1
|
Zhang M, Li Y, Ma Y, Jin Y, Gou X, Yuan Y, Xu F, Wu X. The toxicity of cisplatin derives from effects on renal organic ion transporters expression and serum endogenous substance levels. Food Chem Toxicol 2024; 192:114949. [PMID: 39182635 DOI: 10.1016/j.fct.2024.114949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/04/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Acute kidney injury (AKI) is a worldwide public health problem with high morbidity and mortality. Cisplatin is a widely used chemotherapeutic agent for treating solid tumors, but the induction of AKI restricts its clinical application. In this study, the effect of cisplatin on the expression of organic ion transporters was investigated through in vivo and in vitro experiments. Targeted metabolomics techniques were used to measure the levels of selected endogenous substances in serum. Transmission electron microscopy was used to observe the microstructure of renal tubular epithelial cells. Our results show that the toxicity of cisplatin on HK-2 cells or HEK-293 cells was time- and dose-dependent. Administration of cisplatin decreased the expression of OAT1/3 and OCT2 and increased the expression of MRP2/4. Mitochondrial damage induced by cisplatin lead to renal tubular epithelial cell injury. In addition, administration of cisplatin resulted in significant changes in endogenous substance levels in serum, including amino acids, carnitine, and fatty acids. These serum amino acids and metabolites (α-aminobutyric acid, proline, and alanine), carnitines (tradecanoylcarnitine, hexanylcarnitine, octanoylcarnitine, 2-methylbutyroylcarnitine, palmitoylcarnitine, and linoleylcarnitine) and fatty acids (9E-tetradecenoic acid) represent endogenous substances with diagnostic potential for cisplatin-induced AKI.
Collapse
Affiliation(s)
- Mingkang Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China
| | - Yile Li
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China
| | - Yanrong Ma
- Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China
| | - Yongwen Jin
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China
| | - Xueyan Gou
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China
| | - Yufan Yuan
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China
| | - Fen Xu
- Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China
| | - Xin'an Wu
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China; Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China.
| |
Collapse
|
2
|
Wang XL, Li L, Meng X. Interplay between the Redox System and Renal Tubular Transport. Antioxidants (Basel) 2024; 13:1156. [PMID: 39456410 PMCID: PMC11505102 DOI: 10.3390/antiox13101156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/03/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
The kidney plays a critical role in maintaining the homeostasis of body fluid by filtration of metabolic wastes and reabsorption of nutrients. Due to the overload, a vast of energy is required through aerobic metabolism, which inevitably leads to the generation of reactive oxygen species (ROS) in the kidney. Under unstressed conditions, ROS are counteracted by antioxidant systems and maintained at low levels, which are involved in signal transduction and physiological processes. Accumulating evidence indicates that the reduction-oxidation (redox) system interacts with renal tubular transport. Redox imbalance or dysfunction of tubular transport leads to renal disease. Here, we discuss the ROS and antioxidant systems in the kidney and outline the metabolic dysfunction that is a common feature of renal disease. Importantly, we describe the key molecules involved in renal tubular transport and their relationship to the redox system and, finally, summarize the impact of their dysregulation on the pathogenesis and progression of acute and chronic kidney disease.
Collapse
Affiliation(s)
- Xiao-Lan Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Lianjian Li
- Department of Vascular Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Hubei Academy of Chinese Medicine, Wuhan 430061, China;
| | - Xianfang Meng
- Department of Neurobiology, Institute of Brain Research, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
3
|
Lim YJ, Xiu SG, Kuruvilla MS, Winquist E, Welch S, Black M, Faught LN, Lee J, Rieder MJ, Blydt-Hansen TD, Zappitelli M, Urquhart BL. Metabolomic identification of predictive and early biomarkers of cisplatin-induced acute kidney injury in adult head and neck cancer patients. Br J Clin Pharmacol 2024; 90:1790-1803. [PMID: 36657745 DOI: 10.1111/bcp.15666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/16/2022] [Accepted: 01/03/2023] [Indexed: 01/21/2023] Open
Abstract
AIM Cisplatin causes acute kidney injury (AKI) in approximately one third of patients. Serum creatinine and urinary output are poor markers of cisplatin-induced AKI. Metabolomics was utilized to identify predictive or early diagnostic biomarkers of cisplatin-induced AKI. METHODS Thirty-one adult head and neck cancer patients receiving cisplatin (dose ≥70 mg/m2) were recruited for metabolomics analysis. Urine and serum samples were collected prior to cisplatin (pre), 24-48 h after cisplatin (24-48 h) and 5-14 days (post) after cisplatin. Based on serum creatinine concentrations measured at the post timepoint, 11/31 patients were classified with clinical AKI. Untargeted metabolomics was performed using liquid chromatography-mass spectrometry (LC-MS). RESULTS Metabolic discrimination was observed between "AKI" patients and "no AKI" patients at all timepoints. Urinary glycine, hippuric acid sulfate, 3-hydroxydecanedioc acid and suberate were significantly different between AKI patients and no AKI patients prior to cisplatin infusion. Urinary glycine and hippuric acid sulfate were lower (-2.22-fold and -8.85-fold), whereas 3-hydroxydecanedioc acid and suberate were higher (3.62-fold and 1.91-fold) in AKI patients relative to no AKI patients. Several urine and serum metabolites were found to be altered 24-48 h following cisplatin infusion, particularly metabolites involved with mitochondrial energetics. CONCLUSIONS We propose glycine, hippuric acid sulfate, 3-hydroxydecanedioc acid and suberate as predictive biomarkers of predisposition to cisplatin-induced AKI. Metabolites indicative of mitochondrial dysfunction may serve as early markers of subclinical AKI.
Collapse
Affiliation(s)
- Yong Jin Lim
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Steven G Xiu
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - M Sara Kuruvilla
- Division of Medical Oncology, Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Eric Winquist
- Division of Medical Oncology, Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Stephen Welch
- Division of Medical Oncology, Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Morgan Black
- Division of Medical Oncology, Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Lauren N Faught
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Jasmine Lee
- Division of Nephrology, Department of Pediatrics, Hospital for Sick Children, Toronto, ON, Canada
| | - Michael J Rieder
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Paediatrics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Division of Clinical Pharmacology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Tom D Blydt-Hansen
- Division of Nephrology, Department of Pediatrics, British Columbia Children's Hospital, Vancouver, BC, Canada
| | - Michael Zappitelli
- Division of Nephrology, Department of Pediatrics, Hospital for Sick Children, Toronto, ON, Canada
| | - Bradley L Urquhart
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Division of Nephrology, Department of Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| |
Collapse
|
4
|
Kamal MM, El-Abhar HS, Abdallah DM, Ahmed KA, Aly NES, Rabie MA. Mirabegron, dependent on β3-adrenergic receptor, alleviates mercuric chloride-induced kidney injury by reversing the impact on the inflammatory network, M1/M2 macrophages, and claudin-2. Int Immunopharmacol 2024; 126:111289. [PMID: 38016347 DOI: 10.1016/j.intimp.2023.111289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 11/30/2023]
Abstract
The β3-adrenergic receptor (β3-AR) agonism mirabegron is used to treat overactive urinary bladder syndrome; however, its role against acute kidney injury (AKI) is not unveiled, hence, we aim to repurpose mirabegron in the treatment of mercuric chloride (HgCl2)-induced AKI. Rats were allocated into normal, normal + mirabegron, HgCl2 untreated, HgCl2 + mirabegron, and HgCl2 + the β3-AR blocker SR59230A + mirabegron. The latter increased the mRNA of β3-AR and miR-127 besides downregulating NF-κB p65 protein expression and the contents of its downstream targets iNOS, IL-4, -13, and -17 but increased that of IL-10 to attest its anti-inflammatory capacity. Besides, mirabegron downregulated the protein expression of STAT-6, PI3K, and ERK1/2, the downstream targets of the above cytokines. Additionally, it enhanced the transcription factor PPAR-α but turned off the harmful hub HNF-4α/HNF-1α and the lipid peroxide marker MDA. Mirabegron also downregulated the CD-163 protein expression, which besides the inhibited correlated cytokines of M1 (NF-κB p65, iNOS, IL-17) and M2 (IL-4, IL-13, CD163, STAT6, ERK1/2), inactivated the macrophage phenotypes. The crosstalk between these parameters was echoed in the maintenance of claudin-2, kidney function-related early (cystatin-C, KIM-1, NGAL), and late (creatinine, BUN) injury markers, besides recovering the microscopic structures. Nonetheless, the pre-administration of SR59230A has nullified the beneficial effects of mirabegron on the aforementioned parameters. Here we verified that mirabegron can berepurposedto treat HgCl2-induced AKI by activating the β3-AR. Mirabegron signified its effect by inhibiting inflammation, oxidative stress, and the activated M1/M2 macrophages, events that preserved the proximal tubular tight junction claudin-2 via the intersection of several trajectories.
Collapse
Affiliation(s)
- Mahmoud M Kamal
- Research Institute of Medical Entomology, General Organization for Teaching Hospitals and Institutes, Cairo, Egypt
| | - Hanan S El-Abhar
- Department of Pharmacology, Toxicology, and Biochemistry, Faculty of Pharmacy, Future University in Egypt (FUE), 11835 Cairo, Egypt
| | - Dalaal M Abdallah
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, 11562 Cairo, Egypt.
| | - Kawkab A Ahmed
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Nour Eldin S Aly
- Research Institute of Medical Entomology, General Organization for Teaching Hospitals and Institutes, Cairo, Egypt
| | - Mostafa A Rabie
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, 11562 Cairo, Egypt; Faculty of Pharmacy and Drug Technology, Egyptian Chinese University (ECU), 19346, Egypt
| |
Collapse
|
5
|
Yuan W, Kou S, Ma Y, Qian Y, Li X, Chai Y, Jiang Z, Zhang L, Sun L, Huang X. Hyperoside ameliorates cisplatin-induced acute kidney injury by regulating the expression and function of Oat1. Xenobiotica 2023; 53:559-571. [PMID: 37885225 DOI: 10.1080/00498254.2023.2270046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023]
Abstract
Cisplatin is a widely used chemotherapeutic agent to treat solid tumours in clinics. However, cisplatin-induced acute kidney injury (AKI) limits its clinical application. This study investigated the effect of hyperoside (a flavonol glycoside compound) on regulating AKI.The model of cisplatin-induced AKI was established, and hyperoside was preadministered to investigate its effect on improving kidney injury.Hyperoside ameliorated renal pathological damage, reduced the accumulation of SCr, BUN, Kim-1 and indoxyl sulphate in vivo, increased the excretion of indoxyl sulphate into the urine, and upregulated the expression of renal organic anion transporter 1 (Oat1). Moreover, evaluation of rat kidney slices demonstrated that hyperoside promoted the uptake of PAH (p-aminohippurate, the Oat1 substrate), which was confirmed by transient over-expression of OAT1 in HEK-293T cells. Additionally, hyperoside upregulated the mRNA expression of Oat1 upstream regulators hepatocyte nuclear factor-1α (HNF-1α) and pregnane X receptor (PXR).These findings indicated hyperoside could protect against cisplatin-induced AKI by promoting indoxyl sulphate excretion through regulating the expression and function of Oat1, suggesting hyperoside may offer a potential tactic for cisplatin-induced AKI treatment.
Collapse
Affiliation(s)
- Wenjing Yuan
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China
| | - Shanshan Kou
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China
| | - Ying Ma
- Foreign Language Teaching Department, Nanjing University of Chinese Medicine, Nanjing, PR China
| | - Yusi Qian
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China
| | - Xinyu Li
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China
| | - Yuanyuan Chai
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China
| | - Zhenzhou Jiang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China
| | - Luyong Zhang
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Lixin Sun
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China
| | - Xin Huang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China
| |
Collapse
|
6
|
Chen Y, Li H, Wang K, Wang Y. Recent Advances in Synthetic Drugs and Natural Actives Interacting with OAT3. Molecules 2023; 28:4740. [PMID: 37375294 DOI: 10.3390/molecules28124740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/03/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Organic anion transporter 3 (OAT3) is predominantly expressed in the kidney and plays a vital role in drug clearance. Consequently, co-ingestion of two OAT3 substrates may alter the pharmacokinetics of the substrate. This review summarizes drug-drug interactions (DDIs) and herbal-drug interactions (HDIs) mediated by OAT3, and inhibitors of OAT3 in natural active compounds in the past decade. This provides a valuable reference for the combined use of substrate drugs/herbs for OAT3 in clinical practice in the future and for the screening of OAT3 inhibitors to avoid harmful interactions.
Collapse
Affiliation(s)
- Ying Chen
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China
- Rizhao Huawei Institute of Comprehensive Health Industries, Shandong Keepfit Biotech. Co., Ltd., Rizhao 276800, China
| | - Hongyan Li
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China
- Rizhao Huawei Institute of Comprehensive Health Industries, Shandong Keepfit Biotech. Co., Ltd., Rizhao 276800, China
| | - Ke Wang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China
- Rizhao Huawei Institute of Comprehensive Health Industries, Shandong Keepfit Biotech. Co., Ltd., Rizhao 276800, China
| | - Yousheng Wang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China
- Rizhao Huawei Institute of Comprehensive Health Industries, Shandong Keepfit Biotech. Co., Ltd., Rizhao 276800, China
| |
Collapse
|
7
|
Mandal AK, Leask MP, Sumpter NA, Choi HK, Merriman TR, Mount DB. Genetic and Physiological Effects of Insulin-Like Growth Factor-1 (IGF-1) on Human Urate Homeostasis. J Am Soc Nephrol 2023; 34:451-466. [PMID: 36735516 PMCID: PMC10103387 DOI: 10.1681/asn.0000000000000054] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 10/25/2022] [Indexed: 01/22/2023] Open
Abstract
SIGNIFICANCE STATEMENT Hyperinsulinemia induces hyperuricemia by activating net renal urate reabsorption in the renal proximal tubule. The basolateral reabsorptive urate transporter GLUT9a appears to be the dominant target for insulin. By contrast, IGF-1 infusion reduces serum urate (SU), through mechanisms unknown. Genetic variants of IGF1R associated with reduced SU have increased IGF-1R expression and interact with genes encoding the GLUT9 and ABCG2 urate transporters, in a sex-specific fashion, which controls the SU level. Activation of IGF-1/IGF-1R signaling in Xenopus oocytes modestly activates GLUT9a and inhibits insulin's stimulatory effect on the transporter, which also activates multiple secretory urate transporters-ABCG2, ABCC4, OAT1, and OAT3. The results collectively suggest that IGF-1 reduces SU by activating secretory urate transporters and inhibiting insulin's action on GLUT9a. BACKGROUND Metabolic syndrome and hyperinsulinemia are associated with hyperuricemia. Insulin infusion in healthy volunteers elevates serum urate (SU) by activating net urate reabsorption in the renal proximal tubule, whereas IGF-1 infusion reduces SU by mechanisms unknown. Variation within the IGF1R gene also affects SU levels. METHODS Colocalization analyses of a SU genome-wide association studies signal at IGF1R and expression quantitative trait loci signals in cis using COLOC2, RT-PCR, Western blotting, and urate transport assays in transfected HEK 293T cells and in Xenopus laevis oocytes. RESULTS Genetic association at IGF1R with SU is stronger in women and is mediated by control of IGF1R expression. Inheritance of the urate-lowering homozygous genotype at the SLC2A9 locus is associated with a differential effect of IGF1R genotype between men and women. IGF-1, through IGF-1R, stimulated urate uptake in human renal proximal tubule epithelial cells and transfected HEK 293T cells, through activation of IRS1, PI3/Akt, MEK/ERK, and p38 MAPK; urate uptake was inhibited in the presence of uricosuric drugs, specific inhibitors of protein tyrosine kinase, PI3 kinase (PI3K), ERK, and p38 MAPK. In X. laevis oocytes expressing ten individual urate transporters, IGF-1 through endogenous IGF-1R stimulated urate transport mediated by GLUT9, OAT1, OAT3, ABCG2, and ABCC4 and inhibited insulin's stimulatory action on GLUT9a and OAT3. IGF-1 significantly activated Akt and ERK. Specific inhibitors of PI3K, ERK, and PKC significantly affected IGF-1 stimulation of urate transport in oocytes. CONCLUSIONS The combined results of infusion, genetics, and transport experiments suggest that IGF-1 reduces SU by activating urate secretory transporters and inhibiting insulin's action.
Collapse
Affiliation(s)
- Asim K. Mandal
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Megan P. Leask
- Biochemistry Department, University of Otago, Dunedin, South Island, New Zealand
- Division of Rheumatology and Clinical Immunology, University of Alabama, Birmingham, Alabama
| | - Nicholas A. Sumpter
- Division of Rheumatology and Clinical Immunology, University of Alabama, Birmingham, Alabama
| | - Hyon K. Choi
- Division of Rheumatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tony R. Merriman
- Biochemistry Department, University of Otago, Dunedin, South Island, New Zealand
- Division of Rheumatology and Clinical Immunology, University of Alabama, Birmingham, Alabama
| | - David B. Mount
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Renal Division, VA Boston Healthcare System, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
8
|
The Role of Organic Cation Transporters in the Pharmacokinetics, Pharmacodynamics and Drug-Drug Interactions of Tyrosine Kinase Inhibitors. Int J Mol Sci 2023; 24:ijms24032101. [PMID: 36768423 PMCID: PMC9917293 DOI: 10.3390/ijms24032101] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) decisively contributed in revolutionizing the therapeutic approach to cancer, offering non-invasive, tolerable therapies for a better quality of life. Nonetheless, degree and duration of the response to TKI therapy vary depending on cancer molecular features, the ability of developing resistance to the drug, on pharmacokinetic alterations caused by germline variants and unwanted drug-drug interactions at the level of membrane transporters and metabolizing enzymes. A great deal of approved TKIs are inhibitors of the organic cation transporters (OCTs). A handful are also substrates of them. These transporters are polyspecific and highly expressed in normal epithelia, particularly the intestine, liver and kidney, and are, hence, arguably relevant sites of TKI interactions with other OCT substrates. Moreover, OCTs are often repressed in cancer cells and might contribute to the resistance of cancer cells to TKIs. This article reviews the OCT interactions with approved and in-development TKIs reported in vitro and in vivo and critically discusses the potential clinical ramifications thereof.
Collapse
|
9
|
Gou X, Ran F, Yang J, Ma Y, Wu X. Construction and Evaluation of a Novel Organic Anion Transporter 1/3 CRISPR/Cas9 Double-Knockout Rat Model. Pharmaceutics 2022; 14:2307. [PMID: 36365126 PMCID: PMC9697873 DOI: 10.3390/pharmaceutics14112307] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/13/2022] [Accepted: 10/25/2022] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND Organic anion transporter 1 (OAT1) and OAT3 have an overlapping spectrum of substrates such that one can exert a compensatory effect when the other is dysfunctional. As a result, the knockout of either OAT1 or OAT3 is not reflected in a change in the excretion of organic anionic substrates. To date, only the mOAT1 and mOAT3 individual knockout mouse models have been available. METHODS In this study, we successfully generated a Slc22a6/Slc22a8 double-knockout (KO) rat model using CRISPR/Cas9 technology and evaluated its biological properties. RESULTS The double-knockout rat model did not expression mRNA for rOAT1 or rOAT3 in the kidneys. Consistently, the renal excretion of p-aminohippuric acid (PAH), the classical substrate of OAT1/OAT3, was substantially decreased in the Slc22a6/Slc22a8 double-knockout rats. The relative mRNA level of Slco4c1 was up-regulated in KO rats. No renal pathological phenotype was evident. The renal elimination of the organic anionic drug furosemide was nearly abolished in the Slc22a6/Slc22a8 knockout rats, but elimination of the organic cationic drug metformin was hardly affected. CONCLUSIONS These results demonstrate that this rat model is a useful tool for investigating the functions of OAT1/OAT3 in metabolic diseases, drug metabolism and pharmacokinetics, and OATs-mediated drug interactions.
Collapse
Affiliation(s)
- Xueyan Gou
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730013, China
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Fenglin Ran
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jinru Yang
- School of First Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Yanrong Ma
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730013, China
| | - Xin’an Wu
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730013, China
| |
Collapse
|
10
|
Gales A, Monteiro-Pai S, Hyndman KA. Endothelin system expression in the kidney following cisplatin-induced acute kidney injury in male and female mice. Can J Physiol Pharmacol 2022; 100:868-879. [PMID: 35704945 PMCID: PMC9904337 DOI: 10.1139/cjpp-2022-0126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The chemotherapeutic agent cisplatin accumulates in the kidney and induces acute kidney injury (AKI). Preclinical and clinical studies suggest that young female mice and women show greater recovery from cisplatin-AKI compared to young male mice and men. The endothelin (ET) and ET receptors are enriched in the kidney and may be dysfunctional in cisplatin-AKI; however, there is a gap in our knowledge about the putative effects of sex and cisplatin on the renal ET system. We hypothesized that cisplatin-AKI male and female mice will have increased expression of the renal ET system. As expected, all cisplatin-AKI mice had kidney damage and body weight loss greater than control mice. Cisplatin-AKI mice had greater cortical Edn1, Edn3, Ednra, and Ednrb, while outer medullary Ednra was significantly suppressed in both sexes. Of the ∼25 000 genes sequenced from the inner medulla, only 91 genes (comparing saline mice) and 134 genes (comparing cisplatin-AKI mice) were differentially expressed and they were unrelated to the ET system. However, Edn1 was significantly greater in the inner medulla of male and female cisplatin-AKI mice. Thus, RNA profiles of the ET system were significantly affected by cisplatin-AKI throughout the kidney regardless of sex and this may help determine the therapeutic potential of targeting the ET receptors in cisplatin-AKI.
Collapse
Affiliation(s)
- Anabelle Gales
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sureena Monteiro-Pai
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kelly A. Hyndman
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
11
|
Wu A, Lu J, Zhong G, Lu L, Qu Y, Zhang C. Xanthotoxin (8-methoxypsoralen): A review of its chemistry, pharmacology, pharmacokinetics, and toxicity. Phytother Res 2022; 36:3805-3832. [PMID: 35913174 DOI: 10.1002/ptr.7577] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/30/2022] [Accepted: 07/14/2022] [Indexed: 11/11/2022]
Abstract
Xanthotoxin (XAT) is a natural furanocoumarins, a bioactive psoralen isolated from the fruit of the Rutaceae plant Pepper, which has received increasing attention in recent years due to its wide source and low cost. By collecting and compiling literature on XAT, the results show that XAT exhibits significant activity in the treatment of various diseases, including neuroprotection, skin repair, osteoprotection, organ protection, anticancer, antiinflammatory, antioxidative stress and antibacterial. In this paper, we review the pharmacological activity and potential molecular mechanisms of XAT for the treatment of related diseases. The data suggest that XAT can mechanistically induce ROS production and promote apoptosis through mitochondrial or endoplasmic reticulum pathways, regulate NF-κB, MAPK, JAK/STAT, Nrf2/HO-1, MAPK, AKT/mTOR, and ERK1/2 signaling pathways to exert pharmacological effects. In addition, the pharmacokinetics properties and toxicity of XAT are discussed in this paper, further elucidating the relationship between structure and efficacy. It is worth noting that data from clinical studies of XAT are still scarce, limiting the use of XAT in the clinic, and in the future, more in-depth studies are needed to determine the clinical efficacy of XAT.
Collapse
Affiliation(s)
- Anxin Wu
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.,State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Jing Lu
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.,State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Guofeng Zhong
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.,State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Ling Lu
- Chengdu University of Technology, Chengdu, PR China
| | - Yan Qu
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.,State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Chen Zhang
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.,State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| |
Collapse
|
12
|
Differentiated kidney tubular cell-derived extracellular vesicles enhance maturation of tubuloids. J Nanobiotechnology 2022; 20:326. [PMID: 35841001 PMCID: PMC9284832 DOI: 10.1186/s12951-022-01506-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/09/2022] [Indexed: 12/04/2022] Open
Abstract
The prevalence of end-stage kidney disease (ESKD) is rapidly increasing with the need for regenerative therapies. Adult stem cell derived kidney tubuloids have the potential to functionally mimic the adult kidney tubule, but still lack the expression of important transport proteins needed for waste removal. Here, we investigated the potential of extracellular vesicles (EVs) obtained from matured kidney tubular epithelial cells to modulate in vitro tubuloids functional maturation. We focused on organic anion transporter 1 (OAT1), one of the most important proteins involved in endogenous waste excretion. First, we show that EVs from engineered proximal tubule cells increased the expression of several transcription factors and epithelial transporters, resulting in improved OAT1 transport capacity. Next, a more in-depth proteomic data analysis showed that EVs can trigger various biological pathways, including mesenchymal-to-epithelial transition, which is crucial in the tubular epithelial maturation. Moreover, we demonstrated that the combination of EVs and tubuloid-derived cells can be used as part of a bioartificial kidney to generate a tight polarized epithelial monolayer with formation of dense cilia structures. In conclusion, EVs from kidney tubular epithelial cells can phenotypically improve in vitro tubuloid maturation, thereby enhancing their potential as functional units in regenerative or renal replacement therapies.
Collapse
|
13
|
Xu Y, Chen J, Ruan Z, Jiang B, Yang D, Hu Y, Lou H. Simulation of Febuxostat Pharmacokinetics in Healthy Subjects and Patients with Impaired Kidney Function Using Physiologically Based Pharmacokinetic Modeling. Biopharm Drug Dispos 2022; 43:140-151. [PMID: 35748093 DOI: 10.1002/bdd.2325] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 05/08/2022] [Accepted: 06/03/2022] [Indexed: 11/11/2022]
Abstract
Febuxostat is recommend by the American College of Rheumatology Gout Management Guidelines as a first-line therapy for lowering the level of urate in patients with gout. At present, this drug is being prescribed mainly based on the clinical experience of doctors. The potential effects of clinical and demographic variables on the bioavailability and therapeutic effectiveness of febuxostat are not being considered. In this study, a physiologically based pharmacokinetic (PBPK) model of febuxostat was developed, thereby providing a theoretical basis for the individualized dosing of this drug in gout patients. The plasma concentration-time profiles corresponding to healthy subjects and gout patients with normal kidney function were simulated and validated; then, the model was used to predict the pharmacokinetic (PK) data of the drug in gout patients suffering from varying degrees of impaired kidney function. The error values (the predicted value/observed value) were used to validate the simulated PK parameters predicted by the PBPK model, including the area under the plasma concentration-time curve, the maximum plasma concentration, and time to maximum plasma concentration. Considering that to all error fold changes were smaller than 2 the PBPK model was. In subjects suffering from mild kidney impairment, moderate kidney impairment, severe kidney impairment, and end-stage kidney disease (ESRD), the predicted AUC0-24h values increased by 1.62, 1.74, 2.27, and 2.65-fold, respectively, compared to gout patients with normal kidney function. Overall, the results showed that the PBPK model constructed in this study predict the pharmacokinetic changes in gout patients suffering from varying degrees of impaired kidney function. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yichao Xu
- Center of Clinical Pharmacology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China
| | - Jinliang Chen
- Center of Clinical Pharmacology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China
| | - Zourong Ruan
- Center of Clinical Pharmacology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China
| | - Bo Jiang
- Center of Clinical Pharmacology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China
| | - Dandan Yang
- Center of Clinical Pharmacology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China
| | - Yin Hu
- Center of Clinical Pharmacology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China
| | - Honggang Lou
- Center of Clinical Pharmacology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Brouwer KLR, Evers R, Hayden E, Hu S, Li CY, Meyer Zu Schwabedissen HE, Neuhoff S, Oswald S, Piquette-Miller M, Saran C, Sjöstedt N, Sprowl JA, Stahl SH, Yue W. Regulation of Drug Transport Proteins-From Mechanisms to Clinical Impact: A White Paper on Behalf of the International Transporter Consortium. Clin Pharmacol Ther 2022; 112:461-484. [PMID: 35390174 PMCID: PMC9398928 DOI: 10.1002/cpt.2605] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/20/2022] [Indexed: 12/14/2022]
Abstract
Membrane transport proteins are involved in the absorption, disposition, efficacy, and/or toxicity of many drugs. Numerous mechanisms (e.g., nuclear receptors, epigenetic gene regulation, microRNAs, alternative splicing, post‐translational modifications, and trafficking) regulate transport protein levels, localization, and function. Various factors associated with disease, medications, and dietary constituents, for example, may alter the regulation and activity of transport proteins in the intestine, liver, kidneys, brain, lungs, placenta, and other important sites, such as tumor tissue. This white paper reviews key mechanisms and regulatory factors that alter the function of clinically relevant transport proteins involved in drug disposition. Current considerations with in vitro and in vivo models that are used to investigate transporter regulation are discussed, including strengths, limitations, and the inherent challenges in predicting the impact of changes due to regulation of one transporter on compensatory pathways and overall drug disposition. In addition, translation and scaling of in vitro observations to in vivo outcomes are considered. The importance of incorporating altered transporter regulation in modeling and simulation approaches to predict the clinical impact on drug disposition is also discussed. Regulation of transporters is highly complex and, therefore, identification of knowledge gaps will aid in directing future research to expand our understanding of clinically relevant molecular mechanisms of transporter regulation. This information is critical to the development of tools and approaches to improve therapeutic outcomes by predicting more accurately the impact of regulation‐mediated changes in transporter function on drug disposition and response.
Collapse
Affiliation(s)
- Kim L R Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Raymond Evers
- Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, Pennsylvania, USA
| | - Elizabeth Hayden
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Shuiying Hu
- College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | | | | | | | - Stefan Oswald
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Rostock, Germany
| | | | - Chitra Saran
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Noora Sjöstedt
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jason A Sprowl
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Simone H Stahl
- CVRM Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Wei Yue
- College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
15
|
Louisse J, Dorne JLCM, Dellafiora L. Investigating the interaction between organic anion transporter 1 and ochratoxin A: An in silico structural study to depict early molecular events of substrate recruitment and the impact of single point mutations. Toxicol Lett 2021; 355:19-30. [PMID: 34748852 DOI: 10.1016/j.toxlet.2021.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/18/2021] [Accepted: 11/02/2021] [Indexed: 12/24/2022]
Abstract
Organic anion transporters (OATs) belong to a subgroup of the solute carrier 22 transporter family. OATs have a central role in xenobiotic disposition affecting the toxicokinetics of its substrates and inter-individual differences in their expression, activity and function impact both toxicokinetics and toxicodynamics. Amongst OATs, OAT1 (solute carrier family 22 member 6) is involved in the urinary excretion of many xenobiotics bringing substrates into renal proximal tubular cells which can then be secreted across the apical membrane into the tubule lumen. The mycotoxin ochratoxin A has been shown to have a high affinity for OAT1, which is an important renal transporter involved in its urinary excretion. Nowadays, molecular modeling techniques are widely applied to assess protein-ligand interactions and may provide a tool to depict the mechanic of xenobiotic action be it toxicokinetics or toxicodynamics. This work provides a structured pipeline consisting of docking and molecular dynamic simulations to study OAT1-ligand interactions and the impact of OAT1 polymorphisms on such interactions. Such a computational structure-based analytical framework allowed to: i) model OAT1-substrate complex formation and depict the features correlating its sequence, structure and its capability to recruit substrates; and ii) investigate the impact of OAT1 missense mutations on substrate recruitment. Perspectives on applying such a structured pipeline to xenobiotic-metabolising enzymes are discussed.
Collapse
Affiliation(s)
- Jochem Louisse
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, the Netherlands
| | - Jean Lou C M Dorne
- Scientific Committee and Emerging Risks Unit, European Food Safety Authority, Via Carlo Magno 1A, Parma, 43124, Italy
| | - Luca Dellafiora
- Department of Food and Drug, University of Parma, Parma, 43124, Italy.
| |
Collapse
|
16
|
Molecular Properties of Drugs Handled by Kidney OATs and Liver OATPs Revealed by Chemoinformatics and Machine Learning: Implications for Kidney and Liver Disease. Pharmaceutics 2021; 13:pharmaceutics13101720. [PMID: 34684013 PMCID: PMC8538396 DOI: 10.3390/pharmaceutics13101720] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 12/31/2022] Open
Abstract
In patients with liver or kidney disease, it is especially important to consider the routes of metabolism and elimination of small-molecule pharmaceuticals. Once in the blood, numerous drugs are taken up by the liver for metabolism and/or biliary elimination, or by the kidney for renal elimination. Many common drugs are organic anions. The major liver uptake transporters for organic anion drugs are organic anion transporter polypeptides (OATP1B1 or SLCO1B1; OATP1B3 or SLCO1B3), whereas in the kidney they are organic anion transporters (OAT1 or SLC22A6; OAT3 or SLC22A8). Since these particular OATPs are overwhelmingly found in the liver but not the kidney, and these OATs are overwhelmingly found in the kidney but not liver, it is possible to use chemoinformatics, machine learning (ML) and deep learning to analyze liver OATP-transported drugs versus kidney OAT-transported drugs. Our analysis of >30 quantitative physicochemical properties of OATP- and OAT-interacting drugs revealed eight properties that in combination, indicate a high propensity for interaction with "liver" transporters versus "kidney" ones based on machine learning (e.g., random forest, k-nearest neighbors) and deep-learning classification algorithms. Liver OATPs preferred drugs with greater hydrophobicity, higher complexity, and more ringed structures whereas kidney OATs preferred more polar drugs with more carboxyl groups. The results provide a strong molecular basis for tissue-specific targeting strategies, understanding drug-drug interactions as well as drug-metabolite interactions, and suggest a strategy for how drugs with comparable efficacy might be chosen in chronic liver or kidney disease (CKD) to minimize toxicity.
Collapse
|
17
|
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: 4.3] [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
|
18
|
Droździk M, Oswald S, Droździk A. Impact of kidney dysfunction on hepatic and intestinal drug transporters. Biomed Pharmacother 2021; 143:112125. [PMID: 34474348 DOI: 10.1016/j.biopha.2021.112125] [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] [Received: 06/22/2021] [Revised: 08/11/2021] [Accepted: 08/24/2021] [Indexed: 12/16/2022] Open
Abstract
Emerging information suggests that pathology of the kidney may not only affect expression and function of membrane transporters in the organ, but also in the gastrointestinal tract and the liver. Transporter dysfunction may cause effects on handling of drug as well as endogenous compounds with subsequent clinical consequences. A literature search was conducted on Ovid and PubMed databases to select relevant in vitro, animal and human studies that have reported expression, protein abundance and function of the gastrointestinal and liver localized ABC transporters and SLC carriers in kidney dysfunction or uremia states. The altered function of drug transporters in the liver and intestines in kidney failure subjects may provide compensatory activity in handling endogenous compounds (e.g. uremic toxins), which is expected to affect drug pharmacokinetics and local drug actions.
Collapse
Affiliation(s)
- Marek Droździk
- Department of Pharmacology, Faculty of Medicine and Dentistry, Pomeranian Medical University, Powstancow Wlkp 72, 70-111 Szczecin, Poland.
| | - Stefan Oswald
- Institute of Pharmacology and Toxicology, Faculty of Medicine, Rostock University Medical Center, 18057 Rostock, Germany.
| | - Agnieszka Droździk
- Department of Integrated Dentistry, Faculty of Medicine and Dentistry, Pomeranian Medical University, Powstancow Wlkp 72, 70-111 Szczecin, Poland.
| |
Collapse
|
19
|
Torres AM, Dnyanmote AV, Granados JC, Nigam SK. Renal and non-renal response of ABC and SLC transporters in chronic kidney disease. Expert Opin Drug Metab Toxicol 2021; 17:515-542. [PMID: 33749483 DOI: 10.1080/17425255.2021.1899159] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The solute carrier (SLC) and the ATP-binding cassette (ABC) transporter superfamilies play essential roles in the disposition of small molecules (endogenous metabolites, uremic toxins, drugs) in the blood, kidney, liver, intestine, and other organs. In chronic kidney disease (CKD), the loss of renal function is associated with altered function of remote organs. As renal function declines, many molecules accumulate in the plasma. Many studies now support the view that ABC and SLC transporters as well as drug metabolizing enzymes (DMEs) in renal and non-renal tissues are directly or indirectly affected by the presence of various types of uremic toxins, including those derived from the gut microbiome; this can lead to aberrant inter-organ communication. AREAS COVERED Here, the expression, localization and/or function of various SLC and ABC transporters as well as DMEs in the kidney and other organs are discussed in the context of CKD and systemic pathophysiology. EXPERT OPINION According to the Remote Sensing and Signaling Theory (RSST), a transporter and DME-centric network that optimizes local and systemic metabolism maintains homeostasis in the steady state and resets homeostasis following perturbations due to renal dysfunction. The implications of this view for pharmacotherapy of CKD are also discussed.
Collapse
Affiliation(s)
- Adriana M Torres
- Pharmacology Area, Faculty of Biochemistry and Pharmaceutical Sciences, National University of Rosario, CONICET, Suipacha 531, S2002LRK Rosario, Argentina
| | - Ankur V Dnyanmote
- Department of Pediatrics, IWK Health Centre - Dalhousie University, 5850 University Ave, Halifax, NS, B3K 6R8, Canada
| | - Jeffry C Granados
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093-0693, USA
| | - Sanjay K Nigam
- Departments of Pediatrics and Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093-0693, USA
| |
Collapse
|
20
|
Granados JC, Richelle A, Gutierrez JM, Zhang P, Zhang X, Bhatnagar V, Lewis NE, Nigam SK. Coordinate regulation of systemic and kidney tryptophan metabolism by the drug transporters OAT1 and OAT3. J Biol Chem 2021; 296:100575. [PMID: 33757768 PMCID: PMC8102410 DOI: 10.1016/j.jbc.2021.100575] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/09/2021] [Accepted: 03/19/2021] [Indexed: 12/18/2022] Open
Abstract
How organs sense circulating metabolites is a key question. Here, we show that the multispecific organic anion transporters of drugs, OAT1 (SLC22A6 or NKT) and OAT3 (SLC22A8), play a role in organ sensing. Metabolomics analyses of the serum of Oat1 and Oat3 knockout mice revealed changes in tryptophan derivatives involved in metabolism and signaling. Several of these metabolites are derived from the gut microbiome and are implicated as uremic toxins in chronic kidney disease. Direct interaction with the transporters was supported with cell-based transport assays. To assess the impact of the loss of OAT1 or OAT3 function on the kidney, an organ where these uptake transporters are highly expressed, knockout transcriptomic data were mapped onto a “metabolic task”-based computational model that evaluates over 150 cellular functions. Despite the changes of tryptophan metabolites in both knockouts, only in the Oat1 knockout were multiple tryptophan-related cellular functions increased. Thus, deprived of the ability to take up kynurenine, kynurenate, anthranilate, and N-formylanthranilate through OAT1, the kidney responds by activating its own tryptophan-related biosynthetic pathways. The results support the Remote Sensing and Signaling Theory, which describes how “drug” transporters help optimize levels of metabolites and signaling molecules by facilitating organ cross talk. Since OAT1 and OAT3 are inhibited by many drugs, the data implies potential for drug–metabolite interactions. Indeed, treatment of humans with probenecid, an OAT-inhibitor used to treat gout, elevated circulating tryptophan metabolites. Furthermore, given that regulatory agencies have recommended drugs be tested for OAT1 and OAT3 binding or transport, it follows that these metabolites can be used as endogenous biomarkers to determine if drug candidates interact with OAT1 and/or OAT3.
Collapse
Affiliation(s)
- Jeffry C Granados
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Anne Richelle
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Jahir M Gutierrez
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Patrick Zhang
- Department of Biology, University of California San Diego, La Jolla, California, USA
| | - Xinlian Zhang
- Division of Biostatistics and Bioinformatics, Department of Family Medicine and Public Health, University of California San Diego, La Jolla, California, USA
| | - Vibha Bhatnagar
- Department of Family and Preventative Medicine, University of California San Diego, La Jolla, California, USA
| | - Nathan E Lewis
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA; Department of Pediatrics, University of California San Diego, La Jolla, California, USA; Novo Nordisk Foundation Center for Biosustainability at UC San Diego, University of California San Diego, La Jolla, California, USA
| | - Sanjay K Nigam
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA; Department of Medicine, University of California San Diego, La Jolla, California, USA.
| |
Collapse
|
21
|
Shen WC, Chou YH, Shi LS, Chen ZW, Tu HJ, Lin XY, Wang GJ. AST-120 Improves Cardiac Dysfunction in Acute Kidney Injury Mice via Suppression of Apoptosis and Proinflammatory NF-κB/ICAM-1 Signaling. J Inflamm Res 2021; 14:505-518. [PMID: 33658826 PMCID: PMC7917393 DOI: 10.2147/jir.s283378] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/09/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose Acute kidney injury (AKI) is a devastating disorder associated with considerably high morbidity and mortality. Reports have shown that AST-120, an oral charcoal adsorbent, can reduce oxidative stress by lowering serum indoxyl sulfate levels. The effects of AST-120 and indoxyl sulfate on kidney injury and cardiac dysfunction were investigated in vivo and in vitro. Patients and Methods Patients were tracked for enrollment upon receiving a diagnosis of AKI. Plasma was collected to determine the renal and inflammatory parameters. Renal ischemia/reperfusion (I/R) induced AKI or sham operation was performed in C57BL/6J mice. Animals were divided into sham, AKI+vehicle, and AKI+AST-120 groups. Plasma and tissues were assembled after 48 h to assess apoptotic and inflammatory responses. We also conducted human umbilical vein endothelial cell (HUVECs) and HL-1 cardiomyocyte culture studies to determine the underlying mechanisms of indoxyl sulfate’s effects. Echocardiography, histopathology, biochemical indexes, ELISA, terminal dUTP nick-end labeling (TUNEL) and Western blot analysis were performed. Results The cohort included 25 consecutive patients with AKI and 25 non-AKI. Plasma levels of creatinine, indoxyl sulfate, IL-1β and ICAM-1 were significantly higher in patients with AKI than in non-AKI controls. Plasma levels of blood urea nitrogen, creatinine, indoxyl sulfate, IL-1β and renal tubular injury were increased in mice after renal I/R and were decreased by AST-120 treatment. In addition, AST-120 therapy not only improved the parameters assessed by echocardiography but also substantially attenuated the elevation of plasma BNP. Oral administration of AST-120 significantly downregulated NF-κB/ICAM-1 expression and reduced cell apoptosis in both kidney and heart after renal I/R injury. Conclusion Our investigations demonstrated that AST-120 administration improves cardiac dysfunction in AKI mice via the suppression of apoptosis and proinflammatory NF-κB/ICAM-1 signaling.
Collapse
Affiliation(s)
- Wen-Ching Shen
- Department of Basic Medicine, Putian University, Putian City, Fujian Province, People's Republic of China
| | - Yu-Hsiang Chou
- Department of Internal Medicine, National Taiwan University Hospital Jin-Shan Branch, New Taipei City, Taiwan
| | - Li-Shian Shi
- Department of Biotechnology, National Formosa University, Yun-Lin, Taiwan
| | - Zhi-Wei Chen
- The Affiliated Hospital of Putian University, Putian City, Fujian Province, People's Republic of China
| | - Hai-Jian Tu
- The Affiliated Hospital of Putian University, Putian City, Fujian Province, People's Republic of China
| | - Xin-Yi Lin
- Department of Basic Medicine, Putian University, Putian City, Fujian Province, People's Republic of China
| | - Guei-Jane Wang
- School of Medicine, Graduate Institute of Clinical Medical Science, China Medical University, Taichung, 40402, Taiwan.,School of Medicine, Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 40402, Taiwan.,Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan.,Department of Health and Nutrition Biotechnology, Asia University, Taichung, 41354, Taiwan
| |
Collapse
|
22
|
Yabuuchi N, Hou H, Gunda N, Narita Y, Jono H, Saito H. Suppressed Hepatic Production of Indoxyl Sulfate Attenuates Cisplatin-Induced Acute Kidney Injury in Sulfotransferase 1a1-Deficient Mice. Int J Mol Sci 2021; 22:ijms22041764. [PMID: 33578912 PMCID: PMC7916706 DOI: 10.3390/ijms22041764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/08/2021] [Accepted: 02/08/2021] [Indexed: 12/26/2022] Open
Abstract
Endogenous factors involved in the progression of cisplatin nephropathy remain undetermined. Here, we demonstrate the toxico-pathological roles of indoxyl sulfate (IS), a sulfate-conjugated uremic toxin, and sulfotransferase 1A1 (SULT1A1), an enzyme involved in its synthesis, in cisplatin-induced acute kidney injury using Sult1a1-deficient (Sult1a1-/- KO) mice. With cisplatin administration, severe kidney dysfunction, tissue damage, and apoptosis were attenuated in Sult1a1-/- (KO) mice. Aryl hydrocarbon receptor (AhR) expression was increased by treatment with cisplatin in mouse kidney tissue. Moreover, the downregulation of antioxidant stress enzymes in wild-type (WT) mice was not observed in Sult1a1-/- (KO) mice. To investigate the effect of IS on the reactive oxygen species (ROS) levels, HK-2 cells were treated with cisplatin and IS. The ROS levels were significantly increased compared to cisplatin or IS treatment alone. IS-induced increases in ROS were reversed by downregulation of AhR, xanthine oxidase (XO), and NADPH oxidase 4 (NOX4). These findings suggest that SULT1A1 plays toxico-pathological roles in the progression of cisplatin-induced acute kidney injury, while the IS/AhR/ROS axis brings about oxidative stress.
Collapse
Affiliation(s)
- Nozomi Yabuuchi
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan; (N.Y.); (H.H.); (N.G.); (Y.N.); (H.J.)
| | - Huixian Hou
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan; (N.Y.); (H.H.); (N.G.); (Y.N.); (H.J.)
| | - Nao Gunda
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan; (N.Y.); (H.H.); (N.G.); (Y.N.); (H.J.)
| | - Yuki Narita
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan; (N.Y.); (H.H.); (N.G.); (Y.N.); (H.J.)
- Department of Pharmacy, Kumamoto University Hospital, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Hirofumi Jono
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan; (N.Y.); (H.H.); (N.G.); (Y.N.); (H.J.)
- Department of Pharmacy, Kumamoto University Hospital, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Hideyuki Saito
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan; (N.Y.); (H.H.); (N.G.); (Y.N.); (H.J.)
- Department of Pharmacy, Kumamoto University Hospital, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
- Correspondence: ; Tel./Fax: +81-96-373-5820
| |
Collapse
|
23
|
Chen M, You G, Xie C, Yang R, Hu W, Zheng Z, Liu S, Ye L. Pharmacokinetics of metformin in collagen-induced arthritis rats. Biochem Pharmacol 2021; 185:114413. [PMID: 33434538 DOI: 10.1016/j.bcp.2021.114413] [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] [Received: 11/04/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/22/2022]
Abstract
Due to the elevated presence of cytokines, the expressions of metabolic enzymes and drug transporters are altered in rheumatoid arthritis (RA). Given the high incidence of diabetes in patients with RA, the aim of the present study was to investigate the metformin pharmacokinetics of a single oral dose in rats with collagen-induced arthritis (CIA). Blood and urine samples were collected at different timepoints, and analyzed by ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). Tissue samples were also collected to investigate the expression of metabolic enzymes and drug transporters by quantitative reverse transcription-polymerase chain reaction (RT-qPCR) and western blot. The results indicated that the bioavailability of metformin was markedly decreased in the CIA rats. Moreover, metformin was not metabolized by enzymes of rat liver microsomes, suggesting that the decreased bioavailability of metformin was independent of the liver metabolism. In addition, the mRNA, protein expression level and activity of the renal organic cation transporter 2 (OCT2) was markedly increased, suggesting that the enhanced renal clearance of metformin in CIA rats may be due to the up-regulated activity of OCT2. In conclusion, our study suggested that the reduced bioavailability of metformin in CIA rats is possibly related to the up-regulated function of the renal protein OCT2.
Collapse
Affiliation(s)
- Minghao Chen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Department of Pharmacy, The First Affiliated Hospital of Jinan University (Guangzhou Overseas Chinese Hospital), Guangzhou 510630, China
| | - Guoquan You
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Cong Xie
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ruopeng Yang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wanyu Hu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhijie Zheng
- Clinical Pharmacology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Shuwen Liu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Southern Medical University, Guangzhou 510515, China.
| | - Ling Ye
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
24
|
O’Brien FJ, Mair RD, Plummer NS, Meyer TW, Sutherland SM, Sirich TL. Impaired Tubular Secretion of Organic Solutes in Acute Kidney Injury. KIDNEY360 2020; 1:724-730. [PMID: 35252876 PMCID: PMC8815732 DOI: 10.34067/kid.0001632020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/24/2020] [Indexed: 06/14/2023]
Abstract
BACKGROUND Impairment of kidney function is routinely assessed by measuring the accumulation of creatinine, an organic solute cleared largely by glomerular filtration. We tested whether the clearance of solutes that undergo tubular secretion is reduced in proportion to the clearance of creatinine in humans with AKI. METHODS Four endogenously produced organic solutes (phenylacetylglutamine [PAG], hippurate [HIPP], indoxyl sulfate [IS], and p-cresol sulfate [PCS]) were measured in spot urine and plasma samples from ten patients with AKI and 17 controls. Fractional clearance relative to creatinine was calculated to assess tubular secretion. Fractional clearance values were calculated in terms of the free, unbound levels of HIPP, IS, and PCS that bind to plasma proteins. RESULTS Fractional clearance values for PAG, HIPP, IS, and PCS were >1.0 in patients with AKI as well as controls, indicating that these solutes were still secreted by the tubules of the injured kidneys. Fractional clearance values were, however, significantly lower in patients with AKI than controls, indicating that kidney injury reduced tubular secretion more than glomerular filtration (AKI versus control: PAG, 2.1±0.7 versus 4.6±1.4, P<0.001; HIPP, 10±5 versus 15±7, P=0.02; IS, 10±6 versus 28±7, P<0.001; PCS, 3.3±1.8 versus 10±3, P<0.001). Free plasma levels rose out of proportion to total plasma levels for each of the bound solutes in AKI, so that calculating their fractional clearance in terms of their total plasma levels failed to reveal their impaired secretion. CONCLUSIONS Tubular secretion of organic solutes can be reduced out of proportion to glomerular filtration in AKI. Impaired secretion of protein-bound solutes may be more reliably detected when clearances are expressed in terms of their free, unbound levels in the plasma.
Collapse
Affiliation(s)
- Frank J. O’Brien
- Department of Medicine, Washington University, St. Louis, Missouri
| | - Robert D. Mair
- Department of Medicine, Stanford University, Palo Alto, California
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, California
| | - Natalie S. Plummer
- Department of Medicine, Stanford University, Palo Alto, California
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, California
| | - Timothy W. Meyer
- Department of Medicine, Stanford University, Palo Alto, California
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, California
| | - Scott M. Sutherland
- Department of Pediatrics, Lucile Packard Children’s Hospital, Stanford University, Palo Alto, California
| | - Tammy L. Sirich
- Department of Medicine, Stanford University, Palo Alto, California
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, California
| |
Collapse
|
25
|
Effects of Ischemia-Reperfusion on Tubular Cell Membrane Transporters and Consequences in Kidney Transplantation. J Clin Med 2020; 9:jcm9082610. [PMID: 32806541 PMCID: PMC7464608 DOI: 10.3390/jcm9082610] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/31/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
Ischemia-reperfusion (IR)-induced acute kidney injury (IRI) is an inevitable event in kidney transplantation. It is a complex pathophysiological process associated with numerous structural and metabolic changes that have a profound influence on the early and the late function of the transplanted kidney. Proximal tubular cells are particularly sensitive to IRI. These cells are involved in renal and whole-body homeostasis, detoxification processes and drugs elimination by a transporter-dependent, transcellular transport system involving Solute Carriers (SLCs) and ATP Binding Cassettes (ABCs) transporters. Numerous studies conducted mainly in animal models suggested that IRI causes decreased expression and activity of some major tubular transporters. This could favor uremic toxins accumulation and renal metabolic alterations or impact the pharmacokinetic/toxicity of drugs used in transplantation. It is of particular importance to understand the underlying mechanisms and effects of IR on tubular transporters in order to improve the mechanistic understanding of IRI pathophysiology, identify biomarkers of graft function or promote the design and development of novel and effective therapies. Modulation of transporters’ activity could thus be a new therapeutic opportunity to attenuate kidney injury during IR.
Collapse
|
26
|
Bush KT, Singh P, Nigam SK. Gut-derived uremic toxin handling in vivo requires OAT-mediated tubular secretion in chronic kidney disease. JCI Insight 2020; 5:133817. [PMID: 32271169 DOI: 10.1172/jci.insight.133817] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 02/28/2020] [Indexed: 12/12/2022] Open
Abstract
The role of the renal organic anion transporters OAT1 (also known as SLC22A6, originally identified as NKT) and OAT3 (also known as SLC22A8) in chronic kidney disease (CKD) remains poorly understood. This is particularly so from the viewpoint of residual proximal tubular secretion, a key adaptive mechanism to deal with protein-bound uremic toxins in CKD. Using the subtotal nephrectomy (STN) model, plasma metabolites accumulating in STN rats treated with and without the OAT inhibitor, probenecid, were identified. Comparisons with metabolomics data from Oat1-KO and Oat3-KO mice support the centrality of the OATs in residual tubular secretion of uremic solutes, such as indoxyl sulfate, kynurenate, and anthranilate. Overlapping our data with those of published metabolomics data regarding gut microbiome-derived uremic solutes - which can have dual roles in signaling and toxicity - indicates that OATs play a critical role in determining their plasma levels in CKD. Thus, the OATs, along with other SLC and ABC drug transporters, are critical to the movement of uremic solutes across tissues and into various body fluids, consistent with the remote sensing and signaling theory. The data support a role for OATs in modulating remote interorganismal and interorgan communication (gut microbiota-blood-liver-kidney-urine). The results also have implications for understanding drug-metabolite interactions involving uremic toxins.
Collapse
Affiliation(s)
- Kevin T Bush
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Prabhleen Singh
- Division of Nephrology-Hypertension, University of California, San Diego and Veterans Affairs San Diego Healthcare System, San Diego, California, USA.,Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Sanjay K Nigam
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA.,Department of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
27
|
You X, Wu W, Xu J, Jiao Z, Ke M, Huang P, Lin C. Development of a Physiologically Based Pharmacokinetic Model for Prediction of Pramipexole Pharmacokinetics in Parkinson's Disease Patients With Renal Impairment. J Clin Pharmacol 2020; 60:999-1010. [PMID: 32090332 DOI: 10.1002/jcph.1593] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/23/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Xiang You
- Department of Pharmacythe First Affiliated Hospital of Fujian Medical University Taijiang Fuzhou People's Republic of China
| | - Wanhong Wu
- Department of Pharmacythe First Affiliated Hospital of Fujian Medical University Taijiang Fuzhou People's Republic of China
| | - Jing Xu
- Department of Pharmacythe First Affiliated Hospital of Fujian Medical University Taijiang Fuzhou People's Republic of China
| | - Zheng Jiao
- Department of Pharmacy, Shanghai Chest HospitalShanghai Jiao Tong University Shanghai People's Republic of China
| | - Meng Ke
- Department of Pharmacythe First Affiliated Hospital of Fujian Medical University Taijiang Fuzhou People's Republic of China
| | - Pinfang Huang
- Department of Pharmacythe First Affiliated Hospital of Fujian Medical University Taijiang Fuzhou People's Republic of China
| | - Cuihong Lin
- Department of Pharmacythe First Affiliated Hospital of Fujian Medical University Taijiang Fuzhou People's Republic of China
| |
Collapse
|
28
|
Mahringer A, Bernd A, Miller DS, Fricker G. Aryl hydrocarbon receptor ligands increase ABC transporter activity and protein expression in killifish (Fundulus heteroclitus) renal proximal tubules. Biol Chem 2020; 400:1335-1345. [PMID: 30913027 DOI: 10.1515/hsz-2018-0425] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 03/21/2019] [Indexed: 12/15/2022]
Abstract
Many widespread and persistent organic pollutants, for example, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and some polychlorinated biphenyls, activate the aryl hydrocarbon receptor (AhR) causing it to translocate to the cell nucleus where it transactivates target genes, increasing expression of a number of xenobiotic metabolizing enzymes as well as some transporters. AhR's ability to target transporters within the kidney is essentially unexplored. We show here that exposing isolated killifish (Fundulus heteroclitus) renal proximal tubules to micromolar β-naphthoflavone (BNF) or nanomolar TCDD roughly doubled the transport activity of Multidrug resistance-associated proteins Mrp2 and Mrp4, P-glycoprotein (P-gp) and Breast cancer resistance protein (Bcrp), all ATP-driven xenobiotic efflux pumps and critical determinants of renal xenobiotic excretion. These effects were abolished by actinomycin D and cycloheximide and by the AhR antagonist, α-naphthoflavone, indicating that increased transport activity was dependent on transcription and translation as well as ligand binding to AhR. Quantitative immunostaining of renal tubules exposed to BNF and TCDD showed increased luminal membrane expression of Mrp2, Mrp4, P-gp and Bcrp. Thus, in these renal tubules, the four ABC transporters are targets of AhR action.
Collapse
Affiliation(s)
- Anne Mahringer
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, D-69120 Heidelberg, Germany.,Mount Desert Island Biological Laboratory (MDIBL), Salisbury Cove, ME 04672, USA
| | - Alexandra Bernd
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, D-69120 Heidelberg, Germany.,Mount Desert Island Biological Laboratory (MDIBL), Salisbury Cove, ME 04672, USA
| | - David S Miller
- Mount Desert Island Biological Laboratory (MDIBL), Salisbury Cove, ME 04672, USA.,Laboratory of Toxicology and Pharmacology and Chemistry, National Institutes of Health/National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, D-69120 Heidelberg, Germany.,Mount Desert Island Biological Laboratory, Salisbury Cove, ME 04672, USA
| |
Collapse
|
29
|
Nigam AK, Li JG, Lall K, Shi D, Bush KT, Bhatnagar V, Abagyan R, Nigam SK. Unique metabolite preferences of the drug transporters OAT1 and OAT3 analyzed by machine learning. J Biol Chem 2020; 295:1829-1842. [PMID: 31896576 DOI: 10.1074/jbc.ra119.010729] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/30/2019] [Indexed: 01/04/2023] Open
Abstract
The multispecific organic anion transporters, OAT1 (SLC22A6) and OAT3 (SLC22A8), the main kidney elimination pathways for many common drugs, are often considered to have largely-redundant roles. However, whereas examination of metabolomics data from Oat-knockout mice (Oat1 and Oat3KO) revealed considerable overlap, over a hundred metabolites were increased in the plasma of one or the other of these knockout mice. Many of these relatively unique metabolites are components of distinct biochemical and signaling pathways, including those involving amino acids, lipids, bile acids, and uremic toxins. Cheminformatics, together with a "logical" statistical and machine learning-based approach, identified a number of molecular features distinguishing these unique endogenous substrates. Compared with OAT1, OAT3 tends to interact with more complex substrates possessing more rings and chiral centers. An independent "brute force" approach, analyzing all possible combinations of molecular features, supported the logical approach. Together, the results suggest the potential molecular basis by which OAT1 and OAT3 modulate distinct metabolic and signaling pathways in vivo As suggested by the Remote Sensing and Signaling Theory, the analysis provides a potential mechanism by which "multispecific" kidney proximal tubule transporters exert distinct physiological effects. Furthermore, a strong metabolite-based machine-learning classifier was able to successfully predict unique OAT1 versus OAT3 drugs; this suggests the feasibility of drug design based on knockout metabolomics of drug transporters. The approach can be applied to other SLC and ATP-binding cassette drug transporters to define their nonredundant physiological roles and for analyzing the potential impact of drug-metabolite interactions.
Collapse
Affiliation(s)
- Anisha K Nigam
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093-0693
| | - Julia G Li
- Department of Biology, University of California San Diego, La Jolla, California 92093-0693
| | - Kaustubh Lall
- Department of Computer Engineering, University of California San Diego, La Jolla, California 92093-0693
| | - Da Shi
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093-0693
| | - Kevin T Bush
- Department of Pediatrics, University of California San Diego, La Jolla, California 92093-0693
| | - Vibha Bhatnagar
- Department of Family and Preventative Medicine, University of California San Diego, La Jolla, California 92093-0693
| | - Ruben Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093-0693.
| | - Sanjay K Nigam
- Department of Pediatrics, University of California San Diego, La Jolla, California 92093-0693; Department of Medicine, University of California San Diego, La Jolla, California 92093-0693.
| |
Collapse
|
30
|
Risso MA, Sallustio S, Sueiro V, Bertoni V, Gonzalez-Torres H, Musso CG. The Importance of Tubular Function in Chronic Kidney Disease. Int J Nephrol Renovasc Dis 2019; 12:257-262. [PMID: 31849512 PMCID: PMC6913318 DOI: 10.2147/ijnrd.s216673] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 12/02/2019] [Indexed: 12/19/2022] Open
Abstract
Glomerular filtration rate (GFR) and proteinuria-albuminuria are the renal functional parameters currently used to evaluate chronic kidney disease (CKD) severity. However, tubular secretion is another important renal functional parameter to be taken into account since proximal tubule (PT) secretion, in particular, is a crucial renal mechanism for endogenous organic cations, anions and drug elimination. The residual diuresis is a relevant survival predictor in patients on dialysis, since their urine is produced by the glomerular and tubular functions. It has been hypothesized that drugs which up-regulate some renal tubular transporters could contribute to uremic toxin excretion, and nephroprevention. However, if tubular transporters' down-regulation observed in CKD patients and experimental models is a PT adaptation to avoid intracellular accumulation and damage from uremic toxins, consequently the increase of toxin removal by inducing tubular transporters' up-regulation could be deleterious to the kidney. Therefore, a deeper understanding of this phenomenon is currently needed. In conclusion, tubular function has an important role for endogenous organic cations, anions and drug excretion in CKD patients, and a deeper understanding of its multiple mechanisms could provide new therapeutic alternatives in this population.
Collapse
Affiliation(s)
- Maria A Risso
- Human Physiology Department, Instituto Universitario del Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Sofía Sallustio
- Human Physiology Department, Instituto Universitario del Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Valentin Sueiro
- Human Physiology Department, Instituto Universitario del Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Victoria Bertoni
- Human Physiology Department, Instituto Universitario del Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Henry Gonzalez-Torres
- Facultad de Ciencias de la Salud, Universidad Simon Bolivar, Barranquilla, Colombia.,Ciencias Biomédicas, Universidad del Valle, Cali, Colombia
| | - Carlos G Musso
- Human Physiology Department, Instituto Universitario del Hospital Italiano de Buenos Aires, Buenos Aires, Argentina.,Facultad de Ciencias de la Salud, Universidad Simon Bolivar, Barranquilla, Colombia
| |
Collapse
|
31
|
Rosenthal SB, Bush KT, Nigam SK. A Network of SLC and ABC Transporter and DME Genes Involved in Remote Sensing and Signaling in the Gut-Liver-Kidney Axis. Sci Rep 2019; 9:11879. [PMID: 31417100 PMCID: PMC6695406 DOI: 10.1038/s41598-019-47798-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023] Open
Abstract
Genes central to drug absorption, distribution, metabolism and elimination (ADME) also regulate numerous endogenous molecules. The Remote Sensing and Signaling Hypothesis argues that an ADME gene-centered network-including SLC and ABC "drug" transporters, "drug" metabolizing enzymes (DMEs), and regulatory genes-is essential for inter-organ communication via metabolites, signaling molecules, antioxidants, gut microbiome products, uremic solutes, and uremic toxins. By cross-tissue co-expression network analysis, the gut, liver, and kidney (GLK) formed highly connected tissue-specific clusters of SLC transporters, ABC transporters, and DMEs. SLC22, SLC25 and SLC35 families were network hubs, having more inter-organ and intra-organ connections than other families. Analysis of the GLK network revealed key physiological pathways (e.g., involving bile acids and uric acid). A search for additional genes interacting with the network identified HNF4α, HNF1α, and PXR. Knockout gene expression data confirmed ~60-70% of predictions of ADME gene regulation by these transcription factors. Using the GLK network and known ADME genes, we built a tentative gut-liver-kidney "remote sensing and signaling network" consisting of SLC and ABC transporters, as well as DMEs and regulatory proteins. Together with protein-protein interactions to prioritize likely functional connections, this network suggests how multi-specificity combines with oligo-specificity and mono-specificity to regulate homeostasis of numerous endogenous small molecules.
Collapse
Affiliation(s)
- Sara Brin Rosenthal
- Center for Computational Biology and Bioinformatics, University of California at San Diego, La Jolla, CA, 92093-0693, USA
| | - Kevin T Bush
- Departments of Pediatrics, University of California at San Diego, La Jolla, CA, 92093-0693, USA
| | - Sanjay K Nigam
- Departments of Pediatrics, University of California at San Diego, La Jolla, CA, 92093-0693, USA.
- Departments of Medicine, University of California at San Diego, La Jolla, CA, 92093-0693, USA.
| |
Collapse
|
32
|
Remote sensing and signaling in kidney proximal tubules stimulates gut microbiome-derived organic anion secretion. Proc Natl Acad Sci U S A 2019; 116:16105-16110. [PMID: 31341083 PMCID: PMC6689987 DOI: 10.1073/pnas.1821809116] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Membrane transporters and receptors are responsible for balancing nutrient and metabolite levels to aid body homeostasis. Here, we report that proximal tubule cells in kidneys sense elevated endogenous, gut microbiome-derived, metabolite levels through EGF receptors and downstream signaling to induce their secretion by up-regulating the organic anion transporter-1 (OAT1). Remote metabolite sensing and signaling was observed in kidneys from healthy volunteers and rats in vivo, leading to induced OAT1 expression and increased removal of indoxyl sulfate, a prototypical microbiome-derived metabolite and uremic toxin. Using 2D and 3D human proximal tubule cell models, we show that indoxyl sulfate induces OAT1 via AhR and EGFR signaling, controlled by miR-223. Concomitantly produced reactive oxygen species (ROS) control OAT1 activity and are balanced by the glutathione pathway, as confirmed by cellular metabolomic profiling. Collectively, we demonstrate remote metabolite sensing and signaling as an effective OAT1 regulation mechanism to maintain plasma metabolite levels by controlling their secretion.
Collapse
|
33
|
Nigam SK. The SLC22 Transporter Family: A Paradigm for the Impact of Drug Transporters on Metabolic Pathways, Signaling, and Disease. Annu Rev Pharmacol Toxicol 2019; 58:663-687. [PMID: 29309257 DOI: 10.1146/annurev-pharmtox-010617-052713] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The SLC22 transporter family consists of more than two dozen members, which are expressed in the kidney, the liver, and other tissues. Evolutionary analysis indicates that SLC22 transporters fall into at least six subfamilies: OAT (organic anion transporter), OAT-like, OAT-related, OCT (organic cation transporter), OCTN (organic cation/carnitine transporter), and OCT/OCTN-related. Some-including OAT1 [SLC22A6 or NKT (novel kidney transporter)] and OAT3 (SLC22A8), as well as OCT1 (SLC22A1) and OCT2 (SLC22A2)-are widely studied drug transporters. Nevertheless, analyses of knockout mice and other data indicate that SLC22 transporters regulate key metabolic pathways and levels of signaling molecules (e.g., gut microbiome products, bile acids, tricarboxylic acid cycle intermediates, dietary flavonoids and other nutrients, prostaglandins, vitamins, short-chain fatty acids, urate, and ergothioneine), as well as uremic toxins associated with chronic kidney disease. Certain SLC22 transporters-such as URAT1 (SLC22A12) and OCTN2 (SLC22A5)-are mutated in inherited metabolic diseases. A new systems biology view of transporters is emerging. As proposed in the remote sensing and signaling hypothesis, SLC22 transporters, together with other SLC and ABC transporters, have key roles in interorgan and interorganism small-molecule communication and, together with the neuroendocrine, growth factor-cytokine, and other homeostatic systems, regulate local and whole-body homeostasis.
Collapse
Affiliation(s)
- Sanjay K Nigam
- Departments of Pediatrics and Medicine, University of California, San Diego, La Jolla, California 92093, USA;
| |
Collapse
|
34
|
Reduced Hypoxia-Related Genes in Porcine Limbs in Ex Vivo Hypothermic Perfusion Versus Cold Storage. J Surg Res 2018; 232:137-145. [PMID: 30463709 DOI: 10.1016/j.jss.2018.05.067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 04/15/2018] [Accepted: 05/30/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND Ischemia-reperfusion injury remains the major limiting factor for limb replantation and transplantation. Static cold storage (SCS) on ice currently represents the standard mode of preservation but is limited to 6 h of duration. Ex vivo machine perfusion has evolved as a potential alternative to safely extend the duration of ex vivo preservation by providing continuous supply of oxygen and nutrients. This study aims to evaluate underlying molecular mechanisms of both preservation modalities. METHODS We assessed molecular changes in amputated porcine forelimbs stored on ice at 4°C for 2 h (n = 2) and limbs perfused with Perfadex solution at 10°C for 2 h (n = 3) or 12 h (n = 3) before replantation. Muscle biopsies were examined for histological changes and gene expression levels using H&E staining and a hypoxia-related PCR gene array, respectively. RESULTS Histology revealed only minor differences between the ice (SCS) and perfusion groups after 2 h of preservation, with decreased muscle fiber disruption in the perfusion groups compared with the ice (SCS) group. Perfused limbs demonstrated downregulation of genes coding for glycolytic pathways and glucose transporters after 2 h and 12 h when compared with SCS after 2 h. Similarly, genes that induce angiogenesis and those that are activated on DNA damage were downregulated in both perfusion groups as compared with SCS. CONCLUSIONS Perfusion of porcine limbs resulted in less activation of hypoxia-related gene families when compared with SCS. This may indicate a state more closely resembling physiological conditions during perfusion and potentially limiting ischemic injury. Our study confirms ex vivo perfusion for up to 12 h as a viable alternative for preservation of vascularized composite tissues.
Collapse
|
35
|
Jaikumkao K, Pongchaidecha A, Chueakula N, Thongnak L, Wanchai K, Chatsudthipong V, Chattipakorn N, Lungkaphin A. Renal outcomes with sodium glucose cotransporter 2 (SGLT2) inhibitor, dapagliflozin, in obese insulin-resistant model. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2021-2033. [PMID: 29572114 DOI: 10.1016/j.bbadis.2018.03.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 03/05/2018] [Accepted: 03/19/2018] [Indexed: 02/08/2023]
Abstract
A growing body of evidence indicates that obesity and insulin resistance contribute to the progression of renal disease. This study was performed to determine the effects of dapagliflozin, a novel sodium glucose cotransporter 2 (SGLT2) inhibitor, on renal and renal organic anion transporter 3 (Oat3) functions in high-fat diet fed rats, a model of obese insulin-resistance. Twenty-four male Wistar rats were divided into two groups, and received either a normal diet (ND) (n = 6) or a high-fat diet (HFD) (n = 18) for 16 weeks. At week 17, the HFD-fed rats were subdivided into three subgroups (n = 6/subgroup) and received either a vehicle (HFD), dapagliflozin (HFDAP; 1.0 mg/kg/day) or metformin (HFMET; 30 mg/kg/day), by oral gavage for four weeks. Metabolic parameters, renal function, renal Oat3 function, renal oxidative stress, and renal morphology were determined. The results showed that obese insulin-resistant rats induced by HFD feeding had impaired renal function and renal Oat3 function together with increased renal oxidative injury. Dapagliflozin or metformin treatment decreased insulin resistance, hypercholesterolemia, creatinine clearance and renal oxidative stress leading to improved renal function. However, dapagliflozin treatment decreased blood pressure, serum creatinine, urinary microalbumin and increased glucose excretions, and showed a greater ability to ameliorate impaired renal insulin signaling and glomerular barrier damage than metformin. These data suggest that dapagliflozin had greater efficacy than metformin for attenuating renal dysfunction and improving renal Oat3 function, at least in part by reducing renal oxidative stress and modulating renal insulin signaling pathways, and hence ameliorating renal injury.
Collapse
Affiliation(s)
- Krit Jaikumkao
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Anchalee Pongchaidecha
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nuttawud Chueakula
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Laongdao Thongnak
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Keerati Wanchai
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; School of Medicine, Mae Fah Luang University, Chiang Rai, Thailand
| | | | - Nipon Chattipakorn
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Anusorn Lungkaphin
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center for Research and Development of Natural Products for Health, Chiang Mai University, Thailand.
| |
Collapse
|
36
|
Barin-Le Guellec C, Largeau B, Bon D, Marquet P, Hauet T. Ischemia/reperfusion-associated tubular cells injury in renal transplantation: Can metabolomics inform about mechanisms and help identify new therapeutic targets? Pharmacol Res 2018; 129:34-43. [PMID: 29309901 DOI: 10.1016/j.phrs.2017.12.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/29/2017] [Accepted: 12/29/2017] [Indexed: 12/31/2022]
Abstract
Tubular cells are central targets of ischemia-reperfusion (I/R) injury in kidney transplantation. Inflammation and metabolic disturbances occurring within these cells are deleterious by themselves but also favor secondary events, such as activation of immune response. It is critical to have an in depth understanding of the mechanisms governing tubular cells response to I/R if one wants to define pertinent biomarkers or to elaborate targeted therapeutic interventions. As oxidative damage was shown to be central in the patho-physiological mechanisms, the impact of I/R on proximal tubular cells metabolism has been widely studied, contrary to its effects on expression and activity of membrane transporters of the proximal tubular cells. Yet, temporal modulation of transporters over ischemia and reperfusion periods appears to play a central role, not only in the induction of cells injury but also in graft function recovery. Metabolomics in cell models or diverse biofluids has the potential to provide large pictures of biochemical consequences of I/R. Metabolomic studies conducted in experimental models of I/R or in transplanted patients indeed retrieved metabolites belonging to the pathways known to be particularly affected. Interestingly, they also revealed that metabolic disturbances and transporters activities are in very close mutual interplay. As well as helping to select diagnostic biomarkers, such analyses could also contribute to identify new pharmacological targets and to set up innovative nephroprotective strategies for the future. Even if various therapeutic approaches have been evaluated for a long time to prevent or treat I/R injuries, metabolomics has helped identifying new ones, those related to membrane transporters seeming to be of particular interest. However, considering the very complex and multifactorial effects of I/R in the context of kidney transplantation, all tracks must be followed if one wants to prevent or limit its deleterious consequences.
Collapse
Affiliation(s)
- Chantal Barin-Le Guellec
- INSERM UMR 1248, IPPRITT, Limoges, France; CHU Tours, Laboratory of Biochemistry and Molecular Biology, Tours, France; FHU SUPORT, Limoges, Poitiers, Tours, France.
| | - Bérenger Largeau
- CHU Tours, Laboratory of Biochemistry and Molecular Biology, Tours, France
| | - Delphine Bon
- FHU SUPORT, Limoges, Poitiers, Tours, France; University of Poitiers, Poitiers, France; INSERM UMR 1082, IRTOMIT, Poitiers, France; CHU Poitiers, Laboratory of Biochemistry, Poitiers, France
| | - Pierre Marquet
- INSERM UMR 1248, IPPRITT, Limoges, France; FHU SUPORT, Limoges, Poitiers, Tours, France; University of Limoges, Faculty of Medicine, Limoges, France; CHU Limoges, Department of Pharmacology, Toxicology & Pharmacovigilance, Limoges, France
| | - Thierry Hauet
- FHU SUPORT, Limoges, Poitiers, Tours, France; University of Poitiers, Poitiers, France; INSERM UMR 1082, IRTOMIT, Poitiers, France; CHU Poitiers, Laboratory of Biochemistry, Poitiers, France
| |
Collapse
|
37
|
Zhang R, Zhan S, Li S, Zhu Z, He J, Lorenzo JM, Barba FJ. Anti-hyperuricemic and nephroprotective effects of extracts from Chaenomeles sinensis (Thouin) Koehne in hyperuricemic mice. Food Funct 2018; 9:5778-5790. [DOI: 10.1039/c8fo01480a] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
CSF-E possesses potent anti-hyperuricemic through inhibiting XOD activity.
Collapse
Affiliation(s)
- Rui Zhang
- School of Food Science and Engineering
- Wuhan Polytechnic University
- Wuhan
- China
| | - Shaoying Zhan
- School of Food Science and Engineering
- Wuhan Polytechnic University
- Wuhan
- China
| | - Shuyi Li
- School of Food Science and Engineering
- Wuhan Polytechnic University
- Wuhan
- China
| | - Zhenzhou Zhu
- School of Food Science and Engineering
- Wuhan Polytechnic University
- Wuhan
- China
| | - Jingren He
- School of Food Science and Engineering
- Wuhan Polytechnic University
- Wuhan
- China
| | - Jose M. Lorenzo
- Centro Tecnológico de la Carne de Galicia
- rúa Galicia n° 4
- Parque Tecnológico de Galicia
- San Cibrao das Viñas
- Spain
| | - Francisco J. Barba
- Nutrition and Food Science Area
- Preventive Medicine and Public Health
- Food Sciences
- Toxicology and Forensic Medicine Department
- Faculty of Pharmacy
| |
Collapse
|
38
|
Bush KT, Wu W, Lun C, Nigam SK. The drug transporter OAT3 (SLC22A8) and endogenous metabolite communication via the gut-liver-kidney axis. J Biol Chem 2017; 292:15789-15803. [PMID: 28765282 DOI: 10.1074/jbc.m117.796516] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/27/2017] [Indexed: 12/12/2022] Open
Abstract
The organic anion transporters OAT1 (SLC22A6) and OAT3 (SLC22A8) have similar substrate specificity for drugs, but it is far from clear whether this holds for endogenous substrates. By analysis of more than 600 metabolites in the Oat3KO (Oat3 knockout) by LC/MS, we demonstrate OAT3 involvement in the movement of gut microbiome products, key metabolites, and signaling molecules, including those flowing through the gut-liver-kidney axis. Major pathways affected included those involved in metabolism of bile acids, flavonoids, nutrients, amino acids (including tryptophan-derivatives that are uremic toxins), and lipids. OAT3 is also critical in elimination of liver-derived phase II metabolites, particularly those undergoing glucuronidation. Analysis of physicochemical features revealed nine distinct metabolite groups; at least one member of most clusters has been previously validated in transport assays. In contrast to drugs interacting with the OATs, endogenous metabolites accumulating in the Oat1KO (Oat1 knockout) versus Oat3KO have distinct differences in their physicochemical properties; they are very different in size, number of rings, hydrophobicity, and molecular complexity. Consistent with the Remote Sensing and Signaling Hypothesis, the data support the importance of the OAT transporters in inter-organ and inter-organismal remote communication via transporter-mediated movement of key metabolites and signaling molecules (e.g. gut microbiome-to-intestine-to-blood-to-liver-to-kidney-to-urine). We discuss the possibility of an intimate connection between OATs and metabolite sensing and signaling pathways (e.g. bile acids). Furthermore, the metabolomics and pathway analysis support the view that OAT1 plays a greater role in kidney proximal tubule metabolism and OAT3 appears relatively more important in systemic metabolism, modulating levels of metabolites flowing through intestine, liver, and kidney.
Collapse
Affiliation(s)
| | | | - Christina Lun
- Biology, University of California San Diego, La Jolla, California 92093
| | | |
Collapse
|
39
|
Wu W, Bush KT, Nigam SK. Key Role for the Organic Anion Transporters, OAT1 and OAT3, in the in vivo Handling of Uremic Toxins and Solutes. Sci Rep 2017; 7:4939. [PMID: 28694431 PMCID: PMC5504054 DOI: 10.1038/s41598-017-04949-2] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/30/2017] [Indexed: 01/25/2023] Open
Abstract
In vitro data indicates that the kidney proximal tubule (PT) transporters of uremic toxins and solutes (e.g., indoxyl sulfate, p-cresol sulfate, kynurenine, creatinine, urate) include two “drug” transporters of the organic anion transporter (OAT) family: OAT1 (SLC22A6, originally NKT) and OAT3 (SLC22A8). Here, we have examined new and prior metabolomics data from the Oat1KO and Oat3KO, as well as newly obtained metabolomics data from a “chemical double” knockout (Oat3KO plus probenecid). This gives a picture of the in vivo roles of OAT1 and OAT3 in the regulation of the uremic solutes and supports the centrality of these “drug” transporters in independently and synergistically regulating uremic metabolism. We demonstrate a key in vivo role for OAT1 and/or OAT3 in the handling of over 35 uremic toxins and solutes, including those derived from the gut microbiome (e.g., CMPF, phenylsulfate, indole-3-acetic acid). Although it is not clear whether trimethylamine-N-oxide (TMAO) is directly transported, the Oat3KO had elevated plasma levels of TMAO, which is associated with cardiovascular morbidity in chronic kidney disease (CKD). As described in the Remote Sensing and Signaling (RSS) Hypothesis, many of these molecules are involved in interorgan and interorganismal communication, suggesting that uremia is, at least in part, a disorder of RSS.
Collapse
Affiliation(s)
- Wei Wu
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Kevin T Bush
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Sanjay K Nigam
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA. .,Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA. .,Departments of Medicine, Pediatrics, and Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
40
|
Molecular properties associated with transporter-mediated drug disposition. Adv Drug Deliv Rev 2017; 116:92-99. [PMID: 28554577 DOI: 10.1016/j.addr.2017.05.014] [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: 12/19/2016] [Revised: 03/20/2017] [Accepted: 05/25/2017] [Indexed: 12/18/2022]
Abstract
Membrane transporters play a key role in the absorption, distribution, clearance, elimination, and transport of drugs. Understanding the drug properties and structure activity relationships (SAR) for affinity to membrane transporters is critical to optimize clearance and pharmacokinetics during drug design. To facilitate the early identification of clearance mechanism, a framework named the extended clearance classification system (ECCS) was recently introduced. Using in vitro and physicochemical properties that are readily available in early drug discovery, ECCS has been successfully applied to identify major clearance mechanism and to implicate the role of membrane transporters in determining pharmacokinetics. While the crystal structures for most of the drug transporters are currently not available, ligand-based modeling approaches that use information obtained from the structure and molecular properties of the ligands have been applied to associate the drug-related properties and transporter-mediated disposition. The approach allows prospective prediction of transporter both substrate and/or inhibitor affinity and build quantitative structure-activity relationship (QSAR) to enable early optimization of pharmacokinetics, tissue distribution and drug-drug interaction risk. Drug design applications can be further improved through uncovering transporter protein crystal structure and generation of quality data to refine and develop viable predictive models.
Collapse
|
41
|
Favretto G, Souza LM, Gregório PC, Cunha RS, Maciel RAP, Sassaki GL, Toledo MG, Pecoits-Filho R, Souza WM, Stinghen AEM. Role of Organic Anion Transporters in the Uptake of Protein-Bound Uremic Toxins by Human Endothelial Cells and Monocyte Chemoattractant Protein-1 Expression. J Vasc Res 2017; 54:170-179. [PMID: 28472795 DOI: 10.1159/000468542] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 03/05/2017] [Indexed: 12/12/2022] Open
Abstract
Organic anion transporters (OATs) are involved in the uptake of uremic toxins such as p-cresyl sulfate (PCS) and indoxyl sulfate (IS), which play a role in endothelial dysfunction in patients with chronic kidney diseases (CKD). In this study, we investigated the role of OAT1 and OAT3 in the uptake of PCS and IS into human endothelial cells. PCS was synthesized via p-cresol sulfation and characterized using analytical methods. The cells were treated with PCS and IS in the absence and presence of probenecid (Pb), an OAT inhibitor. Cell viability was assessed using the MTT assay. The absorbed toxins were analyzed using chromatography, OAT expression using immunocytochemistry and western blot, and monocyte chemoattractant protein-1 (MCP-1) expression using enzyme-linked immunosorbent assay. Cell viability decreased after toxin treatment in a dose-dependent manner. PCS and IS showed significant internalization after 60 min treatment, while no internalization was observed in the presence of Pb, suggesting that OATs are involved in the transport of both toxins. Immunocytochemistry and western blot demonstrated OAT1 and OAT3 expression in endothelial cells. MCP-1 expression increased after toxins treatment but decreased after Pb treatment. PCS and IS uptake were mediated by OATs, and OAT blockage could serve as a therapeutic strategy to inhibit MCP-1 expression.
Collapse
Affiliation(s)
- Giane Favretto
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
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: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 10/18/2016] [Indexed: 11/06/2022] Open
|
43
|
Liu HC, Jamshidi N, Chen Y, Eraly SA, Cho SY, Bhatnagar V, Wu W, Bush KT, Abagyan R, Palsson BO, Nigam SK. An Organic Anion Transporter 1 (OAT1)-centered Metabolic Network. J Biol Chem 2016; 291:19474-86. [PMID: 27440044 DOI: 10.1074/jbc.m116.745216] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Indexed: 01/06/2023] Open
Abstract
There has been a recent interest in the broader physiological importance of multispecific "drug" transporters of the SLC and ABC transporter families. Here, a novel multi-tiered systems biology approach was used to predict metabolites and signaling molecules potentially affected by the in vivo deletion of organic anion transporter 1 (Oat1, Slc22a6, originally NKT), a major kidney-expressed drug transporter. Validation of some predictions in wet-lab assays, together with re-evaluation of existing transport and knock-out metabolomics data, generated an experimentally validated, confidence ranked set of OAT1-interacting endogenous compounds enabling construction of an "OAT1-centered metabolic interaction network." Pathway and enrichment analysis indicated an important role for OAT1 in metabolism involving: the TCA cycle, tryptophan and other amino acids, fatty acids, prostaglandins, cyclic nucleotides, odorants, polyamines, and vitamins. The partly validated reconstructed network is also consistent with a major role for OAT1 in modulating metabolic and signaling pathways involving uric acid, gut microbiome products, and so-called uremic toxins accumulating in chronic kidney disease. Together, the findings are compatible with the hypothesized role of drug transporters in remote inter-organ and inter-organismal communication: The Remote Sensing and Signaling Hypothesis (Nigam, S. K. (2015) Nat. Rev. Drug Disc. 14, 29). The fact that OAT1 can affect many systemic biological pathways suggests that drug-metabolite interactions need to be considered beyond simple competition for the drug transporter itself and may explain aspects of drug-induced metabolic syndrome. Our approach should provide novel mechanistic insights into the role of OAT1 and other drug transporters implicated in metabolic diseases like gout, diabetes, and chronic kidney disease.
Collapse
Affiliation(s)
| | | | - Yuchen Chen
- Bioinformatics and Systems Biology Graduate Program
| | | | | | | | | | | | - Ruben Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093
| | | | - Sanjay K Nigam
- Medicine, Pediatrics, and Cellular and Molecular Medicine,
| |
Collapse
|
44
|
Jaikumkao K, Pongchaidecha A, Chattipakorn N, Chatsudthipong V, Promsan S, Arjinajarn P, Lungkaphin A. Atorvastatin improves renal organic anion transporter 3 and renal function in gentamicin-induced nephrotoxicity in rats. Exp Physiol 2016; 101:743-53. [DOI: 10.1113/ep085571] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 03/18/2016] [Indexed: 12/26/2022]
Affiliation(s)
- Krit Jaikumkao
- Department of Physiology, Faculty of Medicine; Chiang Mai University; Chiang Mai Thailand
| | - Anchalee Pongchaidecha
- Department of Physiology, Faculty of Medicine; Chiang Mai University; Chiang Mai Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Department of Physiology, Faculty of Medicine; Chiang Mai University; Chiang Mai Thailand
| | | | - Sasivimon Promsan
- Department of Physiology, Faculty of Medical Sciences; The University of Phayao; Phayao Thailand
| | - Phatchawan Arjinajarn
- Department of Biology, Faculty of Science; Chiang Mai University; Chiang Mai Thailand
| | - Anusorn Lungkaphin
- Department of Physiology, Faculty of Medicine; Chiang Mai University; Chiang Mai Thailand
| |
Collapse
|
45
|
He X, Garza D, Nigam SK, Chang G. Multispecific Organic Cation Transporter 1 (OCT1) from Bos taurus Has High Affinity and Slow Binding Kinetics towards Prostaglandin E2. PLoS One 2016; 11:e0152969. [PMID: 27046168 PMCID: PMC4821612 DOI: 10.1371/journal.pone.0152969] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 03/22/2016] [Indexed: 11/18/2022] Open
Abstract
Organic cation transporter 1 (OCT1, SLC22A1), like many solute carrier 22 (SLC22) family members, is important for the disposition of clinically important drugs, metabolites and signaling molecules. Several studies suggest that SLC22 family (eg. organic anion transporters or OATs and OCTs) bind and possibly transport prostaglandins with relatively high affinity (submicromolar). The affinities of OCT1 and OATs toward PGE2 and PGF2a reported in these cell-based transport studies are considerably greater than for xenobiotics and natural metabolite substrates--in many cases over 100-fold higher. This raises the possibility that prostaglandins are key endogenous substrates and/or that they act on the transporter in a manner different from other substrates such as xenobiotics and lower affinity metabolites. To further investigate OCT1-prostaglandin interactions, we designed biophysical studies using purified bovine OCT1 (Bos taurus, btOCT1/SLC22A1) with PGE2 analogs, in fluorescently labeled and label-free formats. Using fluorescence polarization (FP), we detected a binding of btOCT1 to the PGE2-Rhodamine conjugate at submicromolar affinity, consistent with affinity data for PGE2 from cells over-expressing the related human OCT1. Using purified native btOCT1 as analyte and biotinylated PGE2 analog as ligand, our data from surface plasmon resonance (SPR) revealed that btOCT1 specifically interacts to PGE2 with KD values in the hundred nanomolar range. BtOCT1 also demonstrated a slow association (ka) in the range of 103 M(-1) s(-1) and an even slower dissociation rate (kd) in the range of 10-4 s(-1) for PGE2, suggesting the possibility of a different mode of binding compared to other structurally unrelated transported substrates of low-affinity (eg. drugs, metabolites). Our results complement in vitro transport studies and provide direct evidence that OCT1--which is normally expressed in liver and other tissues--interacts with prostaglandin analogs. While it is not entirely clear from the published literature whether OCTs function as major prostaglandin transporters, the tight binding of the naturally occurring PGE2, as well as the slow dissociation rate, could conceivably affect the transport of lower affinity substrates such as drugs and metabolites by SLC22 transporters. More research is necessary to establish the extent to which individual SLC22 family members actually function as PG transporters in vitro and in vivo and to investigate whether PGs can, independent of being directly transported, alter the ability of SLC22 transporters to handle drugs and other substrates.
Collapse
Affiliation(s)
- Xiao He
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Denisse Garza
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Sanjay K. Nigam
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, United States of America
- Department of Medicine and Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, United States of America
| | - Geoffrey Chang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, United States of America
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92093, United States of America
- * E-mail:
| |
Collapse
|
46
|
Nigam SK, Wu W, Bush KT, Hoenig MP, Blantz RC, Bhatnagar V. Handling of Drugs, Metabolites, and Uremic Toxins by Kidney Proximal Tubule Drug Transporters. Clin J Am Soc Nephrol 2015; 10:2039-49. [PMID: 26490509 DOI: 10.2215/cjn.02440314] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 09/28/2014] [Indexed: 01/22/2023]
Abstract
The proximal tubule of the kidney plays a crucial role in the renal handling of drugs (e.g., diuretics), uremic toxins (e.g., indoxyl sulfate), environmental toxins (e.g., mercury, aristolochic acid), metabolites (e.g., uric acid), dietary compounds, and signaling molecules. This process is dependent on many multispecific transporters of the solute carrier (SLC) superfamily, including organic anion transporter (OAT) and organic cation transporter (OCT) subfamilies, and the ATP-binding cassette (ABC) superfamily. We review the basic physiology of these SLC and ABC transporters, many of which are often called drug transporters. With an emphasis on OAT1 (SLC22A6), the closely related OAT3 (SLC22A8), and OCT2 (SLC22A2), we explore the implications of recent in vitro, in vivo, and clinical data pertinent to the kidney. The analysis of murine knockouts has revealed a key role for these transporters in the renal handling not only of drugs and toxins but also of gut microbiome products, as well as liver-derived phase 1 and phase 2 metabolites, including putative uremic toxins (among other molecules of metabolic and clinical importance). Functional activity of these transporters (and polymorphisms affecting it) plays a key role in drug handling and nephrotoxicity. These transporters may also play a role in remote sensing and signaling, as part of a versatile small molecule communication network operative throughout the body in normal and diseased states, such as AKI and CKD.
Collapse
Affiliation(s)
- Sanjay K Nigam
- Department of Medicine, Department of Pediatrics, Department of Cell & Molecular Medicine,
| | | | | | - Melanie P Hoenig
- Division of Nephrology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Roland C Blantz
- Division of Nephrology-Hypertension, and Veterans Affairs San Diego Healthcare System, San Diego, California; and
| | - Vibha Bhatnagar
- Division of Family & Preventative Medicine, University of California-San Diego, La Jolla, California
| |
Collapse
|
47
|
Singh V, Singla SK, Jha V, Puri V, Puri S. Hepatocyte nuclear factor-1β: A regulator of kidney development and cystogenesis. Indian J Nephrol 2015; 25:70-6. [PMID: 25838642 PMCID: PMC4379628 DOI: 10.4103/0971-4065.139492] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The understanding of the genomics of the renal tissue has gathered a considerable interest and is making rapid progress. The molecular mechanisms as well as the precise function of the associated molecular components toward renal pathophysiology have recently been realized. For the cystic kidney disease, the regulation of gene expression affecting epithelial cells proliferation, apoptosis as well as process of differentiation/de-differentiation represent key molecular targets. For the cystic disorders, molecular targets have been identified, which besides lending heterogeneity to cysts may also provide tools to unravel their functional importance to understand the renal tissue homeostasis. This review focuses on providing comprehensive information about the transcriptional regulatory role of hepatocyte nuclear factor-1β, a homeoprotein, as well as its interacting partners in renal tissue development and pathophysiology.
Collapse
Affiliation(s)
- V Singh
- Department of Biochemistry, Panjab University, Chandigarh, India
| | - S K Singla
- Department of Biochemistry, Panjab University, Chandigarh, India
| | - V Jha
- Department of Nephrology, PGIMER, Chandigarh, India
| | - V Puri
- Centre for Systems Biology and Bioinformatics, Under University Institute of Emerging Areas in Science and Technology, Panjab University, Chandigarh, India
| | - S Puri
- Biotechnology Branch, University Institute of Engineering and Technology, Chandigarh, India ; Centre for Stem Cell and Issue Engineering, University Institute of Emerging Areas in Science and Technology, Panjab University, Chandigarh, India
| |
Collapse
|
48
|
Nigam SK, Bush KT, Martovetsky G, Ahn SY, Liu HC, Richard E, Bhatnagar V, Wu W. The organic anion transporter (OAT) family: a systems biology perspective. Physiol Rev 2015; 95:83-123. [PMID: 25540139 PMCID: PMC4281586 DOI: 10.1152/physrev.00025.2013] [Citation(s) in RCA: 315] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The organic anion transporter (OAT) subfamily, which constitutes roughly half of the SLC22 (solute carrier 22) transporter family, has received a great deal of attention because of its role in handling of common drugs (antibiotics, antivirals, diuretics, nonsteroidal anti-inflammatory drugs), toxins (mercury, aristolochic acid), and nutrients (vitamins, flavonoids). Oats are expressed in many tissues, including kidney, liver, choroid plexus, olfactory mucosa, brain, retina, and placenta. Recent metabolomics and microarray data from Oat1 [Slc22a6, originally identified as NKT (novel kidney transporter)] and Oat3 (Slc22a8) knockouts, as well as systems biology studies, indicate that this pathway plays a central role in the metabolism and handling of gut microbiome metabolites as well as putative uremic toxins of kidney disease. Nuclear receptors and other transcription factors, such as Hnf4α and Hnf1α, appear to regulate the expression of certain Oats in conjunction with phase I and phase II drug metabolizing enzymes. Some Oats have a strong selectivity for particular signaling molecules, including cyclic nucleotides, conjugated sex steroids, odorants, uric acid, and prostaglandins and/or their metabolites. According to the "Remote Sensing and Signaling Hypothesis," which is elaborated in detail here, Oats may function in remote interorgan communication by regulating levels of signaling molecules and key metabolites in tissues and body fluids. Oats may also play a major role in interorganismal communication (via movement of small molecules across the intestine, placental barrier, into breast milk, and volatile odorants into the urine). The role of various Oat isoforms in systems physiology appears quite complex, and their ramifications are discussed in the context of remote sensing and signaling.
Collapse
Affiliation(s)
- Sanjay K Nigam
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Kevin T Bush
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Gleb Martovetsky
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Sun-Young Ahn
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Henry C Liu
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Erin Richard
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Vibha Bhatnagar
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Wei Wu
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
49
|
Abstract
Potential drug-drug interactions mediated by the ATP-binding cassette (ABC) transporter and solute carrier (SLC) transporter families are of clinical and regulatory concern. However, the endogenous functions of these drug transporters are not well understood. Discussed here is evidence for the roles of ABC and SLC transporters in the handling of diverse substrates, including metabolites, antioxidants, signalling molecules, hormones, nutrients and neurotransmitters. It is suggested that these transporters may be part of a larger system of remote communication ('remote sensing and signalling') between cells, organs, body fluid compartments and perhaps even separate organisms. This broader view may help to clarify disease mechanisms, drug-metabolite interactions and drug effects relevant to diabetes, chronic kidney disease, metabolic syndrome, hypertension, gout, liver disease, neuropsychiatric disorders, inflammatory syndromes and organ injury, as well as prenatal and postnatal development.
Collapse
Affiliation(s)
- Sanjay K Nigam
- Departments of Pediatrics, Medicine, and Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0693, USA
| |
Collapse
|
50
|
Saito H, Yoshimura M, Saigo C, Komori M, Nomura Y, Yamamoto Y, Sagata M, Wakida A, Chuman E, Nishi K, Jono H. Hepatic sulfotransferase as a nephropreventing target by suppression of the uremic toxin indoxyl sulfate accumulation in ischemic acute kidney injury. Toxicol Sci 2014; 141:206-17. [PMID: 24958931 DOI: 10.1093/toxsci/kfu119] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Ischemia/reperfusion (I/R)-induced acute kidney injury (AKI) is evoked by diverse pathophysiological conditions and/or surgical procedures. Here, we evaluated the nephropreventive effect of sulfotransferase (SULT) inhibitors, quercetin, and resveratrol, which hamper hepatic indoxyl sulfate (IS) production. I/R of the kidney caused severe renal injury with marked accumulation of serum and renal IS and urinary excretion of kidney injury molecule-1. Oral administration of AST-120 resulted in a significant restoration of kidney injury, suggesting that uremic toxins, which can be suppressed or adsorbed by AST-120 in the intestine, contribute to the progression or development of I/R-induced AKI. Oral administration of resveratrol or quercetin, SULT inhibitors, suppressed IS accumulation, accompanied by significant amelioration of renal dysfunction. The expression of nuclear factor E2-related factor 2 (Nrf2) in the renal nuclear fractions was markedly elevated by renal I/R, but suppressed by treatment with SULT inhibitors. IS is primarily taken up by HK-2 cells derived from human proximal tubular cells via organic anion transporters, which then evokes activation of Nrf2, most likely due to intracellular oxidative stress. Renal basolateral organic anion transporters OAT1 and OAT3, which mediate renal tubular uptake of IS in basolateral membrane, were markedly downregulated by renal I/R, but restored by SULT inhibitors. Our results suggest that renal accumulation of IS in ischemic AKI induces oxidative stress and downregulation of organic anion transporters resulting in kidney damage, which could be restored to some extent by inhibiting hepatic SULT activity as a nephropreventive target.
Collapse
Affiliation(s)
- Hideyuki Saito
- Department of Pharmacy, Kumamoto University Hospital, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Misato Yoshimura
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Chika Saigo
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Megumi Komori
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Yui Nomura
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Yuko Yamamoto
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Masataka Sagata
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Ayaka Wakida
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Erina Chuman
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Kazuhiko Nishi
- Department of Hemo-Dialysis, Kumamoto University Hospital, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan
| | - Hirofumi Jono
- Department of Pharmacy, Kumamoto University Hospital, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556, Japan Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| |
Collapse
|