1
|
Liu M, Yi S, Yu H, Zhang T, Dong F, Zhu L. Underlying Mechanisms for the Sex- and Chemical-Specific Hepatotoxicity of Perfluoroalkyl Phosphinic Acids in Common Carp ( Cyprinus carpio). ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:14515-14525. [PMID: 37728733 DOI: 10.1021/acs.est.3c04964] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
The hepatotoxicities of perfluoroalkyl and polyfluoroalkyl substances (PFASs) have been extensively investigated, while little is known about the sex-specific differences. In this study, common carp were exposed to the emerging perfluoroalkyl phosphinic acids (6:6 and 8:8 PFPiAs) for 14 days to disclose sex-specific hepatotoxicity. Apparent hepatotoxicity, including cell necrosis, apoptosis, and steatosis, was observed in both male and female carp liver. The observed hepatocyte steatosis was predominantly attributed to the dysregulation of hepatic lipid metabolism but was based on sex-specific mechanisms. It was manifested as inhibited oxidative decomposition of fatty acids (FAs) in the female liver, whereas it enhanced the uptake of FAs into the male liver, both of which led to excessive lipid accumulation. Untargeted lipidomics validated that the metabolism pathways of FA, sphingolipid, glycerolipid, and glycerophospholipid were disrupted by both compounds, leading to the generation of reactive oxygen species and oxidative stress. The oxidative stress further evolved into inflammation, manifested as promoted expression of proinflammatory cytokines and repressed expression of anti-inflammatory cytokines. Consistently, all of the changes were more noticeable in male carp, suggesting that male fish were more susceptible to PFPiA disruption. 8:8 PFPiA was less accumulated but caused stronger hepatotoxicity than 6:6 PFPiA, possibly because of the stronger binding capacity of 8:8 PFPiA to nuclear transcription factors mediating lipid metabolism and inflammation. The findings of this study highlight the significance of sex- and chemical-dependent bioaccumulation and the toxicity of PFASs in organisms.
Collapse
Affiliation(s)
- Menglin Liu
- Key Laboratory of Pollution Processes and Environmental Criteria, Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
- School of Environment, Key Laboratory for Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Key Laboratory for Environmental Pollution Control, Henan Normal University, Xinxiang 453007, China
| | - Shujun Yi
- Key Laboratory of Pollution Processes and Environmental Criteria, Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Hao Yu
- School of Environment, Key Laboratory for Yellow River and Huai River Water Environment and Pollution Control, Ministry of Education, Henan Key Laboratory for Environmental Pollution Control, Henan Normal University, Xinxiang 453007, China
| | - Tianxu Zhang
- Key Laboratory of Pollution Processes and Environmental Criteria, Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Fengfeng Dong
- Key Laboratory of Pollution Processes and Environmental Criteria, Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Lingyan Zhu
- Key Laboratory of Pollution Processes and Environmental Criteria, Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| |
Collapse
|
2
|
Liu D, Yan S, Wang P, Chen Q, Liu Y, Cui J, Liang Y, Ren S, Gao Y. Perfluorooctanoic acid (PFOA) exposure in relation to the kidneys: A review of current available literature. Front Physiol 2023; 14:1103141. [PMID: 36776978 PMCID: PMC9909492 DOI: 10.3389/fphys.2023.1103141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/16/2023] [Indexed: 01/27/2023] Open
Abstract
Perfluorooctanoic acid is an artificial and non-degradable chemical. It is widely used due to its stable nature. It can enter the human body through food, drinking water, inhalation of household dust and contact with products containing perfluorooctanoic acid. It accumulates in the human body, causing potential harmful effects on human health. Based on the biodegradability and bioaccumulation of perfluorooctanoic acid in the human body, there are increasing concerns about the adverse effects of perfluorooctanoic acid exposure on kidneys. Research shows that kidney is the main accumulation organ of Perfluorooctanoic acid, and Perfluorooctanoic acid can cause nephrotoxicity and produce adverse effects on kidney function, but the exact mechanism is still unknown. In this review, we summarize the relationship between Perfluorooctanoic acid exposure and kidney health, evaluate risks more clearly, and provide a theoretical basis for subsequent research.
Collapse
Affiliation(s)
- Dongge Liu
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Shuqi Yan
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Pingwei Wang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Qianqian Chen
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Yanping Liu
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Jiajing Cui
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Yujun Liang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Shuping Ren
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Ying Gao
- Department of Endocrinology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
3
|
Puris E, Fricker G, Gynther M. The Role of Solute Carrier Transporters in Efficient Anticancer Drug Delivery and Therapy. Pharmaceutics 2023; 15:pharmaceutics15020364. [PMID: 36839686 PMCID: PMC9966068 DOI: 10.3390/pharmaceutics15020364] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Transporter-mediated drug resistance is a major obstacle in anticancer drug delivery and a key reason for cancer drug therapy failure. Membrane solute carrier (SLC) transporters play a crucial role in the cellular uptake of drugs. The expression and function of the SLC transporters can be down-regulated in cancer cells, which limits the uptake of drugs into the tumor cells, resulting in the inefficiency of the drug therapy. In this review, we summarize the current understanding of low-SLC-transporter-expression-mediated drug resistance in different types of cancers. Recent advances in SLC-transporter-targeting strategies include the development of transporter-utilizing prodrugs and nanocarriers and the modulation of SLC transporter expression in cancer cells. These strategies will play an important role in the future development of anticancer drug therapies by enabling the efficient delivery of drugs into cancer cells.
Collapse
|
4
|
Salem F, Small BG, Johnson TN. Development and application of a pediatric mechanistic kidney model. CPT Pharmacometrics Syst Pharmacol 2022; 11:854-866. [PMID: 35506351 PMCID: PMC9286721 DOI: 10.1002/psp4.12798] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 11/19/2022] Open
Abstract
Pediatric physiologically‐based pharmacokinetic (P‐PBPK) models have been used to predict age related changes in the pharmacokinetics (PKs) of renally cleared drugs mainly in relation to changes in glomerular filtration rate. With emerging data on ontogeny of renal transporters, mechanistic models of renal clearance accounting for the role of active and passive secretion should be developed and evaluated. Data on age‐related physiological changes and ontogeny of renal transporters were applied into a mechanistic kidney within a P‐PBPK model. Plasma concentration–time profile and PK parameters of cimetidine, ciprofloxacin, metformin, tenofovir, and zidovudine were predicted in subjects aged 1 day to 18 years. The predicted and observed plasma concentration–time profiles and PK parameters were compared. The predicted concentration–time profile means and 5th and 95th percent intervals generally captured the observed data and variability in various studies. Overall, based on drugs and age bands, predicted to observed clearance were all within two‐fold and in 11 of 16 cases within 1.5‐fold. Predicted to observed area under the curve (AUC) and maximum plasma concentration (Cmax) were within two‐fold in 12 of 14 and 12 of 15 cases, respectively. Predictions in neonates and early infants (up to 14 weeks postnatal age) were reasonable with 15–20 predicted PK parameters within two‐fold of the observed. ciprofloxacin but not zidovudine PK predictions were sensitive to basal kidney uptake transporter ontogeny. The results indicate that a mechanistic kidney model accounting for physiology and ontogeny of renal processes and transporters can predict the PK of renally excreted drugs in children. Further data especially in neonates are required to verify the model and ontogeny profiles.
Collapse
Affiliation(s)
- Farzaneh Salem
- Drug Metabolism and Pharmacokinetics GlaxoSmithKline R&D Ware UK
| | | | | |
Collapse
|
5
|
Kojima M, Degawa M. Sex, Organ, and Breed Differences in the mRNA Expression of Drug Transporters in the Liver and Kidney of Pigs. Biol Pharm Bull 2022; 45:508-516. [DOI: 10.1248/bpb.b21-01033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Misaki Kojima
- Meat Animal Biosystem Group, Division of Meat Animal and Poultry Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO)
| | - Masakuni Degawa
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
6
|
Weyrich A, Frericks M, Eichenlaub M, Schneider S, Hofmann T, Van Cruchten S, van Ravenzwaay B. Ontogeny of renal, hepatic, and placental expression of ATP-binding cassette and solute carrier transporters in the rat and the rabbit. Reprod Toxicol 2022; 107:1-9. [PMID: 34757165 DOI: 10.1016/j.reprotox.2021.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 09/14/2021] [Accepted: 10/07/2021] [Indexed: 02/08/2023]
Abstract
Species differences in developmental toxicity can be due to varying expression of xenobiotic transporters. Hence, knowledge on the ontogeny of these transporters, especially in human, rat and rabbit, is pivotal. Two superfamilies of transporters, the ATP-binding cassette (ABC) and the solute carrier (SLC) transporters, are well known for their role in the absorption, distribution and/or elimination of xenobiotics and endogenous substances. The aim of this study was to compare the expression levels of these xenobiotic transporters in liver, kidney and placenta of man, Wistar rat and New Zealand White rabbit during pre- and postnatal development. For this purpose, qPCR experiments were performed for rat and rabbit tissues and the gene expression profiles were compared with literature data from man, rat and rabbit. Data analysis showed large differences in transporter expression in development and between species. These results can be used to better understand developmental toxicity findings in non-clinical species and their relevance for man.
Collapse
Affiliation(s)
- Anastasia Weyrich
- Experimental Toxicology and Ecology, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany.
| | - Markus Frericks
- Experimental Toxicology and Ecology, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany
| | - Michael Eichenlaub
- Bioscience Research, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany
| | - Steffen Schneider
- Experimental Toxicology and Ecology, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany
| | - Thomas Hofmann
- Experimental Toxicology and Ecology, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany
| | - Steven Van Cruchten
- Department of Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Bennard van Ravenzwaay
- Experimental Toxicology and Ecology, BASF SE, Carl-Bosch-Straße 38, 67056, Ludwigshafen, Germany
| |
Collapse
|
7
|
Pou Casellas C, Jansen K, Rookmaaker MB, Clevers H, Verhaar MC, Masereeuw R. Regulation of Solute Carriers OCT2 and OAT1/3 in the Kidney: A Phylogenetic, Ontogenetic and Cell Dynamic Perspective. Physiol Rev 2021; 102:993-1024. [PMID: 34486394 DOI: 10.1152/physrev.00009.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Over the course of more than 500 million years, the kidneys have undergone a remarkable evolution from primitive nephric tubes to intricate filtration-reabsorption systems that maintain homeostasis and remove metabolic end products from the body. The evolutionarily conserved solute carriers Organic Cation Transporter 2 (OCT2), and Organic Anion Transporters 1 and 3 (OAT1/3) coordinate the active secretion of a broad range of endogenous and exogenous substances, many of which accumulate in the blood of patients with kidney failure despite dialysis. Harnessing OCT2 and OAT1/3 through functional preservation or regeneration could alleviate the progression of kidney disease. Additionally, it would improve current in vitro test models that lose their expression in culture. With this review, we explore OCT2 and OAT1/3 regulation using different perspectives: phylogenetic, ontogenetic and cell dynamic. Our aim is to identify possible molecular targets to both help prevent or compensate for the loss of transport activity in patients with kidney disease, and to enable endogenous OCT2 and OAT1/3 induction in vitro in order to develop better models for drug development.
Collapse
Affiliation(s)
- Carla Pou Casellas
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands.,Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
| | - Katja Jansen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Maarten B Rookmaaker
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Hans Clevers
- Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
8
|
Frazier KS. The Impact of Functional and Structural Maturation of the Kidney on Susceptibility to Drug and Chemical Toxicity in Neonatal Rodents. Toxicol Pathol 2021; 49:1377-1388. [PMID: 34338059 DOI: 10.1177/01926233211035683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Drug responses are often unpredictable in juvenile animal toxicity studies; hence, optimizing dosages is challenging. Renal functional differences based on age of development will often result in vastly different toxicologic responses. Developmental changes in renal function can alter plasma clearance of compounds with extensive renal elimination. Absorption, distribution, metabolism, and excretion of drugs vary depending on animal age and kidney maturation. Toxicity can result in malformations or renal degeneration. Although renal morphologic development in humans generally occurs in utero, maximal levels of tubular secretion, acid-base equilibrium, concentrating ability, or glomerular filtration rate (GFR) are reached postnatally in humans and animals and subject to drug effects. Maturation of renal metabolism and transporters occurs postnatally and plays a critical role in detoxification and excretion. Maturation times must be considered when designing juvenile toxicity studies and may require cohorts of animals of specific ages to achieve optimal dosing schemes and toxicokinetics. In recent years, critical end points and windows of susceptibility have been established comparatively between species to better model pharmacokinetics and understand pediatric nephrotoxicity. There are examples of agents where toxicity is enhanced in neonates, others where it is diminished, and others where rat nephrotoxicity is expressed as juvenile toxicity, but in humans as gestational toxicity.
Collapse
|
9
|
Commander SJ, Wu H, Boakye-Agyeman F, Melloni C, Hornik CD, Zimmerman K, Al-Uzri A, Mendley SR, Harper B, Cohen-Wolkowiez M, Hornik CP. Pharmacokinetics of Hydrochlorothiazide in Children: A Potential Surrogate for Renal Secretion Maturation. J Clin Pharmacol 2021; 61:368-377. [PMID: 33029806 PMCID: PMC8232568 DOI: 10.1002/jcph.1739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 08/24/2020] [Indexed: 12/26/2022]
Abstract
Hydrochlorothiazide (HCTZ) is a thiazide diuretic used in adults and children for the treatment of hypertension and edema. The pharmacokinetic (PK) properties of HCTZ in children are not well characterized, particularly among children with obesity who frequently suffer from hypertension and may, therefore, benefit from HCTZ therapy. HCTZ is excreted in the kidney via organic anion transporters 1 and 3 (OAT1 and OAT3). The ontogeny of OAT1 and OAT3 remain unknown, but HCTZ clearance may serve as a surrogate marker of OAT1 and OAT3 maturation. Population PK modeling was performed in NONMEM, and the model was leveraged to conduct dose-exposure simulations. This study examined 83 plasma samples from 49 participants (69% male) taking enteral HCTZ. The median (range) postnatal age was 6.7 years (0.03-19.5 years), and 17 (34%) participants were obese or morbidly obese. The median (range) dose of HCTZ was 0.654 mg/kg (0.11-1.8 kg) and the median number of doses recorded per participant was 5 (1-8). HCTZ PK was well characterized by a 1-compartment PK model. Body weight and a maturation model based on postmenstrual age were significant covariates for apparent clearance, but the presence of obesity was not. Dosing simulations were performed with a standardized 1mg/kg. Simulated exposure (area under the curve and maximum HCTZ concentrations) decreased with age and was likely due to older children receiving the maximum absolute doses of HCTZ. Further studies with more patients in each age group are required to confirm these PK findings of HCTZ in the children.
Collapse
Affiliation(s)
- Sarah Jane Commander
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Huali Wu
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Felix Boakye-Agyeman
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Chiara Melloni
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Chi Dang Hornik
- Duke Clinical Research Institute, Durham, North Carolina, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Kanecia Zimmerman
- Duke Clinical Research Institute, Durham, North Carolina, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Amira Al-Uzri
- Department of Pediatrics, Oregon Health and Science University, Portland, Oregon, USA
| | - Susan R Mendley
- Department of Pediatrics and Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Barrie Harper
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Michael Cohen-Wolkowiez
- Duke Clinical Research Institute, Durham, North Carolina, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Christoph P Hornik
- Duke Clinical Research Institute, Durham, North Carolina, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
10
|
Chung SM, Moon JS, Yoon JS, Won KC, Lee HW. The sex-specific effects of blood lead, mercury, and cadmium levels on hepatic steatosis and fibrosis: Korean nationwide cross-sectional study. J Trace Elem Med Biol 2020; 62:126601. [PMID: 32634767 DOI: 10.1016/j.jtemb.2020.126601] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/02/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022]
Abstract
AIM The potential effects of heavy metals on non-alcoholic fatty liver disease (NAFLD) remain unknown. We investigated the sex-specific relationships of blood lead (BPb), mercury (BHg), and cadmium (BCd) levels with hepatic steatosis (HS) and fibrosis (HF). METHOD We included 4420 participants from the 2016-2017 Korea National Health and Nutrition Examination Survey. High-risk alcoholics and patients with chronic hepatitis B or C infections or liver cirrhosis were excluded. We calculated the hepatic steatosis index (HSI) and fibrosis-4 index (FIB-4) values; we defined the presence of HS and HF as an HSI ≥ 36 and FIB-4 score >2.67, respectively. We adjusted for age, smoking and alcohol consumption statuses, hypertension, obesity, diabetes, hypertriglyceridemia, and BPb, BHg, and BCd levels. RESULT In males (n = 1860), the HSI was correlated negatively with the BPb level and positively with the BHg level (both p < 0.01). The FIB-4 score was correlated positively with the BPb and BCd levels (both p < 0.01). In females (n = 2560), the HSI and FIB-4 score were correlated positively with the BPb, BHg, and BCd levels (all p < 0.01). After adjustments, the BHg level increased the risk of HS in both males (OR = 1.065, p = 0.003) and females (OR = 1.061, p = 0.048), and the BCd level increased the risk of HF in females (OR = 1.668, p = 0.012). CONCLUSION Blood heavy metal levels were generally correlated positively with the HSI and FIB4 score, more so in females than males. The BHg level was associated with HS in males and females, and the BCd level was associated with HF in females. Further studies on NAFLD progression according to heavy metal status and sex are warranted.
Collapse
Affiliation(s)
- Seung Min Chung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeungnam College of Medicine, Daegu, Republic of Korea.
| | - Jun Sung Moon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeungnam College of Medicine, Daegu, Republic of Korea.
| | - Ji Sung Yoon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeungnam College of Medicine, Daegu, Republic of Korea.
| | - Kyu Chang Won
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeungnam College of Medicine, Daegu, Republic of Korea.
| | - Hyoung Woo Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeungnam College of Medicine, Daegu, Republic of Korea.
| |
Collapse
|
11
|
Mai Y, Ashiru-Oredope DA, Yao Z, Dou L, Madla CM, Taherali F, Murdan S, Basit AW. Boosting drug bioavailability in men but not women through the action of an excipient. Int J Pharm 2020; 587:119678. [DOI: 10.1016/j.ijpharm.2020.119678] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 12/11/2022]
|
12
|
Bueters R, Bael A, Gasthuys E, Chen C, Schreuder MF, Frazier KS. Ontogeny and Cross-species Comparison of Pathways Involved in Drug Absorption, Distribution, Metabolism, and Excretion in Neonates (Review): Kidney. Drug Metab Dispos 2020; 48:353-367. [PMID: 32114509 DOI: 10.1124/dmd.119.089755] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 02/04/2020] [Indexed: 02/13/2025] Open
Abstract
The kidneys play an important role in many processes, including urine formation, water conservation, acid-base equilibrium, and elimination of waste. The anatomic and functional development of the kidney has different maturation time points in humans versus animals, with critical differences between species in maturation before and after birth. Absorption, distribution, metabolism, and excretion (ADME) of drugs vary depending on age and maturation, which will lead to differences in toxicity and efficacy. When neonate/juvenile laboratory animal studies are designed, a thorough knowledge of the differences in kidney development between newborns/children and laboratory animals is essential. The human and laboratory animal data must be combined to obtain a more complete picture of the development in the kidneys around the neonatal period and the complexity of ADME in newborns and children. This review examines the ontogeny and cross-species differences in ADME processes in the developing kidney in preterm and term laboratory animals and children. It provides an overview of insights into ADME functionality in the kidney by identifying what is currently known and which gaps still exist. Currently important renal function properties such as glomerular filtration rate, renal blood flow, and ability to concentrate are generally well known, while detailed knowledge about transporter and metabolism maturation is growing but is still lacking. Preclinical data in those properties is limited to rodents and generally covers only the expression levels of transporter or enzyme-encoding genes. More knowledge on a functional level is needed to predict the kinetics and toxicity in neonate/juvenile toxicity and efficacy studies. SIGNIFICANCE STATEMENT: This review provides insight in cross-species developmental differences of absorption, distribution, metabolism, and excretion properties in the kidney, which should be considered in neonate/juvenile study interpretation, hypotheses generation, and experimental design.
Collapse
Affiliation(s)
- Ruud Bueters
- Janssen Research & Development, a division of Janssen Pharmaceutica NV, Division of Discovery, Product Development & Supply, Department of Nonclinical Safety, Beerse, Belgium (R.B.); ZNA Queen Paola Children's Hospital, Department of Pediatric Nephrology, Antwerp, Belgium (A.B.); Department of Paediatrics, Faculty of Medicine, Ghent University, Gent, Belgium (E.G.); Health and Environmental Sciences Institute, Washington, DC (C.C.); Radboudumc Amalia Children's Hospital, Department of Pediatric Nephrology, Nijmegen, The Netherlands (M.F.S.); and GlaxoSmithKline, Collegeville, Pennsylvania (K.S.F.)
| | - An Bael
- Janssen Research & Development, a division of Janssen Pharmaceutica NV, Division of Discovery, Product Development & Supply, Department of Nonclinical Safety, Beerse, Belgium (R.B.); ZNA Queen Paola Children's Hospital, Department of Pediatric Nephrology, Antwerp, Belgium (A.B.); Department of Paediatrics, Faculty of Medicine, Ghent University, Gent, Belgium (E.G.); Health and Environmental Sciences Institute, Washington, DC (C.C.); Radboudumc Amalia Children's Hospital, Department of Pediatric Nephrology, Nijmegen, The Netherlands (M.F.S.); and GlaxoSmithKline, Collegeville, Pennsylvania (K.S.F.)
| | - Elke Gasthuys
- Janssen Research & Development, a division of Janssen Pharmaceutica NV, Division of Discovery, Product Development & Supply, Department of Nonclinical Safety, Beerse, Belgium (R.B.); ZNA Queen Paola Children's Hospital, Department of Pediatric Nephrology, Antwerp, Belgium (A.B.); Department of Paediatrics, Faculty of Medicine, Ghent University, Gent, Belgium (E.G.); Health and Environmental Sciences Institute, Washington, DC (C.C.); Radboudumc Amalia Children's Hospital, Department of Pediatric Nephrology, Nijmegen, The Netherlands (M.F.S.); and GlaxoSmithKline, Collegeville, Pennsylvania (K.S.F.)
| | - Connie Chen
- Janssen Research & Development, a division of Janssen Pharmaceutica NV, Division of Discovery, Product Development & Supply, Department of Nonclinical Safety, Beerse, Belgium (R.B.); ZNA Queen Paola Children's Hospital, Department of Pediatric Nephrology, Antwerp, Belgium (A.B.); Department of Paediatrics, Faculty of Medicine, Ghent University, Gent, Belgium (E.G.); Health and Environmental Sciences Institute, Washington, DC (C.C.); Radboudumc Amalia Children's Hospital, Department of Pediatric Nephrology, Nijmegen, The Netherlands (M.F.S.); and GlaxoSmithKline, Collegeville, Pennsylvania (K.S.F.)
| | - Michiel F Schreuder
- Janssen Research & Development, a division of Janssen Pharmaceutica NV, Division of Discovery, Product Development & Supply, Department of Nonclinical Safety, Beerse, Belgium (R.B.); ZNA Queen Paola Children's Hospital, Department of Pediatric Nephrology, Antwerp, Belgium (A.B.); Department of Paediatrics, Faculty of Medicine, Ghent University, Gent, Belgium (E.G.); Health and Environmental Sciences Institute, Washington, DC (C.C.); Radboudumc Amalia Children's Hospital, Department of Pediatric Nephrology, Nijmegen, The Netherlands (M.F.S.); and GlaxoSmithKline, Collegeville, Pennsylvania (K.S.F.)
| | - Kendall S Frazier
- Janssen Research & Development, a division of Janssen Pharmaceutica NV, Division of Discovery, Product Development & Supply, Department of Nonclinical Safety, Beerse, Belgium (R.B.); ZNA Queen Paola Children's Hospital, Department of Pediatric Nephrology, Antwerp, Belgium (A.B.); Department of Paediatrics, Faculty of Medicine, Ghent University, Gent, Belgium (E.G.); Health and Environmental Sciences Institute, Washington, DC (C.C.); Radboudumc Amalia Children's Hospital, Department of Pediatric Nephrology, Nijmegen, The Netherlands (M.F.S.); and GlaxoSmithKline, Collegeville, Pennsylvania (K.S.F.)
| |
Collapse
|
13
|
Gu YZ, Chu X, Houle R, Vlasakova K, Koeplinger KA, Bourgeois I, Palyada K, Anderson KD, Brynczka C, Bhatt B, Chen F, Smith R, Amin R, Glaab WE, Lebron J, Cox K, Sistare FD. Polyethlyene Glycol 200 can Protect Rats against Drug-Induced Kidney Toxicity through Inhibition of the Renal Organic Anion Transporter 3. Toxicol Sci 2019; 172:155-166. [PMID: 31406999 DOI: 10.1093/toxsci/kfz186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/18/2019] [Accepted: 07/19/2019] [Indexed: 01/15/2023] Open
Abstract
MK-7680, a cyclic nucleotide prodrug, caused significant kidney tubule injury in female rats when administered orally at 1000 mg/kg/day for 2 weeks using 10% Polysorbate 80 as vehicle. However, kidney injury was absent when MK-7680 was administered at the same dose regimen using 100% Polyethylene Glycol 200 (PEG 200) as the vehicle. Subsequent investigations revealed that MK-7680 triphosphate concentrations in kidney were much lower in rats treated with MK-7680 using PEG 200 compared to 10% Polysorbate 80 vehicle while plasma exposures of MK-7680 prodrug were similar. In vitro studies demonstrated that PEG 200 is an inhibitor of human renal uptake transporter organic anion transporter 3 (OAT3), of which MK-7680 is a substrate. Furthermore, PEG 200 and PEG 400 were found to interfere in vitro with human renal transporters OAT3, organic cation transporter (OCT) 2, multidrug resistance-associated protein (MRP) 2 and 4, and multidrug and toxin extrusion protein (MATE) 1 and 2K, but not OAT1. These results support a conclusion that PEG 200 may prevent MK-7680-induced kidney injury by inhibiting its active uptake into proximal tubular cells by OAT3. Caution should be exercised therefore when using PEGs as vehicles for toxicity assessment for compounds that are substrates of renal transporters.
Collapse
Affiliation(s)
- Yi-Zhong Gu
- Safety Assessment and Laboratory Animal Resources, Riom, France
| | - Xiaoyan Chu
- Pharmacokinetics, Pharmacodynamics and Metabolism, Riom, France
| | - Robert Houle
- Pharmacokinetics, Pharmacodynamics and Metabolism, Riom, France
| | | | | | - Isabelle Bourgeois
- Safety Assessment and Laboratory Animal Resources, Riom, France.,Merck & Co., Inc., West Point, PA 19486, USA; MSD, Riom, France
| | - Kiran Palyada
- Safety Assessment and Laboratory Animal Resources, Riom, France
| | | | | | - Bhavana Bhatt
- Safety Assessment and Laboratory Animal Resources, Riom, France
| | - Feifei Chen
- Safety Assessment and Laboratory Animal Resources, Riom, France
| | - Roger Smith
- Safety Assessment and Laboratory Animal Resources, Riom, France
| | - Rupesh Amin
- Safety Assessment and Laboratory Animal Resources, Riom, France
| | - Warren E Glaab
- Safety Assessment and Laboratory Animal Resources, Riom, France
| | - Jose Lebron
- Safety Assessment and Laboratory Animal Resources, Riom, France
| | - Kathleen Cox
- Pharmacokinetics, Pharmacodynamics and Metabolism, Riom, France
| | - Frank D Sistare
- Safety Assessment and Laboratory Animal Resources, Riom, France
| |
Collapse
|
14
|
Shi G, Guo H, Sheng N, Cui Q, Pan Y, Wang J, Guo Y, Dai J. Two-generational reproductive toxicity assessment of 6:2 chlorinated polyfluorinated ether sulfonate (F-53B, a novel alternative to perfluorooctane sulfonate) in zebrafish. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 243:1517-1527. [PMID: 30292160 DOI: 10.1016/j.envpol.2018.09.120] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/24/2018] [Accepted: 09/24/2018] [Indexed: 06/08/2023]
Abstract
As an alternative to perfluorooctane sulfonate (PFOS), 6:2 chlorinated polyfluorinated ether sulfonate (commercial name: F-53B) has been used in the Chinese chrome plating industry for over four decades. It has been increasingly detected in environmental matrices in recent years, causing great concern regarding its potential health risks to humans and wildlife. However, its adverse effects on biota remain largely unknown. To explore the chronic toxicity of F-53B on reproduction, a two-generational study was conducted using zebrafish (Danio rerio). Adult zebrafish (F0 generation) were chronically exposed to different concentrations of F-53B (0, 5, 50, and 500 μg/L) for 180 d using a flow-through exposure system, with F1 and F2 generations reared without exposure. The reproductive toxicity endpoints were assessed in F0 and F1 adult fish. Results showed that F-53B accumulated in the F0 gonads and transferred to the F1 generation via maternal eggs, and even remained in F1 adult fish and their eggs (F2) after 180 d depuration. In the F0 generation, F-53B exposure significantly inhibited growth and induced reproductive toxicity, including decreased gonadosomatic index and egg production/female, changes in the histological structure of the gonads, and increased serum testosterone levels. In particular, serum estradiol and vitellogenin levels were significantly increased in 5 μg/L F-53B-exposed adult males. The transcriptional levels of several genes along the hypothalamic-pituitary-gonadal axis were altered in F0 generation fish. Testis transcriptome analysis revealed that F-53B exposure disrupted spermatogenesis in F0 male zebrafish. Maternal transfer of F-53B also induced adverse effects on growth and reproduction in the F1 generation. Furthermore, the higher occurrence of malformation and lower survival in F1 and F2 embryos indicated that parental exposure to F-53B could impair the embryonic development of offspring. Taken together, this study demonstrated that F-53B could induce reproductive toxicity in zebrafish similar to that induced by legacy PFOS, and its potential adverse effects on offspring deserve further investigation.
Collapse
Affiliation(s)
- Guohui Shi
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hua Guo
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Nan Sheng
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qianqian Cui
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yitao Pan
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jinxing Wang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yong Guo
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Jiayin Dai
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
15
|
Knutsen HK, Alexander J, Barregård L, Bignami M, Brüschweiler B, Ceccatelli S, Cottrill B, Dinovi M, Edler L, Grasl-Kraupp B, Hogstrand C, Hoogenboom LR, Nebbia CS, Oswald IP, Petersen A, Rose M, Roudot AC, Vleminckx C, Vollmer G, Wallace H, Bodin L, Cravedi JP, Halldorsson TI, Haug LS, Johansson N, van Loveren H, Gergelova P, Mackay K, Levorato S, van Manen M, Schwerdtle T. Risk to human health related to the presence of perfluorooctane sulfonic acid and perfluorooctanoic acid in food. EFSA J 2018; 16:e05194. [PMID: 32625773 PMCID: PMC7009575 DOI: 10.2903/j.efsa.2018.5194] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The European Commission asked EFSA for a scientific evaluation on the risks to human health related to the presence of perfluorooctane sulfonic acid (PFOS) and perfluorooctanoic acid (PFOA) in food. Regarding PFOS and PFOA occurrence, the final data set available for dietary exposure assessment contained a total of 20,019 analytical results (PFOS n = 10,191 and PFOA n = 9,828). There were large differences between upper and lower bound exposure due to analytical methods with insufficient sensitivity. The CONTAM Panel considered the lower bound estimates to be closer to true exposure levels. Important contributors to the lower bound mean chronic exposure were 'Fish and other seafood', 'Meat and meat products' and 'Eggs and egg products', for PFOS, and 'Milk and dairy products', 'Drinking water' and 'Fish and other seafood' for PFOA. PFOS and PFOA are readily absorbed in the gastrointestinal tract, excreted in urine and faeces, and do not undergo metabolism. Estimated human half-lives for PFOS and PFOA are about 5 years and 2-4 years, respectively. The derivation of a health-based guidance value was based on human epidemiological studies. For PFOS, the increase in serum total cholesterol in adults, and the decrease in antibody response at vaccination in children were identified as the critical effects. For PFOA, the increase in serum total cholesterol was the critical effect. Also reduced birth weight (for both compounds) and increased prevalence of high serum levels of the liver enzyme alanine aminotransferase (ALT) (for PFOA) were considered. After benchmark modelling of serum levels of PFOS and PFOA, and estimating the corresponding daily intakes, the CONTAM Panel established a tolerable weekly intake (TWI) of 13 ng/kg body weight (bw) per week for PFOS and 6 ng/kg bw per week for PFOA. For both compounds, exposure of a considerable proportion of the population exceeds the proposed TWIs.
Collapse
|
16
|
FEMA GRAS assessment of natural flavor complexes: Citrus-derived flavoring ingredients. Food Chem Toxicol 2018; 124:192-218. [PMID: 30481573 DOI: 10.1016/j.fct.2018.11.052] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 11/19/2018] [Accepted: 11/23/2018] [Indexed: 01/06/2023]
Abstract
In 2015, the Expert Panel of the Flavor and Extract Manufacturers Association (FEMA) initiated a re-evaluation of the safety of over 250 natural flavor complexes (NFCs) used as flavoring ingredients. This publication is the first in a series and summarizes the evaluation of 54 Citrus-derived NFCs using the procedure outlined in Smith et al. (2005) and updated in Cohen et al. (2018) to evaluate the safety of naturally-occurring mixtures for their intended use as flavoring ingredients. The procedure relies on a complete chemical characterization of each NFC intended for commerce and organization of each NFC's chemical constituents into well-defined congeneric groups. The safety of the NFC is evaluated using the well-established and conservative threshold of toxicological concern (TTC) concept in addition to data on absorption, metabolism and toxicology of members of the congeneric groups and the NFC under evaluation. As a result of the application of the procedure, 54 natural flavor complexes derived from botanicals of the Citrus genus were affirmed as generally recognized as safe (GRAS) under their conditions of intended use as flavoring ingredients based on an evaluation of each NFC and the constituents and congeneric groups therein.
Collapse
|
17
|
Organic solute carrier 22 (SLC22) family: Potential for interactions with food, herbal/dietary supplements, endogenous compounds, and drugs. J Food Drug Anal 2018; 26:S45-S60. [PMID: 29703386 PMCID: PMC9326878 DOI: 10.1016/j.jfda.2018.03.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 02/07/2023] Open
|
18
|
Cui Q, Pan Y, Zhang H, Sheng N, Wang J, Guo Y, Dai J. Occurrence and Tissue Distribution of Novel Perfluoroether Carboxylic and Sulfonic Acids and Legacy Per/Polyfluoroalkyl Substances in Black-Spotted Frog (Pelophylax nigromaculatus). ENVIRONMENTAL SCIENCE & TECHNOLOGY 2018; 52:982-990. [PMID: 29310433 DOI: 10.1021/acs.est.7b03662] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Research on perfluoroalkyl substances (PFASs) continues to grow. However, very little is known about these substances in amphibians. Here we report for the first time on the occurrence, tissue distribution, and bioaccumulation of two novel PFASs, chlorinated polyfluorinated ether sulfonic acid (6:2 Cl-PFESA) and hexafluoropropylene oxide trimer acid (HFPO-TA), in the black-spotted frog (Pelophylax nigromaculatus) from China. Frogs from cities with large-scale fluorochemical industries had significantly greater liver ∑PFAS levels (mean 54.28 ng/g in Changshu; 31.22 ng/g in Huantai) than those from cities without similar industry (9.91 ng/g in Zhoushan; 7.68 ng/g in Quzhou). Females had significantly lower liver PFAS levels than males, and older frogs tended to have lower PFAS levels than younger frogs. Skin, liver, and muscle contributed nearly 80% to the whole body burden of 6:2 Cl-PFESA in males, whereas the female ovary alone accounted for 58.4%. These results suggest substantial maternal transfer of 6:2 Cl-PFESA to eggs, raising concern regarding its developmental toxicity on frogs and other species. The bioaccumulation factor results (6:2 Cl-PFESA > PFOS; HFPO-TA > PFOA) suggest a stronger accumulative potential in the black-spotted frog for these alternative substances compared to their predecessors. Future studies on their toxicity and ecology risk are warranted.
Collapse
Affiliation(s)
- Qianqian Cui
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing 100101, China
- University of Chinese Academy of Sciences , Beijing 100049, P. R. China
| | - Yitao Pan
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing 100101, China
| | - Hongxia Zhang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing 100101, China
| | - Nan Sheng
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing 100101, China
| | - Jianshe Wang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing 100101, China
| | - Yong Guo
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences , Shanghai 200032, P. R. China
| | - Jiayin Dai
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing 100101, China
| |
Collapse
|
19
|
Soukup ST, Müller DR, Kurrat A, Diel P, Kulling SE. Influence of testosterone on phase II metabolism and availability of soy isoflavones in male Wistar rats. Arch Toxicol 2017; 91:1649-1661. [PMID: 27743010 DOI: 10.1007/s00204-016-1853-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 09/15/2016] [Indexed: 01/16/2023]
Abstract
Genistein and daidzein are the main isoflavones in soy. Their potential beneficial or adverse effects in males like the prevention of prostate cancer or the impact on reproductive functions are controversially discussed. Major determinants of their bioactivity are the absorption and biotransformation of isoflavones. In this study, we focused on the influence of testosterone on plasma availability and phase II metabolism of isoflavones. Male Wistar rats, receiving an isoflavones rich diet, were randomized into three groups: Two groups were orchiectomized (ORX) at postnatal day (PND) 80 and treated for 11 days with testosterone propionate (TP) (ORX TP group) or a vehicle (ORX group) after a 7 days lasting hormonal decline. The third group served as control and remained intact. Rats were sacrificed at PND 98. ORX rats had reduced isoflavones plasma levels. Differently regulated mRNA expressions of transporters relevant for transport of phase II metabolites in liver and kidney may be responsible for this reduction, more precisely Slc10a1 and Slc21a1 in kidney as well as Slc22a8 in liver. While main phase II metabolites in intact rats were disulfates and sulfoglucuronides, the amount of sulfate conjugates was significantly diminished by ORX. In accordance with that, mRNA expression of different sulfotransferases was reduced in liver by ORX. The observed effects could be almost restored by TP treatment. In conclusion, testosterone, and likely further androgens, has a huge impact on phase II metabolism and availability of isoflavones by influencing the expression of different sulfotransferases and transporters.
Collapse
Affiliation(s)
- Sebastian T Soukup
- Department of Safety and Quality of Fruit and Vegetables, Max Rubner-Institut, Haid-und-Neu-Straße 9, 76131, Karlsruhe, Germany
| | - Dennis R Müller
- Department of Molecular and Cellular Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Anne Kurrat
- Department of Molecular and Cellular Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Patrick Diel
- Department of Molecular and Cellular Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Sabine E Kulling
- Department of Safety and Quality of Fruit and Vegetables, Max Rubner-Institut, Haid-und-Neu-Straße 9, 76131, Karlsruhe, Germany.
| |
Collapse
|
20
|
Shen H, Lai Y, Rodrigues AD. Organic Anion Transporter 2: An Enigmatic Human Solute Carrier. Drug Metab Dispos 2017; 45:228-236. [PMID: 27872146 DOI: 10.1124/dmd.116.072264] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 11/17/2016] [Indexed: 12/28/2022] Open
Abstract
As a member of the solute carrier 22A (SLC22A) family, organic anion transporter 2 (OAT2; SLC22A7) is emerging as an important drug transporter because of its expression in both the liver and kidney, two major eliminating organs, and its ability to transport not only a wide variety of xenobiotics but also numerous physiologically important endogenous compounds, like creatinine and cGMP. However, OAT2 has received relatively little attention compared with other OATs and solute carriers (SLCs), like organic cation transporters, sodium-dependent taurocholate cotransporting polypeptide, multidrug and toxin extrusion proteins, and organic anion-transporting polypeptides. Overall, the literature describing OAT2 is rapidly evolving, with numerous publications contradicting each other regarding the transport mechanism, tissue distribution, and transport of creatinine and cGMP, two important endogenous OAT2 substrates. Despite its status as a liver and kidney SLC, tools for assessing its activity and inhibition are lacking, and its role in drug disposition and elimination remains to be defined. The current review focuses on the available and emerging literature describing OAT2. We envision that OAT2 will gain more prominence as its expression, substrate, and inhibitor profile is investigated further and compared with other SLCs.
Collapse
Affiliation(s)
- Hong Shen
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (H.S., Y.L.), and Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer World Wide Research and Development, Groton, Connecticut (A.D.R.)
| | - Yurong Lai
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (H.S., Y.L.), and Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer World Wide Research and Development, Groton, Connecticut (A.D.R.)
| | - A David Rodrigues
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (H.S., Y.L.), and Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer World Wide Research and Development, Groton, Connecticut (A.D.R.)
| |
Collapse
|
21
|
Xu YJ, Wang Y, Lu YF, Xu SF, Wu Q, Liu J. Age-associated differences in transporter gene expression in kidneys of male rats. Mol Med Rep 2016; 15:474-482. [DOI: 10.3892/mmr.2016.5970] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 10/18/2016] [Indexed: 11/06/2022] Open
|
22
|
Breljak D, Ljubojević M, Hagos Y, Micek V, Balen Eror D, Vrhovac Madunić I, Brzica H, Karaica D, Radović N, Kraus O, Anzai N, Koepsell H, Burckhardt G, Burckhardt BC, Sabolić I. Distribution of organic anion transporters NaDC3 and OAT1-3 along the human nephron. Am J Physiol Renal Physiol 2016; 311:F227-38. [PMID: 27053689 DOI: 10.1152/ajprenal.00113.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 03/30/2016] [Indexed: 01/13/2023] Open
Abstract
The initial step in renal secretion of organic anions (OAs) is mediated by transporters in the basolateral membrane (BLM). Contributors to this process are primary active Na(+)-K(+)-ATPase (EC 3.6.3.9), secondary active Na(+)-dicarboxylate cotransporter 3 (NaDC3/SLC13A3), and tertiary active OA transporters (OATs) OAT1/SLC22A6, OAT2/SLC22A7, and OAT3/SLC22A8. In human kidneys, we analyzed the localization of these transporters by immunochemical methods in tissue cryosections and isolated membranes. The specificity of antibodies was validated with human embryonic kidney-293 cells stably transfected with functional OATs. Na(+)-K(+)-ATPase was immunolocalized to the BLM along the entire human nephron. NaDC3-related immunostaining was detected in the BLM of proximal tubules and in the BLM and/or luminal membrane of principal cells in connecting segments and collecting ducts. The thin and thick ascending limbs, macula densa, and distal tubules exhibited no reactivity with the anti-NaDC3 antibody. OAT1-OAT3-related immunostaining in human kidneys was detected only in the BLM of cortical proximal tubules; all three OATs were stained more intensely in S1/S2 segments compared with S3 segment in medullary rays, whereas the S3 segment in the outer stripe remained unstained. Expression of NaDC3, OAT1, OAT2, and OAT3 proteins exhibited considerable interindividual variability in both male and female kidneys, and sex differences in their expression could not be detected. Our experiments provide a side-by-side comparison of basolateral transporters cooperating in renal OA secretion in the human kidney.
Collapse
Affiliation(s)
- Davorka Breljak
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Marija Ljubojević
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Yohannes Hagos
- Center of Physiology and Pathophysiology, Institute of Systemic Physiology and Pathophysiology, University of Göttingen, Göttingen, Germany
| | - Vedran Micek
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Daniela Balen Eror
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Ivana Vrhovac Madunić
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Hrvoje Brzica
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Dean Karaica
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Nikola Radović
- Department of Urology, Clinical Hospital Dubrava, Zagreb, Croatia
| | - Ognjen Kraus
- University Hospital Sisters of Mercy, Zagreb, Croatia
| | - Naohiko Anzai
- Department of Pharmacology and Toxicology, Dokkyo Medical University School of Medicine, Tochigi, Japan; and
| | - Hermann Koepsell
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute and Institute of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Gerhard Burckhardt
- Center of Physiology and Pathophysiology, Institute of Systemic Physiology and Pathophysiology, University of Göttingen, Göttingen, Germany
| | - Birgitta C Burckhardt
- Center of Physiology and Pathophysiology, Institute of Systemic Physiology and Pathophysiology, University of Göttingen, Göttingen, Germany
| | - Ivan Sabolić
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia;
| |
Collapse
|
23
|
Worley RR, Fisher J. Application of physiologically-based pharmacokinetic modeling to explore the role of kidney transporters in renal reabsorption of perfluorooctanoic acid in the rat. Toxicol Appl Pharmacol 2015; 289:428-41. [PMID: 26522833 DOI: 10.1016/j.taap.2015.10.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/20/2015] [Accepted: 10/27/2015] [Indexed: 11/25/2022]
Abstract
Renal elimination and the resulting clearance of perfluorooctanoic acid (PFOA) from the serum exhibit pronounced sex differences in the adult rat. The literature suggests that this is largely due to hormonally regulated expression of organic anion transporters (OATs) on the apical and basolateral membranes of the proximal tubule cells that facilitate excretion and reabsorption of PFOA from the filtrate into the blood. Previously developed PBPK models of PFOA exposure in the rat have not been parameterized to specifically account for transporter-mediated renal elimination. We developed a PBPK model for PFOA in male and female rats to explore the role of Oat1, Oat3, and Oatp1a1 in sex-specific renal reabsorption and excretion of PFOA. Descriptions of the kinetic behavior of these transporters were extrapolated from in vitro studies and the model was used to simulate time-course serum, liver, and urine data for intravenous (IV) and oral exposures in both sexes. Model predicted concentrations of PFOA in the liver, serum, and urine showed good agreement with experimental data for both male and female rats indicating that in vitro derived physiological descriptions of transporter-mediated renal reabsorption can successfully predict sex-dependent excretion of PFOA in the rat. This study supports the hypothesis that sex-specific serum half-lives for PFOA are largely driven by expression of transporters in the kidney and contribute to the development of PBPK modeling as a tool for evaluating the role of transporters in renal clearance.
Collapse
Affiliation(s)
- Rachel Rogers Worley
- Agency for Toxic Substances and Disease Registry, Division of Community Health Investigations, 4770 Buford Highway, Atlanta, GA 30341, United States; Interdisciplinary Toxicology Program, University of Georgia, 341 Pharmacy South, Athens, GA 30602, United States.
| | - Jeffrey Fisher
- Interdisciplinary Toxicology Program, University of Georgia, 341 Pharmacy South, Athens, GA 30602, United States; Food and Drug Administration, National Center for Toxicological Research, 3900 NCTR Road, Jefferson, AR 72079, United States
| |
Collapse
|
24
|
De Sousa Mendes M, Hirt D, Urien S, Valade E, Bouazza N, Foissac F, Blanche S, Treluyer JM, Benaboud S. Physiologically-based pharmacokinetic modeling of renally excreted antiretroviral drugs in pregnant women. Br J Clin Pharmacol 2015; 80:1031-41. [PMID: 26011128 DOI: 10.1111/bcp.12685] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 04/22/2015] [Accepted: 05/07/2015] [Indexed: 12/12/2022] Open
Abstract
AIM Physiological changes during pregnancy can affect drug disposition. Anticipating these changes will help to maximize drug efficacy and safety in pregnant women. Our objective was to determine if physiologically-based pharmacokinetics (PBPK) can accurately predict changes in the disposition of renally excreted antiretroviral drugs during pregnancy. METHODS Whole body PBPK models were developed for three renally excreted antiretroviral drugs, tenofovir (TFV), emtricitabine (FTC) and lamivudine (3TC). To assess the impact of pregnancy on PK, time-varying pregnancy-related physiological parameters available within the p-PBPK Simcyp software package were used. Renal clearance during pregnancy followed glomerular filtration changes with or without alterations in secretion. PK profiles were simulated and compared with observed data, i.e. area under the curves (AUC), peak plasma concentrations (Cmax ) and oral clearances (CL/F). RESULTS PBPK models successfully predicted TFV, FTC and 3TC disposition for non-pregnant and pregnant populations. Both renal secretion and filtration changed during pregnancy. Changes in renal clearance secretion were related to changes in renal plasma flow. The maximum clearance increases were approximately 30% (TFV 33%, FTC 31%, 3TC 29%). CONCLUSIONS Pregnancy PBPK models are useful tools to quantify a priori the drug exposure changes during pregnancy for renally excreted drugs. These models can be applied to evaluate alternative dosing regimens to optimize drug therapy during pregnancy.
Collapse
Affiliation(s)
- Maïlys De Sousa Mendes
- EA08: Evaluation des thérapeutiques et pharmacologie périnatale et pédiatrique, unité de recherche clinique Paris centre, 75006, Paris
| | - Deborah Hirt
- EA08: Evaluation des thérapeutiques et pharmacologie périnatale et pédiatrique, unité de recherche clinique Paris centre, 75006, Paris.,Service de Pharmacologie Clinique, AP-HP, Hôpital Cochin-Broca-Hôtel-Dieu-Dieu, 75014, Paris
| | - Saik Urien
- EA08: Evaluation des thérapeutiques et pharmacologie périnatale et pédiatrique, unité de recherche clinique Paris centre, 75006, Paris.,CIC-1419 Inserm, Cochin-Necker, Paris
| | - Elodie Valade
- EA08: Evaluation des thérapeutiques et pharmacologie périnatale et pédiatrique, unité de recherche clinique Paris centre, 75006, Paris
| | - Naïm Bouazza
- EA08: Evaluation des thérapeutiques et pharmacologie périnatale et pédiatrique, unité de recherche clinique Paris centre, 75006, Paris
| | - Frantz Foissac
- EA08: Evaluation des thérapeutiques et pharmacologie périnatale et pédiatrique, unité de recherche clinique Paris centre, 75006, Paris
| | - Stephane Blanche
- EA08: Evaluation des thérapeutiques et pharmacologie périnatale et pédiatrique, unité de recherche clinique Paris centre, 75006, Paris.,AP-HP, hôpital Necker-Enfants-malades, unité d'immunologie, hématologie et rhumatologie pédiatriques, 75015, Paris, France
| | - Jean-Marc Treluyer
- EA08: Evaluation des thérapeutiques et pharmacologie périnatale et pédiatrique, unité de recherche clinique Paris centre, 75006, Paris.,Service de Pharmacologie Clinique, AP-HP, Hôpital Cochin-Broca-Hôtel-Dieu-Dieu, 75014, Paris
| | - Sihem Benaboud
- EA08: Evaluation des thérapeutiques et pharmacologie périnatale et pédiatrique, unité de recherche clinique Paris centre, 75006, Paris.,Service de Pharmacologie Clinique, AP-HP, Hôpital Cochin-Broca-Hôtel-Dieu-Dieu, 75014, Paris
| |
Collapse
|
25
|
Shen H, Liu T, Morse BL, Zhao Y, Zhang Y, Qiu X, Chen C, Lewin AC, Wang XT, Liu G, Christopher LJ, Marathe P, Lai Y. Characterization of Organic Anion Transporter 2 (SLC22A7): A Highly Efficient Transporter for Creatinine and Species-Dependent Renal Tubular Expression. Drug Metab Dispos 2015; 43:984-93. [PMID: 25904762 DOI: 10.1124/dmd.114.062364] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 04/22/2015] [Indexed: 01/03/2023] Open
Abstract
The contribution of organic anion transporter OAT2 (SLC22A7) to the renal tubular secretion of creatinine and its exact localization in the kidney are reportedly controversial. In the present investigation, the transport of creatinine was assessed in human embryonic kidney (HEK) cells that stably expressed human OAT2 (OAT2-HEK) and isolated human renal proximal tubule cells (HRPTCs). The tubular localization of OAT2 in human, monkey, and rat kidney was characterized. The overexpression of OAT2 significantly enhanced the uptake of creatinine in OAT2-HEK cells. Under physiologic conditions (creatinine concentrations of 41.2 and 123.5 µM), the initial rate of OAT2-mediated creatinine transport was approximately 11-, 80-, and 80-fold higher than OCT2, multidrug and toxin extrusion protein (MATE)1, and MATE2K, respectively, resulting in approximately 37-, 1850-, and 80-fold increase of the intrinsic transport clearance when normalized to the transporter protein concentrations. Creatinine intracellular uptake and transcellular transport in HRPTCs were decreased in the presence of 50 µM bromosulfophthalein and 100 µM indomethacin, which inhibited OAT2 more potently than other known creatinine transporters, OCT2 and multidrug and toxin extrusion proteins MATE1 and MATE2K (IC50: 1.3 µM vs. > 100 µM and 2.1 µM vs. > 200 µM for bromosulfophthalein and indomethacin, respectively) Immunohistochemistry analysis showed that OAT2 protein was localized to both basolateral and apical membranes of human and cynomolgus monkey renal proximal tubules, but appeared only on the apical membrane of rat proximal tubules. Collectively, the findings revealed the important role of OAT2 in renal secretion and possible reabsorption of creatinine and suggested a molecular basis for potential species difference in the transporter handling of creatinine.
Collapse
Affiliation(s)
- Hong Shen
- Departments of Metabolism and Pharmacokinetics (H.S., T.L., B.L.M., Yuep.Z., X.Q., C.C., P.M., Y.L.), Bioanalytical Sciences (Y.Z., G.L.), Oncology Translational Research (A.C.L., X.-T.W.), and Biotransformation (L.J.C.), Bristol-Myers Squibb Research and Development, Princeton, New Jersey
| | - Tongtong Liu
- Departments of Metabolism and Pharmacokinetics (H.S., T.L., B.L.M., Yuep.Z., X.Q., C.C., P.M., Y.L.), Bioanalytical Sciences (Y.Z., G.L.), Oncology Translational Research (A.C.L., X.-T.W.), and Biotransformation (L.J.C.), Bristol-Myers Squibb Research and Development, Princeton, New Jersey
| | - Bridget L Morse
- Departments of Metabolism and Pharmacokinetics (H.S., T.L., B.L.M., Yuep.Z., X.Q., C.C., P.M., Y.L.), Bioanalytical Sciences (Y.Z., G.L.), Oncology Translational Research (A.C.L., X.-T.W.), and Biotransformation (L.J.C.), Bristol-Myers Squibb Research and Development, Princeton, New Jersey
| | - Yue Zhao
- Departments of Metabolism and Pharmacokinetics (H.S., T.L., B.L.M., Yuep.Z., X.Q., C.C., P.M., Y.L.), Bioanalytical Sciences (Y.Z., G.L.), Oncology Translational Research (A.C.L., X.-T.W.), and Biotransformation (L.J.C.), Bristol-Myers Squibb Research and Development, Princeton, New Jersey
| | - Yueping Zhang
- Departments of Metabolism and Pharmacokinetics (H.S., T.L., B.L.M., Yuep.Z., X.Q., C.C., P.M., Y.L.), Bioanalytical Sciences (Y.Z., G.L.), Oncology Translational Research (A.C.L., X.-T.W.), and Biotransformation (L.J.C.), Bristol-Myers Squibb Research and Development, Princeton, New Jersey
| | - Xi Qiu
- Departments of Metabolism and Pharmacokinetics (H.S., T.L., B.L.M., Yuep.Z., X.Q., C.C., P.M., Y.L.), Bioanalytical Sciences (Y.Z., G.L.), Oncology Translational Research (A.C.L., X.-T.W.), and Biotransformation (L.J.C.), Bristol-Myers Squibb Research and Development, Princeton, New Jersey
| | - Cliff Chen
- Departments of Metabolism and Pharmacokinetics (H.S., T.L., B.L.M., Yuep.Z., X.Q., C.C., P.M., Y.L.), Bioanalytical Sciences (Y.Z., G.L.), Oncology Translational Research (A.C.L., X.-T.W.), and Biotransformation (L.J.C.), Bristol-Myers Squibb Research and Development, Princeton, New Jersey
| | - Anne C Lewin
- Departments of Metabolism and Pharmacokinetics (H.S., T.L., B.L.M., Yuep.Z., X.Q., C.C., P.M., Y.L.), Bioanalytical Sciences (Y.Z., G.L.), Oncology Translational Research (A.C.L., X.-T.W.), and Biotransformation (L.J.C.), Bristol-Myers Squibb Research and Development, Princeton, New Jersey
| | - Xi-Tao Wang
- Departments of Metabolism and Pharmacokinetics (H.S., T.L., B.L.M., Yuep.Z., X.Q., C.C., P.M., Y.L.), Bioanalytical Sciences (Y.Z., G.L.), Oncology Translational Research (A.C.L., X.-T.W.), and Biotransformation (L.J.C.), Bristol-Myers Squibb Research and Development, Princeton, New Jersey
| | - Guowen Liu
- Departments of Metabolism and Pharmacokinetics (H.S., T.L., B.L.M., Yuep.Z., X.Q., C.C., P.M., Y.L.), Bioanalytical Sciences (Y.Z., G.L.), Oncology Translational Research (A.C.L., X.-T.W.), and Biotransformation (L.J.C.), Bristol-Myers Squibb Research and Development, Princeton, New Jersey
| | - Lisa J Christopher
- Departments of Metabolism and Pharmacokinetics (H.S., T.L., B.L.M., Yuep.Z., X.Q., C.C., P.M., Y.L.), Bioanalytical Sciences (Y.Z., G.L.), Oncology Translational Research (A.C.L., X.-T.W.), and Biotransformation (L.J.C.), Bristol-Myers Squibb Research and Development, Princeton, New Jersey
| | - Punit Marathe
- Departments of Metabolism and Pharmacokinetics (H.S., T.L., B.L.M., Yuep.Z., X.Q., C.C., P.M., Y.L.), Bioanalytical Sciences (Y.Z., G.L.), Oncology Translational Research (A.C.L., X.-T.W.), and Biotransformation (L.J.C.), Bristol-Myers Squibb Research and Development, Princeton, New Jersey
| | - Yurong Lai
- Departments of Metabolism and Pharmacokinetics (H.S., T.L., B.L.M., Yuep.Z., X.Q., C.C., P.M., Y.L.), Bioanalytical Sciences (Y.Z., G.L.), Oncology Translational Research (A.C.L., X.-T.W.), and Biotransformation (L.J.C.), Bristol-Myers Squibb Research and Development, Princeton, New Jersey
| |
Collapse
|
26
|
Babelova A, Burckhardt BC, Wegner W, Burckhardt G, Henjakovic M. Sex-differences in renal expression of selected transporters and transcription factors in lean and obese Zucker spontaneously hypertensive fatty rats. J Diabetes Res 2015; 2015:483238. [PMID: 25710042 PMCID: PMC4325971 DOI: 10.1155/2015/483238] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 01/22/2023] Open
Abstract
The aim of this study was to identify sex-dependent expression of renal transporter mRNA in lean and obese Zucker spontaneously hypertensive fatty (ZSF1) rats and to investigate the interaction of the most altered transporter, organic anion transporter 2 (Oat2), with diabetes-relevant metabolites and drugs. Higher incidence of glomerulosclerosis, tubulointerstitial fibrosis, and protein casts in Bowman's space and tubular lumen was detected by PAS staining in obese male compared to female ZSF1 rats. Real-time PCR on RNA isolated from kidney cortex revealed that Sglt1-2, Oat1-3, and Oct1 were higher expressed in kidneys of lean females. Oct2 and Mrp2 were higher expressed in obese males. Renal mRNA levels of transporters were reduced with diabetic nephropathy in females and the expression of transcription factors Hnf1β and Hnf4α in both sexes. The highest difference between lean and obese ZSF1 rats was found for Oat2. Therefore, we have tested the interaction of human OAT2 with various substances using tritium-labeled cGMP. Human OAT2 showed no interaction with diabetes-related metabolites, diabetic drugs, and ACE-inhibitors. However, OAT2-dependent uptake of cGMP was inhibited by furosemide. The strongly decreased expression of Oat2 and other transporters in female diabetic ZSF1 rats could possibly impair renal drug excretion, for example, of furosemide.
Collapse
Affiliation(s)
- Andrea Babelova
- Institute for Cardiovascular Physiology (Physiology I), Faculty of Medicine, Goethe-University, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Cancer Research Institute, Slovak Academy of Sciences, Vlarska 7, 83391 Bratislava, Slovakia
| | - Birgitta C. Burckhardt
- Institute for Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | - Waja Wegner
- Institute for Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | - Gerhard Burckhardt
- Institute for Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | - Maja Henjakovic
- Institute for Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
- *Maja Henjakovic:
| |
Collapse
|
27
|
Wegner W, Burckhardt G, Henjakovic M. Transcriptional regulation of human organic anion transporter 1 by B-cell CLL/lymphoma 6. Am J Physiol Renal Physiol 2014; 307:F1283-91. [DOI: 10.1152/ajprenal.00426.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The human organic anion transporter 1 (OAT1) is crucial for the excretion of organic anions in renal proximal tubular cells and has been classified as a clinically relevant transporter in the kidneys. Our previous study indicated that renal male-predominant expression of rat Oat1 and Oat3 appears to be regulated by transcription factor B-cell CLL/lymphoma 6 (BCL6). The aim of this study was to characterize the effect of BCL6 on human OAT1 promoter and on the transcription of OAT1 mediated by hepatocyte nuclear factor-1α (HNF-1α). Luciferase assays were carried out in opossum kidney (OK) cells transiently transfected with promoter constructs of OAT1, expression vectors for BCL6 and HNF-1α, and the empty control vectors. BCL6 and HNF-1α binding on OAT1 promoter was analyzed using electrophoretic mobility shift assay (EMSA). Protein expression of HNF-1α was investigated by Western blot analysis. Site-directed mutagenesis was used to introduce mutations into BCL6 and HNF-1α binding sites within the OAT1 promoter. BCL6 enhanced the promoter activity of OAT1 independently of predicted BCL6 binding sites but was dependent on HNF-1α response element and HNF-1α protein. Coexpression of BCL6 and HNF-1α induced an additive effect on OAT1 promoter activation compared with BCL6 or HNF-1α alone. BCL6 does not bind directly or indirectly to OAT1 promoter but increases the protein expression of HNF-1α and thereby indirectly enhances OAT1 gene transcription. BCL6 constitutes a promising candidate gene for the regulation of human OAT1 transcription and other renal and/or hepatic drug transporters that have been already shown to be activated by HNF-1α.
Collapse
Affiliation(s)
- Waja Wegner
- Department of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Gerhard Burckhardt
- Department of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Maja Henjakovic
- Department of Systemic Physiology and Pathophysiology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
28
|
A population pharmacokinetic approach to describe cephalexin disposition in adult and aged dogs. Vet Med Int 2014; 2014:789353. [PMID: 25431741 PMCID: PMC4241252 DOI: 10.1155/2014/789353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 10/09/2014] [Indexed: 12/30/2022] Open
Abstract
This study was conducted in order to characterize the pharmacokinetics of orally administered cephalexin to healthy adult and aged dogs, using a population pharmacokinetic approach. Two hundred and eighty-six cephalexin plasma concentrations obtained from previous pharmacokinetic studies were used. Sex, age, pharmaceutical formulation, and breed were evaluated as covariates. A one-compartment model with an absorption lag-time (Tlag) best described the data. The final model included age (adult; aged) on apparent volume of distribution (Vd/F), apparent elimination rate (ke/F), and Tlag; sex (female; male) on ke/F, and breed (Beagle; mixed-breed) on Vd/F. Addition of the covariates to the model explained 78% of the interindividal variability (IIV) in Vd/F, 36% in ke/F, and 24% in Tlag, respectively. Formulation did not affect the variability of any of the pharmacokinetic parameters. Tlag was longer, whereas Vd/F and ke/F were lower in aged compared to adult animals; in female aged dogs ke/F was lower than in male aged dogs; however, the differences were of low magnitude. Different disposition of cephalexin may be expected in aged dogs.
Collapse
|
29
|
Klein DM, Wright SH, Cherrington NJ. Xenobiotic transporter expression along the male genital tract. Reprod Toxicol 2014; 47:1-8. [PMID: 24814985 DOI: 10.1016/j.reprotox.2014.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 04/11/2014] [Accepted: 04/29/2014] [Indexed: 12/18/2022]
Abstract
The male genital tract plays an important role in protecting sperm by forming a distinct compartment separate from the body which limits exposure to potentially toxic substrates. Transporters along this tract can influence the distribution of xenobiotics into the male genital tract through efflux back into the blood or facilitating the accumulation of toxicants. The aim of this study was to quantitatively determine the constitutive mRNA expression of 30 xenobiotic transporters in caput and cauda regions of the epididymis, vas deferens, prostate, and seminal vesicles from adult Sprague-Dawley rats. The epididymis was found to express at least moderate levels of 18 transporters, vas deferens 15, seminal vesicles 23, and prostate 18. Constitutive expression of these xenobiotic transporters in the male genital tract may provide insight into the xenobiotics that can potentially be transported into these tissues and may provide the molecular mechanism for site specific toxicity of select agents.
Collapse
Affiliation(s)
- David M Klein
- University of Arizona, Department of Pharmacology and Toxicology, Tucson, AZ 85721, United States
| | - Stephen H Wright
- University of Arizona, Department of Physiology, Tucson, AZ 85721, United States
| | - Nathan J Cherrington
- University of Arizona, Department of Pharmacology and Toxicology, Tucson, AZ 85721, United States.
| |
Collapse
|
30
|
Franconi F, Campesi I. Pharmacogenomics, pharmacokinetics and pharmacodynamics: interaction with biological differences between men and women. Br J Pharmacol 2014; 171:580-94. [PMID: 23981051 PMCID: PMC3969074 DOI: 10.1111/bph.12362] [Citation(s) in RCA: 162] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 08/05/2013] [Accepted: 08/16/2013] [Indexed: 12/16/2022] Open
Abstract
Pharmacological response depends on multiple factors and one of them is sex-gender. Data on the specific effects of sex-gender on pharmacokinetics, as well as the safety and efficacy of numerous medications, are beginning to emerge. Nevertheless, the recruitment of women for clinical research is inadequate, especially during the first phases. In general, pharmacokinetic differences between males and females are more numerous and consistent than disparities in pharmacodynamics. However, sex-gender pharmacodynamic differences are now increasingly being identified at the molecular level. It is now even becoming apparent that sex-gender influences pharmacogenomics and pharmacogenetics. Sex-related differences have been reported for several parameters, and it is consistently shown that women have a worse safety profile, with drug adverse reactions being more frequent and severe in women than in men. Overall, the pharmacological status of women is less well studied than that of men and deserves much more attention. The design of clinical and preclinical studies should have a sex-gender-based approach with the aim of tailoring therapies to an individual's needs and concerns.
Collapse
Affiliation(s)
- Flavia Franconi
- Department of Biomedical Sciences, University of SassariSassari, Italy
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and BiosystemsOsilo, Italy
| | - Ilaria Campesi
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and BiosystemsOsilo, Italy
- Department of Surgical, Microsurgical and Medical Sciences, University of SassariSassari, Italy
| |
Collapse
|
31
|
Abstract
Organic anions and cations (OAs and OCs, respectively) comprise an extraordinarily diverse array of compounds of physiological, pharmacological, and toxicological importance. The kidney, primarily the renal proximal tubule, plays a critical role in regulating the plasma concentrations of these organic electrolytes and in clearing the body of potentially toxic xenobiotics agents, a process that involves active, transepithelial secretion. This transepithelial transport involves separate entry and exit steps at the basolateral and luminal aspects of renal tubular cells. Basolateral and luminal OA and OC transport reflects the concerted activity of a suite of separate proteins arranged in parallel in each pole of proximal tubule cells. The cloning of multiple members of several distinct transport families, the subsequent characterization of their activity, and their subcellular localization within distinct regions of the kidney, now allows the development of models describing the molecular basis of the renal secretion of OAs and OCs. New information on naturally occurring genetic variation of many of these processes provides insight into the basis of observed variability of drug efficacy and unwanted drug-drug interactions in human populations. The present review examines recent work on these issues.
Collapse
Affiliation(s)
- Ryan M Pelis
- Novartis Pharmaceuticals Corp., Translational Sciences, East Hanover, New Jersey, USA
| | | |
Collapse
|
32
|
Saghir SA, Marty MS, Zablotny CL, Passage JK, Perala AW, Neal BH, Hammond L, Bus JS. Life-stage-, sex-, and dose-dependent dietary toxicokinetics and relationship to toxicity of 2,4-dichlorophenoxyacetic acid (2,4-D) in rats: implications for toxicity test dose selection, design, and interpretation. Toxicol Sci 2013; 136:294-307. [PMID: 24105888 PMCID: PMC3858196 DOI: 10.1093/toxsci/kft212] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 08/12/2013] [Indexed: 11/14/2022] Open
Abstract
Life-stage-dependent toxicity and dose-dependent toxicokinetics (TK) were evaluated in Sprague Dawley rats following dietary exposure to 2,4-dichlorophenoxyacetic acid (2,4-D). 2,4-D renal clearance is impacted by dose-dependent saturation of the renal organic anion transporter; thus, this study focused on identifying inflection points of onset of dietary nonlinear TK to inform dose selection decisions for toxicity studies. Male and female rats were fed 2,4-D-fortified diets at doses to 1600 ppm for 4-weeks premating, <2 weeks during mating, and to test day (TD) 71 to parental (P1) males and to P1 females through gestation/lactation to TD 96. F1 offspring were exposed via milk with continuing diet exposure until postnatal day (PND) 35. As assessed by plasma area under the curve for the time-course plasma concentration, nonlinear TK was observed ≥ 1200 ppm (63 mg/kg/day) for P1 males and between 200 and 400 ppm (14-27 mg/kg/day) for P1 females. Dam milk and pup plasma levels were higher on lactation day (LD) 14 than LD 4. Relative to P1 adults, 2,4-D levels were higher in dams during late gestation/lactation and postweaning pups (PND 21-35) and coincided with elevated intake of diet/kg body weight. Using conventional maximum tolerated dose (MTD) criteria based on body weight changes for dose selection would have resulted in excessive top doses approximately 2-fold higher than those identified incorporating critical TK data. These data indicate that demonstration of nonlinear TK, if present at dose levels substantially above real-world human exposures, is a key dose selection consideration for improving the human relevance of toxicity studies compared with studies employing conventional MTD dose selection strategies.
Collapse
Affiliation(s)
- Shakil A. Saghir
- *Intrinsik Environmental Sciences Inc., Mississauga, Ontario, Canada L5T 0A3, and Aga Khan University, Karachi, Pakistan 74800
| | | | | | | | | | | | - Larry Hammond
- §Industry Task Force II on 2,4-D Research Data, Washington, District of Columbia 20006
| | | |
Collapse
|
33
|
Marty MS, Neal BH, Zablotny CL, Yano BL, Andrus AK, Woolhiser MR, Boverhof DR, Saghir SA, Perala AW, Passage JK, Lawson MA, Bus JS, Lamb JC, Hammond L. An F1-extended one-generation reproductive toxicity study in Crl:CD(SD) rats with 2,4-dichlorophenoxyacetic acid. Toxicol Sci 2013; 136:527-47. [PMID: 24072463 PMCID: PMC3858197 DOI: 10.1093/toxsci/kft213] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
2,4-Dichlorophenoxyacetic acid (2,4-D) was assessed for systemic toxicity, reproductive toxicity, developmental neurotoxicity (DNT), developmental immunotoxicity (DIT), and endocrine toxicity. CD rats (27/sex/dose) were exposed to 0, 100, 300, 600 (female), or 800 (male) ppm 2,4-D in diet. Nonlinear toxicokinetic behavior was shown at high doses; the renal clearance saturation threshold for 2,4-D was exceeded markedly in females and slightly exceeded in males. Exposure was 4 weeks premating, 7 weeks postmating for P1 males and through lactation for P1 females. F1 offspring were examined for survival and development, and at weaning, pups were divided in cohorts, by sex and dose, and by systemic toxicity (10), DNT (10), DIT (20), and reproductive toxicity (≥ 23). Remaining weanlings were evaluated for systemic toxicity and neuropathology (10–12). Body weight decreased during lactation in high-dose P1 females and in F1 pups. Kidney was the primary target organ, with slight degeneration of proximal convoluted tubules observed in high-dose P1 males and in high-dose F1 males and females. A slight intergenerational difference in kidney toxicity was attributed to increased intake of 2,4-D in F1 offspring. Decreased weanling testes weights and delayed preputial separation in F1 males were attributed to decreased body weights. Endocrine-related effects were limited to slight thyroid hormone changes and adaptive histopathology in high-dose GD 17 dams seen only at a nonlinear toxicokinetic dose. 2,4-D did not cause reproductive toxicity, DNT, or DIT. The “No Observed Adverse Effect Level” for systemic toxicity was 300 ppm in both males (16.6mg/kg/day) and females (20.6mg/kg/day), which is approximately 6700- to 93 000-fold higher than that reported for 2,4-D exposures in human biomonitoring studies.
Collapse
Affiliation(s)
- Mary Sue Marty
- * Toxicology and Environmental Research & Consulting, The Dow Chemical Company, Midland, Michigan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
How much do we know about drug handling by SLC and ABC drug transporters in children? Clin Pharmacol Ther 2013; 94:27-9. [PMID: 23778708 DOI: 10.1038/clpt.2013.82] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Although solute carrier (SLC) and ATP-binding cassette (ABC) transporters are critical to the absorption, distribution, and elimination of many small-molecule drugs in children, how these transporters regulate pediatric drug handling remains unclear. For proper dosing and to diminish toxicity, we need a better understanding of how organ development and functional maturation, as well as developmental changes in systemic physiology, impact transporter-mediated drug handling at pediatric developmental stages from the preterm infant through adolescence.
Collapse
|
35
|
Rodenburg EM, Hoorn EJ, Ruiter R, Lous JJ, Hofman A, Uitterlinden AG, Stricker BH, Visser LE. Thiazide-Associated Hyponatremia: A Population-Based Study. Am J Kidney Dis 2013; 62:67-72. [DOI: 10.1053/j.ajkd.2013.02.365] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 02/06/2013] [Indexed: 11/11/2022]
|
36
|
Breljak D, Brzica H, Sweet DH, Anzai N, Sabolic I. Sex-dependent expression of Oat3 (Slc22a8) and Oat1 (Slc22a6) proteins in murine kidneys. Am J Physiol Renal Physiol 2013; 304:F1114-26. [PMID: 23389457 DOI: 10.1152/ajprenal.00201.2012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
In the mouse kidney, organic anion transporter 3 (mOat3, Slc22a8) was previously localized to the basolateral membrane (BLM) of proximal tubule (PT), thick ascending limb of Henle, macula densa, distal tubule, and cortical collecting duct. However, the specificity of anti-Oat3 antibodies (Oat3-Ab) used in these studies was not properly verified. Moreover, the sex-dependent expression of mOat3, and of the functionally similar transporter mOat1 (Slc22a6), in the mouse kidney has been studied at mRNA level, whereas their protein expression is poorly documented. Here we investigated 1) specificity of Oat3-Abs by using Oat3 knockout (KO) mice, 2) cell localization of renal mOat3 with a specific mOat3-Ab, 3) sex-dependent expression of renal mOat3 and mOat1 proteins, and 4) hormone(s) responsible for observed sex differences. As previously shown, an Oat3-Ab against the rat protein stained the BLM of various nephron segments in wild-type (WT) mice, but the same staining pattern was noted along the nephron of Oat3 KO mice. However, the mOat3-Ab exclusively stained the BLM of PT in WT mice, where it colocalized with the mOat1 protein, whereas no staining of Oat3 protein was noted in the kidney of Oat3 KO mice. The expression of mOat3 protein was lower in male mice, upregulated by castration, and downregulated by testosterone treatment. The expression of mOat1 protein was stronger in males, downregulated by castration, and upregulated by testosterone treatment. Thus, at the protein level, mOat3 and mOat1 exhibit sex-dependent expression with an opposite pattern; mOat3 is female dominant due to androgen inhibition, while mOat1 is male dominant due to androgen stimulation.
Collapse
Affiliation(s)
- Davorka Breljak
- Molecular Toxicology, Institute for Medical Research and Occupational Health, Ksaverska cesta 2, HR-10000 Zagreb, Croatia.
| | | | | | | | | |
Collapse
|
37
|
Brandoni A, Hazelhoff MH, Bulacio RP, Torres AM. Expression and function of renal and hepatic organic anion transporters in extrahepatic cholestasis. World J Gastroenterol 2012; 18:6387-6397. [PMID: 23197884 PMCID: PMC3508633 DOI: 10.3748/wjg.v18.i44.6387] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 06/18/2012] [Accepted: 06/28/2012] [Indexed: 02/06/2023] Open
Abstract
Obstructive jaundice occurs in patients suffering from cholelithiasis and from neoplasms affecting the pancreas and the common bile duct. The absorption, distribution and elimination of drugs are impaired during this pathology. Prolonged cholestasis may alter both liver and kidney function. Lactam antibiotics, diuretics, non-steroidal anti-inflammatory drugs, several antiviral drugs as well as endogenous compounds are classified as organic anions. The hepatic and renal organic anion transport pathways play a key role in the pharmacokinetics of these compounds. It has been demonstrated that acute extrahepatic cholestasis is associated with increased renal elimination of organic anions. The present work describes the molecular mechanisms involved in the regulation of the expression and function of the renal and hepatic organic anion transporters in extrahepatic cholestasis, such as multidrug resistance-associated protein 2, organic anion transporting polypeptide 1, organic anion transporter 3, bilitranslocase, bromosulfophthalein/bilirubin binding protein, organic anion transporter 1 and sodium dependent bile salt transporter. The modulation in the expression of renal organic anion transporters constitutes a compensatory mechanism to overcome the hepatic dysfunction in the elimination of organic anions.
Collapse
|
38
|
Funk RS, Brown JT, Abdel-Rahman SM. Pediatric pharmacokinetics: human development and drug disposition. Pediatr Clin North Am 2012; 59:1001-16. [PMID: 23036241 DOI: 10.1016/j.pcl.2012.07.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Human development is described by the various anatomic and physiologic changes that occur as the single-celled zygote matures into an adult human being. Concomitant with bodily maturation are changes in the complex interactions between pharmacologic agents and the biologic matrix that is the human body. Profound changes in the manner by which drugs traverse the body during development can have significant implications in drug efficacy and toxicity. Although not a replacement for well-conducted, pediatric, pharmacokinetic studies, an understanding of developmental biology and the mechanisms for drug disposition invariably assists the pediatric clinician with the judicious use of medications in children.
Collapse
Affiliation(s)
- Ryan S Funk
- Division of Clinical Pharmacology and Medical Toxicology, Children's Mercy Hospitals and Clinics, 2401 Gillham Road, Kansas City, MO 64108, USA
| | | | | |
Collapse
|
39
|
El-Kasti MM, Wells T, Carter DA. A novel long-range enhancer regulates postnatal expression of Zeb2: implications for Mowat-Wilson syndrome phenotypes. Hum Mol Genet 2012; 21:5429-42. [PMID: 23001561 DOI: 10.1093/hmg/dds389] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The zinc-finger, E-box-binding homeobox-2 (Zeb2) gene encodes a SMAD-interacting transcription factor that has diverse roles in development and disease. Mutations at the hZeb2 locus cause Mowat-Wilson syndrome (MWS), a genetic disorder that is associated with mental retardation and other, case- and sex-dependent clinical features. Recent studies have detailed microRNA-mediated control of Zeb2, but little is known about the genomic context of this gene or of enhancer sequences that may direct its diverse functions. Here, we describe a novel transgenic rodent model in which Zeb2 regulatory sequence has been disrupted, resulting in a postnatal developmental phenotype that is autosomal dominant. The phenotype exhibits a genotype-by-sex interaction and manifests primarily as an acute attenuation of postnatal kidney development in males. Other aspects of embryonic and neonatal development, including neuronal, are unaffected. The transgene insertion site is associated with a 12 kb deletion, 1.2 Mb upstream of Zeb2, within a 4.1 Mb gene desert. A conserved sequence, derived from the deleted region, enhanced Zeb2 promoter activity in transcription assays. Tissue and temporal restriction of this enhancer activity may involve postnatal changes in proteins that bind this sequence. A control human/mouse VISTA enhancer (62 kb upstream of Zeb2) also up-regulated the Zeb2 promoter, providing evidence of a string of conserved distal enhancers. The phenotype arising from deletion of one copy of the extreme long-range enhancer indicates a critical role for this enhancer at one developmental stage. Haploinsufficiency of Zeb2 in this developmental context reflects inheritance of MWS and may underlie some sex-dependent, non-neural characteristics of this human inherited disorder.
Collapse
Affiliation(s)
- Muna M El-Kasti
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | | | | |
Collapse
|
40
|
Yacovino LL, Aleksunes LM. Endocrine and metabolic regulation of renal drug transporters. J Biochem Mol Toxicol 2012; 26:407-21. [PMID: 22933250 DOI: 10.1002/jbt.21435] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 06/22/2012] [Accepted: 07/21/2012] [Indexed: 12/15/2022]
Abstract
Renal xenobiotic transporters are important determinants of urinary secretion and reabsorption of chemicals. In addition to glomerular filtration, these processes are key to the overall renal clearance of a diverse array of drugs and toxins. Alterations in kidney transporter levels and function can influence the efficacy and toxicity of chemicals. Studies in experimental animals have revealed distinct patterns of renal transporter expression in response to sex hormones, pregnancy, and growth hormone. Likewise, a number of disease states including diabetes, obesity, and cholestasis alter the expression of kidney transporters. The goal of this review is to provide an overview of the major xenobiotic transporters expressed in the kidneys and an understanding of metabolic conditions and hormonal factors that regulate their expression and function.
Collapse
Affiliation(s)
- Lindsay L Yacovino
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ 08854-8020, USA
| | | |
Collapse
|
41
|
Burckhardt G. Drug transport by Organic Anion Transporters (OATs). Pharmacol Ther 2012; 136:106-30. [PMID: 22841915 DOI: 10.1016/j.pharmthera.2012.07.010] [Citation(s) in RCA: 261] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 07/10/2012] [Indexed: 02/08/2023]
Abstract
Common to all so far functionally characterized Organic Anion Transporters (OATs) is their broad substrate specificity and their ability to exchange extracellular against intracellular organic anions. Many OATs occur in renal proximal tubules, the site of active drug secretion. Exceptions are murine Oat6 (nasal epithelium), human OAT7 (liver), and rat Oat8 (renal collecting ducts). In human kidneys, OAT1, OAT2, and OAT3 are localized in the basolateral membrane, and OAT4, OAT10, and URAT1 in the apical cell membrane of proximal tubule cells, respectively. In rats and mice, Oat1 and Oat3 are located basolaterally, and Oat2, Oat5, Oat9, Oat10, and Urat1 apically. Several classes of drugs interact with human OAT1-3, including ACE inhibitors, angiotensin II receptor antagonists, diuretics, HMG CoA reductase inhibitors, β-lactam antibiotics, antineoplastic and antiviral drugs, and uricosuric drugs. For most drugs, interaction was demonstrated in vitro by inhibition of OAT-mediated transport of model substrates; for some drugs, transport by OATs was directly proven. Based on IC₅₀ values reported in the literature, OAT1 and OAT3 show comparable affinities for diuretics, cephalosporins, and nonsteroidal anti-inflammatory drugs whereas OAT2 has a lower affinity to most of these compounds. Drug-drug interactions at OAT1 and OAT3 may retard renal drug secretion and cause untoward effects. OAT4, OAT10, and URAT1 in the apical membrane contribute to proximal tubular urate absorption, and OAT10 to nicotinate absorption. OAT4 is in addition able to release drugs, e.g. diuretics, into the tubule lumen.
Collapse
Affiliation(s)
- Gerhard Burckhardt
- Abteilung Vegetative Physiologie und Pathophysiologie, Zentrum Physiologie und Pathophysiologie, Universitätsmedizin Göttingen, Humboldtallee 23, 37073 Göttingen, Germany.
| |
Collapse
|
42
|
Emami Riedmaier A, Nies AT, Schaeffeler E, Schwab M. Organic anion transporters and their implications in pharmacotherapy. Pharmacol Rev 2012; 64:421-49. [PMID: 22457399 DOI: 10.1124/pr.111.004614] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Organic anion transporters play an essential role in the distribution and excretion of numerous endogenous metabolic products and exogenous organic anions, including a host of widely prescribed drugs. The expression and activity of these transporters is influenced by several conditions, including transcriptional regulation, gender-dependent regulation, and genetic variation. In addition, the interaction of these transporters with several drugs and endogenous substrates has been well documented and may play a significant role in drug disposition and development of various disease states, such as nephrotoxicity and familial idiopathic hypouricemia. Members of this family of transporters have been localized mainly to the renal epithelia of various species. Much of the early research in this field has focused on their role in renal drug transport, yet increasing research on this family of transporters has localized them to various other epithelial tissues, including liver, brain, and placenta. Thus, an understanding of the role of these transporters in drug interaction and disposition in the kidney and other tissues may help in the determination of individual drug response, susceptibility to drug toxicity, and chemical carcinogenesis. This review seeks to summarize current knowledge of the molecular function and substrate profile of cloned organic anion transporters and to discuss recent progress in the understanding of the impact of interindividual variability, transcriptional regulation, and tissue distribution on individual drug response.
Collapse
Affiliation(s)
- Arian Emami Riedmaier
- Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70-376 Stuttgart, Auerbachstr. 112, Germany
| | | | | | | |
Collapse
|
43
|
Falluel-Morel A, Lin L, Sokolowski K, McCandlish E, Buckley B, DiCicco-Bloom E. N-acetyl cysteine treatment reduces mercury-induced neurotoxicity in the developing rat hippocampus. J Neurosci Res 2012; 90:743-50. [PMID: 22420031 DOI: 10.1002/jnr.22819] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mercury is an environmental toxicant that can disrupt brain development. However, although progress has been made in defining its neurotoxic effects, we know far less about available therapies that can effectively protect the brain in exposed individuals. We previously developed an animal model in which we defined the sequence of events underlying neurotoxicity: Methylmercury (MeHg) injection in postnatal rat acutely induced inhibition of mitosis and stimulated apoptosis in the hippocampus, which later resulted in intermediate-term deficits in structure size and cell number. N-acetyl cysteine (NAC) is the N-acetyl derivative of L-cysteine used clinically for treatment of drug intoxication. Here, based on its known efficacy in promoting MeHg urinary excretion, we evaluated NAC for protective effects in the developing brain. In immature neurons and precursors, MeHg (3 μM) induced a >50% decrease in DNA synthesis at 24 hr, an effect that was completely blocked by NAC coincubation. In vivo, injection of MeHg (5 μg/g bw) into 7-day-old rats induced a 22% decrease in DNA synthesis in whole hippocampus and a fourfold increase in activated caspase-3-immunoreactive cells at 24 hr and reduced total cell numbers by 13% at 3 weeks. Treatment of MeHg-exposed rats with repeated injections of NAC abolished MeHg toxicity. NAC prevented the reduction in DNA synthesis and the marked increase in caspase-3 immunoreactivity. Moreover, the intermediate-term decrease in hippocampal cell number provoked by MeHg was fully blocked by NAC. Altogether these results suggest that MeHg toxicity in the perinatal brain can be ameliorated by using NAC, opening potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Anthony Falluel-Morel
- Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | | | | | | | | | |
Collapse
|
44
|
Zhang W, Zhang Y, Zhang H, Wang J, Cui R, Dai J. Sex differences in transcriptional expression of FABPs in zebrafish liver after chronic perfluorononanoic acid exposure. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2012; 46:5175-5182. [PMID: 22500729 DOI: 10.1021/es300147w] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Perfluorononanoic acid (PFNA), a nine carbon backbone of perfluorinated acids (PFAAs), has wide production applications and is found in environmental matrices as a contaminant. To understand the adverse effects of PFNA, adult male and female zebrafish were exposed to differing PFNA dosages (0, 0.01, 0.1, and 1.0 mg/L) for 180 days using a flow-through exposure system. Results showed body weight, body length, and hepatosomatic index (HSI) decreased in both sexes. The HPLC-MS/MS analysis found that PFNA concentrations were higher in male livers than in female livers with increasing significance in a dose-dependent manner. Total cholesterol levels increased in the livers of both sexes, whereas triglyceride (TG) levels increased in males and decreased in females. With the exception of FABP1b, the transcriptional expression levels of fatty acid binding proteins (FABPs) were up-regulated in males and down-regulated in females. A similar trend between sexes occurred for peroxisome proliferator-activated receptors (PPARs) and Ccaat-enhancer-binding proteins (C/EBPs), which may be the upstream regulatory elements of FABPs. The results indicated that PFNA exposure caused opposite adverse effects on liver TG levels between the sexes in zebrafish possibly due to the opposite expression of FABPs and its upstream genes.
Collapse
Affiliation(s)
- Wei Zhang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
| | | | | | | | | | | |
Collapse
|
45
|
The FEMA GRAS assessment of aliphatic and aromatic terpene hydrocarbons used as flavor ingredients. Food Chem Toxicol 2011; 49:2471-94. [DOI: 10.1016/j.fct.2011.06.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 05/20/2011] [Accepted: 06/01/2011] [Indexed: 01/15/2023]
|
46
|
Loccisano AE, Campbell JL, Butenhoff JL, Andersen ME, Clewell HJ. Evaluation of placental and lactational pharmacokinetics of PFOA and PFOS in the pregnant, lactating, fetal and neonatal rat using a physiologically based pharmacokinetic model. Reprod Toxicol 2011; 33:468-490. [PMID: 21872655 DOI: 10.1016/j.reprotox.2011.07.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 06/25/2011] [Accepted: 07/22/2011] [Indexed: 11/24/2022]
Abstract
Perfluoroalkyl carboxylates and sulfonates (PFAAs) have many consumer and industrial applications. Developmental toxicity studies in animals have raised concern about potential developmental effects of PFOA and PFOS in humans. We have developed PBPK models for PFAAs in the rat to help define a relationship between external dose, internal tissue concentrations, and observed adverse effects, and to understand how physiological changes that occur during gestation and lactation affect tissue distribution of PFAAs in the mother, fetus, and neonate. The models developed here expand upon a PBPK model for PFAAs in the adult female rat, and are consistent with available PK data. These models, along with the adult rat PFAA models, published in the companion paper, will help address concerns about possible health effects due to PFAA exposure in the fetus and neonate and will be useful in comparing PK across life stages.
Collapse
Affiliation(s)
- Anne E Loccisano
- Center for Human Health Assessment, The Hamner Institutes for Health Sciences, 6 Davis Drive, P.O. Box 12137, Research Triangle Park, NC 27709, United States.
| | - Jerry L Campbell
- Center for Human Health Assessment, The Hamner Institutes for Health Sciences, 6 Davis Drive, P.O. Box 12137, Research Triangle Park, NC 27709, United States
| | - John L Butenhoff
- 3M Medical Department, Corporate Toxicology, 3M Center 220-2E-02, St. Paul, MN 55144, United States
| | - Melvin E Andersen
- Center for Human Health Assessment, The Hamner Institutes for Health Sciences, 6 Davis Drive, P.O. Box 12137, Research Triangle Park, NC 27709, United States
| | - Harvey J Clewell
- Center for Human Health Assessment, The Hamner Institutes for Health Sciences, 6 Davis Drive, P.O. Box 12137, Research Triangle Park, NC 27709, United States
| |
Collapse
|
47
|
Sweeney DE, Vallon V, Rieg T, Wu W, Gallegos TF, Nigam SK. Functional maturation of drug transporters in the developing, neonatal, and postnatal kidney. Mol Pharmacol 2011; 80:147-54. [PMID: 21493727 PMCID: PMC3127534 DOI: 10.1124/mol.110.070680] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 04/14/2011] [Indexed: 11/22/2022] Open
Abstract
Because renal function in newborns is immature, the pharmacokinetics of drugs administered to neonates vary significantly from adult patients. The establishment of drug transport systems is a key process in the functional maturation of the nephron. However, a thorough examination of the expression of the main drug transporters in the kidney throughout all stages of development (embryonic, postnatal, and mature) has yet to be carried out, and the functional (physiological) impact is not well understood. Using time-series microarray data, we analyzed the temporal behavior of mRNA levels for a wide range of SLC and ABC transporters in the rodent kidney throughout a developmental time series. We find dynamic increases between the postnatal and mature stages of development for a number of transporters, including the proximal tubule-specific drug and organic anion transporters (OATs) OAT1 (SLC22a6) and OAT3 (SLC22a8). The OATs are the major multispecific basolateral drug, toxin, and metabolite transporters in the proximal tubule responsible for handling of many drugs, as well as the prototypical OAT substrate para-aminohippurate (PAH). We therefore performed specific in vivo pharmacokinetic analysis of the transport of PAH in postnatal and maturing rodent kidney. We show that there is a 4-fold increase in PAH clearance during this period. Clearance studies in Oat1 and Oat3 knockouts confirm that, as in the adult, Oat1 is the principle transporter of PAH in the postnatal kidney. The substantial differences observed supports the need for better understanding of pharmacokinetics in the newborn and juvenile kidney compared with the adult kidney at the basic and clinical level.
Collapse
Affiliation(s)
- Derina E Sweeney
- Division of Nephrology & Hypertension, Departments of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | | | | | | | | | | |
Collapse
|
48
|
Loccisano AE, Campbell JL, Butenhoff JL, Andersen ME, Clewell HJ. Comparison and evaluation of pharmacokinetics of PFOA and PFOS in the adult rat using a physiologically based pharmacokinetic model. Reprod Toxicol 2011; 33:452-467. [PMID: 21565266 DOI: 10.1016/j.reprotox.2011.04.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 04/22/2011] [Accepted: 04/23/2011] [Indexed: 01/13/2023]
Abstract
Perfluoroalkyl acid carboxylates and sulfonates (PFAAs) have many consumer and industrial applications. The persistence and widespread distribution of PFAAs have brought them under intense scrutiny. Limited PK data for PFAAs is available for humans; however, toxicological and pharmacokinetic data exist for rats, which can be useful for cross-species extrapolation. In this work, PBPK models were developed for adult male and female rats to describe the pharmacokinetics of PFOA and PFOS. The models contain a description of saturable renal resorption, free fraction of chemical in plasma, and saturable binding in liver. Both male and female rat models for each chemical were consistent with available PK data resulting from IV, oral, and dietary dosing regimens. Predicted plasma concentration curves followed trends observed in experimental data, and model predictions were within a factor of two of experimental values. PFOA and PFOS rat model output is sensitive to parameters governing renal resorption, indicating that renal resorption is responsible for the long-half life. These models, along with the PFAA gestation and lactation models published in this issue, will help address concerns about possible health effects due to PFAA exposure in the fetus and neonate and will be useful in comparing PK across life stages.
Collapse
Affiliation(s)
- Anne E Loccisano
- Center for Human Health Assessment, The Hamner Institutes for Health Sciences, 6 Davis Drive, P.O. Box 12137, Research Triangle Park, NC 27709, United States.
| | - Jerry L Campbell
- Center for Human Health Assessment, The Hamner Institutes for Health Sciences, 6 Davis Drive, P.O. Box 12137, Research Triangle Park, NC 27709, United States
| | - John L Butenhoff
- 3M Medical Department, Corporate Toxicology, 3M Center 220-2E-02, St. Paul, MN 55144, United States
| | - Melvin E Andersen
- Center for Human Health Assessment, The Hamner Institutes for Health Sciences, 6 Davis Drive, P.O. Box 12137, Research Triangle Park, NC 27709, United States
| | - Harvey J Clewell
- Center for Human Health Assessment, The Hamner Institutes for Health Sciences, 6 Davis Drive, P.O. Box 12137, Research Triangle Park, NC 27709, United States
| |
Collapse
|
49
|
Xu H, Li J, Chen R, Zhang B, Wang C, King N, Chen H, Ghishan FK. NHE2X3 DKO mice exhibit gender-specific NHE8 compensation. Am J Physiol Gastrointest Liver Physiol 2011; 300:G647-53. [PMID: 21252044 PMCID: PMC3074987 DOI: 10.1152/ajpgi.00546.2010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
NHE8, the newest member of the sodium/hydrogen exchanger family, is expressed in the epithelial cells of the intestine and the kidney. Intestinal expression of NHE8 is significantly higher than that of NHE2 and NHE3 at a young age, suggesting that NHE8 is an important player for intestinal sodium absorption during early development. The current study was designed to explore if NHE8 plays a compensatory role for the loss of NHE2 and NHE3 function in NHE2X3 double-knockout (NHE2X3 DKO) mice. We further explored the regulatory mechanism(s) responsible for the change in NHE8 expression in NHE2X3 DKO mice. We found that >95% of NHE2X3 DKO mice survived through weanling. However, only 60% of male NHE2X3 DKO mice and 88% of female NHE2X3 DKO mice survived to 6 wk of life. We also found that the expression of NHE8 in wild-type female mice was higher compared with wild-type male mice after puberty. In NHE2X3 KDO mice, NHE8 expression was increased in females but not in males. Using Caco-2 cells as a model of the small intestine, we showed that testosterone inhibited endogenous NHE8 expression by reducing NHE8 mRNA synthesis, whereas estrogen had no effect on NHE8 expression. Thus our data show for the first time that intestinal NHE8 has a compensatory role in NHE2X3 DKO mice and this regulation is gender-dependent.
Collapse
Affiliation(s)
- Hua Xu
- 1Department of Pediatrics, University of Arizona Health Sciences Center, Tucson, Arizona; and
| | - Jing Li
- 1Department of Pediatrics, University of Arizona Health Sciences Center, Tucson, Arizona; and
| | - Rongji Chen
- 1Department of Pediatrics, University of Arizona Health Sciences Center, Tucson, Arizona; and
| | - Bo Zhang
- 1Department of Pediatrics, University of Arizona Health Sciences Center, Tucson, Arizona; and
| | - Chunhui Wang
- 1Department of Pediatrics, University of Arizona Health Sciences Center, Tucson, Arizona; and ,2Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Nolan King
- 1Department of Pediatrics, University of Arizona Health Sciences Center, Tucson, Arizona; and
| | - Huacong Chen
- 1Department of Pediatrics, University of Arizona Health Sciences Center, Tucson, Arizona; and
| | - Fayez K. Ghishan
- 1Department of Pediatrics, University of Arizona Health Sciences Center, Tucson, Arizona; and
| |
Collapse
|
50
|
Burckhardt G, Burckhardt BC. In vitro and in vivo evidence of the importance of organic anion transporters (OATs) in drug therapy. Handb Exp Pharmacol 2011:29-104. [PMID: 21103968 DOI: 10.1007/978-3-642-14541-4_2] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Organic anion transporters 1-10 (OAT1-10) and the urate transporter 1 (URAT1) belong to the SLC22A gene family and accept a huge variety of chemically unrelated endogenous and exogenous organic anions including many frequently described drugs. OAT1 and OAT3 are located in the basolateral membrane of renal proximal tubule cells and are responsible for drug uptake from the blood into the cells. OAT4 in the apical membrane of human proximal tubule cells is related to drug exit into the lumen and to uptake of estrone sulfate and urate from the lumen into the cell. URAT1 is the major urate-absorbing transporter in the apical membrane and is a target for uricosuric drugs. OAT10, also located in the luminal membrane, transports nicotinate with high affinity and interacts with drugs. Major extrarenal locations of OATs include the blood-brain barrier for OAT3, the placenta for OAT4, the nasal epithelium for OAT6, and the liver for OAT2 and OAT7. For all transporters we provide information on cloning, tissue distribution, factors influencing OAT abundance, interaction with endogenous compounds and different drug classes, drug/drug interactions and, if known, single nucleotide polymorphisms.
Collapse
Affiliation(s)
- Gerhard Burckhardt
- Abteilung Vegetative Physiologie und Pathophysiologie, Zentrum Physiologie und Pathophysiologie, Göttingen, Germany.
| | | |
Collapse
|