1
|
Rajpoot R, Rajput S, Koiri RK. Microcystin-LR and its health impacts: Chemistry, transmission routes, mechanisms of toxicity and target organs. Toxicol Rep 2025; 14:101996. [PMID: 40177604 PMCID: PMC11964656 DOI: 10.1016/j.toxrep.2025.101996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/02/2025] [Accepted: 03/09/2025] [Indexed: 04/05/2025] Open
Abstract
Microcystin-LR, a hepatotoxin produced by cyanobacteria, poses significant health risks to humans and other animals through various routes of exposure. This review comprehensively explores the chemistry, transmission pathways, mechanisms of toxicity, and target organs affected by MC-LR to provide a detailed understanding of its health impacts on animals and humans. MC-LR exposure occurs through different transmission routes, including ingesting contaminated water and food, algal dietary supplements, direct body contact with harmful algal blooms, and inhalation of aerosolized toxins. In this review, we explored that the toxic effects of MC-LR are mediated through multiple complex mechanisms. A key mechanism of its toxicity is the inhibition of protein phosphatases PP1 and PP2A which results in abnormal cellular signalling pathways. Additionally, MC-LR induces oxidative stress and disrupts cellular homeostasis. The findings suggest that MC-LR modulates the activity of various antioxidant enzymes and also activates apoptosis pathways by different mechanisms. It also induces cytoskeletal disruption, ultimately compromising cellular integrity and function. MC-LR also induces activation of oncogenes such as Gankyrin, PI3K/AKT, HIF-1α, RAC1/JNK and NEK2 pathway and upregulates the inflammatory molecules such as NF-κβ, and TNF-α, hence leading to carcinogenesis. MC-LR has toxicological effects on multiple organs. The liver is the primary target, where MC-LR accumulates and causes hepatotoxicity, but other organs are affected as well. MC-LR shows neurotoxicity, nephrotoxicity, cardiotoxicity and reproductive toxicity.
Collapse
Affiliation(s)
- Roshni Rajpoot
- Biochemistry Laboratory, Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh 470003, India
| | - Siddharth Rajput
- Biochemistry Laboratory, Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh 470003, India
| | - Raj Kumar Koiri
- Biochemistry Laboratory, Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh 470003, India
| |
Collapse
|
2
|
Cheng J, Zhu J, Chen R, Zhang M, Han B, Zhu M, He Y, Yi H, Tang S. Genetic polymorphisms and anti-tuberculosis drug-induced liver injury: an umbrella review of the evidence. Int J Clin Pharm 2025; 47:624-639. [PMID: 39954223 DOI: 10.1007/s11096-025-01880-9] [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: 10/03/2024] [Accepted: 01/28/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Anti-tuberculosis drug-induced liver injury (ATLI) is a significant adverse drug reaction with genetic susceptibility implications. AIM This study aimed to integrate findings from systematic reviews and meta-analyses on genetic polymorphisms associated with ATLI risk, enhance evidence synthesis, and identify susceptibility gene polymorphisms linked to ATLI occurrence. METHOD The protocol was registered in PROSPERO (CRD42024517311). Systematic searches of PubMed, EMBASE, Web of Science, and Cochrane Library databases were conducted to identify eligible studies from inception to February 21, 2024. Two authors independently reviewed eligibility, extracted data, and assessed quality. Odds ratios (ORs) with 95% confidence intervals (CIs) were used to evaluate associations between genetic polymorphisms and ATLI susceptibility. RESULTS A total of 25 meta-analyses were included, including 57 single nucleotide polymorphisms (SNPs) in 15 candidate genes. Significant associations were found for the glutathione S-transferase M1 (GSTM1) null genotype (OR = 1.43, 95% CI: 1.18-1.73, P < 0.001) and N-acetyltransferase 2 (NAT2) polymorphisms, including rs1799929 (dominant model, OR = 1.35, 95% CI: 1.12-1.63, P < 0.001), rs1799930 (dominant model, OR = 1.43, 95% CI: 1.23-1.66, P < 0.001), rs1799931 (dominant model, OR = 1.22, 95% CI: 1.02-1.46, P = 0.03), and the slow acetylator (SA) phenotype (OR = 2.91, 95% CI: 2.43-3.49, P < 0.001). No significant association was found between the CYP2E1 RsaI/PstI polymorphism (C1/C1 genotype) and ATLI risk (dominant model, OR = 0.79, 95% CI: 0.61-1.02, P = 0.08). CONCLUSION This umbrella review confirms that the GSTM1 null genotype, NAT2 polymorphisms (rs1799929, rs1799930, rs1799931), and the slow acetylator phenotype are associated with increased ATLI risk. These findings provide a foundation for further research on genotype-guided approaches to mitigating ATLI.
Collapse
Affiliation(s)
- Jingru Cheng
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Jia Zhu
- Department of Prevention and Healthcare, Changzhou Xinbei District Sanjing People's Hospital, Changzhou, China
| | - Ruina Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Meiling Zhang
- Department of Infectious Disease, The Jurong Hospital Affiliated to Jiangsu University, Jurong, China
| | - Bing Han
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Min Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Yiwen He
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Honggang Yi
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Shaowen Tang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, People's Republic of China.
| |
Collapse
|
3
|
Sung MW, Hu K, Hurlimann LM, Lees JA, Fennell KF, West MA, Costales C, Rodrigues AD, Zimmermann I, Dawson RJP, Liu S, Han S. Cyclosporine A sterically inhibits statin transport by solute carrier OATP1B1. J Biol Chem 2025; 301:108484. [PMID: 40199401 PMCID: PMC12127550 DOI: 10.1016/j.jbc.2025.108484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 04/10/2025] Open
Abstract
Members of the Organic Anion Transporter Polypeptides (OATP) are integral membrane proteins responsible for facilitating the transport of organic anions across the cell membrane. OATP1B1 (SLCO1B1), the prototypic OATP family member, is the most abundant uptake transporter in the liver and a key mediator of the hepatic uptake and clearance of numerous endogenous and xenobiotic compounds. It serves as a locus of important drug-drug interactions, such as those between statins and cyclosporine A, and carries the potential to enable liver-targeting therapeutics. In this study, we report cryo-EM structures of OATP1B1 and its complexes with one of its statin substrates, atorvastatin, and an inhibitor, cyclosporine A. This structural analysis has yielded insights into the mechanisms underlying the OATP1B1-mediated transport of statins and the inhibitory effect of cyclosporine A. These findings contribute to a better understanding of the molecular processes involved in drug transport and offer potential avenues for the development of targeted medications for liver-related conditions.
Collapse
Affiliation(s)
- Min Woo Sung
- Discovery Sciences, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA
| | - Kuan Hu
- Discovery Sciences, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA
| | | | - Joshua A Lees
- Discovery Sciences, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA
| | - Kimberly F Fennell
- Discovery Sciences, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA
| | - Mark A West
- Pharmacokinetics, Dynamics, and Metabolism, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA
| | - Chester Costales
- Pharmacokinetics, Dynamics, and Metabolism, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA
| | - Amilcar David Rodrigues
- Pharmacokinetics, Dynamics, and Metabolism, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA
| | | | | | - Shenping Liu
- Discovery Sciences, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA.
| | - Seungil Han
- Discovery Sciences, Discovery & Early Development, Pfizer Inc, Groton, Connecticut, USA.
| |
Collapse
|
4
|
Hagenbuch B, Stieger B, Locher KP. Organic anion transporting polypeptides: Pharmacology, toxicology, structure, and transport mechanisms. Pharmacol Rev 2025; 77:100023. [PMID: 40148036 DOI: 10.1016/j.pharmr.2024.100023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/12/2024] [Indexed: 03/29/2025] Open
Abstract
Organic anion transporting polypeptides (OATPs) are membrane proteins that mediate the uptake of a wide range of substrates across the plasma membrane of various cells and tissues. They are classified into 6 subfamilies, OATP1 through OATP6. Humans contain 12 OATPs encoded by 11 solute carrier of organic anion transporting polypeptide (SLCO) genes: OATP1A2, OATP1B1, OATP1B3, the splice variant OATP1B3-1B7, OATP1C1, OATP2A1, OATP2B1, OATP3A1, OATP4A1, OATP4C1, OATP5A1, and OATP6A1. Most of these proteins are expressed in epithelial cells, where they mediate the uptake of structurally unrelated organic anions, cations, and even neutral compounds into the cytoplasm. The best-characterized members are OATP1B1 and OATP1B3, which have an important role in drug metabolism by mediating drug uptake into the liver and are involved in drug-drug interactions. In this review, we aimed to (1) provide a historical perspective on the identification of OATPs and their nomenclature and discuss their phylogenic relationships and molecular characteristics; (2) review the current knowledge of the broad substrate specificity and their role in drug disposition and drug-drug interactions, with a special emphasis on human hepatic OATPs; (3) summarize the different experimental systems that are used to study the function of OATPs and discuss their advantages and disadvantages; (4) review the available experimental 3-dimensional structures and examine how they can help elucidate the transport mechanisms of OATPs; and (5) finally, summarize the current knowledge of the regulation of OATP expression, discuss clinically important single-nucleotide polymorphisms, and outline challenges of physiologically based pharmacokinetic modeling and in vitro to in vivo extrapolation. SIGNIFICANCE STATEMENT: Organic anion transporting polypeptides (OATPs) are a family of 12 uptake transporters in the solute carrier superfamily. Several members, particularly the liver-expressed OATP1B1 and OATP1B3, are important drug transporters. They mediate the uptake of several endobiotics and xenobiotics, including statins and numerous other drugs, into hepatocytes, and their inhibition by other drugs or reduced expression due to single-nucleotide polymorphisms can lead to adverse drug effects. Their recently solved 3-dimensional structures should help to elucidate their transport mechanisms and broad substrate specificities.
Collapse
Affiliation(s)
- Bruno Hagenbuch
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas.
| | - Bruno Stieger
- Institute of Molecular Biology and Biophysics, ETH Zürich, Zürich, Switzerland
| | - Kaspar P Locher
- Institute of Molecular Biology and Biophysics, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
5
|
Uoti A, Järvinen E, Sjöstedt N, Koenderink J, Finel M, Kidron H. Efflux and uptake of androgen sulfates using transporter-overexpressing HEK293 cells and membrane vesicles. J Pharm Sci 2025:103705. [PMID: 39993711 DOI: 10.1016/j.xphs.2025.103705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Hydrophilic steroid conjugates require active and facilitated transport mechanisms for their distribution into tissues and excretion from the body. The ATP-binding cassette (ABC) and solute carrier organic anion (SLCO) transporters involved in androgen sulfate (-S) disposition have been poorly characterized. In this study, we investigated the in vitro transport of testosterone-S, epitestosterone-S, dehydroepiandrosterone-S (DHEA-S), androsterone-S, and etiocholanolone-S by the multidrug resistance-associated proteins 2-4 (MRP2-4, ABCC2-4), breast cancer resistance protein (BCRP, ABCG2), and organic anion-transporting polypeptides (OATP) 1B1, 1B3, and 2B1 (SLCO1B1, SLCO1B3, and SLCO2B1) using human transporter-overexpressing HEK293 cells and membrane vesicles. We found testosterone-S, epitestosterone-S, and DHEA-S to be selectively transported by BCRP and/or MRP4, whereas all studied androgen sulfates were substrates of MRP3, OATP1B1, OATP1B3, and OATP2B1. MRP2 did not transport any of the studied compounds. Evaluation of transport kinetics revealed MRP4 to interact with its substrates at high to moderate affinity, whereas the observed affinities towards MRP3, BCRP, and OATPs were mostly moderate. These results help to build a better mechanistic understanding of the disposition of androgen sulfates in the human body. Additionally, this data may be used to assess the feasibility of androgen sulfates as additional biomarkers in doping detection.
Collapse
Affiliation(s)
- Arttu Uoti
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Erkka Järvinen
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Noora Sjöstedt
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jan Koenderink
- Department of Pharmacy - Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Moshe Finel
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Heidi Kidron
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
6
|
Tambe V, Soderblom EJ, Kayesh R, Aditya V, Xu C, Yue W. Regulation of organic anion transporting polypeptide 1B1 transport function by concurrent phosphorylation and lysine-acetylation: A novel posttranslational regulation mechanism. Mol Pharmacol 2025; 107:100007. [PMID: 40023514 PMCID: PMC11934288 DOI: 10.1016/j.molpha.2024.100007] [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: 07/15/2024] [Accepted: 11/08/2024] [Indexed: 03/04/2025] Open
Abstract
Organic anion transporting polypeptide (OATP) 1B1 is crucial for hepatic uptake of many drugs and endogenous substrates. The clinically relevant OATP1B1 c.521 T>C (V174A) polymorphism exhibits reduced transport activity in vitro and in vivo in humans. Previously, we reported increased total phosphorylation of V174A-OATP1B1 compared to wild-type (WT)-OATP1B1, although the differentially phosphorylated sites remain to be identified. Lysine-acetylation, a key posttranslational modification (PTM), has not been investigated in OATP1B1. This study aimed to identify differential PTMs of WT-OATP1B1 and V174A-OATP1B1 by quantitatively comparing the relative abundance of modified peptides using liquid chromatography-tandem mass spectrometry-based proteomics and to assess the impact of these PTMs on OATP1B1 transport function using [3H]-estradiol-17-β-D-glucuronide as substrate in transporter-expressing human embryonic kidney 293 cells. We discovered that OATP1B1 is lysine-acetylated at 13 residues. Compared to WT-OATP1B1, V174A-OATP1B1 has increased concurrent phosphorylation at S659 and S663 and concurrent phosphorylation (at S659 and S663) and lysine-acetylation (at K650) (P < .05). Variants mimicking concurrent phosphorylation (S659E-S663E-OATP1B1) and concurrent phosphorylation and acetylation (K650Q-659E-S663E-OATP1B1) both demonstrated reduced substrate transport by 0.86 ± 0.055-fold and 0.65 ± 0.047-fold of WT-OATP1B1 (both P < .05), respectively. Single-site mimics of phosphorylation or lysine-acetylation at K650, S659, and S663 did not affect OATP1B1 transport function, indicating cooperative effects on OATP1B1 by concurrent PTMs. All variants and WT-OATP1B1 were primarily localized to the plasma membrane and colocalized with plasma membrane protein Na/K-ATPase as determined by immunofluorescent staining and confocal microscopy. The current study elucidates a novel mechanism in which concurrent serine-phosphorylation and lysine-acetylation impair OATP1B1-mediated transport, suggesting potential interplay between these PTMs in regulating OATP1B1. SIGNIFICANCE STATEMENT: Understanding organic anion transporting polypeptide (OATP1B1) regulation is key to predicting altered drug disposition. The Val174Ala-OATP1B1 polymorphism exhibits reduced transport activity and is the most effective predictor of statin-induced myopathy. Val174Ala-OATP1B1 was found to be associated with increased serine-phosphorylation at Ser659 and Ser663 and lysine-acetylation at Lys650; concurrent PTMs at these sites reduce OATP1B1 function. These findings revealed a novel mechanism involved in transporter regulation, suggesting potential interplay between these PTMs in governing hepatic drug transport and response.
Collapse
Affiliation(s)
- Vishakha Tambe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma
| | - Erik J Soderblom
- Proteomics and Metabolomics Core Facility, Duke University School of Medicine, Durham, North Carolina
| | - Ruhul Kayesh
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma
| | - Vikram Aditya
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma
| | - Chao Xu
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma
| | - Wei Yue
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma.
| |
Collapse
|
7
|
Bintee B, Banerjee R, Hegde M, Vishwa R, Alqahtani MS, Abbas M, Alqahtani A, Rangan L, Sethi G, Kunnumakkara AB. Exploring bile acid transporters as key players in cancer development and treatment: Evidence from preclinical and clinical studies. Cancer Lett 2025; 609:217324. [PMID: 39571783 DOI: 10.1016/j.canlet.2024.217324] [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: 07/11/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 12/01/2024]
Abstract
Bile acid transporters (BATs) are integral membrane proteins belonging to various families, such as solute carriers, organic anion transporters, and ATP-binding cassette families. These transporters play a crucial role in bile acid transportation within the portal and systemic circulations, with expression observed in tissues, including the liver, kidney, and small intestine. Bile acids serve as signaling molecules facilitating the absorption and reabsorption of fats and lipids. Dysregulation of bile acid concentration has been implicated in tumorigenesis, yet the role of BATs in this process remains underexplored. Emerging evidence suggests that BATs may modulate various stages of cancer progression, including initiation, development, proliferation, metastasis, and tumor microenvironment regulation. Targeting BATs using siRNAs, miRNAs, and small compound inhibitors in preclinical models and their polymorphisms are well-studied for transporters like BSEP, MDR1, MRP2, OATP1A2, etc., and have shed light on their involvement in tumorigenesis, particularly in cancers such as those affecting the liver and gastrointestinal tract. While BATs' role in diseases like Alagille syndrome, biliary atresia, and cirrhosis have been extensively studied, their implications in cancer warrant further investigation. This review highlights the expression and function of BATs in cancer development and emphasizes the potential of targeting these transporters as a novel therapeutic strategy for various malignancies.
Collapse
Affiliation(s)
- Bintee Bintee
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Ruchira Banerjee
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India; Applied Biodiversity Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Ravichandran Vishwa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, 61421, Saudi Arabia; BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, LE1 7RH, United Kingdom
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, 61421, Saudi Arabia
| | - Athba Alqahtani
- Research Centre, King Fahad Medical City, P.O. Box: 59046, Riyadh, 11525, Saudi Arabia
| | - Latha Rangan
- Applied Biodiversity Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore; NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117699, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
| |
Collapse
|
8
|
Ramírez-Mejía MM, Castillo-Castañeda SM, Pal SC, Qi X, Méndez-Sánchez N. The Multifaceted Role of Bilirubin in Liver Disease: A Literature Review. J Clin Transl Hepatol 2024; 12:939-948. [PMID: 39544246 PMCID: PMC11557368 DOI: 10.14218/jcth.2024.00156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/19/2024] [Accepted: 10/08/2024] [Indexed: 11/17/2024] Open
Abstract
Bilirubin, the primary breakdown product of hemoproteins, particularly hemoglobin, plays a key role in the diagnosis, prognosis, and monitoring of liver diseases. In acute liver diseases, such as acute liver failure, drug-induced liver injury, and viral hepatitis, bilirubin serves as a biomarker reflecting the extent of hepatocyte loss and liver damage. Chronic liver diseases, including alcohol-related liver disease, chronic hepatitis C virus infection, metabolic dysfunction-associated fatty liver disease, and autoimmune liver diseases, are marked by persistent liver injury and inflammation. Bilirubin levels in chronic liver diseases provide insight into liver function, disease severity, and prognosis. As a versatile biomarker, bilirubin offers valuable information on the pathophysiology of liver diseases and aids in guiding clinical decision-making regarding the treatment of liver diseases and their complications. This review aimed to explore the multifunctional role of bilirubin in liver diseases by analyzing its biological functions beyond its role as a biomarker of liver damage.
Collapse
Affiliation(s)
- Mariana M. Ramírez-Mejía
- Plan of Combined Studies in Medicine (PECEM-MD/PhD), Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| | - Stephany M. Castillo-Castañeda
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
- Medical, Dental and Health Sciences Master and Doctorate Program, National Autonomous University of Mexico, Mexico City, Mexico
| | - Shreya C. Pal
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
- Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Xingshun Qi
- Department of Gastroenterology, General Hospital of Northern Theater Command (formerly General Hospital of Shenyang Military Area), Shenyang, Liaoning, China
| | - Nahum Méndez-Sánchez
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
- Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| |
Collapse
|
9
|
Surrer DB, Schüsser S, König J, Fromm MF, Gessner A. Transport of aromatic amino acids l-tryptophan, l-tyrosine, and l-phenylalanine by the organic anion transporting polypeptide (OATP) 3A1. FEBS J 2024; 291:4732-4743. [PMID: 39206635 DOI: 10.1111/febs.17255] [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: 03/28/2024] [Revised: 07/11/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
Amino acids are important for cellular metabolism. Their uptake across the plasma membrane is mediated by transport proteins. Despite the fact that the organic anion transporting polypeptide 4C1 (OATP4C1, Uniprot: Q6ZQN7) mediates transport of l-arginine and l-arginine derivatives, other members of the OATP family have not been characterized as amino acid transporters. The OATP family member OATP3A1 (gene symbol SLCO3A1, Uniprot: Q9UIG8) is ubiquitously expressed in human cells and highly expressed in many cancer tissues and cell lines. However, only a few substrates are known for OATP3A1. Accordingly, knowledge about its biological relevance is restricted. Our aim was to identify new substrates of OATP3A1 to gain insights into its (patho-)physiological function. In an LC-MS-based untargeted metabolomics assay using untreated OATP3A1-overexpressing HEK293 cells and control cells, we identified several amino acids as potential substrates of OATP3A1. Subsequent uptake experiments using exogenously added substrates revealed OATP3A1-mediated transport of l-tryptophan, l-tyrosine, and l-phenylalanine with 194.8 ± 28.7% (P < 0.05), 226.2 ± 18.7% (P < 0.001), and 235.2 ± 13.5% (P < 0.001), respectively, in OATP3A1-overexpressing cells compared to control cells. Furthermore, kinetic transport parameters (Km values) were determined (Trp = 61.5 ± 14.2 μm, Tyr = 220.8 ± 54.5 μm, Phe = 234.7 ± 20.6 μm). In summary, we identified the amino acids l-tryptophan, l-tyrosine, and l-phenylalanine as new substrates of OATP3A1. These findings could be used for a better understanding of (patho-)physiological processes involving increased demand of amino acids, where OATP3A1 should be considered as an important uptake transporter of l-tryptophan, l-tyrosine, and l-phenylalanine.
Collapse
Affiliation(s)
- Daniela B Surrer
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
| | - Sarah Schüsser
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
| | - Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
- FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
| | - Martin F Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
- FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
| | - Arne Gessner
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
- FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
| |
Collapse
|
10
|
Liang T, Liu H, Li L, Huan R, Gui C. Human organic anion transporting polypeptide 1B3 (OATP1B3) is more heavily N-glycosylated than OATP1B1 in extracellular loops 2 and 5. Int J Biol Macromol 2024; 278:134748. [PMID: 39147348 DOI: 10.1016/j.ijbiomac.2024.134748] [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: 05/29/2024] [Revised: 07/25/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
Human organic anion transporting polypeptide 1B3 (OATP1B3) and 1B1 are two liver-specific and highly homologous uptake transporters, whose structures consist of 12 transmembrane domains. The present study showed that OATP1B3 is more heavily N-glycosylated than OATP1B1 in extracellular loop 2 (EL2) and EL5. OATP1B3 has six N-glycosylation sites, namely N134, N145, N151, N445, N503, and N516, which is twice of that of OATP1B1. Single removal of individual N-glycans seems to have minimal influence on the surface expression and function of OATP1B3. However, simultaneous removal of all N-glycans will lead to OATP1B3's large retention in the endoplasmic reticulum and cellular degradation and thus significantly disrupts its surface expression. While N-glycosylation plays a crucial role in the surface expression of OATP1B3, it also has some effect on the transport function of OATP1B3 per se, which is not due to a decrease of substrate binding affinity but due to a reduced transporter's turnover number. Taken together, N-glycosylation is essential for normal surface expression and function of OATP1B3. Its disruption by some liver diseases such as NASH might alter the pharmacokinetic/pharmacodynamic properties of OATP1B3's substrate drugs.
Collapse
Affiliation(s)
- Ting Liang
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, China
| | - Han Liu
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, China
| | - Lanjing Li
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, China
| | - Ru Huan
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, China
| | - Chunshan Gui
- College of Pharmaceutical Sciences, Soochow University, 199 Renai Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
11
|
Slepnev AA, Abalenikhina YV, Shchulkin AV, Ananyeva PD, Yakusheva EN. Regulation of the Organic Anion Transporting Polypeptide 1B3 (OATP1B3) by Sex Hormones. Bull Exp Biol Med 2024; 177:630-634. [PMID: 39343843 DOI: 10.1007/s10517-024-06238-1] [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: 01/29/2024] [Indexed: 10/01/2024]
Abstract
The mechanisms of regulation of the organic anion transporting polypeptide OATP1B3 by sex hormones were studied using HepG2 cells. Estradiol, progesterone, and testosterone were added to cells at concentrations of 1, 10, 100 μM for 24 h. The relative content of OATP1B3 was evaluated by Western blotting. Estradiol at concentrations of 10 and 100 μM increased the level of OATP1B3 acting through the farnesoid X-receptor, testosterone at concentrations of 1, 10, and 100 μM increased the expression of the transporter protein due to its effect on the liver X-receptor subtype α (LXRα), and progesterone did not affect the expression of OATP1B3.
Collapse
Affiliation(s)
- A A Slepnev
- Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, Russia
| | - Yu V Abalenikhina
- Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, Russia
| | - A V Shchulkin
- Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, Russia.
| | - P D Ananyeva
- Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, Russia
| | - E N Yakusheva
- Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, Russia
| |
Collapse
|
12
|
Choucair I, Mallela DP, Hilser JR, Hartiala JA, Nemet I, Gogonea V, Li L, Lusis AJ, Fischbach MA, Tang WW, Allayee H, Hazen SL. Comprehensive Clinical and Genetic Analyses of Circulating Bile Acids and Their Associations With Diabetes and Its Indices. Diabetes 2024; 73:1215-1228. [PMID: 38701355 PMCID: PMC11262044 DOI: 10.2337/db23-0676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 04/24/2024] [Indexed: 05/05/2024]
Abstract
Bile acids (BAs) are cholesterol-derived compounds that regulate glucose, lipid, and energy metabolism. Despite their significance in glucose homeostasis, the association between specific BA molecular species and their synthetic pathways with diabetes is unclear. Here, we used a recently validated, stable-isotope dilution, high-performance liquid chromatography with tandem mass spectrometry method to quantify a panel of BAs in fasting plasma from 2,145 study participants and explored structural and genetic determinants of BAs linked to diabetes, insulin resistance, and obesity. Multiple 12α-hydroxylated BAs were associated with diabetes (adjusted odds ratio [aOR] range, 1.3-1.9; P < 0.05 for all) and insulin resistance (aOR range, 1.3-2.2; P < 0.05 for all). Conversely, multiple 6α-hydroxylated BAs and isolithocholic acid (iso-LCA) were inversely associated with diabetes and obesity (aOR range, 0.3-0.9; P < 0.05 for all). Genome-wide association studies revealed multiple genome-wide significant loci linked with 9 of the 14 diabetes-associated BAs, including a locus for iso-LCA (rs11866815). Mendelian randomization analyses showed genetically elevated deoxycholic acid levels were causally associated with higher BMI, and iso-LCA levels were causally associated with reduced BMI and diabetes risk. In conclusion, comprehensive, large-scale, quantitative mass spectrometry and genetics analyses show circulating levels of multiple structurally specific BAs, especially DCA and iso-LCA, are clinically associated with and genetically linked to obesity and diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Ibrahim Choucair
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH
| | - Deepthi P. Mallela
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH
| | - James R. Hilser
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Jaana A. Hartiala
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Ina Nemet
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH
| | - Valentin Gogonea
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH
- Department of Chemistry, Cleveland State University, Cleveland, OH
| | - Lin Li
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH
| | - Aldons J. Lusis
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA
| | | | - W.H. Wilson Tang
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH
- Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH
| | - Hooman Allayee
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Stanley L. Hazen
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH
- Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
13
|
Glebavičiūtė G, Vijaya AK, Preta G. Effect of Statin Lipophilicity on the Proliferation of Hepatocellular Carcinoma Cells. BIOLOGY 2024; 13:455. [PMID: 38927335 PMCID: PMC11200858 DOI: 10.3390/biology13060455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/02/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024]
Abstract
The HMG-CoA reductase inhibitors, statins, are drugs used globally for lowering the level of cholesterol in the blood. Different clinical studies of statins in cancer patients have indicated a decrease in cancer mortality, particularly in patients using lipophilic statins compared to those on hydrophilic statins. In this paper, we selected two structurally different statins (simvastatin and pravastatin) with different lipophilicities and investigated their effects on the proliferation and apoptosis of hepatocellular carcinoma cells. Lipophilic simvastatin highly influences cancer cell growth and survival in a time- and concentration-dependent manner, while pravastatin, due to its hydrophilic structure and limited cellular uptake, showed minimal cytotoxic effects.
Collapse
Affiliation(s)
| | | | - Giulio Preta
- Institute of Biochemistry, Life Science Center, Vilnius University, LT-10257 Vilnius, Lithuania; (G.G.); (A.K.V.)
| |
Collapse
|
14
|
Bechtold BJ, Lynch KD, Oyanna VO, Call MR, Graf TN, Oberlies NH, Clarke JD. Rifampin- and Silymarin-Mediated Pharmacokinetic Interactions of Exogenous and Endogenous Substrates in a Transgenic OATP1B Mouse Model. Mol Pharm 2024; 21:2284-2297. [PMID: 38529622 PMCID: PMC11073900 DOI: 10.1021/acs.molpharmaceut.3c01088] [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] [Indexed: 03/27/2024]
Abstract
Organic anion-transporting polypeptides (OATP) 1B1 and OATP1B3, encoded by the SLCO gene family of the solute carrier superfamily, are involved in the disposition of many exogenous and endogenous compounds. Preclinical rodent models help assess risks of pharmacokinetic interactions, but interspecies differences in transporter orthologs and expression limit direct clinical translation. An OATP1B transgenic mouse model comprising a rodent Slco1a/1b gene cluster knockout and human SLCO1B1 and SLCO1B3 gene insertions provides a potential physiologically relevant preclinical tool to predict pharmacokinetic interactions. Pharmacokinetics of exogenous probe substrates, pitavastatin and pravastatin, and endogenous OATP1B biomarkers, coproporphyrin-I and coproporphyrin-III, were determined in the presence and absence of known OATP/Oatp inhibitors, rifampin or silymarin (an extract of milk thistle [Silybum marianum]), in wild-type FVB mice and humanized OATP1B mice. Rifampin increased exposure of pitavastatin (4.6- and 2.8-fold), pravastatin (3.6- and 2.2-fold), and coproporphyrin-III (1.6- and 2.1-fold) in FVB and OATP1B mice, respectively, but increased coproporphyrin-I AUC0-24h only (1.8-fold) in the OATP1B mice. Silymarin did not significantly affect substrate AUC, likely because the silymarin flavonolignan concentrations were at or below their reported IC50 values for the relevant OATPs/Oatps. Silymarin increased the Cmax of pitavastatin 2.7-fold and pravastatin 1.9-fold in the OATP1B mice. The data of the OATP1B mice were similar to those of the pitavastatin and pravastatin clinical data; however, the FVB mice data more closely recapitulated pitavastatin clinical data than the data of the OATP1B mice, suggesting that the OATP1B mice are a reasonable, though costly, preclinical strain for predicting pharmacokinetic interactions when doses are optimized to achieve clinically relevant plasma concentrations.
Collapse
Affiliation(s)
- Baron J. Bechtold
- Department of Pharmaceutical Sciences, Washington State University, 412 E. Spokane Falls Blvd., Spokane, Washington 99202, United States
| | - Katherine D. Lynch
- Department of Pharmaceutical Sciences, Washington State University, 412 E. Spokane Falls Blvd., Spokane, Washington 99202, United States
| | - Victoria O. Oyanna
- Department of Pharmaceutical Sciences, Washington State University, 412 E. Spokane Falls Blvd., Spokane, Washington 99202, United States
| | - M. Ridge Call
- Department of Pharmaceutical Sciences, Washington State University, 412 E. Spokane Falls Blvd., Spokane, Washington 99202, United States
| | - Tyler N. Graf
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, North Carolina, 27412, United States
| | - Nicholas H. Oberlies
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, North Carolina, 27412, United States
| | - John D. Clarke
- Department of Pharmaceutical Sciences, Washington State University, 412 E. Spokane Falls Blvd., Spokane, Washington 99202, United States
| |
Collapse
|
15
|
Zhang Y, Huang J, Huang S, Liu J, Deng L, Liang C, Guo Y, Yao B, Wang X. Construction and characterization of a humanized SLCO1B1 rat model with its application in evaluating the uptake of different statins. Acta Pharm Sin B 2024; 14:1592-1604. [PMID: 38572097 PMCID: PMC10985027 DOI: 10.1016/j.apsb.2023.12.019] [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: 09/21/2023] [Revised: 11/28/2023] [Accepted: 12/06/2023] [Indexed: 04/05/2024] Open
Abstract
Organic anion-transporting polypeptides 1B1 (OATP1B1) plays a crucial role in the transport of statins. However, there are too few animal models related to OATP1B1, especially humanized animal models. In this study, the human SLCO1B1 cDNA was inserted into the second exon of the rat Slco1b2 gene using CRISPR/Cas9 technology. Pharmacokinetic characteristics of statins were conducted in wild-type (WT), humanized OATP1B1 (hOATP1B1), and OATP1B2 knockout (OATP1B2 KO) rats, respectively. The results showed that human OATP1B1 was successfully expressed in rat liver and exhibited transport function. Furthermore, the pharmacokinetic results revealed that OATP1B1 exhibited varying uptake levels of pivastatin, rosuvastatin, and fluvastatin, leading to different levels of exposure within the body. These results were consistent with those obtained from in vitro experiments using overexpressed cell lines. In conclusion, we established a novel humanized SLCO1B1 transgenic rat model to assess the role of human OATP1B1 in the uptake of different statins. The different uptake mediated by OATP1B1 may be an important reason for the different efficacy of statins. The hOATP1B1 rat is a promising model for improving the prediction of human drug transport.
Collapse
Affiliation(s)
- Yuanjin Zhang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai 200241, China
| | - Junze Huang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai 200241, China
| | - Shengbo Huang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai 200241, China
| | - Jie Liu
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai 200241, China
| | - Luyao Deng
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai 200241, China
| | - Chenmeizi Liang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai 200241, China
| | - Yuanqing Guo
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai 200241, China
| | - Bingyi Yao
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai 200241, China
| | - Xin Wang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai 200241, China
| |
Collapse
|
16
|
Takumi S, Tomioka M, Yunoki Y, Eto R, Komatsu Y, Shiozaki K, Komatsu M. Microcystin-LR-induced epithelial-mesenchymal transition-like cells acquire resistance to multi-toxins. Toxicon 2024; 238:107592. [PMID: 38163460 DOI: 10.1016/j.toxicon.2023.107592] [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: 10/31/2023] [Revised: 12/15/2023] [Accepted: 12/29/2023] [Indexed: 01/03/2024]
Abstract
The protein phosphatase inhibitor microcystin-LR (MC-LR), a hepatocyte-selective cyanotoxin, induces phenotypic changes in HEK293 OATP1B3-expressing (HEK293-OATP1B3) cells, which include cytoskeletal reorganization (HEK293-OATP1B3-AD) and anoikis resistance (HEK293-OATP1B3-FL) transformed cells, respectively. These cells acquire resistance to MC-LR and partial epithelial-mesenchymal transition (EMT) characteristics. In cancer cells, EMT is generally involved in multi-drug resistance. Here, we focused on the multi-drug resistance of HEK293-OATP1B3-AD and HEK293-OATP1B3-FL cells. The MTT assay and immunoblotting were conducted to examine the responses of HEK293-OATP1B3, HEK293-OATP1B3-AD, and HEK293-OATP1B3-FL cells to multiple toxins and drugs that function as substrates for OATP1B3, including MC-LR, nodularin (Nod), okadaic acid (OA), and cisplatin (CDDP). HEK293-OATP1B3-AD and HEK293-OATP1B3-FL cells were more resistant to MC-LR, Nod, and OA than HEK293-OATP1B3 cells. Conversely, the three cell types were equivalently sensitive to CDDP. By using protein phosphatase assay, the reduction of the inhibitory effect of MC-LR and Nod on phosphatase activity might be one reason for the resistance to MC-LR and Nod in HEK293-OATP1B3-AD and HEK293-OATP1B3-FL cells. Furthermore, the parental HEK293-OATP1B3 cells showed enhanced p53 phosphorylation and stabilization after MC-LR exposure, while p53 phosphorylation was attenuated in HEK293-OATP1B3-AD and HEK293-OATP1B3-FL cells. Moreover, in HEK293-OATP1B3-AD and HEK293-OATP1B3-FL cells, AKT phosphorylation was higher than that of the parental HEK293-OATP1B3 cell line. These results suggest that the multi-toxin resistance observed in HEK293-OATP1B3-AD and HEK293-OATP1B3-FL cells is associated with AKT activation and p53 inactivation.
Collapse
Affiliation(s)
- Shota Takumi
- Division of Food and Chemical Biology, Faculty of Fisheries, Kagoshima University, 890-0056, Kagoshima, Japan; The United Graduate School of Agricultural Sciences, Kagoshima University, 890-8580, Kagoshima, Japan
| | - Masaru Tomioka
- Division of Food and Chemical Biology, Faculty of Fisheries, Kagoshima University, 890-0056, Kagoshima, Japan
| | - Yasunari Yunoki
- Division of Food and Chemical Biology, Faculty of Fisheries, Kagoshima University, 890-0056, Kagoshima, Japan
| | - Risa Eto
- Division of Food and Chemical Biology, Faculty of Fisheries, Kagoshima University, 890-0056, Kagoshima, Japan
| | - Yumiko Komatsu
- Department of Domestic Science, Kagoshima Women's College, 890-8565, Kagoshima, Japan
| | - Kazuhiro Shiozaki
- Division of Food and Chemical Biology, Faculty of Fisheries, Kagoshima University, 890-0056, Kagoshima, Japan; The United Graduate School of Agricultural Sciences, Kagoshima University, 890-8580, Kagoshima, Japan
| | - Masaharu Komatsu
- Division of Food and Chemical Biology, Faculty of Fisheries, Kagoshima University, 890-0056, Kagoshima, Japan; The United Graduate School of Agricultural Sciences, Kagoshima University, 890-8580, Kagoshima, Japan.
| |
Collapse
|
17
|
Hoste E, Haufroid V, Deldicque L, Balligand JL, Elens L. Atorvastatin-associated myotoxicity: A toxicokinetic review of pharmacogenetic associations to evaluate the feasibility of precision pharmacotherapy. Clin Biochem 2024; 124:110707. [PMID: 38182100 DOI: 10.1016/j.clinbiochem.2024.110707] [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: 10/11/2023] [Revised: 01/02/2024] [Accepted: 01/02/2024] [Indexed: 01/07/2024]
Abstract
Atorvastatin (ATV) and other statins are highly effective in reducing cholesterol levels. However, in some patients, the development of drug-associated muscle side effects remains an issue as it compromises the adherence to treatment. Since the toxicity is dose-dependent, exploring factors modulating pharmacokinetics (PK) appears fundamental. The purpose of this review aims at reporting the current state of knowledge about the singular genetic susceptibilities influencing the risk of developing ATV muscle adverse events through PK modulations. Multiple single nucleotide polymorphisms (SNP) in efflux (ABCB1, ABCC1, ABCC2, ABCC4 and ABCG2) and influx (SLCO1B1, SLCO1B3 and SLCO2B1) transporters have been explored for their association with ATV PK modulation or with statin-related myotoxicities (SRM) development. The most convincing pharmacogenetic association with ATV remains the influence of the rs4149056 (c.521 T > C) in SLCO1B1 on ATV PK and pharmacodynamics. This SNP has been robustly associated with increased ATV systemic exposure and consequently, an increased risk of SRM. Additionally, the SNP rs2231142 (c.421C > A) in ABCG2 has also been associated with increased drug exposure and higher risk of SRM occurrence. SLCO1B1 and ABCG2 pharmacogenetic associations highlight that modulation of ATV systemic exposure is important to explain the risk of developing SRM. However, some novel observations credit the hypothesis that additional genes (e.g. SLCO2B1 or ABCC1) might be important for explaining local PK modulations within the muscle tissue, indicating that studying the local PK directly at the skeletal muscle level might pave the way for additional understanding.
Collapse
Affiliation(s)
- Emilia Hoste
- Integrated PharmacoMetrics, pharmacoGenomics and Pharmacokinetics, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), Brussels 1200, Belgium; Louvain Center for Toxicology and Applied Pharmacology, Institut de recherche expérimentale et clinique (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Vincent Haufroid
- Louvain Center for Toxicology and Applied Pharmacology, Institut de recherche expérimentale et clinique (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium; Department of Clinical Chemistry, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Louise Deldicque
- Institute of Neuroscience (IoNS), Université Catholique de Louvain (UCLouvain), Louvain-la-Neuve 1348, Belgium
| | - Jean-Luc Balligand
- Pole of Pharmacology and Therapeutics (FATH), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Laure Elens
- Integrated PharmacoMetrics, pharmacoGenomics and Pharmacokinetics, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), Brussels 1200, Belgium; Louvain Center for Toxicology and Applied Pharmacology, Institut de recherche expérimentale et clinique (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium.
| |
Collapse
|
18
|
Cho CK, Mo JY, Ko E, Kang P, Jang CG, Lee SY, Lee YJ, Bae JW, Choi CI. Physiologically based pharmacokinetic (PBPK) modeling of pitavastatin in relation to SLCO1B1 genetic polymorphism. Arch Pharm Res 2024; 47:95-110. [PMID: 38159179 DOI: 10.1007/s12272-023-01476-9] [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: 11/10/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024]
Abstract
Pitavastatin, a potent 3-hydroxymethylglutaryl coenzyme A reductase inhibitor, is indicated for the treatment of hypercholesterolemia and mixed dyslipidemia. Hepatic uptake of pitavastatin is predominantly occupied by the organic anion transporting polypeptide 1B1 (OATP1B1) and solute carrier organic anion transporter family member 1B1 (SLCO1B1) gene, which is a polymorphic gene that encodes OATP1B1. SLCO1B1 genetic polymorphism significantly alters the pharmacokinetics of pitavastatin. This study aimed to establish the physiologically based pharmacokinetic (PBPK) model to predict pitavastatin pharmacokinetics according to SLCO1B1 genetic polymorphism. PK-Sim® version 10.0 was used to establish the whole-body PBPK model of pitavastatin. Our pharmacogenomic data and a total of 27 clinical pharmacokinetic data with different dose administration and demographic properties were used to develop and validate the model, respectively. Physicochemical properties and disposition characteristics of pitavastatin were acquired from previously reported data or optimized to capture the plasma concentration-time profiles in different SLCO1B1 diplotypes. Model evaluation was performed by comparing the predicted pharmacokinetic parameters and profiles to the observed data. Predicted plasma concentration-time profiles were visually similar to the observed profiles in the non-genotyped populations and different SLCO1B1 diplotypes. All fold error values for AUC and Cmax were included in the two fold range of observed values. Thus, the PBPK model of pitavastatin in different SLCO1B1 diplotypes was properly established. The present study can be useful to individualize the dose administration strategy of pitavastatin in individuals with various ages, races, and SLCO1B1 diplotypes.
Collapse
Affiliation(s)
- Chang-Keun Cho
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Ju Yeon Mo
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Eunvin Ko
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Pureum Kang
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Choon-Gon Jang
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Seok-Yong Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Yun Jeong Lee
- College of Pharmacy, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jung-Woo Bae
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea
| | - Chang-Ik Choi
- College of Pharmacy, Dongguk University-Seoul, Goyang, 10326, Republic of Korea.
| |
Collapse
|
19
|
Walker LE, Stewart A, Pirmohamed SM, Meschia JF, Kinne FB. Stroke Pharmacogenetics. STROKE GENETICS 2024:423-508. [DOI: 10.1007/978-3-031-41777-1_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
20
|
Powell JT, Kayesh R, Ballesteros-Perez A, Alam K, Niyonshuti P, Soderblom EJ, Ding K, Xu C, Yue W. Assessing Trans-Inhibition of OATP1B1 and OATP1B3 by Calcineurin and/or PPIase Inhibitors and Global Identification of OATP1B1/3-Associated Proteins. Pharmaceutics 2023; 16:63. [PMID: 38258074 PMCID: PMC10818623 DOI: 10.3390/pharmaceutics16010063] [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: 09/09/2023] [Revised: 12/11/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Organic anion transporting polypeptide (OATP) 1B1 and OATP1B3 are key determinants of drug-drug interactions (DDIs). Various drugs including the calcineurin inhibitor (CNI) cyclosporine A (CsA) exert preincubation-induced trans-inhibitory effects upon OATP1B1 and/or OATP1B3 (abbreviated as OATP1B1/3) by unknown mechanism(s). OATP1B1/3 are phosphoproteins; calcineurin, which dephosphorylates and regulates numerous phosphoproteins, has not previously been investigated in the context of preincubation-induced trans-inhibition of OATP1B1/3. Herein, we compare the trans-inhibitory effects exerted on OATP1B1 and OATP1B3 by CsA, the non-analogous CNI tacrolimus, and the non-CNI CsA analogue SCY-635 in transporter-overexpressing human embryonic kidney (HEK) 293 stable cell lines. Preincubation (10-60 min) with tacrolimus (1-10 µM) rapidly and significantly reduces OATP1B1- and OATP1B3-mediated transport up to 0.18 ± 0.03- and 0.20 ± 0.02-fold compared to the control, respectively. Both CsA and SCY-635 can trans-inhibit OATP1B1, with the inhibitory effects progressively increasing over a 60 min preincubation time. At each equivalent preincubation time, CsA has greater trans-inhibitory effects toward OATP1B1 than SCY-635. Preincubation with SCY-635 for 60 min yielded IC50 of 2.2 ± 1.4 µM against OATP1B1, which is ~18 fold greater than that of CsA (0.12 ± 0.04 µM). Furthermore, a proteomics-based screening for protein interactors was used to examine possible proteins and processes contributing to OATP1B1/3 regulation and preincubation-induced inhibition by CNIs and other drugs. A total of 861 and 357 proteins were identified as specifically associated with OATP1B1 and OATP1B3, respectively, including various protein kinases, ubiquitin-related enzymes, the tacrolimus (FK506)-binding proteins FKBP5 and FKBP8, and several known regulatory targets of calcineurin. The current study reports several novel findings that expand our understanding of impaired OATP1B1/3 function; these include preincubation-induced trans-inhibition of OATP1B1/3 by the CNI tacrolimus, greater preincubation-induced inhibition by CsA compared to its non-CNI analogue SCY-635, and association of OATP1B1/3 with various proteins relevant to established and candidate OATP1B1/3 regulatory processes.
Collapse
Affiliation(s)
- John T. Powell
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| | - Ruhul Kayesh
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| | - Alexandra Ballesteros-Perez
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| | - Khondoker Alam
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| | - Pascaline Niyonshuti
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| | - Erik J. Soderblom
- Proteomics and Metabolomics Core Facility, Duke University School of Medicine, Durham, NC 27708, USA
| | - Kai Ding
- Department of Biostatistics & Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.D.); (C.X.)
| | - Chao Xu
- Department of Biostatistics & Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.D.); (C.X.)
| | - Wei Yue
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| |
Collapse
|
21
|
Zhu M, Chen X, Hao Z, He Y, Han B, Tang S. SLCO1B1 variants and the risk of antituberculosis drug-induced hepatotoxicity: a systematic review and meta-analysis. Pharmacogenomics 2023; 24:931-942. [PMID: 38019119 DOI: 10.2217/pgs-2023-0168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023] Open
Abstract
Aims: To evaluate the association between SLCO1B1 gene polymorphisms and susceptibility of antituberculosis drug-induced hepatotoxicity (ATDH). Methods: We searched the PubMed, Cochrane Library, Embase, Web of Science, Wan Fang and China National Knowledge Infrastructure database from inception to 2022. Results: Nine case-control studies with 1129 cases and 2203 controls were included. Among four SNPs reported in two or more studies, the final results indicated that SNP rs4149014 was significantly associated with decreased ATDH risk (dominant model, odds ratio: 0.73; 95% CI: 0.55-0.97; p = 0.03; allele model, odds ratio: 0.69; 95% CI: 0.55-0.86; p = 0.001), and the trial sequential analysis also confirmed this significant association. Conclusion: SLCO1B1 gene SNP rs4149014 was significantly associated with lower risk of ATDH susceptibility.
Collapse
Affiliation(s)
- Min Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xinyu Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Zhuolu Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yiwen He
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Bing Han
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Shaowen Tang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| |
Collapse
|
22
|
Shan Z, Yang X, Liu H, Yuan Y, Xiao Y, Nan J, Zhang W, Song W, Wang J, Wei F, Zhang Y. Cryo-EM structures of human organic anion transporting polypeptide OATP1B1. Cell Res 2023; 33:940-951. [PMID: 37674011 PMCID: PMC10709409 DOI: 10.1038/s41422-023-00870-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 08/17/2023] [Indexed: 09/08/2023] Open
Abstract
Members of the solute carrier organic anion transporting polypeptide (OATPs) family function as transporters for a large variety of amphipathic organic anions including endogenous metabolites and clinical drugs, such as bile salts, steroids, thyroid hormones, statins, antibiotics, antivirals, and anticancer drugs. OATP1B1 plays a vital role in transporting such substances into the liver for hepatic clearance. FDA and EMA recommend conducting in vitro testing of drug-drug interactions (DDIs) involving OATP1B1. However, the structure and working mechanism of OATPs still remains elusive. In this study, we determined cryo-EM structures of human OATP1B1 bound with representative endogenous metabolites (bilirubin and estrone-3-sulfate), a clinical drug (simeprevir), and a fluorescent indicator (2',7'-dichlorofluorescein), in both outward- and inward-open states. These structures reveal major and minor substrate binding pockets and conformational changes during transport. In combination with mutagenesis studies and molecular dynamics simulations, our work comprehensively elucidates the transport mechanism of OATP1B1 and provides the structural basis for DDI predictions involving OATP1B1, which will greatly promote our understanding of OATPs.
Collapse
Affiliation(s)
- Ziyang Shan
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xuemei Yang
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Huihui Liu
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Yafei Yuan
- Beijing Frontier Research Center for Biological Structure, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yuan Xiao
- Beijing Frontier Research Center for Biological Structure, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jing Nan
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wei Zhang
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wenqi Song
- Beijing Frontier Research Center for Biological Structure, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jufang Wang
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Feiwen Wei
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yanqing Zhang
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
23
|
Erokhina PD, Abalenikhina YV, Mylnikov PY, Petrov AV, Ganina SO, Konyakhin EA, Shchulkin AV, Yakusheva EN. The Effect of Original Russian Neurotropic Drugs on Organic Anion Transporting Polypeptides OATP1B1 and OATP1B3. Bull Exp Biol Med 2023; 176:170-175. [PMID: 38198100 DOI: 10.1007/s10517-024-05989-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Indexed: 01/11/2024]
Abstract
In experiments on HepG2 cells, we studied the effect of the original domestic neurotropic drugs omberacetam, fabomotizole, and ethylmethylhydroxypyridine succinate (EMHPS) (1-500 μM) on the activity and content of organic anion transporting polypeptides OATP1B1 and OATP1B3. It was shown that omberacetam (500 μM) increased the content of OATP1B1 and OATP1B3, fabomotizole did not affect the level of both transporters, and EMHPS (500 μM) increased the content of OATP1B1 compared to the control and did not affect the level of OATP1B3. The tested substances also reduced the OATP1B1/OATP1B3 ratio, as evidenced by a decrease in the penetration of atorvastatin, a substrate of the transporters, into HepG2 cells in the presence of omberacetam (100-500 μM), fabomotizole (500 μM), and EMHPS (10-500 μM). Evaluation of clinical significance of the obtained results, according to the FDA approach based on the calculation of the Cmax/IC50 ratio, showed that the effect of the tested substances on OATP1B1/OATP1B3 is clinically insignificant.
Collapse
Affiliation(s)
- P D Erokhina
- Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, Russia
| | - Yu V Abalenikhina
- Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, Russia
| | - P Yu Mylnikov
- Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, Russia
| | - A V Petrov
- Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, Russia
| | - S O Ganina
- Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, Russia
| | - E A Konyakhin
- Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, Russia
| | - A V Shchulkin
- Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, Russia.
| | - E N Yakusheva
- Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, Russia
| |
Collapse
|
24
|
Ungvári O, Bakos É, Kovacsics D, Özvegy-Laczka C. The fluorescence-based competitive counterflow assay developed for organic anion transporting polypeptides 1A2, 1B1, 1B3 and 2B1 identifies pentamidine as a selective OATP1A2 substrate. FASEB J 2023; 37:e23223. [PMID: 37781971 DOI: 10.1096/fj.202300530rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 10/03/2023]
Abstract
Organic anion transporting polypeptides OATP1A2, OATP1B1, OATP1B3 and OATP2B1 are Na+ - and ATP-independent exchangers of large, organic compounds, encompassing structurally diverse xenobiotics, including various drugs. These OATPs influence intestinal absorption (OATP2B1), hepatic clearance (OATP1B1/3) and blood to brain penetration (OATP1A2, OATP2B1) of their drug substrates. Consequently, OATP-mediated drug or food interactions may lead to altered pharmacokinetics and toxicity. During drug development, investigation of hepatic OATP1B1 and OATP1B3 is recommended by international regulatory agencies. Most frequently, OATP-drug interactions are investigated in an indirect assay, i.e., by examining uptake inhibition of a radioactive or fluorescent probe. However, indirect assays do not distinguish between transported substrates and non-transported OATP inhibitors. To fill this hiatus, a novel assay, termed competitive counterflow (CCF) has been developed and has since been applied for several OATPs to differentiate between substrates and non-transported inhibitors. However, previous OATP CCF assays, with the exception of that for OATP1B1, used radioactive probes. In the current study, we demonstrate that sulforhodamine 101 or pyranine can be used as fluorescent probes in a CCF assay to identify transported substrates of OATP1A2, or OATPs 1B1, 1B3 and 2B1, respectively. With the help of the newly developed fluorescence-based CCF method, we identify the FDA-approved anti-protozoal drug, pentamidine as a unique substrate of OATP1A2. Furthermore, we confirm the selective, OATP1A2-mediated uptake of pentamidine in a cytotoxicity assay. Based on our results, OATP1A2 may be an important determinant of pentamidine transport through the blood-brain barrier.
Collapse
Affiliation(s)
- Orsolya Ungvári
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, Budapest, Hungary
- Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, Budapest, Hungary
| | - Éva Bakos
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, Budapest, Hungary
| | - Daniella Kovacsics
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, Budapest, Hungary
| | - Csilla Özvegy-Laczka
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, Budapest, Hungary
| |
Collapse
|
25
|
Marker S, Espinoza AF, King AP, Woodfield SE, Patel RH, Baidoo K, Nix MN, Ciaramicoli LM, Chang YT, Escorcia FE, Vasudevan SA, Schnermann MJ. Development of Iodinated Indocyanine Green Analogs as a Strategy for Targeted Therapy of Liver Cancer. ACS Med Chem Lett 2023; 14:1208-1215. [PMID: 37736195 PMCID: PMC10510512 DOI: 10.1021/acsmedchemlett.3c00213] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/16/2023] [Indexed: 09/23/2023] Open
Abstract
Liver cancer is one of the leading causes of cancer-related deaths, with a significant increase in incidence worldwide. Novel therapies are needed to address this unmet clinical need. Indocyanine green (ICG) is a broadly used fluorescence-guided surgery (FGS) agent for liver tumor resection and has significant potential for conversion to a targeted therapy. Here, we report the design, synthesis, and investigation of a series of iodinated ICG analogs (I-ICG), which can be used to develop ICG-based targeted radiopharmaceutical therapy. We applied a CRISPR-based screen to identify the solute carrier transporter, OATP1B3, as a likely mechanism for ICG uptake. Our lead I-ICG compound specifically localizes to tumors in mice bearing liver cancer xenografts. This study introduces the chemistry needed to incorporate iodine onto the ICG scaffold and defines the impact of these modifications on key properties, including targeting liver cancer in vitro and in vivo.
Collapse
Affiliation(s)
- Sierra
C. Marker
- Chemical
Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Andres F. Espinoza
- Divisions
of Pediatric Surgery and Surgical Research, Michael E. DeBakey Department
of Surgery, Pediatric Surgical Oncology Laboratory, Texas Children’s
Surgical Oncology Program and Liver Tumor Program, Dan L. Duncan Cancer
Center, Baylor College of Medicine, Houston, Texas 77030, United States
| | - A. Paden King
- Molecular
Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20852, United States
| | - Sarah E. Woodfield
- Divisions
of Pediatric Surgery and Surgical Research, Michael E. DeBakey Department
of Surgery, Pediatric Surgical Oncology Laboratory, Texas Children’s
Surgical Oncology Program and Liver Tumor Program, Dan L. Duncan Cancer
Center, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Roma H. Patel
- Divisions
of Pediatric Surgery and Surgical Research, Michael E. DeBakey Department
of Surgery, Pediatric Surgical Oncology Laboratory, Texas Children’s
Surgical Oncology Program and Liver Tumor Program, Dan L. Duncan Cancer
Center, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Kwamena Baidoo
- Molecular
Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20852, United States
| | - Meredith N. Nix
- Chemical
Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Larissa Miasiro Ciaramicoli
- Department
of Chemistry, Pohang University of Science
and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea
| | - Young-Tae Chang
- Department
of Chemistry, Pohang University of Science
and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea
| | - Freddy E. Escorcia
- Molecular
Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20852, United States
| | - Sanjeev A. Vasudevan
- Divisions
of Pediatric Surgery and Surgical Research, Michael E. DeBakey Department
of Surgery, Pediatric Surgical Oncology Laboratory, Texas Children’s
Surgical Oncology Program and Liver Tumor Program, Dan L. Duncan Cancer
Center, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Martin J. Schnermann
- Chemical
Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| |
Collapse
|
26
|
Jang Y, Kang J, Hwang S, Chung JY, Cho JY. Quantification of OATP1B1 endogenous metabolites coproporphyrin I and III in human urine. Transl Clin Pharmacol 2023; 31:139-147. [PMID: 37810628 PMCID: PMC10551744 DOI: 10.12793/tcp.2023.31.e12] [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: 06/06/2023] [Revised: 06/30/2023] [Accepted: 07/12/2023] [Indexed: 10/10/2023] Open
Abstract
Coproporphyrin (CP)-I and CP-III are the markers of organic anion-transporting polypeptides' (OATPs) activities, and they are porphyrin metabolites that originate from heme synthesis. Furthermore, CP-I and CP-III, which are OATP1B endogenous metabolites, have gradually attracted the attention of scientists and researchers in recent years. Previous studies have also observed CP-I and CP-III levels as clinical biomarkers for predicting OATP1B inhibition in drug-drug interaction studies. To establish an accurate ultra-high performance liquid chromatography-mass spectrometry method for the quantitation of CP-I and CP-III, we reviewed previous methodological publications and applied them to a clinical pharmacology study using a human urine matrix. We used 13.25 M formic acid as a working solution for internal standards (CP-I 15N4 and CP-III d8) to avoid isobaric interference. The calibration curve showed good linearity in the range of 1-100 ng/mL, with a correlation coefficient (R2) higher than 0.996 in each validation batch. Both the between-run and within-run assays achieved good precision and accuracy, and we found that both CP-I and CP-III were stable in the pre-study validation. The method exhibited suitable dilution integrity, allowing for the re-analysis of samples with concentrations exceeding the upper limit of quantification through dilution. Overall, the application of the described method in a clinical study revealed that it can be utilized effectively to monitor drug-drug interactions mediated by OATP1B.
Collapse
Affiliation(s)
- Yeonseo Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jihyun Kang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sejung Hwang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jae-Yong Chung
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University Bundang Hospital, Seongnam 13620, Korea
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
27
|
Xue Y, Xiao B, Xia Z, Dai L, Xia Q, Zhong L, Zhu C, Zhu J. A New OATP-Mediated Hepatobiliary-Specific Mn(II)-Based MRI Contrast Agent for Hepatocellular Carcinoma in Mice: A Comparison With Gd-EOB-DTPA. J Magn Reson Imaging 2023; 58:926-933. [PMID: 36609994 DOI: 10.1002/jmri.28590] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/27/2022] [Accepted: 12/27/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Growing concerns about the safety of gadolinium (Gd)-based contrast agents have reinforced the need for the development of Gd-free MRI contrast agents (CAs) that are effective in imaging liver tumors. PURPOSE To evaluate the ability of Mn-BnO-TyEDTA MRI CA to detect hepatocellular carcinoma in a mouse model of implanted liver tumor. STUDY TYPE Prospective. ANIMAL MODEL Thirteen orthotopically implanted liver tumor mice. FIELD STRENGTH/SEQUENCE 3.0 T/precontrast and postcontrast T1-weighted fast spoiled gradient recalled echo and T2-weighted fast recovery fast spin-echo imaging with fat suppression. ASSESSMENT The relative enhancement ratio was calculated and statistically compared. Lesion detection in postcontrast images was analyzed by calculations of area under the curve (AUC, the increases in liver-to-tumor contrast-to-noise ratio [∆CNR] vs. time curve). Mn or Gd levels were measured in the liver and tumoral tissues by inductively coupled plasma-mass spectrometry. Tumor specimens were stained with hematoxylin and eosin (H&E) and the expression of organic anion transfer peptide (OATP)1B1 was evaluated by immunofluorescence (IF) staining and mean fluorescence intensity (MFI) was calculated. STATISTICAL TESTS Unpaired t-test and two-tailed paired t-test. P < 0.05 was considered statistical significance. RESULTS Mn-BnO-TyEDTA and Gd-EOB-DTPA demonstrated nearly identical enhancement patterns in the liver, tumor, and psoas muscle and no difference in lesion detection (AUC10-30, Mn = 851 ∆CR·min, AUC10-30, Gd = 823 ∆CR·min). A Significant higher concentration of metal (Mn or Gd) was found in the liver compared to the tumor ([Mn]liver = 0.88 ± 0.07 μmmol/g, [Mn]tumor = 0.49 ± 0.05 μmmol/g, [Gd]liver = 0.65 ± 0.07 μmmol/g, [Gd]tumor = 0.27 ± 0.04 μmmol/g). IF staining showed significantly decreased expression of OATP1B1 in the tumor core compared to the liver (MFItumor = 5.28 ± 1.54, MFIliver = 25.49 ± 3.41). DATA CONCLUSION Mn-BnO-TyEDTA can provide comparable hepatobiliary tumor contrast enhancement to Gd-EOB-DTPA. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Yuan Xue
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Bin Xiao
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Zhiyang Xia
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Lixiong Dai
- Wenzhou Key Laboratory of Biophysics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, China
| | - Qian Xia
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Lei Zhong
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Chunrong Zhu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jiang Zhu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, China
| |
Collapse
|
28
|
Ontsouka E, Schroeder M, Albrecht C. Revisited role of the placenta in bile acid homeostasis. Front Physiol 2023; 14:1213757. [PMID: 37546542 PMCID: PMC10402276 DOI: 10.3389/fphys.2023.1213757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023] Open
Abstract
To date, the discussion concerning bile acids (BAs) during gestation is almost exclusively linked to pregnancy complications such as intrahepatic cholestasis of pregnancy (ICP) when maternal serum BA levels reach very high concentrations (>100 μM). Generally, the placenta is believed to serve as a protective barrier avoiding exposure of the growing fetus to excessive amounts of maternal BAs that might cause detrimental effects (e.g., intrauterine growth restriction and/or increased vulnerability to metabolic diseases). However, little is known about the precise role of the placenta in BA biosynthesis, transport, and metabolism in healthy pregnancies when serum BAs are at physiological levels (i.e., low maternal and high fetal BA concentrations). It is well known that primary BAs are synthesized from cholesterol in the liver and are later modified to secondary BA species by colonic bacteria. Besides the liver, BA synthesis in extrahepatic sites such as the brain elicits neuroprotective actions through inhibition of apoptosis as well as oxidative and endoplasmic reticulum stress. Even though historically BAs were thought to be only "detergent molecules" required for intestinal absorption of dietary fats, they are nowadays acknowledged as full signaling molecules. They modulate a myriad of signaling pathways with functional consequences on essential processes such as gluconeogenesis -one of the principal energy sources of the fetus- and cellular proliferation. The current manuscript discusses the potential multipotent roles of physiologically circulating BAs on developmental processes during gestation and provides a novel perspective in terms of the importance of the placenta as a previously unknown source of BAs. Since the principle "not too much, not too little" applicable to other signaling molecules may be also true for BAs, the risks associated with fetal exposure to excessive levels of BAs are discussed.
Collapse
|
29
|
Takumi S, Hashimoto K, Tomioka M, Sato M, He W, Komatsu Y, Aoki S, Ikeda R, Shiozaki K, Furukawa T, Komatsu M. Acteoside from Conandron ramondioides Reduces Microcystin-LR Cytotoxicity by Inhibiting Intracellular Uptake Mediated by OATP1B3. PLANTA MEDICA 2023; 89:616-623. [PMID: 36626925 DOI: 10.1055/a-1978-8768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
The hepatotoxin microcystin-LR is a strong inhibitor of serine/threonine protein phosphatase (PP) 1 and PP2A. The onset of its cytotoxicity depends on its selective uptake via the hepatocyte uptake transporters, organic anion transporting polypeptide (OATP) 1B1 and OATP1B3. Understanding and preventing the cytotoxicity of microcystin-LR is crucial to maintain human health. This chemoprevention study demonstrates that the herbal plant extract of iwajisha (20 µg/mL) reduced microcystin-LR cytotoxicity in OATP1B3-expressing cells by approximately six times. In addition, 20 µM acteoside, which is one of the major compounds in iwajisha, reduced microcystin-LR cytotoxicity by approximately 7.4 times. Acteoside could also reduce the cytotoxicity of other compounds, such as okadaic acid and nodularin, which are both substrates of OATP1B3 and inhibitors of PP1/PP2A. To investigate the mechanism by which the cytotoxicity of microcystin-LR is attenuated by acteosides, microcystin-LR and microcystin-LR-binding proteins in cells were examined after microcystin-LR and acteosides were co-exposed. Thus, acteoside noncompetitively inhibited microcystin-LR uptake by OATP1B3-expressing cells. Furthermore, acteoside inhibited the intracellular interaction of microcystin-LR with its binding protein(s), including the 22 kDa protein. Furthermore, using immunoblot analysis, acteoside induced the phosphorylation of extracellular signal-regulated kinase (ERK), which is one of the survival signaling molecules. These results suggest that acteoside reduces microcystin-LR cytotoxicity through several mechanisms, including the inhibition of microcystin-LR uptake via OATP1B3, and decreased interaction between microcystin-LR and its binding protein(s), and that ERK signaling activation contributes to the attenuation effect of acteoside against microcystin-LR cytotoxicity.
Collapse
Affiliation(s)
- Shota Takumi
- Division of Food and Chemical Biology, Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| | - Kairi Hashimoto
- Division of Food and Chemical Biology, Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
| | - Masaru Tomioka
- Division of Food and Chemical Biology, Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
| | - Mina Sato
- Division of Food and Chemical Biology, Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
| | - Weijie He
- Division of Food and Chemical Biology, Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
| | - Yumiko Komatsu
- Department of Domestic Science, Kagoshima Women's College, Kagoshima, Japan
| | - Shunji Aoki
- Department of Pharmacy, School of Pharmacy, Hyogo Medical University, Kobe, Japan
| | - Ryuji Ikeda
- Department of Pharmacy, University of Miyazaki Hospital, Miyazaki, Japan
| | - Kazuhiro Shiozaki
- Division of Food and Chemical Biology, Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| | - Tatsuhiko Furukawa
- Department of Pathology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Masaharu Komatsu
- Division of Food and Chemical Biology, Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
30
|
Du X, Cui Z, Zhang R, Zhao K, Wang L, Yao J, Liu S, Cai C, Cao Y. The Effects of Rumen-Protected Choline and Rumen-Protected Nicotinamide on Liver Transcriptomics in Periparturient Dairy Cows. Metabolites 2023; 13:metabo13050594. [PMID: 37233635 DOI: 10.3390/metabo13050594] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
To investigate the effects of rumen-protected choline (RPC) and rumen-protected nicotinamide (RPM) on liver metabolic function based on transcriptome in periparturient dairy cows, 10 healthy Holstein dairy cows with similar parity were allocated to RPC and RPM groups (n = 5). The cows were fed experimental diets between 14 days before and 21 days after parturition. The RPC diet contained 60 g RPC per day, and the RPM diet contained 18.7 g RPM per day. Liver biopsies were taken 21 days after calving for the transcriptome analysis. A model of fat deposition hepatocytes was constructed using the LO2 cell line with the addition of NEFA (1.6 mmol/L), and the expression level of genes closely related to liver metabolism was validated and divided into a CHO group (75 μmol/L) and a NAM group (2 mmol/L). The results showed that the expression of a total of 11,023 genes was detected and clustered obviously between the RPC and RPM groups. These genes were assigned to 852 Gene Ontology terms, the majority of which were associated with biological process and molecular function. A total of 1123 differentially expressed genes (DEGs), 640 up-regulated and 483 down-regulated, were identified between the RPC and RPM groups. These DEGs were mainly correlated with fat metabolism, oxidative stress and some inflammatory pathways. In addition, compared with the NAM group, the gene expression level of FGF21, CYP26A1, SLC13A5, SLCO1B3, FBP2, MARS1 and CDH11 in the CHO group increased significantly (p < 0.05). We proposed that that RPC could play a prominent role in the liver metabolism of periparturient dairy cows by regulating metabolic processes such as fatty acid synthesis and metabolism and glucose metabolism; yet, RPM was more involved in biological processes such as the TCA cycle, ATP generation and inflammatory signaling.
Collapse
Affiliation(s)
- Xue'er Du
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China
| | - Zhijie Cui
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China
| | - Rui Zhang
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China
| | - Keliang Zhao
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China
| | - Lamei Wang
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China
| | - Shimin Liu
- UWA Institute of Agriculture, The University of Western Australia, Crawley, WA 6009, Australia
| | - Chuanjiang Cai
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China
| | - Yangchun Cao
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China
| |
Collapse
|
31
|
Ramsey LB, Gong L, Lee SB, Wagner JB, Zhou X, Sangkuhl K, Adams SM, Straka RJ, Empey PE, Boone EC, Klein TE, Niemi M, Gaedigk A. PharmVar GeneFocus: SLCO1B1. Clin Pharmacol Ther 2023; 113:782-793. [PMID: 35797228 PMCID: PMC10900141 DOI: 10.1002/cpt.2705] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/24/2022] [Indexed: 11/06/2022]
Abstract
The Pharmacogene Variation Consortium (PharmVar) is now providing star (*) allele nomenclature for the highly polymorphic human SLCO1B1 gene encoding the organic anion transporting polypeptide 1B1 (OATP1B1) drug transporter. Genetic variation within the SLCO1B1 gene locus impacts drug transport, which can lead to altered pharmacokinetic profiles of several commonly prescribed drugs. Variable OATP1B1 function is of particular importance regarding hepatic uptake of statins and the risk of statin-associated musculoskeletal symptoms. To introduce this important drug transporter gene into the PharmVar database and serve as a unified reference of haplotype variation moving forward, an international group of gene experts has performed an extensive review of all published SLCO1B1 star alleles. Previously published star alleles were self-assigned by authors and only loosely followed the star nomenclature system that was first developed for cytochrome P450 genes. This nomenclature system has been standardized by PharmVar and is now applied to other important pharmacogenes such as SLCO1B1. In addition, data from the 1000 Genomes Project and investigator-submitted data were utilized to confirm existing haplotypes, fill knowledge gaps, and/or define novel star alleles. The PharmVar-developed SLCO1B1 nomenclature has been incorporated by the Clinical Pharmacogenetics Implementation Consortium (CPIC) 2022 guideline on statin-associated musculoskeletal symptoms.
Collapse
Affiliation(s)
- Laura B Ramsey
- Divisions of Clinical Pharmacology and Research in Patient Services, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Li Gong
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA
| | - Seung-Been Lee
- Precision Medicine Institute, Macrogen Inc., Seoul, Korea
| | - Jonathan B Wagner
- Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri, USA
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Xujia Zhou
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, USA
| | - Katrin Sangkuhl
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA
| | - Solomon M Adams
- School of Pharmacy, Shenandoah University, Fairfax, Virginia, USA
| | - Robert J Straka
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Philip E Empey
- School of Pharmacy and Institute for Precision Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Erin C Boone
- Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri, USA
| | - Teri E Klein
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA
- Department of Medicine (BMIR), Stanford University, Stanford, California, USA
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Andrea Gaedigk
- Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri, USA
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
| |
Collapse
|
32
|
Häkkinen K, Kiander W, Kidron H, Lähteenvuo M, Urpa L, Lintunen J, Vellonen KS, Auriola S, Holm M, Lahdensuo K, Kampman O, Isometsä E, Kieseppä T, Lönnqvist J, Suvisaari J, Hietala J, Tiihonen J, Palotie A, Ahola-Olli AV, Niemi M. Functional Characterization of Six SLCO1B1 (OATP1B1) Variants Observed in Finnish Individuals with a Psychotic Disorder. Mol Pharm 2023; 20:1500-1508. [PMID: 36779498 PMCID: PMC9996821 DOI: 10.1021/acs.molpharmaceut.2c00715] [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: 02/14/2023]
Abstract
Variants in the SLCO1B1 (solute carrier organic anion transporter family member 1B1) gene encoding the OATP1B1 (organic anion transporting polypeptide 1B1) protein are associated with altered transporter function that can predispose patients to adverse drug effects with statin treatment. We explored the effect of six rare SLCO1B1 single nucleotide variants (SNVs) occurring in Finnish individuals with a psychotic disorder on expression and functionality of the OATP1B1 protein. The SUPER-Finland study has performed exome sequencing on 9381 individuals with at least one psychotic episode during their lifetime. SLCO1B1 SNVs were annotated with PHRED-scaled combined annotation-dependent (CADD) scores and the Ensembl variant effect predictor. In vitro functionality studies were conducted for the SNVs with a PHRED-scaled CADD score of >10 and predicted to be missense. To estimate possible changes in transport activity caused by the variants, transport of 2',7'-dichlorofluorescein (DCF) in OATP1B1-expressing HEK293 cells was measured. According to the findings, additional tests with rosuvastatin and estrone sulfate were conducted. The amount of OATP1B1 in crude membrane fractions was quantified using a liquid chromatography tandem mass spectrometry-based quantitative targeted absolute proteomics analysis. Six rare missense variants of SLCO1B1 were identified in the study population, located in transmembrane helix 3: c.317T>C (p.106I>T), intracellular loop 2: c.629G>T (p.210G>V), c.633A>G (p.211I>M), c.639T>A (p.213N>L), transmembrane helix 6: 820A>G (p.274I>V), and the C-terminal end: 2005A>C (p.669N>H). Of these variants, SLCO1B1 c.629G>T (p.210G>V) resulted in the loss of in vitro function, abolishing the uptake of DCF, estrone sulfate, and rosuvastatin and reducing the membrane protein expression to 31% of reference OATP1B1. Of the six rare missense variants, SLCO1B1 c.629G>T (p.210G>V) causes a loss of function of OATP1B1 transport in vitro and severely decreases membrane protein abundance. Carriers of SLCO1B1 c.629G>T might be susceptible to altered pharmacokinetics of OATP1B1 substrate drugs and might have increased likelihood of adverse drug effects such as statin-associated musculoskeletal symptoms.
Collapse
Affiliation(s)
- Katja Häkkinen
- Department of Forensic Psychiatry, Niuvanniemi Hospital, University of Eastern Finland, Kuopio FI-70240, Finland.,Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki FI-00014, Finland
| | - Wilma Kiander
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland
| | - Heidi Kidron
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00014, Finland
| | - Markku Lähteenvuo
- Department of Forensic Psychiatry, Niuvanniemi Hospital, University of Eastern Finland, Kuopio FI-70240, Finland
| | - Lea Urpa
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki FI-00014, Finland
| | - Jonne Lintunen
- Department of Forensic Psychiatry, Niuvanniemi Hospital, University of Eastern Finland, Kuopio FI-70240, Finland
| | | | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, Kuopio FI-70211, Finland
| | - Minna Holm
- Mental Health Team, Finnish Institute for Health and Welfare, Helsinki FI-00271, Finland
| | | | - Olli Kampman
- Faculty of Medicine and Health Technology, Tampere University, Tampere FI-33100, Finland.,Department of Psychiatry, Pirkanmaa Hospital District, Tampere FI-33521, Finland.,Department of Clinical Sciences (Psychiatry), Faculty of Medicine, Umeå University, Umeå SE-90187, Sweden.,Department of Psychiatry, University Hospital of Umeå, Umeå SE-90187, Sweden.,Department of Clinical Medicine (Psychiatry), Faculty of Medicine, University of Turku, Turku FI-20014, Finland.,Department of Psychiatry, The Wellbeing Services County of Ostrobothnia, Vaasa FI-65101, Finland
| | - Erkki Isometsä
- Department of Psychiatry, University of Helsinki and Helsinki University Hospital, Helsinki FI-00014, Finland
| | - Tuula Kieseppä
- Mental Health Team, Finnish Institute for Health and Welfare, Helsinki FI-00271, Finland.,Department of Psychiatry, University of Helsinki and Helsinki University Hospital, Helsinki FI-00014, Finland
| | - Jouko Lönnqvist
- Mental Health Team, Finnish Institute for Health and Welfare, Helsinki FI-00271, Finland.,Department of Psychiatry, University of Helsinki, Helsinki FI-00014, Finland
| | - Jaana Suvisaari
- Mental Health Team, Finnish Institute for Health and Welfare, Helsinki FI-00271, Finland
| | - Jarmo Hietala
- Department of Psychiatry, University of Turku and Turku University Hospital, Turku FI-20700, Finland
| | - Jari Tiihonen
- Department of Forensic Psychiatry, Niuvanniemi Hospital, University of Eastern Finland, Kuopio FI-70240, Finland.,Department of Clinical Neuroscience, Karolinska Institutet, Stockholm SE-17177, Sweden.,Center for Psychiatry Research, Stockholm City Council, Stockholm SE-11364, Sweden.,Neuroscience Center, University of Helsinki, Helsinki FI-00014, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki FI-00014, Finland.,The Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States.,Analytic and Translational Genetics Unit, Department of Medicine, Department of Neurology and Department of Psychiatry, Massachusetts General Hospital, Boston MA-02114, United States
| | - Ari V Ahola-Olli
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki FI-00014, Finland.,The Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States.,Department of Internal Medicine, Satasairaala Hospital, Pori FI-28500, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki, Helsinki FI-00014, Finland.,Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki FI-00014, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki FI-00029, Finland
| |
Collapse
|
33
|
Özvegy-Laczka C, Ungvári O, Bakos É. Fluorescence-based methods for studying activity and drug-drug interactions of hepatic solute carrier and ATP binding cassette proteins involved in ADME-Tox. Biochem Pharmacol 2023; 209:115448. [PMID: 36758706 DOI: 10.1016/j.bcp.2023.115448] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023]
Abstract
In humans, approximately 70% of drugs are eliminated through the liver. This process is governed by the concerted action of membrane transporters and metabolic enzymes. Transporters mediating hepatocellular uptake of drugs belong to the SLC (Solute carrier) superfamily of transporters. Drug efflux either toward the portal vein or into the bile is mainly mediated by active transporters of the ABC (ATP Binding Cassette) family. Alteration in the function and/or expression of liver transporters due to mutations, disease conditions, or co-administration of drugs or food components can result in altered pharmacokinetics. On the other hand, drugs or food components interacting with liver transporters may also interfere with liver function (e.g., bile acid homeostasis) and may even cause liver toxicity. Accordingly, certain transporters of the liver should be investigated already at an early stage of drug development. Most frequently radioactive probes are applied in these drug-transporter interaction tests. However, fluorescent probes are cost-effective and sensitive alternatives to radioligands, and are gaining wider application in drug-transporter interaction tests. In our review, we summarize our current understanding about hepatocyte ABC and SLC transporters affected by drug interactions. We provide an update of the available fluorescent and fluorogenic/activable probes applicable in in vitro or in vivo testing of these ABC and SLC transporters, including near-infrared transporter probes especially suitable for in vivo imaging. Furthermore, our review gives a comprehensive overview of the available fluorescence-based methods, not directly relying on the transport of the probe, suitable for the investigation of hepatic ABC or SLC-type drug transporters.
Collapse
Affiliation(s)
- Csilla Özvegy-Laczka
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, H-1117 Budapest, Magyar tudósok krt. 2., Hungary.
| | - Orsolya Ungvári
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, H-1117 Budapest, Magyar tudósok krt. 2., Hungary; Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Éva Bakos
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, H-1117 Budapest, Magyar tudósok krt. 2., Hungary
| |
Collapse
|
34
|
Lehtisalo M, Taskinen S, Tarkiainen EK, Neuvonen M, Viinamäki J, Paile-Hyvärinen M, Lilius TO, Tapaninen T, Backman JT, Tornio A, Niemi M. A comprehensive pharmacogenomic study indicates roles for SLCO1B1, ABCG2 and SLCO2B1 in rosuvastatin pharmacokinetics. Br J Clin Pharmacol 2023; 89:242-252. [PMID: 35942816 PMCID: PMC10087178 DOI: 10.1111/bcp.15485] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 07/23/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
AIMS The aim was to comprehensively investigate the effects of genetic variability on the pharmacokinetics of rosuvastatin. METHODS We conducted a genome-wide association study and candidate gene analyses of single dose rosuvastatin pharmacokinetics in a prospective study (n = 159) and a cohort of previously published studies (n = 88). RESULTS In a genome-wide association meta-analysis of the prospective study and the cohort of previously published studies, the SLCO1B1 c.521 T > C (rs4149056) single nucleotide variation (SNV) associated with increased area under the plasma concentration-time curve (AUC) and peak plasma concentration of rosuvastatin (P = 1.8 × 10-12 and P = 3.2 × 10-15 ). The candidate gene analysis suggested that the ABCG2 c.421C > A (rs2231142) SNV associates with increased rosuvastatin AUC (P = .0079), while the SLCO1B1 c.388A > G (rs2306283) and SLCO2B1 c.1457C > T (rs2306168) SNVs associate with decreased rosuvastatin AUC (P = .0041 and P = .0076). Based on SLCO1B1 genotypes, we stratified the participants into poor, decreased, normal, increased and highly increased organic anion transporting polypeptide (OATP) 1B1 function groups. The OATP1B1 poor function phenotype associated with 2.1-fold (90% confidence interval 1.6-2.8, P = 4.69 × 10-5 ) increased AUC of rosuvastatin, whereas the OATP1B1 highly increased function phenotype associated with a 44% (16-62%; P = .019) decreased rosuvastatin AUC. The ABCG2 c.421A/A genotype associated with 2.2-fold (1.5-3.0; P = 2.6 × 10-4 ) increased AUC of rosuvastatin. The SLCO2B1 c.1457C/T genotype associated with 28% decreased rosuvastatin AUC (11-42%; P = .01). CONCLUSION These data suggest roles for SLCO1B1, ABCG2 and SLCO2B1 in rosuvastatin pharmacokinetics. Poor SLCO1B1 or ABCG2 function genotypes may increase the risk of rosuvastatin-induced myotoxicity. Reduced doses of rosuvastatin are advisable for patients with these genotypes.
Collapse
Affiliation(s)
- Minna Lehtisalo
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Suvi Taskinen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - E Katriina Tarkiainen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Mikko Neuvonen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - Jenni Viinamäki
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - Maria Paile-Hyvärinen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Tuomas O Lilius
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Tuija Tapaninen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Janne T Backman
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Aleksi Tornio
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
35
|
Jiang Z, Wu Z, Liu R, Du Q, Fu X, Li M, Kuang Y, Lin S, Wu J, Xie W, Shi G, Peng Y, Zheng F. Effect of polymorphisms in drug metabolism and transportation on plasma concentration of atorvastatin and its metabolites in patients with chronic kidney disease. Front Pharmacol 2023; 14:1102810. [PMID: 36923356 PMCID: PMC10010391 DOI: 10.3389/fphar.2023.1102810] [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: 11/19/2022] [Accepted: 02/15/2023] [Indexed: 03/02/2023] Open
Abstract
Dyslipidemia due to renal insufficiency is a common complication in patients with chronic kidney diseases (CKD), and a major risk factor for the development of cardiovascular events. Atorvastatin (AT) is mainly used in the treatment of dyslipidemia in patients with CKD. However, response to the atorvastatin varies inter-individually in clinical applications. We examined the association between polymorphisms in genes involved in drug metabolism and transport, and plasma concentrations of atorvastatin and its metabolites (2-hydroxy atorvastatin (2-AT), 2-hydroxy atorvastatin lactone (2-ATL), 4-hydroxy atorvastatin (4-AT), 4-hydroxy atorvastatin lactone (4-ATL), atorvastatin lactone (ATL)) in kidney diseases patients. Genotypes were determined using TaqMan real time PCR in 212 CKD patients, treated with 20 mg of atorvastatin daily for 6 weeks. The steady state plasma concentrations of atorvastatin and its metabolites were quantified using ultraperformance liquid chromatography in combination with triple quadrupole mass spectrometry (UPLC-MS/MS). Univariate and multivariate analyses showed the variant in ABCC4 (rs3742106) was associated with decreased concentrations of AT and its metabolites (2-AT+2-ATL: β = -0.162, p = 0.028 in the dominant model; AT+2-AT+4-AT: β = -0.212, p = 0.028 in the genotype model), while patients carrying the variant allele ABCC4-rs868853 (β = 0.177, p = 0.011) or NR1I2-rs6785049 (β = 0.123, p = 0.044) had higher concentrations of 2-AT+2-ATL in plasma compared with homozygous wildtype carriers. Luciferase activity was enhanced in HepG2 cells harboring a construct expressing the rs3742106-T allele or the rs868853-G allele (p < 0.05 for each) compared with a construct expressing the rs3742106G or the rs868853-A allele. These findings suggest that two functional polymorphisms in the ABCC4 gene may affect transcriptional activity, thereby directly or indirectly affecting release of AT and its metabolites from hepatocytes into the circulation.
Collapse
Affiliation(s)
- Zebin Jiang
- Clinical Pharmacology Laboratory, First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Zemin Wu
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Ruixue Liu
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Qin Du
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Xian Fu
- Clinical Pharmacology Laboratory, First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Min Li
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Yongjun Kuang
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Shen Lin
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Jiaxuan Wu
- Department of Anesthesiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Weiji Xie
- Department of Nephrology, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Ganggang Shi
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Yanqiang Peng
- Department of Nephrology, First Affiliated Hospital of Shantou University Medical College, Shantou, China
- *Correspondence: Fuchun Zheng, ; Yanqiang Peng,
| | - Fuchun Zheng
- Clinical Pharmacology Laboratory, First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Pharmacology, Shantou University Medical College, Shantou, China
- *Correspondence: Fuchun Zheng, ; Yanqiang Peng,
| |
Collapse
|
36
|
Kaci H, Bodnárová S, Fliszár-Nyúl E, Lemli B, Pelantová H, Valentová K, Bakos É, Özvegy-Laczka C, Poór M. Interaction of luteolin, naringenin, and their sulfate and glucuronide conjugates with human serum albumin, cytochrome P450 (CYP2C9, CYP2C19, and CYP3A4) enzymes and organic anion transporting polypeptide (OATP1B1 and OATP2B1) transporters. Biomed Pharmacother 2023; 157:114078. [PMID: 36481402 DOI: 10.1016/j.biopha.2022.114078] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/28/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Luteolin and naringenin are flavonoids found in various foods/beverages and present in certain dietary supplements. After a high intake of these flavonoids, their sulfate and glucuronide conjugates reach micromolar concentrations in the bloodstream. Some pharmacokinetic interactions of luteolin and naringenin have been investigated in previous studies; however, only limited data are available in regard to their metabolites. In this study, we aimed to investigate the interactions of the sulfate and glucuronic acid conjugates of luteolin and naringenin with human serum albumin, cytochrome P450 (CYP2C9, 2C19, and 3A4) enzymes, and organic anion transporting polypeptide (OATP1B1 and OATP2B1) transporters. Our main findings are as follows: (1) Sulfate conjugates formed more stable complexes with albumin than the parent flavonoids. (2) Luteolin and naringenin conjugates showed no or only weak inhibitory action on the CYP enzymes examined. (3) Certain conjugates of luteolin and naringenin are potent inhibitors of OATP1B1 and/or OATP2B1 enzymes. (4) Conjugated metabolites of luteolin and naringenin may play an important role in the pharmacokinetic interactions of these flavonoids.
Collapse
Affiliation(s)
- Hana Kaci
- Drug Resistance Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar tudósok krt. 2., H-1117 Budapest, Hungary; Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, Pázmány P. stny. 1/C, H-1117 Budapest, Hungary
| | - Slávka Bodnárová
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary
| | - Eszter Fliszár-Nyúl
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; Food Biotechnology Research Group, János Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary
| | - Beáta Lemli
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; Green Chemistry Research Group, János Szentágothai Research Center, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary
| | - Helena Pelantová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-142 20 Prague, Czech Republic
| | - Kateřina Valentová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-142 20 Prague, Czech Republic
| | - Éva Bakos
- Drug Resistance Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar tudósok krt. 2., H-1117 Budapest, Hungary
| | - Csilla Özvegy-Laczka
- Drug Resistance Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar tudósok krt. 2., H-1117 Budapest, Hungary
| | - Miklós Poór
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; Food Biotechnology Research Group, János Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary.
| |
Collapse
|
37
|
Pivarcsik T, Pósa V, Kovács H, May NV, Spengler G, Pósa SP, Tóth S, Nezafat Yazdi Z, Özvegy-Laczka C, Ugrai I, Szatmári I, Szakács G, Enyedy ÉA. Metal Complexes of a 5-Nitro-8-Hydroxyquinoline-Proline Hybrid with Enhanced Water Solubility Targeting Multidrug Resistant Cancer Cells. Int J Mol Sci 2022; 24:ijms24010593. [PMID: 36614037 PMCID: PMC9820345 DOI: 10.3390/ijms24010593] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Multidrug resistance (MDR) in cancer is one of the major obstacles of chemotherapy. We have recently identified a series of 8-hydroxyquinoline Mannich base derivatives with MDR-selective toxicity, however with limited solubility. In this work, a novel 5-nitro-8-hydroxyquinoline-proline hybrid and its Rh(η5-C5Me5) and Ru(η6-p-cymene) complexes with excellent aqueous solubility were developed, characterized, and tested against sensitive and MDR cells. Complex formation of the ligand with essential metal ions was also investigated using UV-visible, circular dichroism, 1H NMR (Zn(II)), and electron paramagnetic resonance (Cu(II)) spectroscopic methods. Formation of mono and bis complexes was found in all cases with versatile coordination modes, while tris complexes were also formed with Fe(II) and Fe(III) ions, revealing the metal binding affinity of the ligand at pH 7.4: Cu(II) > Zn(II) > Fe(II) > Fe(III). The ligand and its Rh(III) complex displayed enhanced cytotoxicity against the resistant MES-SA/Dx5 and Colo320 human cancer cell lines compared to their chemosensitive counterparts. Both organometallic complexes possess high stability in solution, however the Ru(II) complex has lower chloride ion affinity and slower ligand exchange processes, along with the readiness to lose the arene ring that is likely connected to its inactivity.
Collapse
Affiliation(s)
- Tamás Pivarcsik
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
| | - Vivien Pósa
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
| | - Hilda Kovács
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
| | - Nóra V. May
- Centre for Structural Science, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
| | - Gabriella Spengler
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Szonja P. Pósa
- Drug Resistance Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
- National Laboratory for Drug Research and Development, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
| | - Szilárd Tóth
- Drug Resistance Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
- National Laboratory for Drug Research and Development, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
| | - Zeinab Nezafat Yazdi
- Drug Resistance Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
| | - Csilla Özvegy-Laczka
- Drug Resistance Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
| | - Imre Ugrai
- Institute of Pharmaceutical Chemistry and Stereochemistry Research Group, Eötvös Loránd Research Network, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - István Szatmári
- Institute of Pharmaceutical Chemistry and Stereochemistry Research Group, Eötvös Loránd Research Network, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - Gergely Szakács
- Drug Resistance Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
- Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Éva A. Enyedy
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
- Correspondence:
| |
Collapse
|
38
|
Nies AT, Schaeffeler E, Schwab M. Hepatic solute carrier transporters and drug therapy: Regulation of expression and impact of genetic variation. Pharmacol Ther 2022; 238:108268. [DOI: 10.1016/j.pharmthera.2022.108268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/25/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022]
|
39
|
Morais MB, Machado MV. Benign inheritable disorders of bilirubin metabolism manifested by conjugated hyperbilirubinemia-A narrative review. United European Gastroenterol J 2022; 10:745-753. [PMID: 35860851 PMCID: PMC9486497 DOI: 10.1002/ueg2.12279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/30/2022] [Indexed: 11/08/2022] Open
Abstract
Bilirubin, a breakdown product of heme, is normally glucuronidated and excreted by the liver into bile. Failure of this system can lead to a buildup of conjugated bilirubin in the blood, resulting in jaundice. Hyperbilirubinemia is an important clinical sign that needs to be investigated under a stepwise evaluation. Inherited non-hemolytic conjugated hyperbilirubinemic conditions include Dubin-Johnson syndrome (caused by mutations affecting ABCC2 gene) and Rotor syndrome (caused by the simultaneous presence of mutations in SLCO1B1 and SLCO1B3 genes). Although classically viewed as benign conditions requiring no treatment, they lately gained an increased interest since recent studies suggested that mutations in the responsible genes leading to hyperbilirubinemia, as well as minor genetic variants, may result in an increased susceptibility to drug toxicity. This article provides a comprehensive review on the pathophysiology of Dubin-Johnson and Rotor syndromes, presenting the current knowledge concerning the molecular details and basis of these conditions.
Collapse
Affiliation(s)
- Mariana B Morais
- Centro Hospitalar Universitário Lisboa Norte, Hospital de Santa Maria, Lisbon, Portugal
| | - Mariana Verdelho Machado
- Gastroenterology Department, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Gastroenterology Department, Hospital de Vila Franca de Xira, Lisbon, Portugal
| |
Collapse
|
40
|
Hove VN, Anderson K, Hayden ER, Pasquariello KZ, Gibson AA, Shen S, Qu J, Jin Y, Miecznikowski JC, Hu S, Sprowl JA. Influence of Tyrosine Kinase Inhibition on Organic Anion Transporting Polypeptide 1B3-Mediated Uptake. Mol Pharmacol 2022; 101:381-389. [PMID: 35383108 PMCID: PMC9354029 DOI: 10.1124/molpharm.121.000287] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 03/22/2022] [Indexed: 11/22/2022] Open
Abstract
The organic anion transporting polypeptide family member (OATP) 1B3 is a hepatic uptake transporter that has a broad substrate recognition and plays a significant role in regulating elimination of endogenous biomolecules or xenobiotics. OATP1B3 works in tandem with OATP1B1, with which it shares approximately 80% sequence homology and a high degree of substrate overlap. Despite some substrates being recognized solely by OATP1B3, its ability to compensate for loss of OATP1B1-mediated elimination and recognition by regulatory agencies, little is known about OATP1B3 regulatory factors and how they are involved with drug-drug interaction. It was recently discovered that OATP1B1 function is mediated by the activity of a particular tyrosine kinase that is sensitive to a variety of tyrosine kinase inhibitors (TKIs). This study reports that OATP1B3 is similarly regulated, as at least 50% of its activity is reduced by 20 US Food and Drug Administration -approved TKIs. Nilotinib was assessed as the most potent OATP1B3 inhibitor among the investigated TKIs, which can occur at clinically relevant concentrations and acted predominantly through noncompetitive inhibition without impacting membrane expression. Finally, OATP1B3 function was determined to be sensitive to the knockdown of the Lck/Yes novel tyrosine kinase that is sensitive to nilotinib and has been previously implicated in mediating OATP1B1 activity. Collectively, our findings identify tyrosine kinase activity as a major regulator of OATP1B3 function which is sensitive to kinase inhibition. Given that OATP1B1 is similarly regulated, simultaneous disruption of these transporters can have drastic effects on systemic drug concentrations, which would promote adverse events. SIGNIFICANCE STATEMENT: The organic anion transporting polypeptide family member (OATP) 1B3 is a facilitator of hepatic drug elimination, although much is unknown of how OATP1B3 activity is mediated, or how such regulators contribute to drug-drug interactions. This study reports that OATP1B3 activity is dependent on the Lck/Yes novel tyrosine kinase, which is sensitive to numerous tyrosine kinase inhibitors. These findings provide insight into the occurrence of many clinical drug-drug interactions, and a rationale for future study of tyrosine kinases regulating drug disposition.
Collapse
Affiliation(s)
- Vusumuzi N Hove
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Kenneth Anderson
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Elizabeth R Hayden
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Kyle Z Pasquariello
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Alice A Gibson
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Shichen Shen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Jun Qu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Yan Jin
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Jeffrey C Miecznikowski
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Shuiying Hu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Jason A Sprowl
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| |
Collapse
|
41
|
Green tea polyphenols in cardiometabolic health: A critical appraisal on phytogenomics towards personalized green tea. PHARMANUTRITION 2022. [DOI: 10.1016/j.phanu.2022.100296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
42
|
Järvinen E, Deng F, Kiander W, Sinokki A, Kidron H, Sjöstedt N. The Role of Uptake and Efflux Transporters in the Disposition of Glucuronide and Sulfate Conjugates. Front Pharmacol 2022; 12:802539. [PMID: 35095509 PMCID: PMC8793843 DOI: 10.3389/fphar.2021.802539] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Glucuronidation and sulfation are the most typical phase II metabolic reactions of drugs. The resulting glucuronide and sulfate conjugates are generally considered inactive and safe. They may, however, be the most prominent drug-related material in the circulation and excreta of humans. The glucuronide and sulfate metabolites of drugs typically have limited cell membrane permeability and subsequently, their distribution and excretion from the human body requires transport proteins. Uptake transporters, such as organic anion transporters (OATs and OATPs), mediate the uptake of conjugates into the liver and kidney, while efflux transporters, such as multidrug resistance proteins (MRPs) and breast cancer resistance protein (BCRP), mediate expulsion of conjugates into bile, urine and the intestinal lumen. Understanding the active transport of conjugated drug metabolites is important for predicting the fate of a drug in the body and its safety and efficacy. The aim of this review is to compile the understanding of transporter-mediated disposition of phase II conjugates. We review the literature on hepatic, intestinal and renal uptake transporters participating in the transport of glucuronide and sulfate metabolites of drugs, other xenobiotics and endobiotics. In addition, we provide an update on the involvement of efflux transporters in the disposition of glucuronide and sulfate metabolites. Finally, we discuss the interplay between uptake and efflux transport in the intestine, liver and kidneys as well as the role of transporters in glucuronide and sulfate conjugate toxicity, drug interactions, pharmacogenetics and species differences.
Collapse
Affiliation(s)
- Erkka Järvinen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Feng Deng
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Wilma Kiander
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Alli Sinokki
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Heidi Kidron
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Noora Sjöstedt
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
43
|
Stalke A, Sgodda M, Cantz T, Skawran B, Lainka E, Hartleben B, Baumann U, Pfister ED. KIF12 Variants and Disturbed Hepatocyte Polarity in Children with a Phenotypic Spectrum of Cholestatic Liver Disease. J Pediatr 2022; 240:284-291.e9. [PMID: 34555379 DOI: 10.1016/j.jpeds.2021.09.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/03/2023]
Abstract
KIF12 has been identified as a cholestasis-associated candidate gene. We describe 6 cases from 4 unrelated families with diverse cholestatic phenotypes carrying 2 different homozygous KIF12 truncating variants. Immunofluorescence investigations of paraffin-embedded liver sections suggest that KIF12-associated impaired functional cell polarity may be the underlying cause.
Collapse
Affiliation(s)
- Amelie Stalke
- Pediatric Gastroenterology and Hepatology, Hannover Medical School, Hannover, Germany; Department of Human Genetics, Hannover Medical School, Hannover, Germany.
| | - Malte Sgodda
- Translational Hepatology and Stem Cell Biology, Department of Gastroenterology, Hepatology and Endocrinology, REBIRTH-Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Tobias Cantz
- Translational Hepatology and Stem Cell Biology, Department of Gastroenterology, Hepatology and Endocrinology, REBIRTH-Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Britta Skawran
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Elke Lainka
- Department for Pediatric Nephrology, Gastroenterology, Endocrinology and Transplant Medicine, University Children's Hospital Essen, Essen, Germany
| | - Björn Hartleben
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Ulrich Baumann
- Pediatric Gastroenterology and Hepatology, Hannover Medical School, Hannover, Germany
| | - Eva-Doreen Pfister
- Pediatric Gastroenterology and Hepatology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
44
|
Han JM, Choi KH, Lee HH, Gwak HS. Association between SLCO1B1 polymorphism and methotrexate-induced hepatotoxicity: a systematic review and meta-analysis. Anticancer Drugs 2022; 33:75-79. [PMID: 34726639 DOI: 10.1097/cad.0000000000001125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Reports on the association between the solute carrier organic anion transporter 1B1 (SLCO1B1) T521C polymorphism and methotrexate-induced hepatotoxicity in patients with malignancies are inconsistent. This meta-analysis evaluated the association between the SLCO1B1 T521C polymorphism and methotrexate-induced hepatotoxicity. We performed a systematic review of previous reports from the PubMed, Web of Science, and EMBASE databases, and a meta-analysis was conducted. Odds ratios (ORs) and 95% confidence intervals (CIs) were estimated to evaluate the effect of the SLCO1B1 T521C polymorphism on the occurrence of methotrexate-induced hepatotoxicity. In total, data from five studies including 465 patients were analyzed. Patients had received a high-dose methotrexate regimen (1-5 g/m2). The SLCO1B1 variant allele (C allele) carriers had a 1.9-fold higher risk of hepatotoxicity than wild-type homozygote carriers (TT; OR, 1.94; 95% CI, 1.14-3.31). This meta-analysis demonstrated that C allele carriers of the SLCO1B1 polymorphism had a higher risk of hepatotoxicity than patients with the TT genotype. The SLCO1B1 T521C polymorphism may be a useful predictor for methotrexate-induced hepatotoxicity in patients with malignancies.
Collapse
Affiliation(s)
- Ji Min Han
- College of Pharmacy, Chungbuk National University, Cheongju-si, Chungcheongbuk-do
| | - Kyung Hee Choi
- College of Pharmacy, Sunchon National University, Suncheon, Jeollanam-do
| | | | - Hye Sun Gwak
- Graduate School of Clinical Biohealth
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
45
|
Traj P, Abdolkhaliq AH, Németh A, Dajcs ST, Tömösi F, Lanisnik-Rizner T, Zupkó I, Mernyák E. Transition metal-catalysed A-ring C-H activations and C(sp 2)-C(sp 2) couplings in the 13α-oestrone series and in vitro evaluation of antiproliferative properties. J Enzyme Inhib Med Chem 2021; 36:895-902. [PMID: 33771084 PMCID: PMC8008932 DOI: 10.1080/14756366.2021.1900165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/23/2021] [Accepted: 03/02/2021] [Indexed: 11/06/2022] Open
Abstract
Facile syntheses of 3-O-carbamoyl, -sulfamoyl, or -pivaloyl derivatives of 13α-oestrone and its 17-deoxy counterpart have been carried out. Microwave-induced, Ni-catalysed Suzuki-Miyaura couplings of the newly synthesised phenol esters with phenylboronic acid afforded 3-deoxy-3-phenyl-13α-oestrone derivatives. The carbamate and pivalate esters proved to be suitable for regioselective arylations. 2-(4-Substituted) phenyl derivatives were synthesised via Pd-catalysed, microwave-assisted C-H activation reactions. An efficient, one-pot, tandem methodology was elaborated for the introduction of the carbamoyl or pivaloyl group followed by regioselective C-2-arylation and subsequent removal of the directing group. The antiproliferative properties of the novel 13α-oestrone derivatives were evaluated in vitro on five human adherent cancer cell lines of gynaecological origin. 3-Sulfamate derivatives displayed substantial cell growth inhibitory potential against certain cell lines. The newly identified antiproliferative compounds having hormonally inactive core might be promising candidates for the design of more active anticancer agents.
Collapse
Affiliation(s)
- Péter Traj
- Department of Organic Chemistry, University of Szeged, Szeged, Hungary
| | | | - Anett Németh
- Department of Organic Chemistry, University of Szeged, Szeged, Hungary
| | | | - Ferenc Tömösi
- Department of Medicinal Chemistry, University of Szeged, Szeged, Hungary
| | - Tea Lanisnik-Rizner
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - István Zupkó
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, Szeged, Hungary
| | - Erzsébet Mernyák
- Department of Organic Chemistry, University of Szeged, Szeged, Hungary
| |
Collapse
|
46
|
Weiss J, Foerster KI, Weber M, Burhenne J, Mikus G, Lehr T, Haefeli WE. Does the circulating ketoconazole metabolite N-deacetyl ketoconazole contribute to the drug-drug interaction potential of the parent compound? Eur J Pharm Sci 2021; 169:106076. [PMID: 34856349 DOI: 10.1016/j.ejps.2021.106076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/21/2021] [Accepted: 11/15/2021] [Indexed: 11/15/2022]
Abstract
Ketoconazole is a strong inhibitor of cytochrome P450 3A4 (CYP3A4) and of P-glycoprotein (P-gp) and is often used as an index inhibitor especially for CYP3A4-mediated drug metabolism. A preliminary physiologically based pharmacokinetic (PBPK) model for drug-drug interactions indicated possible involvement of a metabolite to the perpetrator potential of ketoconazole. Still unknown for humans, in rodents, N-deacetyl ketoconazole (DAK) has been identified as the major ketoconazole metabolite. We therefore investigated in vitro, whether DAK also inhibits the human CYPs and drug transporters targeted by ketoconazole and quantified DAK in human plasma from healthy volunteers after receiving a single oral dose of 400 mg ketoconazole. Our data demonstrated that DAK also inhibits CYP3A4 (2.4-fold less potent than ketoconazole), CYP2D6 (13-fold more potent than ketoconazole), CYP2C19 (equally potent), P-gp (3.4-fold less potent than ketoconazole), breast cancer resistance protein (more potent than ketoconazole) and organic anion transporting polypeptide 1B1 and 1B3 (7.8-fold and 2.6-fold less potent than ketoconazole). After a single oral dose of 400 mg ketoconazole, maximum concentrations of DAK in human plasma were only 3.1 ‰ of the parent compound. However, assuming that DAK also highly accumulates in the human liver as demonstrated for rodents, inhibition of the proteins investigated could also be conceivable in vivo. In conclusion, DAK inhibits several CYPs and drug transporters, which might contribute to the perpetrator potential of ketoconazole.
Collapse
Affiliation(s)
- Johanna Weiss
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; German Center for Infection Research (DZIF), Heidelberg Partner Site, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| | - Kathrin Isabelle Foerster
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; German Center for Infection Research (DZIF), Heidelberg Partner Site, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Maria Weber
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; German Center for Infection Research (DZIF), Heidelberg Partner Site, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Jürgen Burhenne
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; German Center for Infection Research (DZIF), Heidelberg Partner Site, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Gerd Mikus
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; German Center for Infection Research (DZIF), Heidelberg Partner Site, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Thorsten Lehr
- Department of Pharmacy, Clinical Pharmacy, Saarland University, Campus C5 3, 66123, Saarbrücken, Germany
| | - Walter Emil Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; German Center for Infection Research (DZIF), Heidelberg Partner Site, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| |
Collapse
|
47
|
Laczkó-Rigó R, Bakos É, Jójárt R, Tömböly C, Mernyák E, Özvegy-Laczka C. Selective antiproliferative effect of C-2 halogenated 13α-estrones on cells expressing Organic anion-transporting polypeptide 2B1 (OATP2B1). Toxicol Appl Pharmacol 2021; 429:115704. [PMID: 34474082 DOI: 10.1016/j.taap.2021.115704] [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: 04/24/2021] [Revised: 08/06/2021] [Accepted: 08/26/2021] [Indexed: 11/18/2022]
Abstract
Organic anion-transporting polypeptide 2B1 (OATP2B1) is a multispecific transporter mediating the cellular uptake of steroids and numerous drugs. OATP2B1 is abundantly expressed in the intestine and is also present in various tumors. Increased steroid hormone uptake by OATP2B1 has been suggested to promote progression of hormone dependent tumors. 13α-estrones are effective inhibitors of endogenous estrogen formation and are potential candidates to inhibit proliferation of hormone dependent cancers. Recently, we have identified a variety of 13α/β-estrone-based inhibitors of OATP2B1. However, the nature of this interaction, whether these inhibitors are potential transported substrates of OATP2B1 and hence may be enriched in OATP2B1-overexpressing cells, has not yet been investigated. In the current study we explored the antiproliferative effect of the most effective OATP2B1 inhibitor 13α/β-estrones in control and OATP2B1-overexpressing A431 carcinoma cells. We found an increased antiproliferative effect of 3-O-benzyl 13α/β-estrones in both mock transfected and OATP2B1-overexpressing cells. However, C-2 halogenated 13α-estrones had a selective OATP2B1-mediated cell growth inhibitory effect. In order to demonstrate that increased sensitization can be attributed to OATP2B1-mediated cellular uptake, tritium labeled 2-bromo-13α-estrone was synthesized for direct transport measurements. These experiments revealed increased accumulation of [3H]2-bromo-13α-estrone due to OATP2B1 function. Our results indicate that C-2 halogenated 13α-estrones are good candidates in the design of anti-cancer drugs targeting OATP2B1.
Collapse
Affiliation(s)
- Réka Laczkó-Rigó
- Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Center, Magyar tudósok körútja 2, H-1117 Budapest, Hungary.
| | - Éva Bakos
- Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Center, Magyar tudósok körútja 2, H-1117 Budapest, Hungary.
| | - Rebeka Jójárt
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Csaba Tömböly
- Laboratory of Chemical Biology, Institute of Biochemistry, Biological Research Centre, Temesvári krt. 62, H-6726 Szeged, Hungary.
| | - Erzsébet Mernyák
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary.
| | - Csilla Özvegy-Laczka
- Institute of Enzymology, Research Centre for Natural Sciences, Eötvös Loránd Research Center, Magyar tudósok körútja 2, H-1117 Budapest, Hungary.
| |
Collapse
|
48
|
Farasyn T, Pahwa S, Xu C, Yue W. Pre-incubation with OATP1B1 and OATP1B3 inhibitors potentiates inhibitory effects in physiologically relevant sandwich-cultured primary human hepatocytes. Eur J Pharm Sci 2021; 165:105951. [PMID: 34311070 PMCID: PMC11005446 DOI: 10.1016/j.ejps.2021.105951] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/11/2021] [Accepted: 07/08/2021] [Indexed: 12/13/2022]
Abstract
Organic anion transporting polypeptides (OATP)1B1 and OATP1B3 are liver-specific transport proteins that express on the basolateral membrane of human hepatocytes and mediate hepatic uptake of many drugs, including statins. They are important determinants of transporter-mediated drug-drug interactions (DDIs). It has been reported that pre-incubation with some OATP1B1 and OATP1B3 inhibitors potentiates the inhibitory effects, yielding reduced IC50 values. The US FDA draft guidance has recently recommended to use the lower IC50 values after inhibitor-preincubation to assess OATP1B1 and OATP1B3-mediated DDIs. However, it remains unknown whether the potentiation effects of inhibitor-preincubation on IC50 values occur in a physiologically relevant cell model. The current study was designed to determine the IC50 values of OATP1B1 and OATP1B3 inhibitors everolimus (EVR), sirolimus (SIR), and dasatinib against OATP1B substrates in physiologically relevant primary human hepatocytes with or without inhibitor-preincubation and to compare the OATP-mediated DDI prediction using data from primary human hepatocytes and that reported previously in transporter-expressing cell lines. Primary human hepatocytes were cultured in a sandwich configuration. Accumulation of [3H]-CCK-8 (1 µM, 1.5 min), [3H]-rosuvastatin (0.5 µM, 2 min) and [3H]-pitavastatin (1 µM, 0.5 min) was determined in human sandwich-cultured hepatocytes (SCH) in the presence of vehicle control or an inhibitor with or without inhibitor-preincubation at designated concentrations, and was utilized to determine the IC50 values for these inhibitors. R-value models were used to predict OATP-mediated DDIs. Pre-incubation with EVR at a clinically relevant concentration of 0.2 µM significantly reduced accumulation of [3H]-CCK-8 and [3H]-rosuvastatin even after washing. Reduced IC50 values following inhibitor pre-incubation were observed for all three inhibitors using [3H]-CCK-8 and [3H]-rosuvastatin as substrates in human SCH. The IC50 values after inhibitor-preincubation were lower or comparable in transporter-expressing cell lines compared with that in human SCH. For dasatinib, R-values from both cell lines and human SCH were greater than the US FDA cut-off value of 1.1. For EVR, R values from cell lines were 1.23 and were lowered to near 1.1 (1.08-1.09) in human SCH. For SIR, R values from either cell type were less than the cut-off values of 1.1. In conclusion, the current study is the first to report that pre-incubation with OATP1B inhibitors potentiates inhibitory effects in physiologically relevant primary human hepatocytes, supporting the rationale of the current US FDA draft guidance of including an inhibitor-preincubation step when assessing OATP-mediated DDIs in vitro. IC50 values after inhibitor-preincubation in transporter-expressing cell lines may be used for DDI prediction for the purpose of mitigating false-negative OATP-mediated DDI prediction.
Collapse
Affiliation(s)
| | | | - Chao Xu
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK US
| | - Wei Yue
- Department of Pharmaceutical Sciences, US.
| |
Collapse
|
49
|
Effect of type 2 diabetes on Gd-EOB-DTPA uptake into liver parenchyma: replication study in human subjects. Abdom Radiol (NY) 2021; 46:4682-4688. [PMID: 34164726 DOI: 10.1007/s00261-021-03184-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Gadolinium ethoxybenzyl diethylenetriamine pentaacetic acid (Gd-EOB-DTPA) is a contrast agent for magnetic resonance imaging (MRI), which specifically taken up by hepatocytes through organic anion-transporting polypeptides (OATPs). Previous research in mice has shown that type 2 diabetes is associated with reduced uptake of Gd-EOB-DTPA into the liver parenchyma, reflecting reduced expression of OATP. Since considerable differences in OATP expression exist between mice and humans, human studies are necessary to clarify the effect of diabetes to Gd-EOB-DTPA uptake. The purpose of this study was to validate the effect of diabetes to Gd-EOB-DTPA liver uptake by a confirmatory study in humans. METHODS Patients who underwent Gd-EOB-DTPA-enhanced MRI were retrospectively reviewed and divided into two groups: severe or uncontrolled diabetic group (patients with insulin therapy and/or HbA1c ≥ 8.4%) and the control group. Liver-to-spleen ratio (LSR) and relative enhancement of the liver (REL) were calculated to represent Gd-EOB-DTPA liver uptake. RESULTS A total of 94 patients fulfilled the criteria. The severe or uncontrolled diabetic group (n = 15) showed significantly lower LSR (1.74 ± 0.26 vs. 1.98 ± 0.31, p = 0.007) and REL (0.69 ± 0.23 vs. 0.87 ± 0.31, p = 0.005), compared to the control group (n = 79). CONCLUSION Our study revealed decreased uptake of Gd-EOB-DTPA into liver parenchyma in the severe or uncontrolled diabetic patients. Further studies to determine the impact of the reduced liver enhancement on clinical diagnostic practice will be needed.
Collapse
|
50
|
Neuvonen M, Tornio A, Hirvensalo P, Backman JT, Niemi M. Performance of Plasma Coproporphyrin I and III as OATP1B1 Biomarkers in Humans. Clin Pharmacol Ther 2021; 110:1622-1632. [PMID: 34580865 PMCID: PMC9292572 DOI: 10.1002/cpt.2429] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/13/2021] [Indexed: 12/20/2022]
Abstract
A previous study in 356 healthy Finnish volunteers showed that glycochenodeoxycholate 3‐O‐glucuronide (GCDCA‐3G) and glycodeoxycholate 3‐O‐glucuronide (GDCA‐3G) are promising biomarkers of organic anion transporting polypeptide 1B1 (OATP1B1). In the same cohort, we now evaluated the performances of two other OATP1B1 biomarkers, coproporphyrin I (CPI) and III (CPIII), and compared them with GCDCA‐3G and GDCA‐3G. Based on decreased (*5 and *15) and increased (*14 and *20) function SLCO1B1 haplotypes, we stratified the participants to poor, decreased, normal, increased, and highly increased OATP1B1 function groups. Fasting plasma CPI concentration was 68% higher in the poor (95% confidence interval, 44%, 97%; P = 1.74 × 10−10), 7% higher in the decreased (0%, 15%; P = 0.0385), 10% lower in the increased (3%, 18%; P = 0.0087), and 23% lower in the highly increased (1%, 40%; P = 0.0387) function group than in the normal function group. CPIII concentration was 27% higher (7%, 51%; P = 0.0071) in the poor function group than in the normal function group. CPI and CPIII detected poor OATP1B1 function with areas under the precision‐recall curve (AUPRC) of 0.388 (95% confidence interval, 0.197, 0.689) and 0.0798 (0.0485, 0.203), and receiver operating characteristic curve (AUROC) of 0.888 (0.851, 0.919) and 0.731 (0.682, 0.776). The AUPRC and AUROC of GCDCA‐3G were, however, 0.389 (0.258, 0.563) and 0.100 (−0.0046, 0.204; P = 0.0610) larger than those of CPI, and 0.697 (0.555, 0.831) and 0.257 (0.141, 0.373; P < 0.0001) larger than those of CPIII. In conclusion, these data indicate that plasma CPI outperforms CPIII in detecting altered OATP1B1 function, but GCDCA‐3G is an even more sensitive OATP1B1 biomarker than CPI.
Collapse
Affiliation(s)
- Mikko Neuvonen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - Aleksi Tornio
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Päivi Hirvensalo
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - Janne T Backman
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|