Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Toxics ; 7(2)2019 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-31167416

RESUMO

Cadmium (Cd) is an environmental toxicant that accumulates in bone and alters bone turnover and metabolism. Periodontal disease is characterized by tooth loss and tissue destruction, specifically, loss of supporting bone around the teeth. We have previously shown that Cd causes loss of dental alveolar (tooth supporting) bone in a rodent model of long-term Cd poisoning. The overall goal of this study was to determine the possible association between levels of Cd in alveolar bone and evidence of periodontal disease in human cadavers. The extent of Cd accumulation in human mandible samples was analyzed. Levels of Cd in mandibular alveolar bone were compared to those in basal bone as well as the renal cortex in samples obtained from the cadavers. Alveolar bone contained significantly higher levels of Cd when compared to basal bone (p < 0.01). Cd levels in mandibular bone were significantly higher in female compared to male cadavers (p < 0.05). The kidney cortex had greater than 15-fold higher Cd levels compared to mandible bone. Additional analyses showed a possible association between levels of Cd in basal bone and the presence of periodontal disease in cadavers from which the samples were obtained. This study shows that Cd accumulates to relatively high levels within alveolar bone as compared to basal bone in the mandible and thus may have a significant and direct effect in the progression of changes in bone associated with periodontal disease.

2.
Artigo em Inglês | MEDLINE | ID: mdl-30988153

RESUMO

Urinary biomarkers are superior to serum creatinine for defining onset and extent of kidney injury. This study classifies the temporal predictive ability of biomarkers for vancomycin-induced kidney injury (VIKI) as defined by histopathologic damage. Male Sprague-Dawley rats (n = 125) were randomized to receive 150 to 400 mg/kg of body weight/day vancomycin via once or twice daily intraperitoneal injection over 1, 3, or 6 days. Urine was collected once during the 24 h prior to euthanasia or twice for rats treated for 6 days. Receiver operating characteristic (ROC) curves were employed to assess the urinary biomarker performances of kidney injury molecule 1 (KIM-1), clusterin, osteopontin (OPN), cystatin C, and neutrophil gelatinase-associated lipocalin (NGAL) to predict histopathologically defined VIKI (using a national standard pathological assessment scheme from hematoxylin and eosin stained kidneys). Urinary KIM-1, clusterin, and OPN outperformed cystatin C and NGAL with regard to sensitivity and specificity. For the earliest injury, urinary KIM-1 (area under the receiver operating characteristic curve [AUC], 0.662; P < 0.001) and clusterin (AUC, 0.706; P < 0.001) were the most sensitive for predicting even low-level histopathologic damage at 24 h compared to NGAL. KIM-1 and clusterin are the earliest and most sensitive predictors of VIKI. As injury progresses, KIM-1, clusterin, and OPN best define the extent of damage.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/urina , Biomarcadores/urina , Moléculas de Adesão Celular/urina , Vancomicina/efeitos adversos , Animais , Cistatina C/urina , Lipocalina-2/urina , Masculino , Osteopontina/urina , Curva ROC , Ratos , Ratos Sprague-Dawley
3.
Toxics ; 6(2)2018 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-29899258

RESUMO

Cadmium (Cd) is an environmental contaminant that damages the kidney, the liver, and bones. Some epidemiological studies showed associations between Cd exposure and periodontal disease. The purpose of this study was to examine the relationship between Cd exposure and periodontal disease in experimental animals. Male Sprague/Dawley rats were given daily subcutaneous injections of Cd (0.6 mg/kg/day) for up to 12 weeks. The animals were euthanized, and their mandibles and maxillae were evaluated for levels of periodontal bone by measuring the distance from the cementoenamel junction (CEJ) to the alveolar bone crest (ABC) of the molar roots. After 12 weeks of Cd exposure in animals, there was a significantly greater distance between the CEJ and ABC in the palatal aspect of the maxillary molars and the lingual aspect of the mandibular molars when compared with controls (p < 0.0001). This study shows that Cd has significant, time-dependent effects on periodontal bone in an animal model of Cd exposure. These findings support the possibility of Cd being a contributing factor to the development of periodontal disease in humans.

4.
Toxics ; 6(1)2018 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-29543730

RESUMO

Cadmium (Cd) is a nephrotoxic environmental pollutant that causes a generalized dysfunction of the proximal tubule characterized by polyuria and proteinuria. Even though the effects of Cd on the kidney have been well-characterized, the molecular mechanisms underlying these effects have not been fully elucidated. MicroRNAs (miRNAs) are small non-coding RNAs that regulate cellular and physiologic function by modulating gene expression at the post-transcriptional level. The goal of the present study was to determine if Cd affects renal cortex miRNA expression in a well-established animal model of Cd-induced kidney injury. Male Sprague-Dawley rats were treated with subcutaneous injections of either isotonic saline or CdCl2 (0.6 mg/kg) 5 days a week for 12 weeks. The 12-week Cd-treatment protocol resulted in kidney injury as determined by the development of polyuria and proteinuria, and a significant increase in the urinary biomarkers Kim-1, ß2 microglobulin and cystatin C. Total RNA was isolated from the renal cortex of the saline control and Cd treated animals, and differentially expressed miRNAs were identified using µParafloTM microRNA microarray analysis. The microarray results demonstrated that the expression of 44 miRNAs were significantly increased and 54 miRNAs were significantly decreased in the Cd treatment group versus the saline control (t-test, p ≤ 0.05, N = 6 per group). miR-21-5p, miR-34a-5p, miR-146b-5p, miR-149-3p, miR-224-5p, miR-451-5p, miR-1949, miR-3084a-3p, and miR-3084c-3p demonstrated more abundant expression and a significant two-fold or greater increased expression in the Cd-treatment group versus the saline control group. miR-193b-3p, miR-455-3p, and miR-342-3p demonstrated more abundant expression and a significant two-fold or greater decreased expression in the Cd-treatment group versus the saline control group. Real-time PCR validation demonstrated (1) a significant (t-test, p ≤ 0.05, N = 6 per group) increase in expression in the Cd-treated group for miR-21-5p (2.7-fold), miR-34a-5p (10.8-fold), miR-146b-5p (2-fold), miR-224-5p (10.2-fold), miR-3084a-3p (2.4-fold), and miR-3084c-3p (3.3-fold) and (2) a significant (t-test, p ≤ 0.05, N = 6 per group) 52% decrease in miR-455-3p expression in the Cd-treatment group. These findings demonstrate that Cd significantly alters the miRNA expression profile in the renal cortex and raises the possibility that dysregulated miRNA expression may play a role in the pathophysiology of Cd-induced kidney injury. In addition, these findings raise the possibility that Cd-dysregulated miRNAs might be used as urinary biomarkers of Cd exposure or Cd-induced kidney injury.

5.
Artigo em Inglês | MEDLINE | ID: mdl-28807910

RESUMO

Vancomycin has been associated with acute kidney injury in preclinical and clinical settings; however, the precise exposure profiles associated with vancomycin-induced acute kidney injury have not been defined. We sought to determine pharmacokinetic/pharmacodynamics indices associated with the development of acute kidney injury using sensitive urinary biomarkers. Male Sprague-Dawley rats received clinical-grade vancomycin or normal saline as an intraperitoneal injection. Total daily doses between 0 and 400 mg/kg of body weight were administered as a single dose or 2 divided doses over a 24-h period. At least five rats were utilized for each dosing protocol. A maximum of 8 plasma samples per rat were obtained, and urine was collected over the 24-h period. Kidney injury molecule-1 (KIM-1), clusterin, osteopontin, cystatin C, and neutrophil gelatinase-associated lipocalin levels were determined using Milliplex multianalyte profiling rat kidney panels. Vancomycin plasma concentrations were determined via a validated high-performance liquid chromatography methodology. Pharmacokinetic analyses were conducted using the Pmetrics package for R. Bayesian maximal a posteriori concentrations were generated and utilized to calculate the 24-h area under the concentration-time curve (AUC), the maximum concentration (Cmax), and the minimum concentration. Spearman's rank correlation coefficient (rs ) was used to assess the correlations between exposure parameters, biomarkers, and histopathological damage. Forty-seven rats contributed pharmacokinetic and toxicodynamic data. KIM-1 was the only urinary biomarker that correlated with both composite histopathological damage (rs = 0.348, P = 0.017) and proximal tubule damage (rs = 0.342, P = 0.019). The vancomycin AUC and Cmax were most predictive of increases in KIM-1 levels (rs = 0.438 and P = 0.002 for AUC and rs = 0.451 and P = 0.002 for Cmax). Novel urinary biomarkers demonstrate that kidney injury can occur within 24 h of vancomycin exposure as a function of either AUC or Cmax.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Antibacterianos/efeitos adversos , Antibacterianos/farmacocinética , Vancomicina , Animais , Área Sob a Curva , Biomarcadores/sangue , Biomarcadores/urina , Moléculas de Adesão Celular/sangue , Clusterina/sangue , Cistatina C/sangue , Lipocalina-2/sangue , Masculino , Osteopontina/sangue , Ratos , Ratos Sprague-Dawley , Vancomicina/efeitos adversos , Vancomicina/sangue , Vancomicina/farmacocinética
6.
Antimicrob Agents Chemother ; 60(10): 5742-51, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27431226

RESUMO

Vancomycin has been associated with acute kidney injury (AKI). However, the pharmacokinetic/toxicodynamic relationship for AKI is not well defined. Allometrically scaled vancomycin exposures were used to assess the relationship between vancomycin exposure and AKI. Male Sprague-Dawley rats received clinical-grade vancomycin in normal saline (NS) as intraperitoneal (i.p.) injections for 24- to 72-h durations with doses ranging 0 to 200 mg/kg of body weight divided once or twice daily. Urine was collected over the protocol's final 24 h. Renal histopathology was qualitatively scored. Urinary biomarkers (e.g., cystatin C, clusterin, kidney injury molecule 1 [KIM-1], osteopontin, lipocalin 2/neutrophil gelatinase-associated lipocalin 2) were assayed using a Luminex xMAP system. Plasma vancomycin concentrations were assayed by high-performance liquid chromatography with UV detection. A three-compartment vancomycin pharmacokinetic model was fit to the data with the Pmetrics package for R. The exposure-response in the first 24 h was evaluated using Spearman's nonparametric correlation coefficient (rs) values for the area under the concentration-time curve during the first 24 h (AUC0-24), the maximum concentration in plasma during the first 24 h (Cmax0-24 ), and the lowest (minimum) concentration in plasma after the dose closest to 24 h (Cmin0-24 ). A total of 52 rats received vancomycin (n = 42) or NS (n = 10). The strongest exposure-response correlations were observed between AUC0-24 and Cmax0-24 and urinary AKI biomarkers. Exposure-response correlations (rs values) for AUC0-24, Cmax0-24 , and Cmin0-24 were 0.37, 0.39, and 0.22, respectively, for clusterin; 0.42, 0.45, and 0.26, respectively, for KIM-1; and 0.52, 0.55, and 0.42, respectively, for osteopontin. However, no differences in histopathological scores were observed. Optimal sampling times after administration of the i.p. dose were 0.25, 0.75, 2.75, and 8 h for the once-daily dosing schemes and 0.25, 1.25, 14.5, and 17.25 h for the twice-daily dosing schemes. Our observations suggest that AUC0-24 or Cmax0-24 correlates with increases in urinary AKI biomarkers.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/urina , Biomarcadores/urina , Vancomicina/efeitos adversos , Vancomicina/farmacocinética , Animais , Rim/efeitos dos fármacos , Rim/patologia , Masculino , Ratos Sprague-Dawley
7.
Biometals ; 29(1): 131-46, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26715107

RESUMO

Cadmium (Cd) is a nephrotoxic environmental pollutant that causes insidious injury to the proximal tubule that results in severe polyuria and proteinuria. Cystatin C is a low molecular weight protein that is being evaluated as a serum and urinary biomarker for various types of ischemic and nephrotoxic renal injury. The objective of the present study was to determine if cystatin C might be a useful early biomarker of Cd nephrotoxicity. Male Sprague-Dawley rats were given daily injections of Cd for up to 12 weeks. At 3, 6, 9 and 12 weeks, urine samples were analyzed for cystatin C, protein, creatinine, ß2 microglobulin and kidney injury molecule-1. The results showed that Cd caused a significant increase in the urinary excretion of cystatin C that occurred 3-4 weeks before the onset of polyuria and proteinuria. Serum levels of cystatin C were not altered by Cd. Immunolabeling studies showed that Cd caused the relocalization of cystatin C from the cytoplasm to the apical surface of the epithelial cells of the proximal tubule. The Cd-induced changes in cystatin C labelling paralleled those of the brush border transport protein, megalin, which has been implicated as a mediator of cystatin C uptake in the proximal tubule. These results indicate that Cd increases the urinary excretion of cystatin C, and they suggest that this effect may involve disruption of megalin-mediated uptake of cystatin C by epithelial cells of the proximal tubule.


Assuntos
Biomarcadores/urina , Cádmio/toxicidade , Cistatina C/urina , Túbulos Renais Proximais/metabolismo , Animais , Biomarcadores/sangue , Cádmio/administração & dosagem , Moléculas de Adesão Celular/sangue , Creatinina/sangue , Cistatina C/sangue , Poluentes Ambientais , Humanos , Túbulos Renais Proximais/lesões , Túbulos Renais Proximais/patologia , Masculino , Ratos
8.
Toxicol Rep ; 3: 740-746, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28959600

RESUMO

Cd (Cd) is a nephrotoxic environmental pollutant that causes generalized proximal tubule dysfunction. Even though the specific mechanisms by which Cd damages the kidney have yet to be fully elucidated, there is evidence to suggest that some of these nephrotoxic effects may result from the ability of Cd to alter the levels and function of metals such as Cu, Se, Zn and Fe within the kidney. In order to further explore this issue, we examined the effects of subchronic Cd exposure on tissue levels of a panel of metals (Ca, Cu, Fe, K, Mg, Na, Se and Zn) in the rat renal cortex. Adult male Sprague-Dawley rats were treated with CdCl2 (0.6 mg Cd/kg body weight in isotonic saline by subcutaneous injection, 5 days per week for 6, 9 or 12 weeks). At each time point, 24 h urine samples were collected and assayed for levels of protein, creatinine, ß2 microglobulin and cystatin C. Samples of renal cortex were removed and assayed for levels of the metals of interest by inductively-coupled mass spectrometry at Michigan State University. Results showed that at 9 and 12 weeks, Cd caused significant increases in urine volume and urinary protein with no change in creatinine excretion. Increases in the excretion of the urinary biomarkers ß2 microglobulin and cystatin C were evident after 6 weeks of Cd exposure. Results of the metal analyses showed that Cd caused significant increases in tissue levels of Cu and Se at all of the time points examined. Tissue levels of Zn were transiently elevated at 6 weeks but declined to control levels at 9 and 12 weeks. Cd caused a significant decrease in levels of Fe at 9 and 12 weeks. Cd had no effects on any of the other metals. Tissue levels of Cd were 530 ± 52, 863 ± 23, 837 ± 23 ppm dry weight at 6, 9 and 12 weeks, respectively. These results indicate that the early stages of Cd nephrotoxicity are associated with alterations in renal tissue levels of Cu, Se, Zn and Fe. The fact that the changes in levels of the metals occurred during the early stages of Cd toxicity raises the possibility that the alterations in renal cortical metal content may play some role in the pathophysiology or Cd-induced injury.

9.
J Toxicol Environ Health A ; 78(12): 711-24, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26090557

RESUMO

Nanoparticles (NP) are pervasive in many areas of modern life, with little known about their potential toxicities. One commercially important NP is cadmium oxide (CdO), which is used to synthesize other Cd-containing NP, such as quantum dots. Cadmium (Cd) is a well-known nephrotoxicant, but the nephrotoxic potential of CdO NP remains unknown, particularly when exposure occurs during pregnancy. Therefore, pregnant CD-1 mice were used to examine the effects of inhaled CdO NP (230 µg CdO NP/m(3)) on maternal and neonatal renal function by examining urinary creatinine and urinary biomarkers of kidney injury, including kidney injury molecule-1 (Kim-1) and neutrophil gelatinase-associated lipocalin (NGAL). Inhalation of CdO NP by dams produced a fivefold increase in urinary Kim-1 with no marked effect on urinary creatinine levels. Kim-1 mRNA expression peaked by gestational day (GD) 10.5, and NGAL expression increased from GD 10.5 to 17.5. In addition, histological analyses revealed proximal tubular pathology at GD 10.5. Neonatal Kim-1 mRNA expression rose between postnatal days (PND) 7 and 14, with mammary glands/milk being the apparent source of Cd for offspring. These studies demonstrate that, similar to what is seen with other Cd forms, Cd associated with inhaled CdO NP results in renal injury to both directly exposed dam and offspring. As commercial uses for nanotechnology continue to expand throughout the world, risks for unintentional exposure in the workplace increase. Given the large number of women in the industrial workforce, care needs to be taken to protect these already vulnerable populations.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/congênito , Compostos de Cádmio/toxicidade , Nanopartículas/toxicidade , Óxidos/toxicidade , Injúria Renal Aguda/patologia , Proteínas de Fase Aguda/biossíntese , Proteínas de Fase Aguda/genética , Animais , Animais Recém-Nascidos , Biomarcadores/urina , Compostos de Cádmio/farmacocinética , Creatinina/urina , Feminino , Glicosúria/induzido quimicamente , Glicosúria/urina , Receptor Celular 1 do Vírus da Hepatite A , Exposição por Inalação , Rim/patologia , Lipocalina-2 , Lipocalinas/biossíntese , Lipocalinas/genética , Glândulas Mamárias Animais/metabolismo , Exposição Materna , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Camundongos , Proteínas Oncogênicas/biossíntese , Proteínas Oncogênicas/genética , Óxidos/farmacocinética , Gravidez , RNA Mensageiro/biossíntese
10.
Biometals ; 26(1): 33-42, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23080431

RESUMO

The E-cadherin/ß-catenin complex is a structural component of adherens-type junctions in epithelial cells. Moreover, ß-catenin acts as an intracellular signaling molecule that can influence the expression of a variety of genes that regulate apoptosis and cell cycle control. Cadmium (Cd) is an environmental toxicant that causes renal dysfunction and disrupts cadherin-dependent cell-cell adhesion in various types of epithelial cells. In this study, we examined the effects of Cd on the subcellular localization of ß-catenin, the cadherin/ß-catenin complex and ß-catenin-mediated gene transcription in rat proximal tubule NRK-52E cells. Exposure to 5-10 µM Cd for 4 h caused the NRK cells to separate from each other without killing the cells or causing them to detach from the growing surface. This effect was associated with the loss of ß-catenin and E-cadherin from the cell-cell contacts and apparent changes in the accumulation of ß-catenin in the nuclear cell subfraction. The expression of the ß-catenin-sensitive gene, c-jun was significantly increased in cells exposed to 5 µM Cd. However, there was no change in the expression of several other ß-catenin-regulated genes including: c-myc, cyclin D1 and matrilysin. Additional studies utilizing the TOPFLASH ß-catenin reporter gene construct showed that Cd caused a 2-3 fold increase in the expression of the luciferase reporter gene. Overall, these results indicate that Cd disrupts the cadherin/ß-catenin complex in NRK-52E cells, but this effect leads to only partial activation of ß-catenin-mediated gene transcription.


Assuntos
Cloreto de Cádmio/toxicidade , Poluentes Ambientais/toxicidade , beta Catenina/metabolismo , Animais , Caderinas/genética , Caderinas/metabolismo , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Rim/citologia , Lítio/farmacologia , Transporte Proteico/efeitos dos fármacos , Ratos , Ativação Transcricional/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , beta Catenina/genética
11.
J Pharmacol Exp Ther ; 343(1): 2-12, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22669569

RESUMO

Cadmium is an important industrial agent and environmental pollutant that is a major cause of kidney disease. With chronic exposure, cadmium accumulates in the epithelial cells of the proximal tubule, resulting in a generalized reabsorptive dysfunction characterized by polyuria and low-molecular-weight proteinuria. The traditional view has been that as cadmium accumulates in proximal tubule cells, it produces a variety of relatively nonspecific toxic effects that result in the death of renal epithelial cells through necrotic or apoptotic mechanisms. However, a growing volume of evidence suggests that rather than merely being a consequence of cell death, the early stages of cadmium-induced proximal tubule injury may involve much more specific changes in cell-cell adhesion, cellular signaling pathways, and autophagic responses that occur well before the onset of necrosis or apoptosis. In this commentary, we summarize these recent findings, and we offer our own perspectives as to how they relate to the toxic actions of cadmium in the kidney. In addition, we highlight recent findings, suggesting that it may be possible to detect the early stages of cadmium toxicity through the use of improved biomarkers. Finally, some of the therapeutic implications of these findings will be considered. Because cadmium is, in many respects, a model cumulative nephrotoxicant, these insights may have broader implications regarding the general mechanisms through which a variety of drugs and toxic chemicals damage the kidney.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/metabolismo , Intoxicação por Cádmio/metabolismo , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Injúria Renal Aguda/patologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Biomarcadores/metabolismo , Cádmio/metabolismo , Intoxicação por Cádmio/patologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Poluentes Ambientais/toxicidade , Humanos , Túbulos Renais Proximais/patologia
12.
Biometals ; 23(5): 793-809, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20107869

RESUMO

As the risks of cadmium (Cd)-induced kidney disease have become increasingly apparent, much attention has been focused on the development and use of sensitive biomarkers of Cd nephrotoxicity. The purpose of this review is to briefly summarize the current state of Cd biomarker research. The review includes overviews of the toxicokinetics of Cd, the mechanisms of Cd-induced proximal tubule injury, and mechanistic summaries of some of the biomarkers (N-acetyl-ß-D-glucosamidase; ß(2)-microglubulin, metallothionein, etc.) that have been most widely used in monitoring of human populations for Cd exposure and nephrotoxicity. In addition, several novel biomarkers (kidney injury molecule-1, α-glutathione-S-transferase and insulin) that offer the potential for improved biomonitoring of Cd-exposed populations are discussed.


Assuntos
Biomarcadores/metabolismo , Cádmio/toxicidade , Rim/efeitos dos fármacos , Cádmio/administração & dosagem , Cádmio/farmacocinética , Exposição Ambiental , Saúde Ambiental , Monitoramento Ambiental , Humanos , Rim/lesões , Rim/fisiopatologia , Metalotioneína/metabolismo , Modelos Biológicos , Distribuição Tecidual
13.
Toxicol Appl Pharmacol ; 238(3): 301-5, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19371616

RESUMO

As a result of the widespread use of Cd in industry and its extensive dissemination in the environment, there has been considerable interest in the identification of early biomarkers of Cd-induced kidney injury. Kim-1 is a transmembrane glycoprotein that is not detectable in normal kidney, but is up-regulated and shed into the urine following ischemic or nephrotoxic injury. Recent studies utilizing a sub-chronic model of Cd exposure in the rat have shown that Kim-1 is an early urinary marker of Cd-induced kidney injury. Kim-1 was detected in the urine 4-5 weeks before the onset of proteinuria and 1-3 weeks before the appearance of urinary metallothionein and Clara cell protein 16, which are standard markers of Cd nephrotoxicity. In the present study, we have compared the time course for the appearance of Kim-1 in the urine with the time course for the appearance of alpha glutathione-S-transferase (alpha-GST), N-acetyl-beta-D-glucose amidase (NAG) and Cd, each of which have been used or proposed as urinary markers of Cd nephrotoxicity. Adult male Sprague-Dawley rats were given daily subcutaneous injections of 0.6 mg (5.36 micromoles)/kg Cd, 5 days per week for up to 12 weeks. One day each week, 24 h urine samples were collected and analyzed for protein, creatinine and the various markers. The results showed that significant levels of Kim-1 appeared in the urine as early as 6 weeks into the treatment protocol and then continued to rise for the remainder of the 12 week treatment period. By contrast, significant levels of alpha-GST and NAG did not appear in the urine until 8 and 12 weeks, respectively, while proteinuria was not evident until 10 weeks. The urinary excretion of Cd was below the level of detection until week 4 and then showed a slow, linear increase over the next 6 weeks before increasing markedly between weeks 10 and 12. These results provide additional evidence that Kim-1 is a sensitive biomarker of the early stages of Cd-induced proximal tubule injury.


Assuntos
Acetilglucosaminidase/urina , Cádmio/toxicidade , Moléculas de Adesão Celular/urina , Poluentes Ambientais/toxicidade , Glutationa Transferase/urina , Isoenzimas/urina , Proteinúria/diagnóstico , Animais , Biomarcadores/urina , Cádmio/urina , Diagnóstico Precoce , Poluentes Ambientais/urina , Masculino , Valor Preditivo dos Testes , Proteinúria/induzido quimicamente , Proteinúria/urina , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Regulação para Cima
14.
Kidney Int ; 76(1): 108-14, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19387469

RESUMO

Kidney injury molecule-1 (Kim-1) has been qualified by the Food and Drug Administration and European Medicines Agency as a highly sensitive and specific urinary biomarker to monitor drug-induced kidney injury in preclinical studies and on a case-by-case basis in clinical trials. Here we report the development and evaluation of a rapid direct immunochromatographic lateral flow 15-min assay for detection of urinary Kim-1 (rat) or KIM-1 (human). The urinary Kim-1 band intensity using the rat Kim-1 dipstick significantly correlated with levels of Kim-1 as measured by a microbead-based assay, histopathological damage, and immunohistochemical assessment of renal Kim-1 in a dose- and time-dependent manner. Kim-1 was detected following kidney injury induced in rats by cadmium, gentamicin, or bilateral renal ischemia/reperfusion. In humans, the urinary KIM-1 band intensity was significantly greater in urine from patients with acute kidney injury than in urine from healthy volunteers. The KIM-1 dipstick also enabled temporal evaluation of kidney injury and recovery in two patients who developed postoperative acute kidney injury following cytoreductive surgery for malignant mesothelioma with intraoperative local cisplatin administration. We hope that future, more extensive studies will confirm the utility of these results, which show that the Kim-1/KIM-1 dipsticks can provide a sensitive and accurate detection of Kim-1/KIM-1, thereby providing a rapid diagnostic assay for kidney damage and facilitating the rapid and early detection of kidney injury in preclinical and clinical studies.


Assuntos
Moléculas de Adesão Celular/urina , Nefropatias/diagnóstico , Nefropatias/urina , Rim/química , Glicoproteínas de Membrana/urina , Animais , Bioensaio , Biomarcadores/urina , Cádmio/efeitos adversos , Estudos de Casos e Controles , Cisplatino/efeitos adversos , Estudos Transversais , Diagnóstico Precoce , Receptor Celular 1 do Vírus da Hepatite A , Humanos , Imuno-Histoquímica , Rim/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Receptores Virais , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/urina , Sensibilidade e Especificidade , Fatores de Tempo , Urinálise
15.
Toxicol Appl Pharmacol ; 238(3): 289-93, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19327375

RESUMO

Recent epidemiological studies suggest a positive association between exposure to the environmental pollutant cadmium (Cd) and the incidence and severity of diabetes. In this review, we examine the literature suggesting a relationship between Cd exposure, elevated blood glucose levels, and the development of diabetes. In addition we review human and animal studies indicating that Cd potentiates or exacerbates diabetic nephropathy. We also review the various possible cellular mechanisms by which Cd may alter blood glucose levels. In addition, we present some novel findings from our own laboratories showing that Cd elevates fasting blood glucose levels in an animal model of subchronic Cd exposure before overt signs of renal dysfunction are evident. These studies also show that Cd reduces insulin levels and has direct cytotoxic effects on the pancreas. Together, these findings indicate that Cd may be a factor in the development of some types of diabetes and they raise the possibility that Cd and diabetes-related hyperglycemia may act synergistically to damage the kidney.


Assuntos
Cádmio/toxicidade , Diabetes Mellitus/induzido quimicamente , Nefropatias Diabéticas/etiologia , Poluentes Ambientais/toxicidade , Pâncreas/efeitos dos fármacos , Animais , Glicemia/efeitos dos fármacos , Doença Crônica , Diabetes Mellitus/epidemiologia , Diabetes Mellitus/metabolismo , Nefropatias Diabéticas/induzido quimicamente , Nefropatias Diabéticas/epidemiologia , Nefropatias Diabéticas/metabolismo , Humanos , Insulina/sangue , Modelos Animais , Pâncreas/metabolismo , Pâncreas/patologia , Medição de Risco , Fatores de Risco
16.
Toxicol Sci ; 102(2): 207-18, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17947343

RESUMO

Vascular system function involves complex interactions among the vascular endothelium, smooth muscle, the immune system, and the nervous system. The toxic metals cadmium (Cd), arsenic (As), and lead (Pb) can target the vascular system in a variety of ways, ranging from hemorrhagic injury to subtle pathogenic remodeling and metabolic changes. Acute Cd exposure results in hemorrhagic injury to the testis, although some strains of animals are resistant to this effect. A comparison of Cd-sensitive with Cd-resistant mouse strains showed that expression of the Slc39a8 gene, encoding the ZIP8 transporter, in the testis vasculature endothelium is responsible for this difference. Endogenously, ZIP8 is a Mn(2+)/HCO(3)(-)symporter that may also contribute to Cd damage in the kidney. Chronic Cd exposure is associated with various cardiovascular disorders such as hypertension and cardiomyopathy and it is reported to have both carcinogenic and anticarcinogenic activities. At noncytotoxic concentrations of 10-100nM, Cd can inhibit chemotaxis and tube formation of vascular endothelial cells. These angiostatic effects may be mediated through disruption of vascular endothelial cadherin, a Ca(2+)-dependent cell adhesion molecule. With regard to As, ingestion of water containing disease-promoting concentrations of As promotes capillarization of the liver sinusoidal endothelium. Because capillarization is a hallmark precursor for liver fibrosis and contributes to an imbalance of lipid metabolism, this As effect on hepatic endothelial cells may be a pathogenic mechanism underlying As-related vascular diseases. With regard to Pb, perinatal exposure may cause sustained elevations in adult blood pressure, and genetically susceptible animals may show enhanced sensitivity to this effect. Taken together, these data indicate that the vascular system is a critical target of metal toxicity and that actions of metals on the vascular system may play important roles in mediating the pathophysiologic effects of metals in specific target organs.


Assuntos
Arsenicais/efeitos adversos , Vasos Sanguíneos/efeitos dos fármacos , Compostos de Cádmio/toxicidade , Intoxicação por Chumbo , Chumbo , Animais , Vasos Sanguíneos/patologia , Relação Dose-Resposta a Droga , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Humanos , Neovascularização Patológica/induzido quimicamente , Neovascularização Patológica/patologia
17.
Pharmacol Ther ; 114(1): 74-93, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17316817

RESUMO

Cell adhesion molecules are integral cell-membrane proteins that maintain cell-cell and cell-substrate adhesion and in some cases act as regulators of intracellular signaling cascades. In the kidney, cell adhesion molecules, such as the cadherins, the catenins, the zonula occludens protein-1 (ZO-1), occludin and the claudins are essential for maintaining the epithelial polarity and barrier integrity that are necessary for the normal absorption/excretion of fluid and solutes. A growing volume of evidence indicates that these cell adhesion molecules are important early targets for a variety of nephrotoxic substances including metals, drugs, and venom components. In addition, it is now widely appreciated that molecules, such as intracellular adhesion molecule-1 (ICAM-1), integrins, and selectins play important roles in the recruitment of leukocytes and inflammatory responses that are associated with nephrotoxic injury. This review summarizes the results of recent in vitro and in vivo studies indicating that these cell adhesion molecules may be primary molecular targets in many types of chemically-induced renal injury. Some of the specific agents that are discussed include cadmium (Cd), mercury (Hg), bismuth (Bi), cisplatin, aminoglycoside antibiotics, S-(1,2-dichlorovinyl)-l-cysteine (DCVC), and various venom toxins. This review also includes a discussion of the various mechanisms, by which these substances can affect cell adhesion molecules in the kidney.


Assuntos
Moléculas de Adesão Celular/fisiologia , Nefropatias/induzido quimicamente , Nefropatias/fisiopatologia , Animais , Carcinógenos/toxicidade , Junções Comunicantes/fisiologia , Humanos , Glomérulos Renais/fisiologia , Túbulos Renais/fisiologia , Xenobióticos/toxicidade
19.
Life Sci ; 79(16): 1493-506, 2006 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-16765992

RESUMO

Cadmium (Cd) is an important industrial and environmental pollutant that can produce a wide variety of adverse effects in humans and animals. A growing volume of evidence indicates that the vascular endothelium may be one of the primary targets of Cd toxicity in vivo. Studies over the past 20 years have shown that Cd, at relatively low, sublethal concentrations, can target vascular endothelial cells at a variety of molecular levels, including cell adhesion molecules, metal ion transporters and protein kinase signaling pathways. The purpose of this review is to summarize the results of these recent studies and to discuss the implications of these findings with regard to the mechanisms of Cd toxicity in specific organs including the lung, liver, kidney, testis and heart. In addition the possible roles of the vascular endothelium in mediating the tumor promoting and anticarcinogenic effects of Cd are discussed.


Assuntos
Intoxicação por Cádmio/metabolismo , Cádmio/toxicidade , Endotélio Vascular/efeitos dos fármacos , Animais , Intoxicação por Cádmio/patologia , Endotélio Vascular/patologia , Humanos
20.
Cancer Cell Int ; 5: 24, 2005 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-16053526

RESUMO

BACKGROUND: This study was undertaken to examine the relationship between the tumor suppressor gene p53 and the nuclear signaling protein beta-catenin during bone differentiation. Cross talk between p53 and beta-catenin pathways has been demonstrated and is important during tumorigenesis and DNA damage, where deregulation of beta catenin activates p53. In this study, we used estrogen treatment of osteoblasts as a paradigm to study the relationship between the two proteins during osteoblast differentiation. RESULTS: We exposed osteoblast-like ROS17/2.8 cells to 17-beta estradiol (E2), in a short term assay, and studied the cellular distribution and expression of beta-catenin. We found beta-catenin to be up regulated several fold following E2 treatment. Levels of p53 and its functional activity mirrored the quantitative changes seen in beta-catenin. Alkaline phosphatase, an early marker of osteoblast differentiation, was increased in a manner similar to beta-catenin and p53. In order to determine if there was a direct relationship between alkaline phosphatase expression and beta-catenin, we used two different approaches. In the first approach, treatment with LiCl, which is known to activate beta-catenin, caused a several fold increase in alkaline phosphatase activity. In the second approach, transient transfection of wild type beta-catenin into osteoblasts increased alkaline phosphatase activity two fold over basal levels, showing that beta catenin expression can directly affect alkaline phosphatase expression. However increase in beta catenin activity was not associated with an increase in its signaling activity through TCF/LEF mediated transcription. Immunofluorescence analyses of p53 and beta-catenin localization showed that E2 first caused an increase in cytosolic beta-catenin followed by the accumulation of beta-catenin in the nucleus. Nuclear p53 localization was detected in several cells. Expression of p53 was accompanied by distribution of beta-catenin to the cytoplasm and cell borders. A sub population of cells staining strongly for both proteins appeared to be apoptotic. CONCLUSION: These results suggest that interactions between p53 and beta-catenin signaling pathways may play a key role in osteoblast differentiation and maintenance of tissue homeostasis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA