1
|
Pujol-Giménez J, Baumann SP, Ho TM, Augustynek B, Hediger MA. Functional Characterization of the Lysosomal Peptide/Histidine Transporter PHT1 ( SLC15A4) by Solid Supported Membrane Electrophysiology (SSME). Biomolecules 2024; 14:771. [PMID: 39062485 PMCID: PMC11275134 DOI: 10.3390/biom14070771] [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: 06/07/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
The peptide/histidine transporter PHT1 (SLC15A4) is expressed in the lysosomal membranes of immune cells where it plays an important role in metabolic and inflammatory signaling. PHT1 is an H+-coupled/histidine symporter that can transport a wide range of oligopeptides, including a variety of bacterial-derived peptides. Moreover, it enables the scaffolding of various metabolic signaling molecules and interacts with key regulatory elements of the immune response. Not surprisingly, PHT1 has been implicated in the pathogenesis of autoimmune diseases such as systemic lupus erythematosus (SLE). Unfortunately, the pharmacological development of PHT1 modulators has been hampered by the lack of suitable transport assays. To address this shortcoming, a novel transport assay based on solid-supported membrane-based electrophysiology (SSME) is presented. Key findings of the present SSME studies include the first recordings of electrophysiological properties, a pH dependence analysis, an assessment of PHT1 substrate selectivity, as well as the transport kinetics of the identified substrates. In contrast to previous work, PHT1 is studied in its native lysosomal environment. Moreover, observed substrate selectivity is validated by molecular docking. Overall, this new SSME-based assay is expected to contribute to unlocking the pharmacological potential of PHT1 and to deepen the understanding of its functional properties.
Collapse
Affiliation(s)
- Jonai Pujol-Giménez
- Department of Nephrology and Hypertension, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland (T.M.H.); (B.A.); (M.A.H.)
- Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Sven P. Baumann
- Department of Nephrology and Hypertension, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland (T.M.H.); (B.A.); (M.A.H.)
- Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Tin Manh Ho
- Department of Nephrology and Hypertension, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland (T.M.H.); (B.A.); (M.A.H.)
- Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Bartlomiej Augustynek
- Department of Nephrology and Hypertension, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland (T.M.H.); (B.A.); (M.A.H.)
- Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Matthias A. Hediger
- Department of Nephrology and Hypertension, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland (T.M.H.); (B.A.); (M.A.H.)
- Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Freiburgstrasse 15, 3010 Bern, Switzerland
| |
Collapse
|
2
|
Liu K, Chen Y, Yang D, Cai Y, Yang Z, Jin J. Betaine-Based and Polyguanidine-Inserted Zwitterionic Micelle as a Promising Platform to Conquer the Intestinal Mucosal Barrier. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37878752 DOI: 10.1021/acsami.3c07658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
Developing nanocarriers for oral drug delivery is often hampered by the dilemma of balancing mucus permeation and epithelium absorption, since huge differences in surface properties are required for sequentially overcoming these two processes. Inspired by mucus-penetrating viruses that universally possess a dense charge distribution with equal opposite charges on their surfaces, we rationally designed and constructed a poly(carboxybetaine)-based and polyguanidine-inserted cationic micelle platform (hybrid micelle) for oral drug delivery. The optimized hybrid micelle exhibited a great capacity for sequentially overcoming the mucus and villi barriers. It was demonstrated that a longer zwitterionic chain was favorable for mucus diffusion for hybrid micelles but not conducive to cellular uptake. In addition, the significantly enhanced internalization absorption of hybrid micelles was attributed to the synergistic effect of polyguanidine and proton-assisted amine acid transporter 1 (PAT1). Moreover, the retrograde pathway was mainly involved in the intracellular transport of hybrid micelles and transcytosis delivery. Furthermore, the prominent intestinal mucosa absorption in situ and in vivo liver distribution of the oral hybrid micelle were both detected. The results of this study indicated that the hybrid micelles were capable of conquering the intestinal mucosal barrier, having a great potential for oral application of drugs with poor oral bioavailability.
Collapse
Affiliation(s)
- Kedong Liu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
- School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
| | - Yun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Dutao Yang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Yanfei Cai
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Zhaoqi Yang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Jian Jin
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
3
|
Deng Y, Cheng H, Li J, Han H, Qi M, Wang N, Tan B, Li J, Wang J. Effects of glutamine, glutamate, and aspartate on intestinal barrier integrity and amino acid pool of the small intestine in piglets with normal or low energy diet. Front Vet Sci 2023; 10:1202369. [PMID: 37576837 PMCID: PMC10414990 DOI: 10.3389/fvets.2023.1202369] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 07/03/2023] [Indexed: 08/15/2023] Open
Abstract
Aspartate (asp), glutamate (glu), and glutamine (gln) are the major energy fuels for the small intestine, and it had been indicated in our previous study that the mix of these three amino acid supplementations could maintain intestinal energy homeostasis. This study aimed to further investigate whether the treatment of gln, glu, and asp in low energy diet affects the intestinal barrier integrity and amino acid pool in weaning piglets. A total of 198 weaned piglets were assigned to 3 treatments: control (basal diet + 1.59% L-Ala); T1 (basal diet + 1% L-Gln + 0.5% L-Glu + 0.1% L-Asp); and T2 (low energy diet + 1% L-Gln + 0.5% L-Glu + 0.1% L-Asp). The blood, jejunum, and ileum were obtained on day 5 or on day 21 post-weaning, respectively. Our results showed that T1 and T2 treatments increased the abundances of occludin, claudin-1, and claudin-3 in the small intestine while decreasing the serum diamine oxidase (DAO) and D-lactate levels in weaning piglets. Meanwhile, T1 and T2 treatments significantly increased the positive rate of proliferating cell nuclear antigen (PCNA) of the small intestine, promoting intestinal cell proliferation. We also found that supplementation with glu, gln, and asp improved the serum amino acid pool and promoted ileal amino acid transporter gene expression of slc3a2, slc6a14, and slc7a11 in weaned piglets. Additionally, on day 21 post-weaning, T1 and T2 treatments stimulated the phosphorylation of the mTOR-S6K1-4EBP1 signaling pathway in the small intestine, which may implicate the enhanced protein synthesis rate. In summary, dietary supplementation of gln, glu, and asp was beneficial to the intestinal barrier function and amino acid pool regulation, while the benefits of gln, glu, and asp treatment might be diminished by the low-energy diet. The results demonstrated that the supplementation of gln, glu, and asp under low energy levels was preferentially supplied as the energy fuel to restore the gut barrier function in piglets on day 5 post-weaning. With the increase in age and intestinal maturation (on day 21 post-weaning), gln, glu, and asp supplementation could also show an effect on the regulation of the amino acid pool and protein synthesis.
Collapse
Affiliation(s)
- Yuankun Deng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Hao Cheng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Junyao Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Hui Han
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agroecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Ming Qi
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agroecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Nan Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Bi'e Tan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Jianjun Li
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agroecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Jing Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| |
Collapse
|
4
|
Liu K, Chen Y, Yang Z, Jin J. zwitterionic Pluronic analog-coated PLGA nanoparticles for oral insulin delivery. Int J Biol Macromol 2023; 236:123870. [PMID: 36870645 DOI: 10.1016/j.ijbiomac.2023.123870] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023]
Abstract
In recent years, zwitterionic materials have drawn great attention in oral drug delivery system due to their capacity for rapid mucus diffusion and enhanced cellular internalization. However, zwitterionic materials tend to show strong polarity that was hard to directly coat hydrophobic nanoparticles (NPs). Inspired by Pluronic coating, a simple and convenient strategy to coat NPs with zwitterionic materials using zwitterionic Pluronic analogs was developed in this investigation. Poly(carboxybetaine)-poly(propylene oxide)-Poly(carboxybetaine) (PCB-PPO-PCB, PPP), containing PPO segments with MW > 2.0 kDa, can effectively adsorb on the surface of PLGA NPs with typical core-shell spherical in shape. The PLGA@PPP4K NPs were stable in gastrointestinal physiological environment and sequentially conquered mucus and epithelium barriers. Proton-assisted amine acid transporter 1 (PAT1) was verified to contribute to the enhanced internalization of PLGA@PPP4K NPs, and the NPs could partially evade lysosomal degradation pathway and utilize retrograde pathway for intracellular transport. In addition, the enhanced villi absorption in situ and oral liver distribution in vivo were also observed compared to PLGA@F127 NPs. Moreover, insulin-loaded PLGA@PPP4K NPs as an oral delivery application for diabetes induce a fine hypoglycemic response in diabetic rats after oral administration. The results of this study demonstrated that zwitterionic Pluronic analogs-coated NPs might provide a new perspective for zwitterionic materials application as well as oral delivery of biotherapeutics.
Collapse
Affiliation(s)
- Kedong Liu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China; School of Chemical and Material Engineering, Jiangnan University, Wuxi 214122, China
| | - Yun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Zhaoqi Yang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China.
| | - Jian Jin
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
5
|
Canlas J, Myers AL. Interactions of Betel Quid Constituents with Drug Disposition Pathways: An Overview. Curr Drug Metab 2023; 24:92-105. [PMID: 36852799 PMCID: PMC11271041 DOI: 10.2174/1389200224666230228142052] [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: 07/18/2022] [Revised: 01/10/2023] [Accepted: 01/25/2023] [Indexed: 03/01/2023]
Abstract
Global estimates indicate that over 600 million individuals worldwide consume the areca (betel) nut in some form. Nonetheless, its consumption is associated with a myriad of oral and systemic ailments, such as precancerous oral lesions, oropharyngeal cancers, liver toxicity and hepatic carcinoma, cardiovascular distress, and addiction. Users commonly chew slivers of areca nut in a complex consumable preparation called betel quid (BQ). Consequently, the user is exposed to a wide array of chemicals with diverse pharmacokinetic behavior in the body. However, a comprehensive understanding of the metabolic pathways significant to BQ chemicals is lacking. Henceforth, we performed a literature search to identify prominent BQ constituents and examine each chemical's interplay with drug disposition proteins. In total, we uncovered over 20 major chemicals (e.g., arecoline, nicotine, menthol, quercetin, tannic acid) present in the BQ mixture that were substrates, inhibitors, and/or inducers of various phase I (e.g., CYP, FMO, hydrolases) and phase II (e.g., GST, UGT, SULT) drug metabolizing enzymes, along with several transporters (e.g., P-gp, BCRP, MRP). Altogether, over 80 potential interactivities were found. Utilizing this new information, we generated theoretical predictions of drug interactions precipitated by BQ consumption. Data suggests that BQ consumers are at risk for drug interactions (and possible adverse effects) when co-ingesting other substances (multiple therapeutic classes) with overlapping elimination mechanisms. Until now, prediction about interactions is not widely known among BQ consumers and their clinicians. Further research is necessary based on our speculations to elucidate the biological ramifications of specific BQ-induced interactions and to take measures that improve the health of BQ consumers.
Collapse
Affiliation(s)
- Jasmine Canlas
- Department of Pharmaceutical & Biomedical Sciences, The University of Georgia, Athens, GA 30602, United States
| | - Alan L. Myers
- Department of Diagnostic & Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77030, United States
| |
Collapse
|
6
|
Diet evolution of carnivorous and herbivorous mammals in Laurasiatheria. BMC Ecol Evol 2022; 22:82. [PMID: 35729512 PMCID: PMC9210794 DOI: 10.1186/s12862-022-02033-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/09/2021] [Indexed: 11/15/2022] Open
Abstract
Background Laurasiatheria contains taxa with diverse diets, while the molecular basis and evolutionary history underlying their dietary diversification are less clear. Results In this study, we used the recently developed molecular phyloecological approach to examine the adaptive evolution of digestive system-related genes across both carnivorous and herbivorous mammals within Laurasiatheria. Our results show an intensified selection of fat and/or protein utilization across all examined carnivorous lineages, which is consistent with their high-protein and high-fat diets. Intriguingly, for herbivorous lineages (ungulates), which have a high-carbohydrate diet, they show a similar selection pattern as that of carnivorous lineages. Our results suggest that for the ungulates, which have a specialized digestive system, the selection intensity of their digestive system-related genes does not necessarily reflect loads of the nutrient components in their diets but appears to be positively related to the loads of the nutrient components that are capable of being directly utilized by the herbivores themselves. Based on these findings, we reconstructed the dietary evolution within Laurasiatheria, and our results reveal the dominant carnivory during the early diversification of Laurasiatheria. In particular, our results suggest that the ancestral bats and the common ancestor of ruminants and cetaceans may be carnivorous as well. We also found evidence of the convergent evolution of one fat utilization-related gene, APOB, across carnivorous taxa. Conclusions Our molecular phyloecological results suggest that digestive system-related genes can be used to determine the molecular basis of diet differentiations and to reconstruct ancestral diets. Supplementary Information The online version contains supplementary material available at 10.1186/s12862-022-02033-6.
Collapse
|
7
|
Nielsen CU, Krog NF, Sjekirica I, Nielsen SS, Pedersen ML. SNAT2 is responsible for hyperosmotic induced sarcosine and glycine uptake in human prostate PC-3 cells. Pflugers Arch 2022; 474:1249-1262. [PMID: 36175560 DOI: 10.1007/s00424-022-02752-1] [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: 08/15/2022] [Revised: 09/02/2022] [Accepted: 09/19/2022] [Indexed: 10/14/2022]
Abstract
Solute carriers (SLC) are important membrane transport proteins in normal and pathophysiological cells. The aim was to identify amino acid SLC(s) responsible for uptake of sarcosine and glycine in prostate cancer cells and investigate the impact hereon of hyperosmotic stress. Uptake of 14C-sarcosine and 3H-glycine was measured in human prostate cancer (PC-3) cells cultured under isosmotic (300 mOsm/kg) and hyperosmotic (500 mOsm/kg) conditions for 24 h. Hyperosmotic culture medium was obtained by supplementing the medium with 200 mM of the trisaccharide raffinose. Amino acid SLC expression was studied using RT-PCR, real-time PCR, and western blotting. siRNA knockdown of SNAT2 was performed. Experiments were conducted in at least 3 independent cell passages. The uptake of Sar and Gly was increased approximately 8-ninefold in PC-3 cells after 24 h hyperosmotic culture. PAT1 mRNA and protein could not be detected, while SNAT2 was upregulated at the mRNA and protein level. Transfection with SNAT2-specific siRNA reduced Vmax of Sar uptake from 2653 ± 38 to 513 ± 38 nmol mg protein-1 min-1, without altering the Km value (3.19 ± 0.13 vs. 3.42 ± 0.71 mM), indicating that SNAT2 is responsible for at least 80% of Sar uptake in hyperosmotic cultured PC-3 cells. SNAT2 is upregulated in hyperosmotic stressed prostate cancer cells and SNAT2 is responsible for cellular sarcosine and glycine uptake in hyperosmotic cultured PC-3 cells. Sar is identified as a substrate for SNAT2, and this has physiological implications for understanding cellular solute transport in prostate cancer cells.
Collapse
Affiliation(s)
- Carsten Uhd Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark.
| | - Nanna Friberg Krog
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Ilham Sjekirica
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Sidsel Strandgaard Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Maria L Pedersen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| |
Collapse
|
8
|
Santos L, Gonçalves LS, Bagheri-Hanei S, Möller GB, Sale C, James RM, Artioli GG. Insulin stimulates β-alanine uptake in skeletal muscle cells in vitro. Amino Acids 2021; 53:1763-1766. [PMID: 34676442 PMCID: PMC8592947 DOI: 10.1007/s00726-021-03090-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 10/07/2021] [Indexed: 10/26/2022]
Abstract
We evaluated whether insulin could stimulate β-alanine uptake by skeletal muscle cells in vitro. Mouse myoblasts (C2C12) (n = 3 wells per condition) were cultured with β-alanine (350 or 700 µmol·L-1), with insulin (100 µU·mL-1) either added to the media or not. Insulin stimulated the β-alanine uptake at the lower (350 µmol·L-1) but not higher (700 µmol·L-1) β-alanine concentration in culture medium, indicating that transporter saturation might blunt the stimulatory effects of insulin.
Collapse
Affiliation(s)
- Lívia Santos
- Musculoskeletal Physiology Research Group, Sport, Health and Performance Enhancement Research Centre, Nottingham Trent University, Nottingham, UK
| | - L S Gonçalves
- Applied Physiology and Nutrition Research GroupRheumatology DivisionFaculdade de Medicina FMUSPEscola de Educação Física E Esporte, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Shirin Bagheri-Hanei
- College of Engineering and Physical Science, Aston University, Birmingham, B4 7ET, UK
| | - Gabriella Berwig Möller
- Applied Physiology and Nutrition Research GroupRheumatology DivisionFaculdade de Medicina FMUSPEscola de Educação Física E Esporte, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Craig Sale
- Musculoskeletal Physiology Research Group, Sport, Health and Performance Enhancement Research Centre, Nottingham Trent University, Nottingham, UK
| | - Ruth M James
- Musculoskeletal Physiology Research Group, Sport, Health and Performance Enhancement Research Centre, Nottingham Trent University, Nottingham, UK
| | - Guilherme Giannini Artioli
- Department of Life Sciences, Manchester Metropolitan University, John Dalton Building, Manchester, M1 5GD, UK.
| |
Collapse
|
9
|
Measuring the oral bioavailability of protein hydrolysates derived from food sources: A critical review of current bioassays. Biomed Pharmacother 2021; 144:112275. [PMID: 34628165 DOI: 10.1016/j.biopha.2021.112275] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/21/2021] [Accepted: 09/28/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Food proteins are a source of hydrolysates with potentially useful biological attributes. Bioactive peptides from food-derived proteins are released from hydrolysates using exogenous industrial processes or endogenous intestinal enzymes. Current in vitro permeability assays have limitations in predicting the oral bioavailability (BA) of bioactive peptides in humans. There are also difficulties in relating the low blood levels of food-derived bioactive peptides detected in preclinical in vivo models to pharmacodynamic read-outs relevant for humans. SCOPE AND APPROACH In this review, we describe in vitro assays of digestion, permeation, and metabolism as indirect predictors of the potential oral BA of hydrolysates and their constituent bioactive peptides. We discuss the relationship between industrial hydrolysis processes and the oral BA of hydrolysates and their peptide by-products. KEY FINDINGS Hydrolysates are challenging for analytical detection methods due to capacity for enzymatic generation of peptides with novel sequences and also new modifications of these peptides during digestion. Mass spectrometry and peptidomics can improve the capacity to detect individual peptides released from complex hydrolysates in biological milieu.
Collapse
|
10
|
Rapid and Sensitive Quantification of Intracellular Glycyl-Sarcosine for Semi-High-Throughput Screening for Inhibitors of PEPT-1. Pharmaceutics 2021; 13:pharmaceutics13071019. [PMID: 34371711 PMCID: PMC8309108 DOI: 10.3390/pharmaceutics13071019] [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] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/23/2022] Open
Abstract
The peptide transporter PEPT-1 (SLC15A1) plays a major role in nutritional supply with amino acids by mediating the intestinal influx of dipeptides and tripeptides generated during food digestion. Its role in the uptake of small bioactive peptides and various therapeutics makes it an important target for the investigation of the systemic absorption of small peptide-like active compounds and prodrug strategies of poorly absorbed therapeutics. The dipeptide glycyl-sarcosine (Gly-Sar), which comprises an N-methylated peptide bond that increases stability against enzymatic degradation, is widely utilized for studying PEPT-1-mediated transport. To support experiments on PEPT-1 inhibitor screening to identify potential substrates, we developed a highly sensitive Gly-Sar quantification assay for Caco-2 cell lysates with a dynamic range of 0.1 to 1000 ng/mL (lower limit of quantification 0.68 nM) in 50 µL of cell lysate. The assay was validated following the applicable recommendations for bioanalytic method validation of the FDA and EMA. Sample preparation and quantification were established in 96-well cell culture plates that were also used for the cellular uptake studies, resulting in a rapid and robust screening assay for PEPT-1 inhibitors. This sample preparation principle, combined with the high sensitivity of the UPLC-MS/MS quantification, is suitable for screening assays for PEPT-1 inhibitors and substrates in high-throughput formats and holds the potential for automation. Applicability was demonstrated by IC50 determinations of the known PEPT-1 inhibitor losartan, the known substrates glycyl-proline (Gly-Pro), and valaciclovir, the prodrug of aciclovir, which itself is no substrate of PEPT-1 and consequently showed no inhibition in our assay.
Collapse
|
11
|
Wu Y. Molecular phyloecology suggests a trophic shift concurrent with the evolution of the first birds. Commun Biol 2021; 4:547. [PMID: 33986452 PMCID: PMC8119460 DOI: 10.1038/s42003-021-02067-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 03/31/2021] [Indexed: 02/03/2023] Open
Abstract
Birds are characterized by evolutionary specializations of both locomotion (e.g., flapping flight) and digestive system (toothless, crop, and gizzard), while the potential selection pressures responsible for these evolutionary specializations remain unclear. Here we used a recently developed molecular phyloecological method to reconstruct the diets of the ancestral archosaur and of the common ancestor of living birds (CALB). Our results suggest a trophic shift from carnivory to herbivory (fruit, seed, and/or nut eater) at the archosaur-to-bird transition. The evolutionary shift of the CALB to herbivory may have essentially made them become a low-level consumer and, consequently, subject to relatively high predation risk from potential predators such as gliding non-avian maniraptorans, from which birds descended. Under the relatively high predation pressure, ancestral birds with gliding capability may have then evolved not only flapping flight as a possible anti-predator strategy against gliding predatory non-avian maniraptorans but also the specialized digestive system as an evolutionary tradeoff of maximizing foraging efficiency and minimizing predation risk. Our results suggest that the powered flight and specialized digestive system of birds may have evolved as a result of their tropic shift-associated predation pressure.
Collapse
Affiliation(s)
- Yonghua Wu
- School of Life Sciences, Northeast Normal University, Changchun, China.
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China.
| |
Collapse
|
12
|
Nielsen CU, Pedersen M, Müller S, Kæstel T, Bjerg M, Ulaganathan N, Nielsen S, Carlsen KL, Nøhr MK, Holm R. Inhibitory Effects of 17-α-Ethinyl-Estradiol and 17-β-Estradiol on Transport Via the Intestinal Proton-Coupled Amino Acid Transporter (PAT1) Investigated In Vitro and In Vivo. J Pharm Sci 2020; 110:354-364. [PMID: 32835702 DOI: 10.1016/j.xphs.2020.08.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/12/2020] [Accepted: 08/18/2020] [Indexed: 11/24/2022]
Abstract
The proton-coupled amino acid transporter, PAT1, is known to be responsible for intestinal absorption drug substances such as gaboxadol and vigabatrin. The aim of the present study was to investigate, if 17-α-ethinyl-estradiol (E-E2) and 17-β-estradiol (E) inhibit PAT1-mediated intestinal absorption of proline and taurine in vitro in Caco-2 cells and in vivo using Sprague-Dawley rats to assess the potential for taurine-drug interactions. E and E-E2 inhibited the PAT1-mediated uptake of proline and taurine in Caco-2 cells with IC50 values of 10.0-50.0 μM without major effect on other solute carriers such as the taurine transporter (TauT), di/tri-peptide transporter (PEPT1), and serotonin transporter (SERT1). In PAT1-expressing oocytes E and E-E2 were non-translocated inhibitors. In Caco-2 cells, E and E-E2 lowered the maximal uptake capacity of PAT1 in a non-competitive manner. Likewise, the transepithelial permeability of proline and taurine was reduced in presence of E and E-E2. In male Sprague Dawley rats pre-dosed with E-E2 a decreased maximal plasma concentration (Cmax) of taurine and increased the time (tmax) to reach this was indicated, suggesting the possibility for an in vivo effect on the absorption of PAT1 substrates. In conclusion, 17-α-ethinyl-estradiol and 17-β-estradiol were identified as non-translocated and non-competitive inhibitors of PAT1.
Collapse
Affiliation(s)
- Carsten Uhd Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark.
| | - Maria Pedersen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Stefanie Müller
- Department of Pharmacy, Faculty of Health and Medical Sciences, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Thea Kæstel
- Department of Pharmacy, Faculty of Health and Medical Sciences, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Maria Bjerg
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Nithiya Ulaganathan
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Salli Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Krestine Lundgaard Carlsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Martha Kampp Nøhr
- Department of Pharmacy, Faculty of Health and Medical Sciences, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - René Holm
- Drug Product Development, Janssen R&D, Johnson & Johnson, Turnhoutseweg 30, 2340 Beerse, Belgium; Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark
| |
Collapse
|
13
|
Han X, Lu Y, Xie J, Zhang E, Zhu H, Du H, Wang K, Song B, Yang C, Shi Y, Cao Z. Zwitterionic micelles efficiently deliver oral insulin without opening tight junctions. NATURE NANOTECHNOLOGY 2020; 15:605-614. [PMID: 32483319 PMCID: PMC7534179 DOI: 10.1038/s41565-020-0693-6] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 04/15/2020] [Indexed: 05/06/2023]
Abstract
Oral delivery of protein drugs is considered a life-changing solution for patients who require regular needle injections. However, clinical translation of oral protein formulations has been hampered by inefficient penetration of drugs through the intestinal mucus and epithelial cell layer, leading to low absorption and bioavailability, and safety concerns owing to tight junction openings. Here we report a zwitterionic micelle platform featuring a virus-mimetic zwitterionic surface, a betaine side chain and an ultralow critical micelle concentration, enabling drug penetration through the mucus and efficient transporter-mediated epithelial absorption without the need for tight junction opening. This micelle platform was used to fabricate a prototype oral insulin formulation by encapsulating a freeze-dried powder of zwitterionic micelle insulin into an enteric-coated capsule. The biocompatible oral insulin formulation shows a high oral bioavailability of >40%, offers the possibility to fine tune insulin acting profiles and provides long-term safety, enabling the oral delivery of protein drugs.
Collapse
Affiliation(s)
- Xiangfei Han
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Yang Lu
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Jinbing Xie
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Ershuai Zhang
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Hui Zhu
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Hong Du
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Ke Wang
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Boyi Song
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Chengbiao Yang
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Yuanjie Shi
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Zhiqiang Cao
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
14
|
Higuchi-Sanabria R, Shen K, Kelet N, Frankino PA, Durieux J, Bar-Ziv R, Sing CN, Garcia EJ, Homentcovschi S, Sanchez M, Wu R, Tronnes SU, Joe L, Webster B, Ahilon-Jeronimo A, Monshietehadi S, Dallarda S, Pender C, Pon LA, Zoncu R, Dillin A. Lysosomal recycling of amino acids affects ER quality control. SCIENCE ADVANCES 2020; 6:eaaz9805. [PMID: 32637599 PMCID: PMC7319768 DOI: 10.1126/sciadv.aaz9805] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 05/13/2020] [Indexed: 06/11/2023]
Abstract
Recent work has highlighted the fact that lysosomes are a critical signaling hub of metabolic processes, providing fundamental building blocks crucial for anabolic functions. How lysosomal functions affect other cellular compartments is not fully understood. Here, we find that lysosomal recycling of the amino acids lysine and arginine is essential for proper ER quality control through the UPRER. Specifically, loss of the lysine and arginine amino acid transporter LAAT-1 results in increased sensitivity to proteotoxic stress in the ER and decreased animal physiology. We find that these LAAT-1-dependent effects are linked to glycine metabolism and transport and that the loss of function of the glycine transporter SKAT-1 also increases sensitivity to ER stress. Direct lysine and arginine supplementation, or glycine supplementation alone, can ameliorate increased ER stress sensitivity found in laat-1 mutants. These data implicate a crucial role in recycling lysine, arginine, and glycine in communication between the lysosome and ER.
Collapse
Affiliation(s)
- Ryo Higuchi-Sanabria
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Koning Shen
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Naame Kelet
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Phillip A. Frankino
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Jenni Durieux
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Raz Bar-Ziv
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Cierra N. Sing
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Enrique J. Garcia
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Stefan Homentcovschi
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Melissa Sanchez
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Rui Wu
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Sarah U. Tronnes
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Larry Joe
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Brant Webster
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Alex Ahilon-Jeronimo
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Samira Monshietehadi
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Sofia Dallarda
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Corinne Pender
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Liza A. Pon
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Roberto Zoncu
- Department of Biochemistry, Biophysics, and Structural Biology, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Andrew Dillin
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| |
Collapse
|
15
|
Gonçalves LDS, Kratz C, Santos L, Carvalho VH, Sales LP, Nemezio K, Longobardi I, Riani LA, Lima MMDO, Saito T, Fernandes AL, Rodrigues J, James RM, Sale C, Gualano B, Geloneze B, de Medeiros MHG, Artioli GG. Insulin does not stimulate β-alanine transport into human skeletal muscle. Am J Physiol Cell Physiol 2020; 318:C777-C786. [DOI: 10.1152/ajpcell.00550.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To test whether high circulating insulin concentrations influence the transport of β-alanine into skeletal muscle at either saturating or subsaturating β-alanine concentrations, we conducted two experiments whereby β-alanine and insulin concentrations were controlled. In experiment 1, 12 men received supraphysiological amounts of β-alanine intravenously (0.11 g·kg−1·min−1for 150 min), with or without insulin infusion. β-Alanine and carnosine were measured in muscle before and 30 min after infusion. Blood samples were taken throughout the infusion protocol for plasma insulin and β-alanine analyses. β-Alanine content in 24-h urine was assessed. In experiment 2, six men ingested typical doses of β-alanine (10 mg/kg) before insulin infusion or no infusion. β-Alanine was assessed in muscle before and 120 min following ingestion. In experiment 1, no differences between conditions were shown for plasma β-alanine, muscle β-alanine, muscle carnosine and urinary β-alanine concentrations (all P > 0.05). In experiment 2, no differences between conditions were shown for plasma β-alanine or muscle β-alanine concentrations (all P > 0.05). Hyperinsulinemia did not increase β-alanine uptake by skeletal muscle cells, neither when substrate concentrations exceed the Vmaxof β-alanine transporter TauT nor when it was below saturation. These results suggest that increasing insulin concentration is not necessary to maximize β-alanine transport into muscle following β-alanine intake.
Collapse
Affiliation(s)
- Lívia de Souza Gonçalves
- Applied Physiology and Nutrition Research Group; School of Physical Education and Sport, Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Caroline Kratz
- Applied Physiology and Nutrition Research Group; School of Physical Education and Sport, Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Lívia Santos
- Musculoskeletal Physiology Research Group, Sport, Health, and Performance Enhancement Research Centre, Nottingham Trent University, Nottingham, United Kingdom
| | | | - Lucas Peixoto Sales
- Applied Physiology and Nutrition Research Group; School of Physical Education and Sport, Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Kleiner Nemezio
- Applied Physiology and Nutrition Research Group; School of Physical Education and Sport, Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Igor Longobardi
- Applied Physiology and Nutrition Research Group; School of Physical Education and Sport, Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Luiz Augusto Riani
- Applied Physiology and Nutrition Research Group; School of Physical Education and Sport, Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Marcelo Miranda de Oliveira Lima
- Laboratory of Investigation in Metabolism and Diabetes (LIMED)/Gastrocentro Departamento de Cirurgia, Universidade de Campinas (UNICAMP), Campinas, Brazil
| | - Tiemi Saito
- Applied Physiology and Nutrition Research Group; School of Physical Education and Sport, Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Alan Lins Fernandes
- Applied Physiology and Nutrition Research Group; School of Physical Education and Sport, Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Joice Rodrigues
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Ruth Margaret James
- Musculoskeletal Physiology Research Group, Sport, Health, and Performance Enhancement Research Centre, Nottingham Trent University, Nottingham, United Kingdom
| | - Craig Sale
- Musculoskeletal Physiology Research Group, Sport, Health, and Performance Enhancement Research Centre, Nottingham Trent University, Nottingham, United Kingdom
| | - Bruno Gualano
- Applied Physiology and Nutrition Research Group; School of Physical Education and Sport, Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Bruno Geloneze
- Laboratory of Investigation in Metabolism and Diabetes (LIMED)/Gastrocentro Departamento de Cirurgia, Universidade de Campinas (UNICAMP), Campinas, Brazil
| | | | - Guilherme Giannini Artioli
- Applied Physiology and Nutrition Research Group; School of Physical Education and Sport, Rheumatology Division, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
16
|
Antonescu IE, Rasmussen KF, Neuhoff S, Fretté X, Karlgren M, Bergström CAS, Nielsen CU, Steffansen B. The Permeation of Acamprosate Is Predominantly Caused by Paracellular Diffusion across Caco-2 Cell Monolayers: A Paracellular Modeling Approach. Mol Pharm 2019; 16:4636-4650. [DOI: 10.1021/acs.molpharmaceut.9b00733] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | | | | | | | - Maria Karlgren
- Department of Pharmacy, Uppsala University, P.O. Box 580, SE-751 23 Uppsala, Sweden
| | | | | | | |
Collapse
|
17
|
Koyambo-Konzapa SJ, Minguirbara A, Nsangou M. Solvent effects on the structures and vibrational features of zwitterionic dipeptides: L-diglycine and L-dialanine. J Mol Model 2015; 21:189. [DOI: 10.1007/s00894-015-2718-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 06/01/2015] [Indexed: 11/24/2022]
|
18
|
Nielsen CU, Frølund S, Abdulhadi S, Sari H, Langthaler L, Nøhr MK, Kall MA, Brodin B, Holm R. Sertraline inhibits the transport of PAT1 substrates in vivo and in vitro. Br J Pharmacol 2014; 170:1041-52. [PMID: 23962042 DOI: 10.1111/bph.12341] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 07/02/2013] [Accepted: 08/03/2013] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND AND PURPOSE Intestinal nutrient transporters may mediate the uptake of drugs. The aim of this study was to investigate whether sertraline interacts with the intestinal proton-coupled amino acid transporter 1 PAT1 (SLC36A1). EXPERIMENTAL APPROACH In vitro investigations of interactions between sertraline and human (h)PAT1, hSGLT1 (sodium-glucose linked transporter 1) and hPepT1 (proton-coupled di-/tri-peptide transporter 1) were conducted in Caco-2 cells using radiolabelled substrates. In vivo pharmacokinetic investigations were conducted in male Sprague-Dawley rats using gaboxadol (10 mg·kg(-1), p.o.) as a PAT1 substrate and sertraline (0-30.6 mg·kg(-1)). Gaboxadol was quantified by hydrophilic interaction chromatography followed by MS/MS detection. KEY RESULTS Sertraline inhibited hPAT1-mediated L-[(3)H]-Pro uptake in Caco-2 cells. This interaction between sertraline and PAT1 appeared to be non-competitive. The uptake of the hSGLT1 substrate [(14)C]-α-methyl-D-glycopyranoside and the hPepT1 substrate [(14)C]-Gly-Sar in Caco-2 cells was also decreased in the presence of 0.3 mM sertraline. In rats, the administration of sertraline (0.1-10 mM, corresponding to 0.3-30.6 mg·kg(-1), p.o.) significantly reduced the maximal gaboxadol plasma concentration and AUC after its administration p.o. CONCLUSIONS AND IMPLICATIONS Sertraline is an apparent non-competitive inhibitor of hPAT1-mediated transport in vitro. This inhibitory effect of sertraline is not specific to hPAT1 as substrate transport via hPepT1 and hSGLT1 was also reduced in the presence of sertraline. In vivo, sertraline reduced the amount of gaboxadol absorbed, suggesting that the inhibitory effect of sertraline on PAT1 occurs both in vitro and in vivo. Hence, sertraline could alter the bioavailability of drugs absorbed via PAT1.
Collapse
Affiliation(s)
- C U Nielsen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Pharmacokinetic aspects of the anti-epileptic drug substance vigabatrin: focus on transporter interactions. Ther Deliv 2014; 5:927-42. [DOI: 10.4155/tde.14.55] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Drug transporters in various tissues, such as intestine, kidney, liver and brain, are recognized as important mediators of absorption, distribution, metabolism and excretion of drug substances. This review gives a current status on the transporter(s) mediating the absorption, distribution, metabolism and excretion properties of the anti-epileptic drug substance vigabatrin. For orally administered drugs, like vigabatrin, the absorption from the intestine is a prerequisite for the bioavailability. Therefore, transporter(s) involved in the intestinal absorption of vigabatrin in vitro and in vivo are discussed in detail. Special focus is on the contribution of the proton-coupled amino acid transporter 1 (PAT1) for intestinal vigabatrin absorption. Furthermore, the review gives an overview of the pharmacokinetic parameters of vigabatrin across different species and drug–food and drug–drug interactions involving vigabatrin.
Collapse
|
20
|
Abstract
In vertebrates and invertebrates, morphological and functional features of gastrointestinal (GI) tracts generally reflect food chemistry, such as content of carbohydrates, proteins, fats, and material(s) refractory to rapid digestion (e.g., cellulose). The expression of digestive enzymes and nutrient transporters approximately matches the dietary load of their respective substrates, with relatively modest excess capacity. Mechanisms explaining differences in hydrolase activity between populations and species include gene copy number variations and single-nucleotide polymorphisms. Transcriptional and posttranscriptional adjustments mediate phenotypic changes in the expression of hydrolases and transporters in response to dietary signals. Many species respond to higher food intake by flexibly increasing digestive compartment size. Fermentative processes by symbiotic microorganisms are important for cellulose degradation but are relatively slow, so animals that rely on those processes typically possess special enlarged compartment(s) to maintain a microbiota and other GI structures that slow digesta flow. The taxon richness of the gut microbiota, usually identified by 16S rRNA gene sequencing, is typically an order of magnitude greater in vertebrates than invertebrates, and the interspecific variation in microbial composition is strongly influenced by diet. Many of the nutrient transporters are orthologous across different animal phyla, though functional details may vary (e.g., glucose and amino acid transport with K+ rather than Na+ as a counter ion). Paracellular absorption is important in many birds. Natural toxins are ubiquitous in foods and may influence key features such as digesta transit, enzymatic breakdown, microbial fermentation, and absorption.
Collapse
Affiliation(s)
- William H Karasov
- Forest and Wildlife Ecology, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| | | |
Collapse
|
21
|
Frølund S, Nøhr M, Holm R, Brodin B, Nielsen C. Potential involvement of the proton-coupled amino acid transporter PAT1 (SLC36A1) in the delivery of pharmaceutical agents. J Drug Deliv Sci Technol 2013. [DOI: 10.1016/s1773-2247(13)50046-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
22
|
Broberg ML, Holm R, Tønsberg H, Frølund S, Ewon KB, Nielsen AL, Brodin B, Jensen A, Kall MA, Christensen KV, Nielsen CU. Function and expression of the proton-coupled amino acid transporter PAT1 along the rat gastrointestinal tract: implications for intestinal absorption of gaboxadol. Br J Pharmacol 2012; 167:654-65. [PMID: 22577815 PMCID: PMC3449268 DOI: 10.1111/j.1476-5381.2012.02030.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 04/12/2012] [Accepted: 05/02/2012] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Intestinal absorption via membrane transporters may determine the pharmacokinetics of drug compounds. The hypothesis is that oral absorption of gaboxadol (4,5,6,7-tetrahydroisoxazolo [5,4-c] pyridine-3-ol) in rats occurs via the proton-coupled amino acid transporter, rPAT1 (encoded by the gene rSlc36a1). Consequently, we aimed to elucidate the in vivo role of rPAT1 in the absorption of gaboxadol from various intestinal segments obtained from Sprague-Dawley rats. EXPERIMENTAL APPROACH The absorption of gaboxadol was investigated following its administration into four different intestinal segments. The intestinal expression of rSlc36a1 mRNA was measured by quantitative real-time PCR. Furthermore, the hPAT1-/rPAT1-mediated transport of gaboxadol or L-proline was studied in hPAT1-expressing Xenopus laevis oocytes, Caco-2 cell monolayers and excised segments of the rat intestine. KEY RESULTS The absorption fraction of gaboxadol was high (81.3-91.3%) following its administration into the stomach, duodenum and jejunum, but low (4.2%) after administration into the colon. The pharmacokinetics of gaboxadol were modified by the co-administration of L-tryptophan (an hPAT1 inhibitor) and L-proline (an hPAT1 substrate). The in vitro carrier-mediated uptake rate of L-proline in the excised intestinal segments was highest in the mid jejunum and lowest in the colon. The in vitro uptake and the in vivo absorption correlated with the expression of rSlc36a1 mRNA along the rat intestine. CONCLUSIONS AND IMPLICATIONS These results suggest that PAT1 mediates the intestinal absorption of gaboxadol and therefore determines its oral bioavailability. This has implications for the in vivo role of PAT1 and may have an influence on the design of pharmaceutical formulations of PAT1 substrates.
Collapse
Affiliation(s)
- M l Broberg
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Frølund S, Langthaler L, Kall MA, Holm R, Nielsen CU. Intestinal Drug Transport via the Proton-Coupled Amino Acid Transporter PAT1 (SLC36A1) Is Inhibited by Gly-Xaa Dipeptides. Mol Pharm 2012; 9:2761-9. [DOI: 10.1021/mp300345e] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Sidsel Frølund
- Department of Pharmacy, Faculty
of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Louise Langthaler
- Department of Pharmacy, Faculty
of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
- Department of Preformulation, H. Lundbeck A/S, Ottiliavej 9, DK-2500 Valby, Denmark
| | - Morten A. Kall
- Department of Bioanalysis, H. Lundbeck A/S, Ottiliavej 9, DK-2500 Valby, Denmark
| | - René Holm
- Department of Pharmacy, Faculty
of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
- Department of Preformulation, H. Lundbeck A/S, Ottiliavej 9, DK-2500 Valby, Denmark
| | - Carsten Uhd Nielsen
- Department of Pharmacy, Faculty
of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| |
Collapse
|
24
|
Moravek M, Fisseha S, Swain JE. Dipeptide forms of glycine support mouse preimplantation embryo development in vitro and provide protection against high media osmolality. J Assist Reprod Genet 2012; 29:283-90. [PMID: 22246224 DOI: 10.1007/s10815-011-9705-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 12/26/2011] [Indexed: 01/23/2023] Open
Abstract
PURPOSE To examine potential benefits of dipeptide forms of amino acids for embryo culture by determining ability of dipeptide glycine forms to support embryo development, act as osmolytes, and reduce ammonia production. METHODS Frozen thawed 1-cell mouse embryos were cultured in media with varying osmolality with glycine and dipeptide forms of glycine and development assessed. Ammonia levels were measured in various media. RESULTS Dipeptide forms of glycine, alanyl- and glycyl-glycine, can support mouse embryo development in vitro. Additionally, dipeptide glycine can act as an organic osmolyte in developing embryos, permitting blastocyst formation in high osmolality media. Interestingly, as evidenced by decreased embryo development, dipeptides are not as efficient as osmolytes as their constituent individual amino acids. Dipeptide glycine produced less ammonia than glycine. CONCLUSION Though dipeptides can provide osmoregulation in preimplantation embryos, efficacy may be lower than individual amino acids. The mechanism by which embryos transport and utilize dipeptide amino acids remains to be identified.
Collapse
Affiliation(s)
- Molly Moravek
- Department of OB/GYN, Reproductive Sciences Program, University of Michigan, Ann Arbor, MI 48108, USA
| | | | | |
Collapse
|
25
|
Rectal Absorption of Vigabatrin, a Substrate of the Proton Coupled Amino Acid Transporter (PAT1, Slc36a1), in Rats. Pharm Res 2012; 29:1134-42. [DOI: 10.1007/s11095-012-0673-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 01/03/2012] [Indexed: 01/16/2023]
|
26
|
Damon C, Vallon L, Zimmermann S, Haider MZ, Galeote V, Dequin S, Luis P, Fraissinet-Tachet L, Marmeisse R. A novel fungal family of oligopeptide transporters identified by functional metatranscriptomics of soil eukaryotes. THE ISME JOURNAL 2011; 5:1871-80. [PMID: 21654847 PMCID: PMC3223307 DOI: 10.1038/ismej.2011.67] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 04/26/2011] [Accepted: 04/29/2011] [Indexed: 11/08/2022]
Abstract
Functional environmental genomics has the potential to identify novel biological functions that the systematic sequencing of microbial genomes or environmental DNA may fail to uncover. We targeted the functions expressed by soil eukaryotes using a metatranscriptomic approach based on the use of soil-extracted polyadenylated messenger RNA to construct environmental complementary DNA expression libraries. Functional complementation of a yeast mutant defective in di/tripeptide uptake identified a novel family of oligopeptide transporters expressed by fungi. This family has a patchy distribution in the Basidiomycota and Ascomycota and is present in the genome of a Saccharomyces cerevisiae wine strain. High throughput phenotyping of yeast mutants expressing two environmental transporters showed that they both displayed broad substrate specificity and could transport more than 60-80 dipeptides. When expressed in Xenopus oocytes one environmental transporter induced currents upon dipeptide addition, suggesting proton-coupled co-transport of dipeptides. This transporter was also able to transport specifically cysteine. Deletion of the two copies of the corresponding gene family members in the genome of the wine yeast strain severely reduced the number of dipeptides that it could assimilate. These results demonstrate that these genes are functional and can be used by fungi to efficiently scavenge the numerous, low concentration, oligopeptides continuously generated in soils by proteolysis.
Collapse
Affiliation(s)
- Coralie Damon
- Université de Lyon, Université Lyon 1, CNRS, UMR5557 d'Ecologie Microbienne, Villeurbanne, France
| | - Laurent Vallon
- Université de Lyon, Université Lyon 1, CNRS, UMR5557 d'Ecologie Microbienne, Villeurbanne, France
| | - Sabine Zimmermann
- CNRS, UMR5004 Biochimie et Physiologie Moléculaire des Plantes, Montpellier, France
| | - Muhammad Z Haider
- CNRS, UMR5004 Biochimie et Physiologie Moléculaire des Plantes, Montpellier, France
| | | | - Sylvie Dequin
- INRA, UMR1083 Sciences pour l'Oenologie, Montpellier, France
| | - Patricia Luis
- Université de Lyon, Université Lyon 1, CNRS, UMR5557 d'Ecologie Microbienne, Villeurbanne, France
| | | | - Roland Marmeisse
- Université de Lyon, Université Lyon 1, CNRS, UMR5557 d'Ecologie Microbienne, Villeurbanne, France
| |
Collapse
|
27
|
Frølund S, Rapin N, Nielsen CU. Gaboxadol has affinity for the proton-coupled amino acid transporter 1, SLC36A1 (hPAT1)—A modelling approach to determine IC50 values of the three ionic species of gaboxadol. Eur J Pharm Sci 2011; 42:192-8. [DOI: 10.1016/j.ejps.2010.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 10/20/2010] [Accepted: 11/18/2010] [Indexed: 10/18/2022]
|