Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Birth Defects Res ; 109(15): 1228-1235, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28696058

ABSTRACT

BACKGROUND: Up to two-thirds of premature born neonates are treated for infections with aminoglycosides such as gentamicin. Although acute toxicities are well described, there is uncertainty on developmental changes after treatment of premature born neonates. We studied the effect of gentamicin and ceftazidime on kidney development in the rat. Additionally, we evaluated the modulating effect of extrauterine growth restriction. METHODS: On postnatal day (PND) 2, Wistar rats were cross-fostered into normal sized litters (12 pups) or large litters (20 pups) to create normal food (NF) or food restricted (FR) litters to simulate growth restriction and dosed daily intraperitoneally with placebo, 4 mg/kg of gentamicin or 50 mg/kg ceftazidime until PND 8. Gentamicin pharmacokinetics were studied in a separate group of animals. Kidneys were weighed. Renal expression of 18 developmental genes was evaluated by quantitative PCR on PND 8. On PND 35, glomerular number was assessed by stereology and glomerular generations were counted. RESULTS: Food restricted litters showed 22% less body weight compared with controls by day 35 (p < 0.001), 1.4- to 1.5-fold down regulation of Renin, Oat1, and Agtr1a (p < 0.05) expression and a 12% reduction in glomerular numbers (mean 30841 vs. 35187, p < 0.001), whereas glomerular generation count was unaffected. Gentamicin pharmacokinetic parameters were found to be in a human clinical range (mean maximum concentration in plasma of 4.88 mg/L and mean area under the plasma-concentration time curve up to the last measured concentration after 4 hr of 10.71 mg.h/L for sexes combined) and all endpoints were unaffected. Ceftazidime reduced Renin expression by 1.7-fold (p < 0.01). CONCLUSION: Our experiments showed that gentamicin at clinical levels did not disturb kidney development, ceftazidime can affect Renin expression, and extrauterine growth restriction impairs kidney development, but did not modulate potential drug toxicity. Birth Defects Research 109:1228-1235, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Ceftazidime/adverse effects , Gentamicins/adverse effects , Kidney/drug effects , Animals , Animals, Newborn , Body Weight/drug effects , Ceftazidime/metabolism , Ceftazidime/pharmacology , Diet, Reducing , Female , Food , Gentamicins/metabolism , Gentamicins/pharmacology , Kidney/growth & development , Kidney Glomerulus/drug effects , Kidney Glomerulus/growth & development , Litter Size/drug effects , Male , Organogenesis/drug effects , Pregnancy , Rats , Rats, Wistar
2.
Nephron ; 132(2): 110-8, 2016.
Article in English | MEDLINE | ID: mdl-26807737

ABSTRACT

BACKGROUND: Diuretics are administered to neonates to control fluid balance. We studied whether clinical doses affected kidney development and function and whether extrauterine growth retardation (EUGR) could be a modulator. METHODS: Wistar rats were cross-fostered in normal food or food restricted litters at postnatal day (PND) 2 and treated daily with 0.9% NaCl, 5 mg/kg furosemide or 5 mg/kg hydrochlorothiazide (HCTZ) up to PND 8. Kidneys were evaluated on proliferation, apoptosis and a set of mRNA target genes at PND 8, glomerular- and glomerular generation count at PND 35, clinical pathology parameters at 3- and 9 months, neutrophil gelatinase-associated lipocalin at PND 8, 3 and 6 months, monthly blood pressure from 3 months onward and histopathology at study end. RESULTS: Treatment with furosemide or HCTZ did not have relevant effects on measured parameters. EUGR resulted in lower body weight from day 3 onwards (-29% at weaning; p < 0.001, -10% at necropsy; p < 0.001), less glomerular generations (4.4 ± 0.32 vs. 5.0 ± 0.423; p = 0.025, males only), decreased glomerular numbers (27,861 ± 3,468 vs. 30,527 ± 4,096; p = 0.026), higher creatinine clearance (0.84 ± 0.1 vs. 0.77 ± 0.09 ml/min/kg; p = 0.047) at 3 months and lower plasma creatinine (25.7 ± 1.8 vs. 27.5 ± 2.8 µmol/l; p = 0.043) at 9 months. CONCLUSION: Furosemide and HCTZ did not influence kidney development or function when administered in a clinically relevant dose to rat pups at a stage of ongoing nephrogenesis. EUGR led to impaired kidney development but did not modify furosemide or HCTZ findings.


Subject(s)
Diuretics/pharmacology , Kidney/drug effects , Kidney/growth & development , Acute-Phase Proteins/metabolism , Aging/metabolism , Animals , Apoptosis/drug effects , Blood Pressure , Body Weight/drug effects , Cell Proliferation/drug effects , Female , Fetal Growth Retardation/physiopathology , Furosemide/pharmacology , Hydrochlorothiazide/pharmacology , Kidney/physiology , Kidney Glomerulus/cytology , Kidney Glomerulus/drug effects , Kidney Glomerulus/growth & development , Lipocalin-2 , Lipocalins/metabolism , Male , Pregnancy , Proto-Oncogene Proteins/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Sodium Chloride/pharmacology
3.
Birth Defects Res B Dev Reprod Toxicol ; 104(5): 218-26, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26375421

ABSTRACT

BACKGROUND: Prematurely born children with patent ductus arteriosus are treated with ibuprofen or indomethacin, which may inhibit kidney development. We determined whether clinical doses affected kidney development and function, with or without extrauterine growth retardation. METHODS: Wistar rats were cross-fostered in normal food (NF) or food restricted (FR) litters at postnatal day (PND) 2. On PND 3 to 4, three doses of 0.9% NaCl, 0.1 mg/kg indomethacin, or 10 mg/kg ibuprofen were administered via intraperitoneal injection with 12-hr intervals. Kidneys were evaluated for apoptosis, proliferation, and gene expression at PND 8; stereological assessment of nephron number at PND 35; and clinical pathology and neutrophil gelatinase-associated lipocalin at 4 and 9 months. Blood pressure was measured at the ages of 4, 6, and 9 months. RESULTS: NF and FR bodyweight differed from PND 3 onwards, ranging from 16.5 g at weaning (p < 0.001) to 39 g at necropsy (p = 0.019). Kidney proliferation/apoptosis ratios were 7:1 and 3:1 (p = 0.001), respectively and different expression of Wnt4 (0.7x), Oat1 (1.3x), Nphs1 (1.7x), and Aqp4 (1.3x) was noted (but its biological relevance doubted). Nephron numbers were decreased by 12% (p = 0.109) in the ibuprofen-NF group and 7.5% (p = 0.237) in FR groups. This coincided with a tendency to increased neutrophil gelatinase-associated lipocalin at 9 months. No differences were noted in electrolytes, creatinine, or urea clearance. No valid blood pressure results could be obtained. CONCLUSION: A clinical Ibuprofen dose showed potential to inhibit kidney development in neonatal rats. FR did not modulate these effects.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Kidney/growth & development , Organogenesis/drug effects , Animals , Animals, Newborn , Apoptosis/drug effects , Body Weight/drug effects , Cell Proliferation/drug effects , Female , Follow-Up Studies , Gene Expression Regulation, Developmental/drug effects , Kidney/drug effects , Rats, Wistar , Time Factors
4.
Pediatr Res ; 76(6): 508-14, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25167203

ABSTRACT

BACKGROUND: Many premature born neonates receive antibiotic drugs to treat infections, which are applied during active nephrogenesis. We studied the impact of clinical concentrations of gentamicin and alternatives, ceftazidime and meropenem, on ureteric branching. METHODS: Mice metanephroi were dissected at embryonic day 13 and cultured in media with or without various concentrations of gentamicin, ceftazidime, or meropenem. Zero and 24 h kidney size were assessed by surface area measurements, and the ureteric tree was visualized by whole mount staining and confocal microscopy. Branching was evaluated by counting and gene expression levels of Wt1, Sox9, Bmp7, Fgf8, and Gdnf were investigated. RESULTS: A concentration of 2,000 µmol/l ceftazidime impaired ureteric development. In addition, a 4.5-fold and a 2.5-fold downregulation was noted in Fgf8 and Gdnf, respectively. No adverse effects were noted after gentamicin or meropenem treatment. No relationship was noted between surface area expansion and ureteric bud formation, but surface area at explantation related to bud count after 24 h of culture. CONCLUSION: Ceftazidime, but not gentamicin or meropenem reduced ureteric branching in mice and suggest a role for Fgf8 and Gdnf in its mechanism. Metanephros surface area measurements can be used to reduce intra- and inter-litter variation.


Subject(s)
Anti-Bacterial Agents/toxicity , Nephrons/drug effects , Ureter/drug effects , Animals , Ceftazidime/toxicity , Dose-Response Relationship, Drug , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism , Gene Expression Regulation, Developmental , Gentamicins/toxicity , Gestational Age , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Meropenem , Mice, Inbred ICR , Morphogenesis , Nephrons/embryology , Nephrons/metabolism , Thienamycins/toxicity , Tissue Culture Techniques , Ureter/embryology , Ureter/metabolism
5.
Pediatr Nephrol ; 29(11): 2083-91, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24217783

ABSTRACT

The kidney plays a central role in the clearance of drugs. However, renal drug handling entails more than glomerular filtration and includes tubular excretion and reabsorption, and intracellular metabolization by cellular enzyme systems, such as the Cytochrome P450 isoenzymes. All these processes show maturation from birth onwards, which is one of the reasons why drug dosing in children is not simply similar to dosing in small adults. As kidney development normally finishes around the 36th week of gestation, being born prematurely will result in even more immature renal drug handling. Environmental effects, such as extra-uterine growth restriction, sepsis, asphyxia, or drug treatments like caffeine, aminoglycosides, or non-steroidal anti-inflammatory drugs, may further hamper drug handling in the kidney. Dosing in preterm neonates is therefore dependent on many factors that need to be taken into account. Drug treatment may significantly hamper postnatal kidney development in preterm neonates, just like renal immaturity has an impact on drug handling. The restricted kidney development results in a lower number of nephrons that may have several long-term sequelae, such as hypertension, albuminuria, and renal failure. This review focuses on the interplay between drugs and the kidney in premature neonates.


Subject(s)
Infant, Premature/metabolism , Kidney/metabolism , Pharmaceutical Preparations/metabolism , Child , Child, Preschool , Humans , Infant , Infant, Newborn , Infant, Very Low Birth Weight , Kidney/growth & development , Kidney/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...