Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Food Funct ; 13(2): 891-903, 2022 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-34994761

RESUMO

A high-salt (HS) diet leads to metabolic disorders in Dahl salt-sensitive (SS) rats, and promotes the development of hypertension. According to the changes in the metabolites of SS rats, a set of combined dietary supplements containing amino acids and organic acids (AO) were designed. The purpose of the present study was to evaluate the effect of AO supplementation on the blood pressure of SS rats after the HS diet and clarify the mechanism of AO by metabolomics and biochemical analyses. The results showed that AO supplementation avoided the elevation of blood pressure induced by the HS diet in SS rats, increased the renal antioxidant enzyme activities (catalase, superoxide dismutase, glutathione reductase, and glutathione S-transferase), reduced the H2O2 and MDA levels, and restored the normal antioxidant status of the serum and kidneys. AO also reversed the decrease in the nitric oxide (NO) levels and NO synthase activity induced by the HS feed, which involved the L-arginine/NO pathway. Metabolomics analysis showed that AO administration increased the levels of amino acids such as cysteine, glycine, hypotaurine, and lysine in the renal medulla and the levels of leucine, isoleucine, and serine in the renal cortex. Of note, lysine, hypotaurine and glycine had higher metabolic centrality in the metabolic correlation network of the renal medulla after AO administration. In conclusion, AO intervention could prevent HS diet-induced hypertension in SS rats by restoring the metabolic homeostasis of the kidneys. Hence, AO has the potential to become a functional food additive to improve salt-sensitive hypertension.


Assuntos
Aminoácidos/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Hipertensão/induzido quimicamente , Cloreto de Sódio na Dieta/administração & dosagem , Aminoácidos/química , Animais , Suplementos Nutricionais , Glutationa/metabolismo , Hipertensão/prevenção & controle , Córtex Renal/efeitos dos fármacos , Córtex Renal/metabolismo , Medula Renal/efeitos dos fármacos , Medula Renal/metabolismo , Masculino , Ratos , Ratos Endogâmicos Dahl , Cloreto de Sódio na Dieta/efeitos adversos
2.
Sci Rep ; 11(1): 23452, 2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34873190

RESUMO

Diabetic nephropathy (DN), the leading cause of end-stage renal disease, has become a massive global health burden. Despite considerable efforts, the underlying mechanisms have not yet been comprehensively understood. In this study, a systematic approach was utilized to identify the microRNA signature in DN and to introduce novel drug targets (DTs) in DN. Using microarray profiling followed by qPCR confirmation, 13 and 6 differentially expressed (DE) microRNAs were identified in the kidney cortex and medulla, respectively. The microRNA-target interaction networks for each anatomical compartment were constructed and central nodes were identified. Moreover, enrichment analysis was performed to identify key signaling pathways. To develop a strategy for DT prediction, the human proteome was annotated with 65 biochemical characteristics and 23 network topology parameters. Furthermore, all proteins targeted by at least one FDA-approved drug were identified. Next, mGMDH-AFS, a high-performance machine learning algorithm capable of tolerating massive imbalanced size of the classes, was developed to classify DT and non-DT proteins. The sensitivity, specificity, accuracy, and precision of the proposed method were 90%, 86%, 88%, and 89%, respectively. Moreover, it significantly outperformed the state-of-the-art (P-value ≤ 0.05) and showed very good diagnostic accuracy and high agreement between predicted and observed class labels. The cortex and medulla networks were then analyzed with this validated machine to identify potential DTs. Among the high-rank DT candidates are Egfr, Prkce, clic5, Kit, and Agtr1a which is a current well-known target in DN. In conclusion, a combination of experimental and computational approaches was exploited to provide a holistic insight into the disorder for introducing novel therapeutic targets.


Assuntos
Nefropatias Diabéticas/tratamento farmacológico , Aprendizado de Máquina , Biologia de Sistemas , Algoritmos , Animais , Química Farmacêutica/métodos , Análise por Conglomerados , Biologia Computacional/métodos , Desenho de Fármacos , Epigênese Genética , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Saúde Global , Humanos , Córtex Renal/efeitos dos fármacos , Medula Renal/efeitos dos fármacos , Modelos Lineares , Masculino , Camundongos , Camundongos Endogâmicos DBA , MicroRNAs/genética , Análise em Microsséries , Análise de Sequência com Séries de Oligonucleotídeos , Análise de Componente Principal , Análise de Regressão , Transdução de Sinais , Máquina de Vetores de Suporte
3.
Cells ; 9(4)2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32295252

RESUMO

Aldosterone indirectly regulates water reabsorption in the distal tubule by regulating sodium reabsorption. However, the direct effect of aldosterone on vasopressin-regulated water and urea permeability in the rat inner medullary collecting duct (IMCD) has not been tested. We investigated whether aldosterone regulates osmotic water permeability in isolated perfused rat IMCDs. Adding aldosterone (500 nM) to the bath significantly decreased osmotic water permeability in the presence of vasopressin (50 pM) in both male and female rat IMCDs. Aldosterone significantly decreased aquaporin-2 (AQP2) phosphorylation at S256 but did not change it at S261. Previous studies show that aldosterone can act both genomically and non-genomically. We tested the mechanism by which aldosterone attenuates osmotic water permeability. Blockade of gene transcription with actinomycin D did not reverse aldosterone-attenuated osmotic water permeability. In addition to AQP2, the urea transporter UT-A1 contributes to vasopressin-regulated urine concentrating ability. We tested aldosterone-regulated urea permeability in vasopressin-treated IMCDs. Blockade of gene transcription did not reverse aldosterone-attenuated urea permeability. In conclusion, aldosterone directly regulates water reabsorption through a non-genomic mechanism. Aldosterone-attenuated water reabsorption may be related to decreased trafficking of AQP2 to the plasma membrane. There may be a sex difference apparent in the inhibitory effect of aldosterone on water reabsorption in the inner medullary collecting duct. This study is the first to show a direct effect of aldosterone to inhibit vasopressin-stimulated osmotic water permeability and urea permeability in perfused rat IMCDs.


Assuntos
Aldosterona/uso terapêutico , Transporte Biológico/fisiologia , Medula Renal/efeitos dos fármacos , Túbulos Renais Coletores/efeitos dos fármacos , Proteínas de Membrana Transportadoras/metabolismo , Vasopressinas/efeitos adversos , Aldosterona/farmacologia , Animais , Células Cultivadas , Feminino , Masculino , Ratos
4.
Food Funct ; 10(2): 849-858, 2019 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-30681096

RESUMO

In the present study, the renal-protective effect of hawthorn fruit extract (HW) on high-salt hypertension and its effect on metabolic patterns are determined. High salt causes hypertension in Dahl salt sensitive (SS) rats, while HW can effectively attenuate high-salt induced hypertension, and, various antihypertensive ingredients of HW have also been successfully identified using GC/MS. Of note, the biochemical assay indicates that HW significantly increases the concentration of nitric oxide (NO) and decreases the concentration of H2O2 and malonaldehyde. Especially, HW increases the activities of NO synthase and catalase in the renal medulla. Simultaneously, the renal cortex and medulla, harvested from SS rats, are used to perform the metabolomics analysis, and then, 11 and 8 differential metabolites are identified in the renal medulla and cortex with the HW gavage, respectively. All differential metabolites are then used to perform the pathway enrichment analysis. The results show that many metabolic pathways are enriched in both the renal medulla and cortex, especially those in the medulla including 23 enriched pathways. Therefore, it provides evidence that HW confers an antioxidant effect on high-salt induced hypertension and dramatically alters the metabolic patterns of SS rats, and the antihypertensive ingredients of HW also further indicate that it may be used as a nutritional supplemental therapeutic drug to protect against high-salt induced hypertension in the renal medulla.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Crataegus/química , Frutas/química , Extratos Vegetais/farmacologia , Animais , Catalase/genética , Catalase/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Medula Renal/efeitos dos fármacos , Medula Renal/enzimologia , Masculino , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Estresse Oxidativo , Extratos Vegetais/química , Distribuição Aleatória , Ratos , Ratos Endogâmicos Dahl , Cloreto de Sódio na Dieta/toxicidade
5.
J Appl Toxicol ; 33(7): 685-94, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23619997

RESUMO

Renal toxicity is the principal health concern after uranium exposure. Children are particularly vulnerable to uranium exposure; with contact with depleted uranium in war zones or groundwater contamination the most likely exposure scenarios. To investigate renal sensitivity to uranium exposure during development, we examined uranium distribution and uranium-induced apoptosis in the kidneys of neonate (7-day-old), prepubertal (25-day-old) and adult (70-day-old) male Wistar rats. Mean renal uranium concentrations increased with both age-at-exposure and exposure level after subcutaneous administration of uranium acetate (UA) (0.1-2 mg kg(-1) body weight). Although less of the injected uranium was deposited in the kidneys of the two younger rat groups, the proportion of the peak uranium content remaining in the kidneys after 2 weeks declined with age-at-exposure, suggesting reduced clearance in younger animals. In situ high-energy synchrotron radiation X-ray fluorescence analysis revealed site-specific accumulation of uranium in the S3 segment of the proximal tubules, distributed in the inner cortex and outer stripe of the outer medulla. Apoptosis and cell loss in the proximal tubules increased with age-at-exposure to 0.5 mg kg(-1) UA. Surprisingly, prepubertal rats were uniquely sensitive to uranium-induced lethality from the higher exposure levels. Observations of increased apoptosis in generating/re-generating tubules particularly in prepubertal rats could help to explain their high mortality rate. Together, our findings suggest that age-at-exposure and exposure level are important parameters for uranium toxicity; uranium tends to persist in developing kidneys after low-level exposures, although renal toxicity is more pronounced in adults.


Assuntos
Rim/crescimento & desenvolvimento , Compostos Organometálicos/toxicidade , Envelhecimento/fisiologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Feminino , Glutamato-Amônia Ligase/metabolismo , Marcação In Situ das Extremidades Cortadas , Rim/efeitos dos fármacos , Rim/metabolismo , Medula Renal/efeitos dos fármacos , Medula Renal/patologia , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/patologia , Compostos Organometálicos/farmacocinética , Gravidez , Ratos , Ratos Wistar , Espectrometria por Raios X , Síncrotrons , Distribuição Tecidual , Urânio/análise , Urânio/metabolismo
6.
Inflammopharmacology ; 21(2): 147-59, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22644380

RESUMO

The up-regulation of angiotensin II AT1 receptors has been implicated as a major mediator in the development of hypertension and progressive nephropathy in experimental diabetes. In spite of the documented potential of garlic treatments in ameliorating diabetic complications, the possible involvement of the angiotensin II AT1 receptor, as a central target for the anti-diabetic potential of garlic, has not been explored. Three groups of rats were studied after 8 weeks following diabetes induction: normal, streptozotocin-induced diabetic (control diabetic), and garlic-treated diabetic rats. A polyclonal antibody of proven specificity to the AT1 receptor, as verified by western blotting, indicated in immunohistochemical assays that AT1 receptor labeling was significantly increased in adrenal and renal tissues of control diabetic rats compared to the normal group. The increased AT1 receptor labeling involved all cortical zones and medullary chromaffin cells of the adrenal gland. Except for glomerulii, increased AT1 receptor labeling was also evident in proximal convoluted tubules in the renal cortex, and all tubular segments and interstitial cells outlining the vasa recta bundles in the inner stripe of the outer renal medulla. Compared with control diabetic rats, the labeling of the AT1 receptor in the garlic-treated diabetic group was significantly reduced throughout adrenal and renal regions to levels comparable to those observed in normal rats. The capacity of garlic to modulate diabetes-induced AT1 receptor up-regulation may be implicated in reversing the detrimental consequences of excessive Ang II signaling, manifested by the development of hypertension and nephropathy.


Assuntos
Glândulas Suprarrenais/efeitos dos fármacos , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Alho , Medula Renal/efeitos dos fármacos , Túbulos Renais Proximais/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/biossíntese , Glândulas Suprarrenais/metabolismo , Animais , Glicemia/efeitos dos fármacos , Glicemia/genética , Glicemia/metabolismo , Células Cromafins/efeitos dos fármacos , Células Cromafins/metabolismo , Diabetes Mellitus Experimental/genética , Regulação para Baixo/efeitos dos fármacos , Medula Renal/metabolismo , Túbulos Renais Proximais/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Regulação para Cima/efeitos dos fármacos
7.
PLoS One ; 7(10): e47738, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23091645

RESUMO

PURPOSE: We have previously shown that production of reactive oxygen species (ROS) is an important contributor to renal injury and inflammation following exposure to oxalate (Ox) or calcium-oxalate (CaOx) crystals. The present study was conducted, utilizing global transcriptome analyses, to determine the effect of Apocynin on changes in the NADPH oxidase system activated in kidneys of rats fed a diet leading to hyperoxaluria and CaOx crystal deposition. APPROACH: Age-, sex- and weight-matched rats were either fed regular rat chow or regular rat chow supplemented with 5% w/w hydroxy-L-proline (HLP). Half of the rats on the HLP diet were also placed on Apocynin-supplemented H(2)O. After 28 days, each rat was euthanized, their kidneys freshly explanted and dissected to obtain both cortex and medulla tissues. Total RNA was extracted from each tissue and subjected to genomic microarrays to obtain global transcriptome data. KEGG was used to identify gene clusters with differentially expressed genes. Immunohistochemistry was used to confirm protein expressions of selected genes. RESULTS: Genes encoding both membrane- and cytosolic-NADPH oxidase complex-associated proteins, together with p21rac and Rap1a, were coordinately up-regulated significantly in both renal medulla and cortex tissues in the HLP-fed rats compared to normal healthy untreated controls. Activation of NADPH oxidase appears to occur via the angiotensin-II/angiotensin-II receptor-2 pathway, although the DAG-PKC pathway of neutrophils may also contribute. Immuno histochemical staining confirmed up-regulated gene expressions. Simultaneously, genes encoding ROS scavenger proteins were down-regulated. HLP-fed rats receiving Apocynin had a complete reversal in the differential-expression of the NADPH oxidase system genes, despite showing similar levels of hyperoxaluria. CONCLUSIONS: A strong up-regulation of an oxidative/respiratory burst involving the NADPH oxidase system, activated via the angiotensin-II and most likely the DAG-PKC pathways, occurs in kidneys of hyperoxaluric rats. Apocynin treatment reversed this activation without affecting the levels of hyperoxaluria.


Assuntos
Acetofenonas/farmacologia , Hiperoxalúria/genética , Rim/efeitos dos fármacos , Rim/metabolismo , NADPH Oxidases/genética , Transcrição Gênica , Animais , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Hiperoxalúria/metabolismo , Rim/patologia , Córtex Renal/efeitos dos fármacos , Córtex Renal/metabolismo , Medula Renal/efeitos dos fármacos , Medula Renal/metabolismo , Masculino , NADPH Oxidases/metabolismo , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Transdução de Sinais
8.
Am J Physiol Renal Physiol ; 301(1): F94-100, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21511694

RESUMO

The effects of TNF gene deletion on renal Na(+)-K(+)-2Cl(-) cotransporter (NKCC2) expression and activity were determined. Outer medulla from TNF(-/-) mice exhibited a twofold increase in total NKCC2 protein expression compared with wild-type (WT) mice. This increase was not observed in TNF(-/-) mice treated with recombinant human TNF (hTNF) for 7 days. Administration of hTNF had no effect on total NKCC2 expression in WT mice. A fourfold increase in NKCC2A mRNA accumulation was observed in outer medulla from TNF(-/-) compared with WT mice; NKCC2F and NKCC2B mRNA accumulation was similar between genotypes. The increase in NKCC2A mRNA accumulation was attenuated when TNF(-/-) mice were treated with hTNF. Bumetanide-sensitive O(2) consumption, an in vitro correlate of NKCC2 activity, was 2.8 ± 0.2 nmol·min(-1)·mg(-1) in medullary thick ascending limb tubules from WT, representing ∼40% of total O(2) consumption, whereas, in medullary thick ascending limb tubules from TNF(-/-) mice, it was 5.6 ± 0.3 nmol·min(-1)·mg(-1), representing ∼60% of total O(2) consumption. Administration of hTNF to TNF(-/-) mice restored the bumetanide-sensitive component to ∼30% of total O(2) consumption. Ambient urine osmolality was higher in TNF(-/-) compared with WT mice (2,072 ± 104 vs. 1,696 ± 153 mosmol/kgH(2)O, P < 0.05). The diluting ability of the kidney, assessed by measuring urine osmolality before and after 1 h of water loading also was greater in TNF(-/-) compared with WT mice (174 ± 38 and 465 ± 81 mosmol/kgH(2)O, respectively, P < 0.01). Collectively, these findings suggest that TNF plays a role as an endogenous inhibitor of NKCC2 expression and function.


Assuntos
Medula Renal/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Animais , Western Blotting , Cloretos/metabolismo , Fragmentação do DNA , DNA Complementar/biossíntese , Indicadores e Reagentes , Isomerismo , Capacidade de Concentração Renal/fisiologia , Testes de Função Renal , Medula Renal/citologia , Medula Renal/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Concentração Osmolar , Consumo de Oxigênio/efeitos dos fármacos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Simportadores de Cloreto de Sódio-Potássio/genética , Membro 1 da Família 12 de Carreador de Soluto , Fator de Necrose Tumoral alfa/genética
9.
Cytoskeleton (Hoboken) ; 68(3): 188-203, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21360831

RESUMO

Cilia are motile and sensory organelles with critical roles in physiology. Ciliary defects can cause numerous human disease symptoms including polycystic kidneys, hydrocephalus, and retinal degeneration. Despite the importance of these organelles, their assembly and function is not fully understood. The unicellular green alga Chlamydomonas reinhardtii has many advantages as a model system for studies of ciliary assembly and function. Here we describe our initial efforts to build a chemical-biology toolkit to augment the genetic tools available for studying cilia in this organism, with the goal of being able to reversibly perturb ciliary function on a rapid time-scale compared to that available with traditional genetic methods. We screened a set of 5520 compounds from which we identified four candidate compounds with reproducible effects on flagella at nontoxic doses. Three of these compounds resulted in flagellar paralysis and one induced flagellar shortening in a reversible and dose-dependent fashion, accompanied by a reduction in the speed of intraflagellar transport. This latter compound also reduced the length of cilia in mammalian cells, hence we named the compound "ciliabrevin" due to its ability to shorten cilia. This compound also robustly and reversibly inhibited microtubule movement and retrograde actin flow in Drosophila S2 cells. Ciliabrevin may prove especially useful for the study of retrograde actin flow at the leading edge of cells, as it slows the retrograde flow in a tunable dose-dependent fashion until flow completely stops at high concentrations, and these effects are quickly reversed upon washout of the drug.


Assuntos
Benzimidazóis/farmacologia , Benzilaminas/farmacologia , Movimento Celular/efeitos dos fármacos , Chlamydomonas/citologia , Chlamydomonas/efeitos dos fármacos , Cílios/metabolismo , Flagelos/efeitos dos fármacos , Medula Renal/efeitos dos fármacos , Traqueia/efeitos dos fármacos , Actinas/metabolismo , Animais , Movimento Celular/fisiologia , Células Cultivadas , Chlamydomonas/fisiologia , Cílios/efeitos dos fármacos , Citoesqueleto/metabolismo , Avaliação Pré-Clínica de Medicamentos , Flagelos/metabolismo , Humanos , Medula Renal/citologia , Medula Renal/metabolismo , Camundongos , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Traqueia/citologia , Traqueia/metabolismo
10.
Steroids ; 75(11): 760-5, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20580730

RESUMO

The Na-K-2Cl cotransporter (NKCC2) regulates sodium transport along the thick ascending limb of Henle's loop and is important in control of sodium balance, renal concentrating ability and renin release. To determine if there are sex differences in NKCC2 abundance and/or distribution, and to evaluate the contribution of ovarian hormones to any such differences, we performed semiquantitative immunoblotting and immunoperoxidase immunohistochemistry for NKCC2 in the kidney of Sprague Dawley male, female and ovariectomized (OVX) rats with and without 17-beta estradiol or progesterone supplementation. Intact females demonstrated greater NKCC2 protein in homogenates of whole kidney (334+/-29%), cortex (219+/-20%) and outer medulla (133+/-9%) compared to males. Ovarian hormone supplementation to OVX rats regulated NKCC2 in the outer medulla only, with NKCC2 protein abundance decreasing slightly in response to progesterone but increasing in response to 17-beta estradiol. Immunohistochemistry demonstrated prominent NKCC2 labeling in the apical membrane of thick ascending limb cells. Kidney section NKCC2 labeling confirmed regionalized regulation of NKCC2 by ovarian hormones. Localized regulation of NKCC2 by ovarian hormones may have importance in controlling sodium and water balance over the lifetime of women as the milieu of sex hormones varies.


Assuntos
Bumetanida/farmacologia , Estradiol/farmacologia , Ovário/metabolismo , Progesterona/farmacologia , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Animais , Feminino , Immunoblotting , Córtex Renal/citologia , Córtex Renal/efeitos dos fármacos , Córtex Renal/metabolismo , Medula Renal/citologia , Medula Renal/efeitos dos fármacos , Medula Renal/metabolismo , Masculino , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Caracteres Sexuais , Membro 1 da Família 12 de Carreador de Soluto
11.
J Ethnopharmacol ; 128(2): 433-7, 2010 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-20083182

RESUMO

AIM OF THE STUDY: Zhuling, sclerotia of Polyporus umbellatus FRIES, a Traditional Chinese Medicine, has long been used as a diuretic. The aim of the present study was to evaluate the diuretic effect on the urinary electrolyte concentration (Na(+), K(+), and Cl(-)) and regulation of the relative mRNA expression of aquaporin-1 (AQP1), aquaporin-2 (AQP2), aquaporin-3 (AQP3) and vasopressin V(2) receptor (V(2)R) post-oral administration of sclerotia of Polyporus umbellata aqueous extract in normal rats. MATERIALS AND METHODS: Aqueous extract of sclerotia of Polyporus umbellatus (50 mg/kg, 250 mg/kg, 500 mg/kg) or the reference drug, furosemide (10mg/kg) were administrated orally to male SD rats and their urine output was quantified and collected 24h and 8 days after the treatment. The kidney medulla AQP1, AQP2, AQP3 and V(2)R mRNA relative expressions were measured with RT-PCR. RESULTS: After single dose of the exact of sclerotia of Polyporus umbellata, urine output was found to be significantly increased, which began at 4h, and at 24h after the treatment, the sclerotia of Polyporus umbellatus extract and furosemide treatment produced the similar total volume of urine excreted. The extract increases urinary levels of Na(+), K(+), and Cl(-), to about the same extent, while furosemide increased urinary levels of Na(+) and Cl(-). After the 8-day doses, all two substances induced significant diuresis, natriuresis and chloriuresis. These two substances do not regulate the AQP1 and AQP3 mRNA level in normal rat kidney medulla. The AQP2 mRNA level of sclerotia of Polyporus umbellata extract was down-regulated significantly, the V(2)R mRNA level of sclerotia of Polyporus umbellata extract 50mg/kg dose group and 250 mg/kg dose group were down-regulated significantly too. Interestingly, the low-dose group had higher effect on regulation of AQP2 and V(2)R mRNA level. CONCLUSION: Aqueous extract of sclerotia of Polyporus umbellatus has conspicuous diuretic effect confirming its ethnopharmacological use. From the pattern of excretion of water, sodium, potassium, chlorine, AQP2 and V2R mRNA level, it may be logically concluded that it has effect from down-regulating AQP2, and down-regulate AQP2 by down-regulating V(2)R.


Assuntos
Aquaporinas/metabolismo , Arginina Vasopressina/metabolismo , Diuréticos/farmacologia , Polyporus/química , Receptores de Vasopressinas/metabolismo , Animais , Aquaporina 1/metabolismo , Aquaporina 2/metabolismo , Aquaporina 3/metabolismo , Diurese/efeitos dos fármacos , Furosemida/farmacologia , Medula Renal/efeitos dos fármacos , Medula Renal/metabolismo , Masculino , Natriurese/efeitos dos fármacos , Poliúria/metabolismo , Potássio/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sódio/metabolismo , Água/farmacologia
12.
Hypertension ; 54(2): 255-60, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19546378

RESUMO

In a previous proteomic study, we found dramatic differences in fumarase in the kidney between Dahl salt-sensitive rats and salt-insensitive consomic SS-13(BN) rats. Fumarase catalyzes the conversion between fumarate and l-malate in the tricarboxylic acid cycle. Little is known about the pathophysiological significance of fumarate metabolism in cardiovascular and renal functions, including salt-induced hypertension. The fumarase gene is located on the chromosome substituted in the SS-13(BN) rat. Sequencing of fumarase cDNA indicated the presence of lysine at amino acid position 481 in Dahl salt-sensitive rats and glutamic acid in Brown Norway and SS-13(BN) rats. Total fumarase activity was significantly lower in the kidneys of Dahl salt-sensitive rats compared with SS-13(BN) rats, despite an apparent compensatory increase in fumarase abundance in Dahl salt-sensitive rats. Intravenous infusion of a fumarate precursor in SS-13(BN) rats resulted in a fumarate excess in the renal medulla comparable to that seen in Dahl salt-sensitive rats. The infusion significantly exacerbated salt-induced hypertension in SS-13(BN) rats (140+/-3 vs125+/-2 mm Hg in vehicle control at day 5 on a 4% NaCl diet; P<0.05). In addition, the fumarate infusion increased renal medullary tissue levels of H2O2. Treatment of cultured human renal epithelial cells with the fumarate precursor also increased cellular levels of H2O2. These data suggest a novel role for fumarate metabolism in salt-induced hypertension and renal medullary oxidative stress.


Assuntos
Fumarato Hidratase/metabolismo , Hipertensão/enzimologia , Hipertensão/genética , Malatos/metabolismo , Ácido Succínico/metabolismo , Análise de Variância , Animais , Biomarcadores/metabolismo , Western Blotting , DNA Complementar/análise , Modelos Animais de Doenças , Hipertensão/fisiopatologia , Medula Renal/efeitos dos fármacos , Medula Renal/metabolismo , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/metabolismo , Masculino , Estresse Oxidativo/fisiologia , Distribuição Aleatória , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos Dahl , Cloreto de Sódio na Dieta/farmacologia
13.
Artigo em Inglês | MEDLINE | ID: mdl-18556188

RESUMO

Gentamicin (GM)-induced nephrotoxicity limits its long-term clinical use. Several agents/strategies were attempted to prevent GM nephrotoxicity but were not found suitable for clinical practice. Dietary fish oil (FO) retard the progression of certain types of cancers, cardiovascular and renal disorders. We aimed to evaluate protective effect of FO on GM-induced renal proximal tubular damage. The rats were pre-fed experimental diets for 10 days and then received GM (80 mg/kg body weight/day) treatment for 10 days while still on diet. Serum/urine parameters, enzymes of carbohydrate metabolism, brush border membrane (BBM), oxidative stress and phosphate transport in rat kidney were analyzed. GM nephrotoxicity was recorded by increased serum creatinine and blood urea nitrogen. GM increased the activities of lactate and glucose-6-phosphate dehydrogenases whereas decreased malate, isocitrate dehydrogenases; glucose-6 and fructose-1,6-bisphosphatases; superoxide dismutase, catalase, glutathione peroxidase and BBM enzymes. In contrast, FO alone increased enzyme activities of carbohydrate metabolism, BBM and oxidative stress. FO feeding to GM treated rats markedly enhanced resistance to GM elicited deleterious effects and prevented GM-induced decrease in 32Pi uptake across BBM. Dietary FO supplementation ameliorated GM-induced specific metabolic alterations and oxidative damage due to its intrinsic biochemical/antioxidant properties.


Assuntos
Gorduras Insaturadas na Dieta/farmacologia , Óleos de Peixe/farmacologia , Nefropatias/prevenção & controle , Rim/efeitos dos fármacos , Animais , Antioxidantes/metabolismo , Transporte Biológico Ativo/efeitos dos fármacos , Glicemia/metabolismo , Nitrogênio da Ureia Sanguínea , Metabolismo dos Carboidratos/efeitos dos fármacos , Óleo de Milho/administração & dosagem , Óleo de Milho/farmacologia , Óleo de Milho/uso terapêutico , Creatinina/sangue , Gorduras Insaturadas na Dieta/administração & dosagem , Gorduras Insaturadas na Dieta/uso terapêutico , Enzimas/metabolismo , Óleos de Peixe/administração & dosagem , Óleos de Peixe/uso terapêutico , Gentamicinas/toxicidade , Rim/enzimologia , Rim/metabolismo , Córtex Renal/efeitos dos fármacos , Córtex Renal/enzimologia , Córtex Renal/metabolismo , Nefropatias/induzido quimicamente , Nefropatias/metabolismo , Medula Renal/efeitos dos fármacos , Medula Renal/enzimologia , Medula Renal/metabolismo , Lipídeos/sangue , Masculino , Microvilosidades/efeitos dos fármacos , Microvilosidades/enzimologia , Microvilosidades/metabolismo , Modelos Biológicos , Estresse Oxidativo/efeitos dos fármacos , Fosfatos/sangue , Fosfatos/metabolismo , Fosfatos/urina , Ratos , Ratos Wistar , Transtornos Urinários/metabolismo , Transtornos Urinários/patologia , Transtornos Urinários/prevenção & controle
14.
Drug Metab Dispos ; 35(7): 1217-22, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17392392

RESUMO

Ingestion of herbal remedies containing aristolochic acids (AAs) is associated with the development of a syndrome, designated aristolochic acid nephropathy (AAN), which is characterized by chronic renal failure, tubulointerstitial fibrosis, and urothelial cancer. To distinguish the component(s) of AA responsible for these varied toxic effects, we administered 2.5 mg/kg/day of AA-I or AA-II for 9 days, either i.p. or p.o., to male C3H/He mice. Tissues were then collected and subjected to biochemical and histopathologic examination. Genotoxicity was assessed by determining quantitatively the level of aristolactam-DNA adducts in various tissues using (32)P-postlabeling/polyacrylamide gel electrophoresis and an internal standard. In the primary target tissues, represented by the renal cortex, medulla, and bladder, we found similar levels of DNA adducts derived from AA-I and AA-II. However, in nontarget tissues, the liver, stomach, intestine, and lung, the levels of aristolactam-DNA adducts derived from AA-I were significantly higher than those derived from AA-II. Histopathologic analysis revealed tubular cell necrosis and interstitial fibrosis in the renal cortex of AA-I-treated mice but only minimal changes in the renal cortex of mice treated with AA-II. We conclude that AA-I and AA-II have similar genotoxic and carcinogenic potential, and, although both compounds are cytotoxic, AA-I is solely responsible for the nephrotoxicity associated with AAN.


Assuntos
Ácidos Aristolóquicos/toxicidade , Carcinógenos/toxicidade , Transformação Celular Neoplásica/efeitos dos fármacos , Nefropatias/induzido quimicamente , Mutagênicos/toxicidade , Preparações de Plantas/toxicidade , Animais , Transformação Celular Neoplásica/metabolismo , Adutos de DNA/metabolismo , Fibrose , Córtex Renal/efeitos dos fármacos , Córtex Renal/metabolismo , Nefropatias/patologia , Medula Renal/efeitos dos fármacos , Medula Renal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C3H , Bexiga Urinária/efeitos dos fármacos , Bexiga Urinária/metabolismo
16.
Kidney Int ; 70(6): 1026-37, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16850027

RESUMO

We aimed to investigate the molecular mechanisms underlying the renal wasting of Na(+), K(+), Ca(2+), and Mg(2+) in gentamicin (GM)-treated rats. Male Wistar rats were injected with GM (40 or 80 mg/kg/day for 7 days, respectively; GM-40 or GM-80). The expression of NHE3, Na-K-ATPase, NKCC2, ROMK, NCC, alpha-, beta- and gamma-ENaC, and CaSR was examined in the kidney by immunoblotting and immunohistochemistry. Urinary fractional excretion of Na(+), K(+), Ca(2+), and Mg(2+) was increased and urinary concentration was decreased in both GM-40 and GM-80 rats. In cortex and outer stripe of outer medulla (cortex) in GM-80 rats, the expression of NHE3, Na-K-ATPase, and NKCC2 was decreased; NCC expression was unchanged; and CaSR was upregulated compared to controls. In the inner stripe of outer medulla (ISOM) in GM-80 rats, NKCC2 and Na-K-ATPase expression was decreased, whereas CaSR was upregulated, and NHE3 and ROMK expression remained unchanged. In GM-40 rats, NKCC2 expression was decreased in the cortex and ISOM, whereas NHE3, Na-K-ATPase, CaSR, ROMK, and NCC abundance was unchanged in both cortex and ISOM. Immunoperoxidase labeling confirmed decreased expression of NKCC2 in the thick ascending limb (TAL) in both GM-80- and GM-40-treated rats. Immunoblotting and immunohistochemical analysis revealed increased expression of alpha-, beta-, and gamma-ENaC in cortex in GM-80 rats, but not in GM-40 rats. These findings suggest that the decrease in NKCC2 in TAL seen in response to low-dose (40 mg/kg/day) gentamicin treatment may play an essential role for the increased urinary excretion of Mg(2+) and Ca(2+), and play a significant role for the development of the urinary concentrating defect, and increased urinary excretion of Na(+) and K(+). At high-dose gentamicin, both proximal and TAL sodium transporter downregulation is likely to contribute to this.


Assuntos
Antibacterianos/farmacologia , Gentamicinas/farmacologia , Rim/metabolismo , Canais de Sódio/efeitos dos fármacos , Sódio/metabolismo , Animais , Antibacterianos/farmacocinética , Cálcio/urina , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Gentamicinas/farmacocinética , Imuno-Histoquímica , Rim/efeitos dos fármacos , Córtex Renal/efeitos dos fármacos , Córtex Renal/metabolismo , Medula Renal/efeitos dos fármacos , Medula Renal/metabolismo , Magnésio/urina , Masculino , Potássio/urina , Ratos , Ratos Wistar , Receptores de Detecção de Cálcio/metabolismo , Sódio/urina , Simportadores de Cloreto de Sódio/metabolismo , Simportadores de Sódio-Bicarbonato/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo
17.
J Pharm Biomed Anal ; 40(2): 375-81, 2006 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-16146678

RESUMO

Mercury II chloride (HgCl2) toxicity was investigated in Sprague-Dawley rats using high-resolution magic angle spinning (HRMAS) 1H NMR spectroscopy in conjunction with principal component analysis (PCA). Intact renal cortex and papilla samples from Sprague-Dawley rats treated with HgCl2 at two dose levels (0.5 and 2 mg/kg) and from matched controls (n=5 per group) were assessed at 48 h p.d. HgCl2) caused depletion of renal osmolytes such as glycerophosphocholine (GPC), betaine, trimethylamine N-oxide (TMAO), myo-inositol and taurine in both the renal cortex and the papilla. In addition, relatively higher concentrations of valine, isobutyrate, threonine and glutamate were observed in HgCl2-treated rats, particularly in the renal cortex, which may reflect a counterbalance response to the observed loss of other classes of renal osmolytes. Increased levels of glutamate were present in the cortex of treated rats, which may be associated with HgCl2-induced renal acidosis and disruption of the tricarboxylic acid cycle. A dose response was observed in both cortical and papillary tissue with increasing severity of metabolic disruption in the high dose group. 1H HRMAS NMR profiles of individual animals correlated well with conventional clinical chemistry and histology confirming the reproducibility of the technology and generating complementary molecular pathway information.


Assuntos
Rim/efeitos dos fármacos , Cloreto de Mercúrio/toxicidade , Aminoácidos/metabolismo , Animais , Relação Dose-Resposta a Droga , Rim/metabolismo , Córtex Renal/efeitos dos fármacos , Córtex Renal/metabolismo , Medula Renal/efeitos dos fármacos , Medula Renal/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Masculino , Metilaminas/metabolismo , Análise de Componente Principal , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Taurina/metabolismo
18.
Am J Physiol Renal Physiol ; 289(4): F768-76, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15914777

RESUMO

Activation of P2Y2 receptor (P2Y2-R) in inner medullary collecting duct (IMCD) of rat decreases AVP-induced water flow and releases PGE(2). We observed that dehydration of rats decreases the expression of P2Y2 receptor in inner medulla (IM) and P2Y2-R-mediated PGE(2) release by IMCD. Because circulating vasopressin (AVP) levels are increased in dehydrated condition, we examined whether chronic infusion of desmopressin (dDAVP) has a similar effect on the expression and activity of P2Y2-R. Groups of rats were infused with saline or dDAVP (5 or 20 ng/h sc, 5 or 6 days) via osmotic minipumps and euthanized. Urine volume, osmolality, and PGE(2) metabolite content were determined. AQP2- and P2Y2- and V2-R mRNA and/or protein in IM were quantified by real-time RT-PCR and immunoblotting, respectively. P2Y2-R-mediated PGE(2) release by freshly prepared IMCD was assayed using ATPgammaS as a ligand. Chronic dDAVP infusion resulted in low-output of concentrated urine and significantly increased the AQP2 protein abundance in IM. On the contrary, dDAVP infusion at 5 or 20 ng/h significantly decreased P2Y2-R protein abundance (approximately 40% of saline-treated group). In parallel, the relative expression of P2Y2-R vs. AQP2- or V2-R mRNA was significantly decreased. Furthermore, the P2Y2-R-mediated PGE(2) release by IMCD was significantly decreased in rats infused 20 ng/h but not 5 ng/h of dDAVP. Urinary PGE(2) metabolite excretion, however, did not change with dDAVP infusion. In conclusion, chronic dDAVP infusion decreases the expression and activity of P2Y2-R in IM. This may be due to a direct effect of dDAVP or dDAVP-induced increase in medullary tonicity.


Assuntos
Desamino Arginina Vasopressina/farmacologia , Dinoprostona/metabolismo , Medula Renal/metabolismo , Túbulos Renais Coletores/metabolismo , Antagonistas do Receptor Purinérgico P2 , Receptores Purinérgicos P2/biossíntese , Fármacos Renais/farmacologia , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Animais , Aquaporina 2 , Aquaporinas/metabolismo , Western Blotting , Primers do DNA , DNA Complementar/biossíntese , Desamino Arginina Vasopressina/administração & dosagem , Infusões Intravenosas , Medula Renal/efeitos dos fármacos , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y2 , Fármacos Renais/administração & dosagem , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Urodinâmica/efeitos dos fármacos
19.
Am J Physiol Renal Physiol ; 288(4): F642-9, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15585669

RESUMO

The use of LiCl in clinical psychiatry is routinely complicated by overt nephrogenic diabetes insipidus (NDI), the mechanism of which is incompletely understood. In vitro studies indicate that lithium can induce renal medullary interstitial cell cyclooxygenase 2 (COX2) protein expression via inhibition of glycogen synthase kinase-3beta (GSK-3beta). Both COX1 and COX2 are expressed in the kidney. Renal prostaglandins have been suggested to play an important role in lithium-induced polyuria. The present studies examined whether induction of the COX2 isoform contributes to LiCl-induced polyuria. Four days after initiation of lithium treatment in C57 BL/6J mice, urine volume increased in LiCl-treated mice by fourfold compared with controls (P < 0.0001) and was accompanied by decreased urine osmolality. This was temporally associated with increased renal COX2 protein expression and increased urinary PGE(2) excretion, whereas COX1 levels remained unchanged. COX2 inhibition significantly blunted lithium-induced polyuria (P < 0.0001) and reduced urinary PGE(2) levels. Lithium-associated polyuria was also seen in COX1-/- mice and was associated with increased urinary PGE(2). COX2 inhibition completely prevented polyuria and PGE(2) excretion in COX1-/- mice, suggesting that COX2, but not COX1, plays a critical role in lithium-induced polyuria. Lithium also induced renal medullary COX2 protein expression in congenitally polyuric antidiuretic hormone (AHD)-deficient rats, demonstrating that lithium-induced COX2 protein expression is not secondary to altered ADH levels or polyuria. Lithium also decreased renal medullary GSK-3beta activity, and this was temporally related to increased COX2 expression in the kidney from lithium-treated mice, consistent with a tonic in vivo suppression of COX2 expression by GSK-3 activity. In conclusion, these findings temporally link decreased GSK-3 activity to enhanced renal COX2 expression and COX2-derived urine PGE(2) excretion. Suppression of COX2-derived PGE(2) blunts lithium-associated polyuria.


Assuntos
Adjuvantes Imunológicos/farmacologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Cloreto de Lítio/farmacologia , Poliúria/induzido quimicamente , Prostaglandina-Endoperóxido Sintases/metabolismo , Animais , Células Cultivadas , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Diabetes Insípido/induzido quimicamente , Diabetes Insípido/metabolismo , Dinoprostona/urina , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Medula Renal/citologia , Medula Renal/efeitos dos fármacos , Medula Renal/enzimologia , Masculino , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Microssomos/efeitos dos fármacos , Microssomos/enzimologia , Concentração Osmolar , Poliúria/metabolismo , Prostaglandina-Endoperóxido Sintases/genética , Ratos , Ratos Brattleboro
20.
Ann N Y Acad Sci ; 1030: 144-9, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15659791

RESUMO

Oxalate, a metabolic end product and a major constituent of the majority of renal stones, has been shown to be toxic to renal epithelial cells of cortical origin. However, it is unknown whether inner medullary collecting duct (IMCD) cells that are physiologically exposed to higher concentrations of oxalate also behave in a similar manner. In the present study, we examined the effects of oxalate on IMCD cells. IMCD cells from the mouse were maintained in DMEM/F12 media supplemented with fetal bovine serum and antibiotics. Exposure of IMCD cells to oxalate produced time- and concentration-dependent changes in the light microscopic appearance of the cells. Long-term exposure to oxalate resulted in alterations in cell viability, with net cell loss after exposure to concentrations of 2 mM or greater. The production of free radicals was directly related to the exposure time and the concentration of oxalate. Crystal formation occurred in less than 1 h and cells in proximity to crystals would lose membrane integrity. Compared with IMCD cells, LLC-PK1 cells as well as HK-2 cells showed significant toxicity starting at lower oxalate concentrations (0.4 mM or greater). These results provide the first direct demonstration of toxic effects of oxalate in IMCD cells, a line of renal epithelial cells of the inner medullary collecting duct, and suggest that the cells lining the collecting duct are relatively resistant to oxalate toxicity.


Assuntos
Medula Renal/efeitos dos fármacos , Ácido Oxálico/farmacologia , Animais , Linhagem Celular , Medula Renal/citologia , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA