1
|
Zuo Y, Li T, Yang S, Chen X, Tao X, Dong D, Liu F, Zhu Y. Contribution and expression of renal drug transporters in renal cell carcinoma. Front Pharmacol 2025; 15:1466877. [PMID: 40034145 PMCID: PMC11873565 DOI: 10.3389/fphar.2024.1466877] [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: 07/18/2024] [Accepted: 12/23/2024] [Indexed: 03/05/2025] Open
Abstract
Renal cell carcinoma (RCC) is a common substantive tumor. According to incomplete statistics, RCC incidence accounts for approximately 90% of renal malignant tumors, and is the second most prevalent major malignant tumor in the genitourinary system, following bladder cancer. Only 10%-15% of chemotherapy regimens for metastatic renal cell carcinoma (mRCC) are effective, and mRCC has a high mortality. Drug transporters are proteins located on the cell membrane that are responsible for the absorption, distribution, and excretion of drugs. Lots of drug transporters are expressed in the kidneys. Changes in carrier function weaken balance, cause disease, or modify the effectiveness of drug treatment. The changes in expression of these transporters during cancer pathology results in multi-drug resistance to cancer chemotherapy. In the treatment of RCC, the study of drug transporters helps to optimize treatment regimens, improve therapeutic effects, and reduce drug side effects. In this review, we summarize advances in the role of renal drug transporters in the genesis, progression, and treatment of RCC.
Collapse
Affiliation(s)
- Yawen Zuo
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tong Li
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shilei Yang
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xuyang Chen
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xufeng Tao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Deshi Dong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fang Liu
- Department of Medical Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yanna Zhu
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
2
|
Dong S, Yang F, Zhang Y, Teng Y, Tang W, Liu J, Fan H. Effect of X-ray irradiation on renal excretion of bestatin through down-regulating organic anion transporters via the vitamin D receptor in rats. Chem Biol Interact 2024; 399:111123. [PMID: 38964638 DOI: 10.1016/j.cbi.2024.111123] [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: 01/14/2024] [Revised: 06/14/2024] [Accepted: 06/27/2024] [Indexed: 07/06/2024]
Abstract
Pharmacokinetic changes induced by radiation following radiotherapy ("RT-PK" phenomenon) are of great significance to the effectiveness and safety of chemotherapeutic agents in clinical settings. The aims of this study were to clarify the organic anion transporters (Oats) involved in the "RT-PK" phenomenon of bestatin in rats following X-ray irradiation and to elucidate its potential mechanism via vitamin D signalling. Pharmacokinetic studies, uptake assays using rat kidney slices and primary proximal tubule cells, and molecular biological studies were performed. Significantly increased plasma concentrations and systemic exposure to bestatin were observed at 24 and 48 h following abdominal X-ray irradiation, regardless of oral or intravenous administration of the drugs in rats. Reduced renal clearance and cumulative urinary excretion of bestatin were observed at 24 and 48 h post-irradiation in rats following intravenous administration. The uptake of the probe substrates p-aminohippuric acid and oestrone 3-sulfate sodium in vitro and the expression of Oat1 and Oat3 in vivo were reduced in the corresponding models following irradiation. Moreover, the upregulation of the vitamin D receptor (Vdr) in mRNA and protein levels negatively correlated with the expressions and functions of Oat1 and Oat3 following irradiation. Additionally, elevated plasma urea nitrogen levels and histopathological changes were observed in rats after exposure to irradiation. The "RT-PK" phenomenon of bestatin occurs in rats after exposure to irradiation, possibly resulting in the regulation of the expressions and activities of renal Oats via activation of the Vdr signalling pathway.
Collapse
Affiliation(s)
- Shiqi Dong
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 238, Baidi Road, Tianjin, 300192, China
| | - Fanlong Yang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 238, Baidi Road, Tianjin, 300192, China
| | - Yufeng Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Yunhua Teng
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 238, Baidi Road, Tianjin, 300192, China
| | - Weisheng Tang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 238, Baidi Road, Tianjin, 300192, China
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 238, Baidi Road, Tianjin, 300192, China
| | - Huirong Fan
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 238, Baidi Road, Tianjin, 300192, China.
| |
Collapse
|
3
|
Luo Y, Gao J, Jiang X, Zhu L, Zhou QT, Murray M, Li J, Zhou F. Molecular Insights to the Structure-Interaction Relationships of Human Proton-Coupled Oligopeptide Transporters (PepTs). Pharmaceutics 2023; 15:2517. [PMID: 37896276 PMCID: PMC10609898 DOI: 10.3390/pharmaceutics15102517] [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: 08/29/2023] [Revised: 10/06/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Human proton-coupled oligopeptide transporters (PepTs) are important membrane influx transporters that facilitate the cellular uptake of many drugs including ACE inhibitors and antibiotics. PepTs mediate the absorption of di- and tri-peptides from dietary proteins or gastrointestinal secretions, facilitate the reabsorption of peptide-bound amino acids in the kidney, and regulate neuropeptide homeostasis in extracellular fluids. PepT1 and PepT2 have been the most intensively investigated of all PepT isoforms. Modulating the interactions of PepTs and their drug substrates could influence treatment outcomes and adverse effects with certain therapies. In recent studies, topology models and protein structures of PepTs have been developed. The aim of this review was to summarise the current knowledge regarding structure-interaction relationships (SIRs) of PepTs and their substrates as well as the potential applications of this information in therapeutic optimisation and drug development. Such information may provide insights into the efficacy of PepT drug substrates in patients, mechanisms of drug-drug/food interactions and the potential role of PepTs targeting in drug design and development strategies.
Collapse
Affiliation(s)
- Yining Luo
- Molecular Drug Development Group, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia; (Y.L.); (J.G.); (M.M.)
| | - Jingchun Gao
- Molecular Drug Development Group, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia; (Y.L.); (J.G.); (M.M.)
| | - Xukai Jiang
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, China;
| | - Ling Zhu
- Macular Research Group, Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia;
| | - Qi Tony Zhou
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA;
| | - Michael Murray
- Molecular Drug Development Group, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia; (Y.L.); (J.G.); (M.M.)
| | - Jian Li
- Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne 3800, Australia;
| | - Fanfan Zhou
- Molecular Drug Development Group, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia; (Y.L.); (J.G.); (M.M.)
| |
Collapse
|
4
|
Chen X, Xie M, Zhang S, Monguió-Tortajada M, Yin J, Liu C, Zhang Y, Delacrétaz M, Song M, Wang Y, Dong L, Ding Q, Zhou B, Tian X, Deng H, Xu L, Liu X, Yang Z, Chang Q, Na J, Zeng W, Superti-Furga G, Rebsamen M, Yang M. Structural basis for recruitment of TASL by SLC15A4 in human endolysosomal TLR signaling. Nat Commun 2023; 14:6627. [PMID: 37863913 PMCID: PMC10589346 DOI: 10.1038/s41467-023-42210-9] [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: 03/24/2023] [Accepted: 10/04/2023] [Indexed: 10/22/2023] Open
Abstract
Toll-like receptors (TLRs) are a class of proteins that play critical roles in recognizing pathogens and initiating innate immune responses. TASL, a recently identified innate immune adaptor protein for endolysosomal TLR7/8/9 signaling, is recruited by the lysosomal proton-coupled amino-acid transporter SLC15A4, and then activates IRF5, which in turn triggers the transcription of type I interferons and cytokines. Here, we report three cryo-electron microscopy (cryo-EM) structures of human SLC15A4 in the apo monomeric and dimeric state and as a TASL-bound complex. The apo forms are in an outward-facing conformation, with the dimeric form showing an extensive interface involving four cholesterol molecules. The structure of the TASL-bound complex reveals an unprecedented interaction mode with solute carriers. During the recruitment of TASL, SLC15A4 undergoes a conformational change from an outward-facing, lysosomal lumen-exposed state to an inward-facing state to form a binding pocket, allowing the N-terminal helix of TASL to be inserted into. Our findings provide insights into the molecular basis of regulatory switch involving a human solute carrier and offers an important framework for structure-guided drug discovery targeting SLC15A4-TASL-related human autoimmune diseases.
Collapse
Affiliation(s)
- Xudong Chen
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Min Xie
- School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Sensen Zhang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | | | - Jian Yin
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Chang Liu
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, 100084, Beijing, China
- Beijing Life Science Academy, 102209, Beijing, China
| | - Youqi Zhang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, 100853, Beijing, China
| | - Maeva Delacrétaz
- Department of Immunobiology, University of Lausanne, 1066, Epalinges, Switzerland
| | - Mingyue Song
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Yixue Wang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Lin Dong
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Qiang Ding
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Boda Zhou
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, 102218, Beijing, China
| | - Xiaolin Tian
- MOE Key laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Haiteng Deng
- MOE Key laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Lina Xu
- Metabolomics and Lipidomics Center at Tsinghua-National Protein Science Facility, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Xiaohui Liu
- Metabolomics and Lipidomics Center at Tsinghua-National Protein Science Facility, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Zi Yang
- Beijing Advanced Innovation Center for Structural Biology, Technology for Protein Research, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Qing Chang
- Beijing Advanced Innovation Center for Structural Biology, Technology for Protein Research, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Jie Na
- School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Wenwen Zeng
- School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Manuele Rebsamen
- Department of Immunobiology, University of Lausanne, 1066, Epalinges, Switzerland
| | - Maojun Yang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, 100084, Beijing, China.
- Cryo-EM Facility Center, Southern University of Science & Technology, 518055, Shenzhen, Guangdong, China.
| |
Collapse
|
5
|
Aleti G, Troyer EA, Hong S. G protein-coupled receptors: A target for microbial metabolites and a mechanistic link to microbiome-immune-brain interactions. Brain Behav Immun Health 2023; 32:100671. [PMID: 37560037 PMCID: PMC10407893 DOI: 10.1016/j.bbih.2023.100671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 07/30/2023] [Indexed: 08/11/2023] Open
Abstract
Human-microorganism interactions play a key role in human health. However, the underlying molecular mechanisms remain poorly understood. Small-molecules that offer a functional readout of microbe-microbe-human relationship are of great interest for deeper understanding of the inter-kingdom crosstalk at the molecular level. Recent studies have demonstrated that small-molecules from gut microbiota act as ligands for specific human G protein-coupled receptors (GPCRs) and modulate a range of human physiological functions, offering a mechanistic insight into the microbe-human interaction. To this end, we focused on analysis of bacterial metabolites that are currently recognized to bind to GPCRs and are found to activate the known downstream signaling pathways. We further mapped the distribution of these molecules across the public mass spectrometry-based metabolomics data, to identify the presence of these molecules across body sites and their association with health status. By combining this with RNA-Seq expression and spatial localization of GPCRs from a public human protein atlas database, we inferred the most predominant GPCR-mediated microbial metabolite-human cell interactions regulating gut-immune-brain axis. Furthermore, by evaluating the intestinal absorption properties and blood-brain barrier permeability of the small-molecules we elucidated their molecular interactions with specific human cell receptors, particularly expressed on human intestinal epithelial cells, immune cells and the nervous system that are shown to hold much promise for clinical translational potential. Furthermore, we provide an overview of an open-source resource for simultaneous interrogation of bioactive molecules across the druggable human GPCRome, a useful framework for integration of microbiome and metabolite cataloging with mechanistic studies for an improved understanding of gut microbiota-immune-brain molecular interactions and their potential therapeutic use.
Collapse
Affiliation(s)
- Gajender Aleti
- Department of Food and Animal Sciences, Tennessee State University, Nashville, TN, 37209, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Emily A. Troyer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Suzi Hong
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
6
|
Bhatt M, Di Iacovo A, Romanazzi T, Roseti C, Cinquetti R, Bossi E. The "www" of Xenopus laevis Oocytes: The Why, When, What of Xenopus laevis Oocytes in Membrane Transporters Research. MEMBRANES 2022; 12:membranes12100927. [PMID: 36295686 PMCID: PMC9610376 DOI: 10.3390/membranes12100927] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 05/16/2023]
Abstract
After 50 years, the heterologous expression of proteins in Xenopus laevis oocytes is still essential in many research fields. New approaches and revised protocols, but also classical methods, such as the two-electrode voltage clamp, are applied in studying membrane transporters. New and old methods for investigating the activity and the expression of Solute Carriers (SLC) are reviewed, and the kinds of experiment that are still useful to perform with this kind of cell are reported. Xenopus laevis oocytes at the full-grown stage have a highly efficient biosynthetic apparatus that correctly targets functional proteins at the defined compartment. This small protein factory can produce, fold, and localize almost any kind of wild-type or recombinant protein; some tricks are required to obtain high expression and to verify the functionality. The methodologies examined here are mainly related to research in the field of membrane transporters. This work is certainly not exhaustive; it has been carried out to be helpful to researchers who want to quickly find suggestions and detailed indications when investigating the functionality and expression of the different members of the solute carrier families.
Collapse
Affiliation(s)
- Manan Bhatt
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, Via Dunant 3, 21100 Varese, Italy
- Experimental and Translational Medicine, University of Insubria, Via Ottorino Rossi 9, 21100 Varese, Italy
| | - Angela Di Iacovo
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, Via Dunant 3, 21100 Varese, Italy
- Experimental and Translational Medicine, University of Insubria, Via Ottorino Rossi 9, 21100 Varese, Italy
| | - Tiziana Romanazzi
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, Via Dunant 3, 21100 Varese, Italy
- Experimental and Translational Medicine, University of Insubria, Via Ottorino Rossi 9, 21100 Varese, Italy
| | - Cristina Roseti
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, Via Dunant 3, 21100 Varese, Italy
- Centre for Neuroscience—Via Manara 7, University of Insubria, 21052 Busto Arsizio, Italy
| | - Raffaella Cinquetti
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, Via Dunant 3, 21100 Varese, Italy
| | - Elena Bossi
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, Via Dunant 3, 21100 Varese, Italy
- Centre for Neuroscience—Via Manara 7, University of Insubria, 21052 Busto Arsizio, Italy
- Correspondence:
| |
Collapse
|
7
|
Wang C, Chu C, Ji X, Luo G, Xu C, He H, Yao J, Wu J, Hu J, Jin Y. Biology of Peptide Transporter 2 in Mammals: New Insights into Its Function, Structure and Regulation. Cells 2022; 11:cells11182874. [PMID: 36139448 PMCID: PMC9497230 DOI: 10.3390/cells11182874] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
Peptide transporter 2 (PepT2) in mammals plays essential roles in the reabsorption and conservation of peptide-bound amino acids in the kidney and in maintaining neuropeptide homeostasis in the brain. It is also of significant medical and pharmacological significance in the absorption and disposing of peptide-like drugs, including angiotensin-converting enzyme inhibitors, β-lactam antibiotics and antiviral prodrugs. Understanding the structure, function and regulation of PepT2 is of emerging interest in nutrition, medical and pharmacological research. In this review, we provide a comprehensive overview of the structure, substrate preferences and localization of PepT2 in mammals. As PepT2 is expressed in various organs, its function in the liver, kidney, brain, heart, lung and mammary gland has also been addressed. Finally, the regulatory factors that affect the expression and function of PepT2, such as transcriptional activation and posttranslational modification, are also discussed.
Collapse
Affiliation(s)
- Caihong Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- Zhejiang Conba Pharmaceutical Limited Company, Hangzhou 310052, China
| | - Chu Chu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Xiang Ji
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Guoliang Luo
- Zhejiang Conba Pharmaceutical Limited Company, Hangzhou 310052, China
- Zhejiang Institute of Modern Chinese Medicine and Natural Medicine, Hangzhou 310052, China
| | - Chunling Xu
- Zhejiang Conba Pharmaceutical Limited Company, Hangzhou 310052, China
- Zhejiang Institute of Modern Chinese Medicine and Natural Medicine, Hangzhou 310052, China
| | - Houhong He
- Zhejiang Conba Pharmaceutical Limited Company, Hangzhou 310052, China
- Zhejiang Institute of Modern Chinese Medicine and Natural Medicine, Hangzhou 310052, China
| | - Jianbiao Yao
- Zhejiang Conba Pharmaceutical Limited Company, Hangzhou 310052, China
- Zhejiang Institute of Modern Chinese Medicine and Natural Medicine, Hangzhou 310052, China
| | - Jian Wu
- Zhejiang Conba Pharmaceutical Limited Company, Hangzhou 310052, China
- Zhejiang Institute of Modern Chinese Medicine and Natural Medicine, Hangzhou 310052, China
| | - Jiangning Hu
- Zhejiang Conba Pharmaceutical Limited Company, Hangzhou 310052, China
- Zhejiang Institute of Modern Chinese Medicine and Natural Medicine, Hangzhou 310052, China
- Correspondence: (J.H.); (Y.J.)
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
- Correspondence: (J.H.); (Y.J.)
| |
Collapse
|
8
|
Sala-Rabanal M. Peptide transporters in teleost fish: a look into the distant past for a more sustainable future. J Physiol 2022; 600:3015. [PMID: 35653271 DOI: 10.1113/jp283171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Monica Sala-Rabanal
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, Missouri, 63110, United States of America
| |
Collapse
|
9
|
Vacca F, Gomes AS, Murashita K, Cinquetti R, Roseti C, Barca A, Rønnestad I, Verri T, Bossi E. Functional characterization of Atlantic salmon (Salmo salar L.) PepT2 transporters. J Physiol 2022; 600:2377-2400. [PMID: 35413133 PMCID: PMC9321897 DOI: 10.1113/jp282781] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/16/2022] [Indexed: 11/24/2022] Open
Abstract
Abstract The high‐affinity/low‐capacity system Slc15a2 (PepT2) is responsible for the reuptake of di/tripeptides from the renal proximal tubule, but it also operates in many other tissues and organs. Information regarding PepT2 in teleost fish is limited and, to date, functional data are available from the zebrafish (Danio rerio) only. Here, we report the identification of two slc15a2 genes in the Atlantic salmon (Salmo salar) genome, namely slc15a2a and slc15a2b. The two encoded PepT2 proteins share 87% identity and resemble both structurally and functionally the canonical vertebrate PepT2 system. The mRNA tissue distribution analyses reveal a widespread distribution of slc15a2a transcripts, being more abundant in the brain and gills, while slc15a2b transcripts are mainly expressed in the kidney and the distal part of the gastrointestinal tract. The function of the two transporters was investigated by heterologous expression in Xenopus laevis oocytes and two‐electrode voltage‐clamp recordings of transport and presteady‐state currents. Both PepT2a and PepT2b in the presence of Gly‐Gln elicit pH‐dependent and Na+ independent inward currents. The biophysical and kinetic analysis of the recorded currents defined the transport properties, confirming that the two Atlantic salmon PepT2 proteins behave as high‐affinity/low‐capacity transporters. The recent structures and the previous kinetic schemes of rat and human PepT2 qualitatively account for the characteristics of the two Atlantic salmon proteins. This study is the first to report on the functional expression of two PepT2‐type transporters that operate in the same vertebrate organism as a result of (a) gene duplication process(es). Key points Two slc15a2‐type genes, slc15a2a and slc15a2b coding for PepT2‐type peptide transporters were found in the Atlantic salmon. slc15a2a
transcripts, widely distributed in the fish tissues, are abundant in the brain and gills, while slc15a2b transcripts are mainly expressed in the kidney and distal gastrointestinal tract. Amino acids involved in vertebrate Slc15 transport function are conserved in PepT2a and PepT2b proteins. Detailed kinetic analysis indicates that both PepT2a and PepT2b operate as high‐affinity transporters. The kinetic schemes and structures proposed for the mammalian models of PepT2 are suitable to explain the function of the two Atlantic salmon transporters.
Collapse
Affiliation(s)
- Francesca Vacca
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, via Dunant 3, Varese, I-21100, Italy
| | - Ana S Gomes
- Department of Biological Sciences, University of Bergen, Po. Box 7803, Bergen, NO-5020, Norway
| | - Koji Murashita
- Research Center for Aquaculture Systems, National Research Institute of Aquaculture, Japan Fisheries Research and Education Agency, Minami-ise, Mie, 516-0193, Japan
| | - Raffella Cinquetti
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, via Dunant 3, Varese, I-21100, Italy
| | - Cristina Roseti
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, via Dunant 3, Varese, I-21100, Italy
| | - Amilcare Barca
- Laboratory of Applied Physiology, Department of Biological and Environmental Sciences and Technologies, University of Salento, via Provinciale Lecce-Monteroni, Lecce, I-73100, Italy
| | - Ivar Rønnestad
- Department of Biological Sciences, University of Bergen, Po. Box 7803, Bergen, NO-5020, Norway
| | - Tiziano Verri
- Laboratory of Applied Physiology, Department of Biological and Environmental Sciences and Technologies, University of Salento, via Provinciale Lecce-Monteroni, Lecce, I-73100, Italy
| | - Elena Bossi
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, via Dunant 3, Varese, I-21100, Italy
| |
Collapse
|
10
|
Robison SW, Li J, Viera L, Blackburn JP, Patel RP, Blalock JE, Gaggar A, Xu X. A mechanism for matrikine regulation in acute inflammatory lung injury. JCI Insight 2021; 6:140750. [PMID: 33830084 PMCID: PMC8119180 DOI: 10.1172/jci.insight.140750] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
Proline-glycine-proline (PGP) and its acetylated form (Ac-PGP) are neutrophil chemoattractants generated by collagen degradation, and they have been shown to play a role in chronic inflammatory disease. However, the mechanism for matrikine regulation in acute inflammation has not been well established. Here, we show that these peptides are actively transported from the lung by the oligopeptide transporter, PEPT2. Following intratracheal instillation of Ac-PGP in a mouse model, there was a rapid decline in concentration of the labeled peptide in the bronchoalveolar lavage (BAL) over time and redistribution to extrapulmonary sites. In vitro knockdown of the PEPT2 transporter in airway epithelia or use of a competitive inhibitor of PEPT2, cefadroxil, significantly reduced uptake of Ac-PGP. Animals that received intratracheal Ac-PGP plus cefadroxil had higher levels of Ac-PGP in BAL and lung tissue. Utilizing an acute LPS-induced lung injury model, we demonstrate that PEPT2 blockade enhanced pulmonary Ac-PGP levels and lung inflammation. We further validated this effect using clinical samples from patients with acute lung injury in coculture with airway epithelia. This is the first study to our knowledge to determine the in vitro and in vivo significance of active matrikine transport as a mechanism of modulating acute inflammation and to demonstrate that it may serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Sarah W Robison
- Department of Medicine, Division of Pulmonology, Allergy and Critical Care Medicine, and.,Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - JinDong Li
- Department of Medicine, Division of Pulmonology, Allergy and Critical Care Medicine, and.,Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Birmingham VA Medical Center, Birmingham, Alabama, USA
| | - Liliana Viera
- Department of Medicine, Division of Pulmonology, Allergy and Critical Care Medicine, and.,Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jonathan P Blackburn
- Department of Medicine, Division of Pulmonology, Allergy and Critical Care Medicine, and.,Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rakesh P Patel
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of Pathology, Division of Molecular and Cellular Pathology, and.,Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - J Edwin Blalock
- Department of Medicine, Division of Pulmonology, Allergy and Critical Care Medicine, and.,Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Gregory Fleming James Cystic Fibrosis Research Center, Birmingham, Alabama, USA.,Lung Health Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Amit Gaggar
- Department of Medicine, Division of Pulmonology, Allergy and Critical Care Medicine, and.,Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Birmingham VA Medical Center, Birmingham, Alabama, USA.,Gregory Fleming James Cystic Fibrosis Research Center, Birmingham, Alabama, USA.,Lung Health Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Xin Xu
- Department of Medicine, Division of Pulmonology, Allergy and Critical Care Medicine, and.,Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Birmingham VA Medical Center, Birmingham, Alabama, USA.,Gregory Fleming James Cystic Fibrosis Research Center, Birmingham, Alabama, USA.,Lung Health Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
11
|
Recent advances in understanding prodrug transport through the SLC15 family of proton-coupled transporters. Biochem Soc Trans 2021; 48:337-346. [PMID: 32219385 PMCID: PMC7200629 DOI: 10.1042/bst20180302] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/15/2020] [Accepted: 03/04/2020] [Indexed: 12/28/2022]
Abstract
Solute carrier (SLC) transporters play important roles in regulating the movement of small molecules and ions across cellular membranes. In mammals, they play an important role in regulating the uptake of nutrients and vitamins from the diet, and in controlling the distribution of their metabolic intermediates within the cell. Several SLC families also play an important role in drug transport and strategies are being developed to hijack SLC transporters to control and regulate drug transport within the body. Through the addition of amino acid and peptide moieties several novel antiviral and anticancer agents have been developed that hijack the proton-coupled oligopeptide transporters, PepT1 (SCL15A1) and PepT2 (SLC15A2), for improved intestinal absorption and renal retention in the body. A major goal is to understand the rationale behind these successes and expand the library of prodrug molecules that utilise SLC transporters. Recent co-crystal structures of prokaryotic homologues of the human PepT1 and PepT2 transporters have shed important new insights into the mechanism of prodrug recognition. Here, I will review recent developments in our understanding of ligand recognition and binding promiscuity within the SLC15 family, and discuss current models for prodrug recognition.
Collapse
|
12
|
Identification of PEPT2 as an important candidate molecule in 5-ALA-mediated fluorescence-guided surgery in WHO grade II/III gliomas. J Neurooncol 2019; 143:197-206. [PMID: 30929128 DOI: 10.1007/s11060-019-03158-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/25/2019] [Indexed: 10/27/2022]
Abstract
PURPOSE 5-aminolevulinic acid (5-ALA) fluorescence-guided surgery (FGS) appears to be a promising treatment for glioma. However, 5-ALA-mediated fluorescence cannot always be detected in grade II/III gliomas. We hypothesized that gene expression patterns in the Protoporphyrin IX (PpIX) synthesis pathway may be associated with intraoperative fluorescence status of grade II/III gliomas, and then attempted to identify the key molecule of 5-ALA-mediated fluorescence. METHODS Using 50 surgically obtained specimens, which were diagnosed as grade II and III gliomas, we analyzed gene expression within the PpIX synthesis pathway to identify candidate molecules according to intraoperative 5-ALA-mediated fluorescence status. The most likely candidate gene was selected and confirmed by protein expression analysis. To evaluate the biological function of the molecule in PpIX synthesis, functional analysis was performed using specific, small interference (si)RNA in the SW-1783 human grade III glioma cell line. RESULTS Among the genes involved in the porphyrin synthesis pathway, the mRNA expression of Peptide transporter 2 (PEPT2) in FGS fluorescence-positive gliomas was significantly higher than that in fluorescence-negative gliomas. Protein expression of PEPT2 was also significantly higher in the fluorescence-positive gliomas, which was confirmed by western blot analysis and immunofluorescence analysis. The siRNA-mediated downregulation of the mRNA and protein expression of PEPT2 led to decreased PpIX fluorescence intensity, as confirmed by fluorescence spectrum analysis. CONCLUSIONS The results suggest PEPT2 is an important candidate molecule in 5-ALA-mediated FGS in grade II/III gliomas. As the overexpression of PEPT2 was associated with higher PpIX fluorescence intensity, PEPT2 may improve fluorescence-guided resection in grade II/III gliomas.
Collapse
|
13
|
Structural basis for prodrug recognition by the SLC15 family of proton-coupled peptide transporters. Proc Natl Acad Sci U S A 2019; 116:804-809. [PMID: 30602453 PMCID: PMC6338836 DOI: 10.1073/pnas.1813715116] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Poor oral bioavailability is one of the leading causes of compound failure in drug development and a major challenge for the pharmaceutical industry. A successful approach to address this challenge has been the development of prodrugs that target the intestinal peptide transporter, PepT1 (SLC15A1). PepT1 exhibits a remarkably promiscuous binding site and is known to transport many different drug molecules, making it an excellent target for prodrug design and delivery. However, the structural basis for drug recognition remains largely unknown. Here we present the structure of a bacterial homolog of PepT1 bound to both an antiviral prodrug, valacyclovir, and anticancer drug 5-aminolevulinic acid. These structures enable a pharmacophore model to be developed that will aid future prodrug design. A major challenge in drug development is the optimization of intestinal absorption and cellular uptake. A successful strategy has been to develop prodrug molecules, which hijack solute carrier (SLC) transporters for active transport into the body. The proton-coupled oligopeptide transporters, PepT1 and PepT2, have been successfully targeted using this approach. Peptide transporters display a remarkable capacity to recognize a diverse library of di- and tripeptides, making them extremely promiscuous and major contributors to the pharmacokinetic profile of several important drug classes, including beta-lactam antibiotics and antiviral and antineoplastic agents. Of particular interest has been their ability to recognize amino acid and peptide-based prodrug molecules, thereby providing a rational approach to improving drug transport into the body. However, the structural basis for prodrug recognition has remained elusive. Here we present crystal structures of a prokaryotic homolog of the mammalian transporters in complex with the antiviral prodrug valacyclovir and the peptide-based photodynamic therapy agent, 5-aminolevulinic acid. The valacyclovir structure reveals that prodrug recognition is mediated through both the amino acid scaffold and the ester bond, which is commonly used to link drug molecules to the carrier’s physiological ligand, whereas 5-aminolevulinic acid makes far fewer interactions compared with physiological peptides. These structures provide a unique insight into how peptide transporters interact with xenobiotic molecules and provide a template for further prodrug development.
Collapse
|
14
|
Maestri E, Pavlicevic M, Montorsi M, Marmiroli N. Meta-Analysis for Correlating Structure of Bioactive Peptides in Foods of Animal Origin with Regard to Effect and Stability. Compr Rev Food Sci Food Saf 2018; 18:3-30. [PMID: 33337011 DOI: 10.1111/1541-4337.12402] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 09/28/2018] [Accepted: 09/29/2018] [Indexed: 01/09/2023]
Abstract
Amino acid (AA) sequences of 807 bioactive peptides from foods of animal origin were examined in order to correlate peptide structure with activity (antihypertensive, antioxidative, immunomodulatory, antimicrobial, hypolipidemic, antithrombotic, and opioid) and stability in vivo. Food sources, such as milk, meat, eggs, and marine products, show different frequencies of bioactive peptides exhibiting specific effects. There is a correlation of peptide structure and effect, depending on type and position of AA. Opioid peptides contain a high percentage of aromatic AA residues, while antimicrobial peptides show an excess of positively charged AAs. AA residue position is significant, with those in the first and penultimate positions having the biggest effects on peptide activity. Peptides that have activity in vivo contain a high percentage (67%) of proline residues, but the positions of proline in the sequence depend on the length of the peptide. We also discuss the influence of processing on activity of these peptides, as well as methods for predicting release from the source protein and activity of peptides.
Collapse
Affiliation(s)
- Elena Maestri
- Dept. of Chemistry, Life Sciences and Environmental Sustainability, Univ. of Parma, Parco Area delle Scienze 11/A, 43124, Parma, Italy.,Interdepartmental Centre for Food Safety, Technologies and Innovation for Agri-food (SITEIA.PARMA), Univ. of Parma, Parco Area delle Scienze, 43124, Parma, Italy
| | - Milica Pavlicevic
- Inst. for Food Technology and Biochemistry, Faculty of Agriculture, Univ. of Belgrade, Belgrade, Serbia
| | - Michela Montorsi
- Dept. of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open Univ., Via F. Daverio 7, 20122, Milan, Italy.,Consorzio Italbiotec, Via Fantoli, 16/15, 20138, Milano, Italy.,Inst. of Bioimaging and Molecular Physiology, National Council of Research (CNR), Via Fratelli Cervi 93, 20090, Segrate, Italy
| | - Nelson Marmiroli
- Dept. of Chemistry, Life Sciences and Environmental Sustainability, Univ. of Parma, Parco Area delle Scienze 11/A, 43124, Parma, Italy.,Interdepartmental Centre for Food Safety, Technologies and Innovation for Agri-food (SITEIA.PARMA), Univ. of Parma, Parco Area delle Scienze, 43124, Parma, Italy.,Consorzio Italbiotec, Via Fantoli, 16/15, 20138, Milano, Italy
| |
Collapse
|
15
|
Abstract
Transporters in proximal renal tubules contribute to the disposition of numerous drugs. Furthermore, the molecular mechanisms of tubular secretion have been progressively elucidated during the past decades. Organic anions tend to be secreted by the transport proteins OAT1, OAT3 and OATP4C1 on the basolateral side of tubular cells, and multidrug resistance protein (MRP) 2, MRP4, OATP1A2 and breast cancer resistance protein (BCRP) on the apical side. Organic cations are secreted by organic cation transporter (OCT) 2 on the basolateral side, and multidrug and toxic compound extrusion (MATE) proteins MATE1, MATE2/2-K, P-glycoprotein, organic cation and carnitine transporter (OCTN) 1 and OCTN2 on the apical side. Significant drug-drug interactions (DDIs) may affect any of these transporters, altering the clearance and, consequently, the efficacy and/or toxicity of substrate drugs. Interactions at the level of basolateral transporters typically decrease the clearance of the victim drug, causing higher systemic exposure. Interactions at the apical level can also lower drug clearance, but may be associated with higher renal toxicity, due to intracellular accumulation. Whereas the importance of glomerular filtration in drug disposition is largely appreciated among clinicians, DDIs involving renal transporters are less well recognized. This review summarizes current knowledge on the roles, quantitative importance and clinical relevance of these transporters in drug therapy. It proposes an approach based on substrate-inhibitor associations for predicting potential tubular-based DDIs and preventing their adverse consequences. We provide a comprehensive list of known drug interactions with renally-expressed transporters. While many of these interactions have limited clinical consequences, some involving high-risk drugs (e.g. methotrexate) definitely deserve the attention of prescribers.
Collapse
Affiliation(s)
- Anton Ivanyuk
- Division of Clinical Pharmacology, Lausanne University Hospital (CHUV), Bugnon 17, 1011, Lausanne, Switzerland.
| | - Françoise Livio
- Division of Clinical Pharmacology, Lausanne University Hospital (CHUV), Bugnon 17, 1011, Lausanne, Switzerland
| | - Jérôme Biollaz
- Division of Clinical Pharmacology, Lausanne University Hospital (CHUV), Bugnon 17, 1011, Lausanne, Switzerland
| | - Thierry Buclin
- Division of Clinical Pharmacology, Lausanne University Hospital (CHUV), Bugnon 17, 1011, Lausanne, Switzerland
| |
Collapse
|
16
|
Tchernitchko D, Tavernier Q, Lamoril J, Schmitt C, Talbi N, Lyoumi S, Robreau AM, Karim Z, Gouya L, Thervet E, Karras A, Puy H, Pallet N. A Variant of Peptide Transporter 2 Predicts the Severity of Porphyria-Associated Kidney Disease. J Am Soc Nephrol 2016; 28:1924-1932. [PMID: 28031405 DOI: 10.1681/asn.2016080918] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 11/29/2016] [Indexed: 01/15/2023] Open
Abstract
CKD occurs in most patients with acute intermittent porphyria (AIP). During AIP, δ-aminolevulinic acid (ALA) accumulates and promotes tubular cell death and tubulointerstitial damage. The human peptide transporter 2 (PEPT2) expressed by proximal tubular cells mediates the reabsorption of ALA, and variants of PEPT2 have different affinities for ALA. We tested the hypothesis that PEPT2 genotypes affect the severity and prognosis of porphyria-associated kidney disease. We analyzed data from 122 individuals with AIP who were followed from 2003 to 2013 and genotyped for PEPT2 At last follow-up, carriers of the PEPT2*1*1 genotype (higher affinity variant) exhibited worse renal function than carriers of the lower affinity variants PEPT2*1/*2 and PEPT2*2/*2 (mean±SD eGFR: 54.4±19.1, 66.6±23.8, and 78.1±19.9 ml/min per 1.73 m2, respectively). Change in eGFR (mean±SD) over the 10-year period was -11.0±3.3, -2.4±1.9, and 3.4±2.6 ml/min per 1.73 m2 for PEPT2*1/*1, PEPT2*1*2, and PEPT*2*2*2 carriers, respectively. At the end of follow-up, 68% of PEPT2*1*1 carriers had an eGFR<60 ml/min per 1.73 m2, compared with 37% of PEPT2*1*2 carriers and 15% of PEPT2*2*2 carriers. Multiple regression models including all confounders indicated that the PEPT2*1*1 genotype independently associated with an eGFR<60 ml/min per 1.73 m2 (odds ratio, 6.85; 95% confidence interval, 1.34 to 46.20) and an annual decrease in eGFR of >1 ml/min per 1.73 m2 (odds ratio, 3.64; 95% confidence interval, 1.37 to 9.91). Thus, a gene variant is predictive of the severity of a chronic complication of AIP. The therapeutic value of PEPT2 inhibitors in preventing porphyria-associated kidney disease warrants investigation.
Collapse
Affiliation(s)
- Dimitri Tchernitchko
- Centre Français des Porphyries, Hôpital Louis Mourier, Assistance Publique-Hôpitaux de Paris, Colombes, Paris, France.,Institut National pour la Santé et la Recherche Médicale (INSERM) U1149, Center for Research on Inflammation (CRI), Site Bichat, Paris, France
| | - Quentin Tavernier
- Institut National pour la Santé et la Recherche Médicale (INSERM) U1147, Centre Universitaire des Saints Pères, Paris, France.,Université Paris Descartes, Paris, France
| | - Jérôme Lamoril
- Centre Français des Porphyries, Hôpital Louis Mourier, Assistance Publique-Hôpitaux de Paris, Colombes, Paris, France.,Institut National pour la Santé et la Recherche Médicale (INSERM) U1149, Center for Research on Inflammation (CRI), Site Bichat, Paris, France
| | - Caroline Schmitt
- Centre Français des Porphyries, Hôpital Louis Mourier, Assistance Publique-Hôpitaux de Paris, Colombes, Paris, France.,Institut National pour la Santé et la Recherche Médicale (INSERM) U1149, Center for Research on Inflammation (CRI), Site Bichat, Paris, France.,Université Paris Diderot, Paris, France.,Laboratory of Excellence, GR-Ex, Paris, France
| | - Neila Talbi
- Centre Français des Porphyries, Hôpital Louis Mourier, Assistance Publique-Hôpitaux de Paris, Colombes, Paris, France
| | - Said Lyoumi
- Institut National pour la Santé et la Recherche Médicale (INSERM) U1149, Center for Research on Inflammation (CRI), Site Bichat, Paris, France.,Université Versailles Saint Quentin, Versailles, France; and
| | - Anne-Marie Robreau
- Centre Français des Porphyries, Hôpital Louis Mourier, Assistance Publique-Hôpitaux de Paris, Colombes, Paris, France.,Institut National pour la Santé et la Recherche Médicale (INSERM) U1149, Center for Research on Inflammation (CRI), Site Bichat, Paris, France
| | - Zoubida Karim
- Institut National pour la Santé et la Recherche Médicale (INSERM) U1149, Center for Research on Inflammation (CRI), Site Bichat, Paris, France
| | - Laurent Gouya
- Centre Français des Porphyries, Hôpital Louis Mourier, Assistance Publique-Hôpitaux de Paris, Colombes, Paris, France.,Institut National pour la Santé et la Recherche Médicale (INSERM) U1149, Center for Research on Inflammation (CRI), Site Bichat, Paris, France.,Université Paris Diderot, Paris, France.,Laboratory of Excellence, GR-Ex, Paris, France
| | - Eric Thervet
- Institut National pour la Santé et la Recherche Médicale (INSERM) U1147, Centre Universitaire des Saints Pères, Paris, France.,Université Paris Diderot, Paris, France.,Laboratory of Excellence, GR-Ex, Paris, France
| | - Alexandre Karras
- Institut National pour la Santé et la Recherche Médicale (INSERM) U1147, Centre Universitaire des Saints Pères, Paris, France.,Université Paris Diderot, Paris, France.,Laboratory of Excellence, GR-Ex, Paris, France
| | - Hervé Puy
- Centre Français des Porphyries, Hôpital Louis Mourier, Assistance Publique-Hôpitaux de Paris, Colombes, Paris, France.,Institut National pour la Santé et la Recherche Médicale (INSERM) U1149, Center for Research on Inflammation (CRI), Site Bichat, Paris, France.,Université Paris Diderot, Paris, France.,Laboratory of Excellence, GR-Ex, Paris, France
| | - Nicolas Pallet
- Institut National pour la Santé et la Recherche Médicale (INSERM) U1147, Centre Universitaire des Saints Pères, Paris, France; .,Université Paris Descartes, Paris, France.,Service de Néphrologie and.,Service de Biochimie, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
17
|
Abstract
Oligopeptide transporters serve important functions in nutrition and pharmacology. In particular, these transporters help maintain the homeostasis of peptides. The peptide-transporter PEPT2 is a high-affinity and low-capacity type oligopeptide transporter from the proton-coupled oligopeptide transporter family. PEPT2 has recently received attention because of its potential application in targeted drug delivery. PEPT2 is widely distributed in kidney, central nervous system, and lung of organisms. In general, all dipeptides, tripeptides, and peptide-like drugs such as β-lactam antibiotics and angiotensin-converting enzyme inhibitors could be mediated and transported as a substrate of PEPT2. The design of many extant drugs and prodrugs is based on the substrate structure of PEPT2 to accelerate absorption via peptide transporters. Thus, this paper summarizes the substrate features of PEPT2 to promote the rational design of drugs and prodrugs that target peptide transporters.
Collapse
Affiliation(s)
- Dongxin Zhao
- School of Chemistry and Chemical Engineering, Henan University of Technology
| | | |
Collapse
|
18
|
Qiao M, Sanes JR. Genetic Method for Labeling Electrically Coupled Cells: Application to Retina. Front Mol Neurosci 2016; 8:81. [PMID: 26778956 PMCID: PMC4703850 DOI: 10.3389/fnmol.2015.00081] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 12/11/2015] [Indexed: 01/08/2023] Open
Abstract
Understanding how the nervous system functions requires mapping synaptic connections between neurons. Several methods are available for imaging neurons connected by chemical synapses, but few enable marking neurons connected by electrical synapses. Here, we demonstrate that a peptide transporter, Pept2, can be used for this purpose. Pept2 transports a gap junction-permeable fluorophore-coupled dipeptide, beta-alanine-lysine-N-7-amino-4-methyl coumarin-3-acid (βALA). Cre-dependent expression of pept2 in specific neurons followed by incubation in βALA labeled electrically coupled synaptic partners. Using this method, we analyze light-dependent modulation of electrical connectivity among retinal horizontal cells.
Collapse
Affiliation(s)
- Mu Qiao
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge MA, USA
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge MA, USA
| |
Collapse
|
19
|
Takano M, Sugimoto N, Ehrhardt C, Yumoto R. Functional Expression of PEPT2 in the Human Distal Lung Epithelial Cell Line NCl-H441. Pharm Res 2015; 32:3916-26. [PMID: 26168863 DOI: 10.1007/s11095-015-1751-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/06/2015] [Indexed: 12/31/2022]
Abstract
PURPOSE The peptide transporter PEPT2 is expressed in alveolar type II epithelial cells. So far, however, no appropriate alveolar epithelial cell line for studying PEPT2 function has been known. In this study, we examined the functional expression of PEPT2 in the human distal lung epithelial cell line NCl-H441 (H441). METHODS Expression of PEPT2 mRNA and protein was examined in H441 cells. Transport function of PEPT2 was studied using glycylsarcosine (Gly-Sar) as a substrate. RESULTS Lamellar bodies were well developed in H441 cells and mRNA expression of type II cell markers and PEPT2 increased during time in culture. PEPT2 protein expression was confirmed in H441 cells, but not in A549 cells, by immunostaining and Western blotting. The uptake of Gly-Sar in H441 cells was inhibited by cefadroxil, and the cefadroxil-sensitive uptake was pH-dependent and peaked at pH 6.5. Gly-Sar uptake in H441 cells showed saturation kinetics with a Km value of 112.5 μM. In addition, apical-to-basal, but not basal-to-apical, transport of cephalexin across H441 cell monolayers was sensitive to cefadroxil. CONCLUSIONS PEPT2 is functionally expressed in H441 cells, making the cell line a good in vitro model to study PEPT2 function and its regulation in human distal lung.
Collapse
Affiliation(s)
- Mikihisa Takano
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| | - Natsumi Sugimoto
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Carsten Ehrhardt
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Panoz Institute, Dublin 2, Ireland
| | - Ryoko Yumoto
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| |
Collapse
|
20
|
Study of protein haptenation by amoxicillin through the use of a biotinylated antibiotic. PLoS One 2014; 9:e90891. [PMID: 24595455 PMCID: PMC3940954 DOI: 10.1371/journal.pone.0090891] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 02/06/2014] [Indexed: 12/28/2022] Open
Abstract
Allergic reactions towards β-lactam antibiotics pose an important clinical problem. The ability of small molecules, such as a β-lactams, to bind covalently to proteins, in a process known as haptenation, is considered necessary for induction of a specific immunological response. Identification of the proteins modified by β-lactams and elucidation of the relevance of this process in allergic reactions requires sensitive tools. Here we describe the preparation and characterization of a biotinylated amoxicillin analog (AX-B) as a tool for the study of protein haptenation by amoxicillin (AX). AX-B, obtained by the inclusion of a biotin moiety at the lateral chain of AX, showed a chemical reactivity identical to AX. Covalent modification of proteins by AX-B was reduced by excess AX and vice versa, suggesting competition for binding to the same targets. From an immunological point of view, AX and AX-B behaved similarly in RAST inhibition studies with sera of patients with non-selective allergy towards β-lactams, whereas, as expected, competition by AX-B was poorer with sera of AX-selective patients, which recognize AX lateral chain. Use of AX-B followed by biotin detection allowed the observation of human serum albumin (HSA) modification by concentrations 100-fold lower that when using AX followed by immunological detection. Incubation of human serum with AX-B led to the haptenation of all of the previously identified major AX targets. In addition, some new targets could be detected. Interestingly, AX-B allowed the detection of intracellular protein adducts, which showed a cell type-specific pattern. This opens the possibility of following the formation and fate of AX-B adducts in cells. Thus, AX-B may constitute a valuable tool for the identification of AX targets with high sensitivity as well as for the elucidation of the mechanisms involved in allergy towards β-lactams.
Collapse
|
21
|
Alternating access mechanism in the POT family of oligopeptide transporters. EMBO J 2012; 31:3411-21. [PMID: 22659829 PMCID: PMC3419923 DOI: 10.1038/emboj.2012.157] [Citation(s) in RCA: 182] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 05/03/2012] [Indexed: 01/16/2023] Open
Abstract
Proton-dependent oligopeptide transporters are required for the uptake of diet-derived peptides in all kingdoms of life. The crystal structure of a bacterial transporter in the inward open conformation, together with a published structure in an occluded conformation, reveals the peptide transport mechanism. Short chain peptides are actively transported across membranes as an efficient route for dietary protein absorption and for maintaining cellular homeostasis. In mammals, peptide transport occurs via PepT1 and PepT2, which belong to the proton-dependent oligopeptide transporter, or POT family. The recent crystal structure of a bacterial POT transporter confirmed that they belong to the major facilitator superfamily of secondary active transporters. Despite the functional characterization of POT family members in bacteria, fungi and mammals, a detailed model for peptide recognition and transport remains unavailable. In this study, we report the 3.3-Å resolution crystal structure and functional characterization of a POT family transporter from the bacterium Streptococcus thermophilus. Crystallized in an inward open conformation the structure identifies a hinge-like movement within the C-terminal half of the transporter that facilitates opening of an intracellular gate controlling access to a central peptide-binding site. Our associated functional data support a model for peptide transport that highlights the importance of salt bridge interactions in orchestrating alternating access within the POT family.
Collapse
|
22
|
Liu R, Tang AMY, Tan YL, Limenta LMG, Lee EJD. Effects of Sodium Bicarbonate and Ammonium Chloride Pre-treatments on PEPT2 (SLC15A2) Mediated Renal Clearance of Cephalexin in Healthy Subjects. Drug Metab Pharmacokinet 2011; 26:87-93. [DOI: 10.2133/dmpk.dmpk-10-rg-039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
23
|
Zhou MM, Wu YM, Liu HY, Zhao K, Liu JX. Effects of tripeptides and lactogenic hormones on oligopeptide transporter 2 in bovine mammary gland. J Anim Physiol Anim Nutr (Berl) 2010; 95:781-9. [PMID: 21198960 DOI: 10.1111/j.1439-0396.2010.01110.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
This study was conducted to investigate the expression of oligopeptide transporter 2 (PepT2) and its potential function in bovine mammary gland. First, the PepT2 mRNA and protein were determined in cultured mammary epithelial cells. Then the effects of lactogenic hormones (prolactin, hydrocortisone or insulin) and substrate (threonyl-phenylalanyl-phenylalanine) on PepT2 were investigated. The PepT2 mRNA and protein were successfully detected in bovine mammary epithelial cells. PepT2 gene expression was enhanced by the addition of 50, 500 and 5000 ng/ml prolactin, 10 and 100 ng/ml hydrocortisone, and 50, 500, 5000 and 50,000 ng/ml insulin. PepT2 mRNA abundance was increased when 5, 10 and 15% of threonyl-phenylalanyl-phenylalanine was included. Responses of PepT2 to lactogenic hormones and oligopeptide inferred that it may play an important role in bovine mammary gland.
Collapse
Affiliation(s)
- M M Zhou
- Institute of Dairy Science, Ministry of Education Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, China
| | | | | | | | | |
Collapse
|
24
|
Peterson LW, Sala-Rabanal M, Krylov IS, Serpi M, Kashemirov BA, McKenna CE. Serine side chain-linked peptidomimetic conjugates of cyclic HPMPC and HPMPA: synthesis and interaction with hPEPT1. Mol Pharm 2010; 7:2349-61. [PMID: 20929265 DOI: 10.1021/mp100186b] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cidofovir (HPMPC), a broad spectrum antiviral agent, cannot be administered orally due to ionization of its phosphonic acid group at physiological pH. One prodrug approach involves conversion to the cyclic form (cHPMPC, 1) and esterification by the side chain hydroxyl group of a peptidomimetic serine. Transport studies in a rat model have shown enhanced levels of total cidofovir species in the plasma after oral dosing with L-Val-L-Ser-OMe cHPMPC, 2a. To explore the possibility that 2a and its three L/D stereoisomers 2b-d undergo active transport mediated by the peptide-specific intestinal transporter PEPT1, we performed radiotracer uptake and electrophysiology experiments applying the two-electrode voltage clamp technique in Xenopus laevis oocytes overexpressing human PEPT1 (hPEPT1, SLC15A1). 2a-d did not induce inward currents, indicating that they are not transported, but the stereoisomers with an L-configuration at the N-terminal valine (2a and 2b) potently inhibited transport of the hPEPT1 substrate glycylsarcosine (Gly-Sar). A "reversed" dipeptide conjugate, L-Ser-L-Ala-OiPr cHPMPC (4), also did not exhibit detectable transport, but completely abolished the Gly-Sar signal, suggesting that affinity of the transporter for these prodrugs is not impaired by a proximate linkage to the drug in the N-terminal amino acid of the dipeptide. Single amino acid conjugates of cHPMPC (3a and 3b) or cHPMPA (5, 6a and 6b) were not transported and only weakly inhibited Gly-Sar transport. The known hPEPT1 prodrug substrate valacyclovir (7) and its L-Val-L-Val dipeptide analogue (8) were used to verify coupled transport by the oocyte model. The results indicate that the previously observed enhanced oral bioavailability of 2a relative to the parent drug is unlikely to be due to active transport by hPEPT1. Syntheses of the novel compounds 2b-d and 3-6 are described, including a convenient solid-phase method to prepare 5, 6a and 6b.
Collapse
Affiliation(s)
- Larryn W Peterson
- Department of Chemistry, University of Southern California, Los Angeles, California 90089-0744, USA
| | | | | | | | | | | |
Collapse
|
25
|
Effects of Na+ and H+ on steady-state and presteady-state currents of the human concentrative nucleoside transporter 3 (hCNT3). Pflugers Arch 2010; 460:617-32. [DOI: 10.1007/s00424-010-0846-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 04/13/2010] [Accepted: 05/04/2010] [Indexed: 11/26/2022]
|
26
|
Akamatsu M, Fujikawa M, Nakao K, Shimizu R. In silico prediction of human oral absorption based on QSAR analyses of PAMPA permeability. Chem Biodivers 2010; 6:1845-66. [PMID: 19937826 DOI: 10.1002/cbdv.200900112] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The parallel artificial membrane permeation assay (PAMPA) was developed as a model for the prediction of transcellular permeation in the process of drug absorption. Our research group has measured the PAMPA permeability of peptide-related compounds, diverse drugs, and agrochemicals. This work led to a classical quantitative structure-activity relationship (QSAR) equation for PAMPA permeability coefficients of structurally diverse compounds based on simple physicochemical parameters such as lipophilicity at a particular pH (log P(oct) and |pKa-pH|), H-bond acceptor ability (SA(HA)), and H-bond donor ability (SA(HD)). Since the PAMPA permeability of lipophilic compounds decreased with their apparent lipophilicity due to the unstirred water layer (UWL) barrier on membrane surfaces and to membrane retention, a bilinear QSAR model was introduced to explain the permeability of a broader set of compounds using the same physicochemical parameters as those used for the linear model. We also compared PAMPA and Caco-2 cell permeability coefficients of compounds transported by various absorption mechanisms. The compounds were classified according to their absorption pathway (passively transported compounds, actively transported compounds, and compounds excreted by efflux systems) in the plot of Caco-2 vs. PAMPA permeability. Finally, based on the QSAR analyses of PAMPA permeability, an in silico prediction model of human oral absorption for possibly transported compounds was proposed, and the usefulness of the model was examined.
Collapse
Affiliation(s)
- Miki Akamatsu
- Laboratory of Comparative Agricultural Science, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan.
| | | | | | | |
Collapse
|
27
|
Knütter I, Wollesky C, Kottra G, Hahn MG, Fischer W, Zebisch K, Neubert RHH, Daniel H, Brandsch M. Transport of angiotensin-converting enzyme inhibitors by H+/peptide transporters revisited. J Pharmacol Exp Ther 2008; 327:432-41. [PMID: 18713951 DOI: 10.1124/jpet.108.143339] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Angiotensin-converting enzyme (ACE) inhibitors are often regarded as substrates for the H+/peptide transporters (PEPT)1 and PEPT2. Even though the conclusions drawn from published data are quite inconsistent, in most review articles PEPT1 is claimed to mediate the intestinal absorption of ACE inhibitors and thus to determine their oral availability. We systematically investigated the interaction of a series of ACE inhibitors with PEPT1 and PEPT2. First, we studied the effect of 14 ACE inhibitors including new drugs on the uptake of the dipeptide [14C]glycylsarcosine into human intestinal Caco-2 cells constitutively expressing PEPT1 and rat renal SKPT cells expressing PEPT2. In a second approach, the interaction of ACE inhibitors with heterologously expressed human PEPT1 and PEPT2 was determined. In both assay systems, zofenopril and fosinopril were found to have very high affinity for binding to peptide transporters. Medium to low affinity for transporter interaction was found for benazepril, quinapril, trandolapril, spirapril, cilazapril, ramipril, moexipril, quinaprilat, and perindopril. For enalapril, lisinopril, and captopril, very weak affinity or lack of interaction was found. Transport currents of PEPT1 and PEPT2 expressed in Xenopus laevis oocytes were recorded by the two-electrode voltage-clamp technique. Statistically significant, but very low currents were only observed for lisinopril, enalapril, quinapril, and benazepril at PEPT1 and for spirapril at PEPT2. For the other ACE inhibitors, electrogenic transport activity was extremely low or not measurable at all. The present results suggest that peptide transporters do not control intestinal absorption and renal reabsorption of ACE inhibitors.
Collapse
Affiliation(s)
- Ilka Knütter
- Biozentrum of the Martin-Luther-University Halle-Wittenberg, Membrane Transport Group, Halle, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Loo DDF, Hirayama BA, Sala-Rabanal M, Wright EM. How drugs interact with transporters: SGLT1 as a model. J Membr Biol 2008; 223:87-106. [PMID: 18592293 DOI: 10.1007/s00232-008-9116-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2008] [Accepted: 05/20/2008] [Indexed: 11/29/2022]
Abstract
Drugs are transported by cotransporters with widely different turnover rates. We have examined the underlying mechanism using, as a model system, glucose and indican (indoxyl-beta-D-glucopyranoside) transport by human Na+/glucose cotransporter (hSGLT1). Indican is transported by hSGLT1 at 10% of the rate for glucose but with a fivefold higher apparent affinity. We expressed wild-type hSGLT1 and mutant G507C in Xenopus oocytes and used electrical and optical methods to measure the kinetics of glucose (using nonmetabolized glucose analogue alpha-methyl-D-glucopyranoside, alphaMDG) and indican transport, alone and together. Indican behaved as a competitive inhibitor of alphaMDG transport. To examine protein conformations, we recorded SGLT1 capacitive currents (charge movements) and fluorescence changes in response to step jumps in membrane voltage, in the presence and absence of indican and/or alphaMDG. In the absence of sugar, voltage jumps elicited capacitive SGLT currents that decayed to steady state with time constants (tau) of 3-20 ms. These transient currents were abolished in saturating alphaMDG but only slightly reduced (10%) in saturating indican. SGLT1 G507C rhodamine fluorescence intensity increased with depolarizing and decreased with hyperpolarizing voltages. Maximal fluorescence increased approximately 150% in saturating indican but decreased approximately 50% in saturating alphaMDG. Modeling indicated that the rate-limiting step for indican transport is sugar translocation, whereas for alphaMDG it is dissociation of Na+ from the internal binding sites. The inhibitory effects of indican on alphaMDG transport are due to its higher affinity and a 100-fold lower translocation rate. Our results indicate that competition between substrates and drugs should be taken into consideration when targeting transporters as drug delivery systems.
Collapse
Affiliation(s)
- Donald D F Loo
- Department of Physiology, David Geffen School of Medicine at UCLA, 10833 LeConte Avenue, Los Angeles, CA 90095-1751, USA.
| | | | | | | |
Collapse
|