidora 50 , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos ICR , Cultura Primária de Células , Reabsorção Renal/efeitos dos fármacos , Esquizofrenia/tratamento farmacológico , Testes de Toxicidade Aguda , Testes de Toxicidade Crônica
11.
Am J Physiol Renal Physiol ; 316(2): F253-F262, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30427219

RESUMO

Hypertonicity increases water permeability, independently of vasopressin, in the inner medullary collecting duct (IMCD) by increasing aquaporin-2 (AQP2) membrane accumulation. We investigated whether protein kinase C (PKC) and adenosine monophosphate kinase (AMPK) are involved in hypertonicity-regulated water permeability. Increasing perfusate osmolality from 150 to 290 mosmol/kgH2O and bath osmolality from 290 to 430 mosmol/kgH2O significantly stimulated osmotic water permeability. The PKC inhibitors chelerythrine (10 µM) and rottlerin (50 µM) significantly reversed the increase in osmotic water permeability stimulated by hypertonicity in perfused rat terminal IMCDs. Chelerythrine significantly increased phosphorylation of AQP2 at S261 but not at S256. Previous studies show that AMPK is stimulated by osmotic stress. We tested AMPK phosphorylation under hypertonic conditions. Hypertonicity significantly increased AMPK phosphorylation in inner medullary tissues. Blockade of AMPK with Compound C decreased hypertonicity-stimulated water permeability but did not alter phosphorylation of AQP2 at S256 and S261. AICAR, an AMPK stimulator, caused a transient increase in osmotic water permeability and increased phosphorylation of AQP2 at S256. When inner medullary tissue was treated with the PKC activator phorbol dibutyrate (PDBu), the AMPK activator metformin, or both, AQP2 phosphorylation at S261 was decreased with PDBu or metformin alone, but there was no additive effect on phosphorylation with PDBu and metformin together. In conclusion, hypertonicity regulates water reabsorption by activating PKC. Hypertonicity-stimulated water reabsorption by PKC may be related to the decrease in endocytosis of AQP2. AMPK activation promotes water reabsorption, but the mechanism remains to be determined. PKC and AMPK do not appear to act synergistically to regulate water reabsorption.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Água Corporal/metabolismo , Túbulos Renais Coletores/efeitos dos fármacos , Proteína Quinase C/metabolismo , Reabsorção Renal/efeitos dos fármacos , Solução Salina Hipertônica/farmacologia , Animais , Aquaporina 2/metabolismo , Endocitose , Feminino , Túbulos Renais Coletores/enzimologia , Masculino , Concentração Osmolar , Osmorregulação , Permeabilidade , Fosforilação , Ratos
12.
Am J Physiol Renal Physiol ; 316(1): F173-F185, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30427222

RESUMO

Inhibitors of the Na+-glucose cotransporter SGLT2 enhance urinary glucose and urate excretion and lower plasma urate levels. The mechanisms remain unclear, but a role for enhanced glucose in the tubular fluid, which may interact with tubular urate transporters, such as the glucose transporter GLUT9 or the urate transporter URAT1, has been proposed. Studies were performed in nondiabetic mice treated with the SGLT2 inhibitor canagliflozin and in gene-targeted mice lacking the urate transporter Glut9 in the tubule or in mice with whole body knockout of Sglt2, Sglt1, or Urat1. Renal urate handling was assessed by analysis of urate in spontaneous plasma and urine samples and normalization to creatinine concentrations or by renal clearance studies with assessment of glomerular filtration rate by FITC-sinistrin. The experiments confirmed the contribution of URAT1 and GLUT9 to renal urate reabsorption, showing a greater contribution of the latter and additive effects. Genetic and pharmacological inhibition of SGLT2 enhanced fractional renal urate excretion (FE-urate), indicating that a direct effect of the SGLT2 inhibitor on urate transporters is not absolutely necessary. Consistent with a proposed role of increased luminal glucose delivery, the absence of Sglt1, which by itself had no effect on FE-urate, enhanced the glycosuric and uricosuric effects of the SGLT2 inhibitor. The SGLT2 inhibitor enhanced renal mRNA expression of Glut9 in wild-type mice, but tubular GLUT9 seemed dispensable for the increase in FE-urate in response to canagliflozin. First evidence is presented that URAT1 is required for the acute uricosuric effect of the SGLT2 inhibitor in mice.


Assuntos
Canagliflozina/farmacologia , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Túbulos Renais Proximais/efeitos dos fármacos , Transportadores de Ânions Orgânicos/metabolismo , Eliminação Renal/efeitos dos fármacos , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Transportador 2 de Glucose-Sódio/efeitos dos fármacos , Ácido Úrico/urina , Uricosúricos/farmacologia , Animais , Genótipo , Proteínas Facilitadoras de Transporte de Glucose/deficiência , Proteínas Facilitadoras de Transporte de Glucose/genética , Túbulos Renais Proximais/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transportadores de Ânions Orgânicos/deficiência , Transportadores de Ânions Orgânicos/genética , Fenótipo , Reabsorção Renal/efeitos dos fármacos , Transportador 2 de Glucose-Sódio/deficiência , Transportador 2 de Glucose-Sódio/genética , Transportador 2 de Glucose-Sódio/metabolismo
13.
Drug Metab Dispos ; 47(2): 104-113, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30442650

RESUMO

Lesinurad [Zurampic; 2-(5-bromo-4-(4-cyclopropylnaphthalen-1-yl)-4H-1,2,4-triazol-3-ylthio)], a selective inhibitor of uric acid reabsorption transporters approved for the treatment of gout, is a racemate of two atropisomers. The objective of this investigation was to evaluate the stereoselectivity of metabolism, the inhibitory potency on kidney uric acid reabsorption transporters (URAT1 and OAT4), and the clinical pharmacokinetics of the lesinurad atropisomers. Incubations with human liver microsomes (HLM), recombinant CYP2C9, and recombinant CYP3A4 were carried out to characterize the stereoselective formation of three metabolites: M3 (hydroxylation), M4 (a dihydrodiol metabolite), and M6 (S-dealkylation). The formation of M3 in HLM with atropisomer 1 was approximately twice as much as that with atropisomer 2, whereas formation of M4 with atropisomer 1 was 8- to 12-fold greater than that with atropisomer 2. There were no significant differences in the plasma protein binding among lesinurad and the atropisomers. Following oral administration of 400 mg lesinurad once daily for 14 days to healthy human volunteers, the systemic exposure (C max at steady state and area under the concentration-time curve from time zero to the time of dosing interval) of atropisomer 1 was approximately 30% lower than that of atropisomer 2, whereas renal clearance was similar. In vitro cell-based assays using HEK293 stable cells expressing URAT1 and OAT4 demonstrated that atropisomer 2 was approximately 4-fold more potent against URAT1 than atropisomer 1 and equally active against OAT4. In conclusion, lesinurad atropisomers showed stereoselectivity in clinical pharmacokinetics, metabolism, and inhibitory potency against URAT1.


Assuntos
Transportadores de Ânions Orgânicos/antagonistas & inibidores , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Reabsorção Renal/efeitos dos fármacos , Tioglicolatos/farmacologia , Triazóis/farmacologia , Ácido Úrico/metabolismo , Uricosúricos/farmacologia , Administração Oral , Adulto , Gota/tratamento farmacológico , Células HEK293 , Voluntários Saudáveis , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Masculino , Microssomos Hepáticos , Pessoa de Meia-Idade , Transportadores de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/antagonistas & inibidores , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Estereoisomerismo , Relação Estrutura-Atividade , Tioglicolatos/química , Tioglicolatos/metabolismo , Tioglicolatos/uso terapêutico , Triazóis/química , Triazóis/metabolismo , Triazóis/uso terapêutico , Ácido Úrico/sangue , Ácido Úrico/urina , Uricosúricos/química , Uricosúricos/metabolismo , Uricosúricos/uso terapêutico , Adulto Jovem
14.
Nutrients ; 10(9)2018 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-30200571

RESUMO

Dietary fructose causes salt-sensitive hypertension. Proximal tubules (PTs) reabsorb 70% of the filtered NaCl. Angiotensin II (Ang II), atrial natriuretic peptide (ANP) and norepinephrine (NE) regulate this process. Although Ang II signaling blockade ameliorates fructose-induced salt-sensitive hypertension, basal PT Na⁺ reabsorption and its sensitivity to the aforementioned factors have not been studied in this model. We hypothesized consuming fructose with a high-salt diet selectively enhances the sensitivity of PT transport to Ang II. We investigated the effects of Ang II, ANP and NE on PT Na reabsorption in rats fed a high-salt diet drinking tap water (HS) or 20% fructose (HS-FRU). Oxygen consumption (QO2) was used as a measure of all ATP-dependent transport processes. Na⁺/K⁺-ATPase and Na⁺/H⁺-exchange (NHE) activities were studied because they represent primary apical and basolateral transporters in this segment. The effect of 10-12 mol/L Ang II in QO2 by PTs from HS-FRU was larger than HS (p < 0.02; n = 7). In PTs from HS-FRU 10-12 mol/L Ang II stimulated NHE activity by 2.6 ± 0.7 arbitrary fluorescence units/s (p < 0.01; n = 5) but not in those from HS. The stimulatory effect of Ang II on PT Na⁺/K⁺-ATPase activity was not affected by HS-FRU. Responses of QO2 and NHE activity to ANP did not differ between groups. The response of QO2 to NE was unaltered by HS-FRU. We concluded that the sensitivity of PT Na⁺ reabsorption specifically to Ang II is enhanced by HS-FRU. This maintains high rates of transport even in the presence of low concentrations of the peptide, and likely contributes to the hypertension.


Assuntos
Angiotensina II/farmacologia , Açúcares da Dieta , Frutose , Hipertensão/metabolismo , Túbulos Renais Proximais/efeitos dos fármacos , Reabsorção Renal/efeitos dos fármacos , Cloreto de Sódio na Dieta , Sódio/metabolismo , Animais , Fator Natriurético Atrial/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Modelos Animais de Doenças , Hipertensão/induzido quimicamente , Hipertensão/fisiopatologia , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/fisiopatologia , Masculino , Norepinefrina/farmacologia , Consumo de Oxigênio/efeitos dos fármacos , Ratos Sprague-Dawley , Trocadores de Sódio-Hidrogênio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo
15.
Nutrients ; 10(10)2018 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-30241394

RESUMO

A chronic magnesium deficiency may be one of the causes of lifestyle-related diseases such as hypertension and diabetes. Serum Mg2+ concentration is strictly controlled by the reabsorption pathway in the renal tubules, but little is known about how Mg2+ reabsorption is upregulated. We searched for food compounds which can increase the expression levels of Mg2+ transport carriers including transient receptor potential melastatin 6 (TRPM6) channel and cyclin M2 (CNNM2). Sodium citrate (SC) increased the mRNA levels of TRPM6 and CNNM2 in renal tubular epithelial NRK-52E cells. The SC-induced elevation of TRPM6 was inhibited by U0126, a mitogen-activated protein kinase kinase (MEK) inhibitor, but the CNNM2 was not. SC increased the levels of p-ERK1/2 and p-c-Fos, which were inhibited by U0126. SC induced alkalization of culture medium. Both SC and alkalization enhanced Mg2+ influx, which was inhibited by U0126 and introduction of TRPM6 siRNA. The reporter activity of TRPM6 was increased by SC and alkalization, which was suppressed by mutation in an AP-1-binding site. The SC-induced elevation of p-ERK1/2 and p-EGFR was inhibited by diphenylene iodonium, a nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitor, and erlotinib, an epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor. SC did not change the level of acetyl histone H3, but increased the association of c-Fos with the promoter region of TRPM6. These results suggest that SC increases TRPM6 expression and Mg2+ influx mediated by the activation of NADPH oxidase and an EGFR/ERK/c-Fos pathway in the renal tubules.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Células Epiteliais/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Túbulos Renais/efeitos dos fármacos , MAP Quinase Quinase Quinases/metabolismo , Magnésio/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Citrato de Sódio/farmacologia , Canais de Cátion TRPM/metabolismo , Animais , Sítios de Ligação , Proteínas de Transporte de Cátions/genética , Linhagem Celular , Células Epiteliais/enzimologia , Túbulos Renais/enzimologia , Cinética , NADPH Oxidases/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Ratos , Reabsorção Renal/efeitos dos fármacos , Transdução de Sinais , Canais de Cátion TRPM/genética , Regulação para Cima
16.
Am J Physiol Renal Physiol ; 315(3): F711-F725, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29741098

RESUMO

Metabolic acidosis is the most common acid-base disorder in septic patients and is associated with increased mortality. Previously, we demonstrated that sepsis induced by cecal ligation and puncture (CLP) impairs [Formula: see text] absorption in the medullary thick ascending limb (MTAL) by 1) decreasing the intrinsic [Formula: see text] absorptive capacity and 2) enhancing inhibition of [Formula: see text] absorption by LPS through upregulation of Toll-like receptor (TLR) 4 signaling. Both effects depend on ERK activation. Monophosphoryl lipid A (MPLA) is a detoxified TLR4 agonist that enhances innate antimicrobial immunity and improves survival following sepsis. Pretreatment of MTALs with MPLA in vitro prevents LPS inhibition of [Formula: see text] absorption. Here we examined whether pretreatment with MPLA would protect the MTAL against sepsis. Vehicle or MPLA was administered to mice 48 h before sham or CLP surgery, and MTALs were studied in vitro 18 h postsurgery. Pretreatment with MPLA prevented the effects of sepsis to decrease the basal [Formula: see text] absorption rate and enhance inhibition by LPS. These protective effects were mediated through MPLA stimulation of a Toll/IL-1 receptor domain-containing adaptor-inducing IFN-ß-(TRIF)-dependent phosphatidylinositol 3-kinase-Akt pathway that prevents sepsis- and LPS-induced ERK activation. The effects of MPLA to improve MTAL [Formula: see text] absorption were associated with marked improvement in plasma [Formula: see text] concentration, supporting a role for the kidneys in the pathogenesis of sepsis-induced metabolic acidosis. These studies support detoxified TLR4-based immunomodulators, such as MPLA, that enhance antimicrobial responses as a safe and effective approach to prevent or treat sepsis-induced renal tubule dysfunction and identify cell signaling pathways that can be targeted to preserve MTAL [Formula: see text] absorption and attenuate metabolic acidosis during sepsis.


Assuntos
Acidose/prevenção & controle , Bicarbonatos/metabolismo , Lipídeo A/análogos & derivados , Alça do Néfron/efeitos dos fármacos , Reabsorção Renal/efeitos dos fármacos , Sepse/tratamento farmacológico , Equilíbrio Ácido-Base/efeitos dos fármacos , Acidose/metabolismo , Acidose/fisiopatologia , Proteínas Adaptadoras de Transporte Vesicular/deficiência , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Bicarbonatos/sangue , Bicarbonatos/urina , Modelos Animais de Doenças , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Lipídeo A/farmacologia , Alça do Néfron/metabolismo , Alça do Néfron/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sepse/metabolismo , Sepse/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/agonistas
17.
J Am Soc Nephrol ; 29(6): 1706-1719, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29773687

RESUMO

BackgroundNedd4-2 is an E3 ubiquitin-protein ligase that associates with transport proteins, causing their ubiquitylation, and then internalization and degradation. Previous research has suggested a correlation between Nedd4-2 and BP. In this study, we explored the effect of intercalated cell (IC) Nedd4-2 gene ablation on IC transporter abundance and function and on BP.Methods We generated IC Nedd4-2 knockout mice using Cre-lox technology and produced global pendrin/Nedd4-2 null mice by breeding global Nedd4-2 null (Nedd4-2-/- ) mice with global pendrin null (Slc26a4-/- ) mice. Mice ate a diet with 1%-4% NaCl; BP was measured by tail cuff and radiotelemetry. We measured transepithelial transport of Cl- and total CO2 and transepithelial voltage in cortical collecting ducts perfused in vitro Transporter abundance was detected with immunoblots, immunohistochemistry, and immunogold cytochemistry.Results IC Nedd4-2 gene ablation markedly increased electroneutral Cl-/HCO3- exchange in the cortical collecting duct, although benzamil-, thiazide-, and bafilomycin-sensitive ion flux changed very little. IC Nedd4-2 gene ablation did not increase the abundance of type B IC transporters, such as AE4 (Slc4a9), H+-ATPase, barttin, or the Na+-dependent Cl-/HCO3- exchanger (Slc4a8). However, IC Nedd4-2 gene ablation increased CIC-5 total protein abundance, apical plasma membrane pendrin abundance, and the ratio of pendrin expression on the apical membrane to the cytoplasm. IC Nedd4-2 gene ablation increased BP by approximately 10 mm Hg. Moreover, pendrin gene ablation eliminated the increase in BP observed in global Nedd4-2 knockout mice.Conclusions IC Nedd4-2 regulates Cl-/HCO3- exchange in ICs., Nedd4-2 gene ablation increases BP in part through its action in these cells.


Assuntos
Pressão Sanguínea/genética , Canais Epiteliais de Sódio/metabolismo , Transporte de Íons/genética , Ubiquitina-Proteína Ligases Nedd4/genética , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Amilorida/análogos & derivados , Amilorida/farmacologia , Animais , Bicarbonatos/metabolismo , Membrana Celular/metabolismo , Canais de Cloreto/metabolismo , Antiportadores de Cloreto-Bicarbonato/metabolismo , Cloretos/metabolismo , Troca Iônica , Túbulos Renais Coletores/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , ATPases Translocadoras de Prótons/metabolismo , Prótons , Reabsorção Renal/efeitos dos fármacos , Simportadores de Sódio-Bicarbonato/metabolismo , Transportadores de Sulfato/genética , Transportadores de Sulfato/metabolismo , Tiazidas/farmacologia
18.
Am J Physiol Renal Physiol ; 314(2): F190-F195, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28971992

RESUMO

The nephron segments in the inner medulla are part of the urine concentrating mechanism. Depending on the diuretic state, they are facing a large range of extracellular osmolality. We investigated whether water homeostasis affects tubular transport and permeability properties in inner medullary descending thin limb (IMdTL) and ascending thin limb (IMaTL). Three experimental groups of rats under different diuretic states were investigated on metabolic cages: waterload, furosemide-induced diuresis, and control (antidiuresis). Urine production and osmolalities reflected the 3-day treatment. To functionally investigate tubular epithelial properties, we performed experiments in freshly isolated inner medullary thin limbs from these animals. Tubular segments were acutely dissected and investigated for trans- and paracellular properties by in vitro perfusion and electrophysiological analysis. IMdTL and IMaTL were distinguished by morphological criteria. We confirmed absence of transepithelial electrogenic transport in thin limbs. Although diffusion potential measurements showed no differences between treatments in IMdTLs, we observed increased paracellular cation selectivity under waterload in IMaTLs. NaCl diffusion potential was -5.64 ± 1.93 mV under waterload, -1.99 ± 1.72 mV under furosemide-induced diuresis, and 0.27 ± 0.40 mV under control. The corresponding permeability ratio PNa/Cl was 1.53 ± 0.21 (waterload), 1.22 ± 0.18 (furosemide-induced diuresis), and 0.99 ± 0.02 (control), respectively. Claudins are main constituents of the tight junction responsible for paracellular selectivity; however, immunofluorescence did not show qualitative differences in claudin 4, 10, and 16 localization. Our results show that IMaTLs change tight junction properties in response to diuretic state to allow adaptation of NaCl reabsorption.


Assuntos
Diurese/efeitos dos fármacos , Diuréticos/farmacologia , Ingestão de Líquidos , Células Epiteliais/efeitos dos fármacos , Furosemida/farmacologia , Alça do Néfron/efeitos dos fármacos , Junções Íntimas/efeitos dos fármacos , Água/metabolismo , Animais , Claudinas/metabolismo , Difusão , Células Epiteliais/metabolismo , Feminino , Alça do Néfron/metabolismo , Masculino , Concentração Osmolar , Permeabilidade , Ratos Sprague-Dawley , Reabsorção Renal/efeitos dos fármacos , Cloreto de Sódio/urina , Junções Íntimas/metabolismo
19.
Nat Commun ; 8(1): 1920, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29203863

RESUMO

Impaired albumin reabsorption by proximal tubular epithelial cells (PTECs) has been highlighted in diabetic nephropathy (DN), but little is known about the underlying molecular mechanisms. Here we find that ORAI1-3, are preferentially expressed in PTECs and downregulated in patients with DN. Hyperglycemia or blockade of insulin signaling reduces the expression of ORAI1-3. Inhibition of ORAI channels by BTP2 and diethylstilbestrol or silencing of ORAI expression impairs albumin uptake. Transgenic mice expressing a dominant-negative Orai1 mutant (E108Q) increases albuminuria, and in vivo injection of BTP2 exacerbates albuminuria in streptozotocin-induced and Akita diabetic mice. The albumin endocytosis is Ca2+-dependent and accompanied by ORAI1 internalization. Amnionless (AMN) associates with ORAIs and forms STIM/ORAI/AMN complexes after Ca2+ store depletion. STIM1/ORAI1 colocalizes with clathrin, but not with caveolin, at the apical membrane of PTECs, which determines clathrin-mediated endocytosis. These findings provide insights into the mechanisms of protein reabsorption and potential targets for treating diabetic proteinuria.


Assuntos
Albuminas/metabolismo , Albuminúria/genética , Canais de Cálcio/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Nefropatias Diabéticas/metabolismo , Células Epiteliais/metabolismo , Túbulos Renais Proximais/metabolismo , Proteína ORAI1/genética , Proteína ORAI2/genética , Albuminas/efeitos dos fármacos , Albuminúria/metabolismo , Anilidas/farmacologia , Animais , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio , Canais de Cálcio/metabolismo , Estudos de Casos e Controles , Caveolinas/metabolismo , Linhagem Celular , Clatrina/metabolismo , Dietilestilbestrol/farmacologia , Regulação para Baixo , Endocitose , Células Epiteliais/efeitos dos fármacos , Estrogênios não Esteroides/farmacologia , Feminino , Humanos , Hiperglicemia/genética , Hiperglicemia/metabolismo , Rim/metabolismo , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/efeitos dos fármacos , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Mutação , Proteína ORAI1/antagonistas & inibidores , Proteína ORAI1/metabolismo , Proteína ORAI2/antagonistas & inibidores , Proteína ORAI2/metabolismo , Reabsorção Renal/efeitos dos fármacos , Reabsorção Renal/genética , Molécula 1 de Interação Estromal/metabolismo , Tiadiazóis/farmacologia
20.
PLoS One ; 12(10): e0184330, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29020011

RESUMO

In the kidney, the renal proximal tubule (PT) reabsorbs solutes into the peritubular capillaries through active transport. Here, we replicate this reabsorptive function in vitro by engineering a microfluidic PT. The microfluidic PT architecture comprises a porous membrane with user-defined submicron surface topography separating two microchannels representing a PT filtrate lumen and a peritubular capillary lumen. Human PT epithelial cells and microvascular endothelial cells in respective microchannels created a PT-like reabsorptive barrier. Co-culturing epithelial and endothelial cells in the microfluidic architecture enhanced viability, metabolic activity, and compactness of the epithelial layer. The resulting tissue expressed tight junctions, kidney-specific morphology, and polarized expression of kidney markers. The microfluidic PT actively performed sodium-coupled glucose transport, which could be modulated by administration of a sodium-transport inhibiting drug. The microfluidic PT reproduces human physiology at the cellular and tissue levels, and measurable tissue function which can quantify kidney pharmaceutical efficacy and toxicity.


Assuntos
Túbulos Renais Proximais/metabolismo , Microfluídica/métodos , Reabsorção Renal , Técnicas de Cocultura , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Glucose/análogos & derivados , Humanos , Imageamento Tridimensional , Túbulos Renais Proximais/efeitos dos fármacos , Modelos Teóricos , Ouabaína/farmacologia , Reabsorção Renal/efeitos dos fármacos , Sódio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA