1
|
Palmen R, Walton M, Wagner J. Pediatric flecainide pharmacogenomics: a roadmap to delivering precision-based care to pediatrics arrhythmias. Front Pharmacol 2024; 15:1477485. [PMID: 39741635 PMCID: PMC11686437 DOI: 10.3389/fphar.2024.1477485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025] Open
Abstract
Flecainide acetate is a Class 1c anti-arrhythmic with a potent sodium voltage gated channel blockade which is utilized for the second-line treatment of tachyarrhythmias in children and adults. Given its narrow therapeutic index, the individualization of drug therapy is of utmost importance for clinicians. Despite efforts to improve anti-arrhythmic drug therapy, there remain knowledge gaps regarding the impact of variation in the genes relevant to flecainide's disposition and response. This variability is compounded in developing children whose drug disposition and response pathways may remain immature. The purpose of this comprehensive review is to outline flecainide's disposition and response pathways while simultaneously highlighting opportunities for prospective investigation in the pediatric population.
Collapse
Affiliation(s)
- Ronald Palmen
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
| | - Mollie Walton
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
- Division of Cardiology, Kansas City, MO, United States
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children’s Mercy, Kansas City, MO, United States
| | - Jonathan Wagner
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
- Division of Cardiology, Kansas City, MO, United States
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children’s Mercy, Kansas City, MO, United States
| |
Collapse
|
2
|
Parker JL, Deme JC, Lichtinger SM, Kuteyi G, Biggin PC, Lea SM, Newstead S. Structural basis for antibiotic transport and inhibition in PepT2. Nat Commun 2024; 15:8755. [PMID: 39384780 PMCID: PMC11464717 DOI: 10.1038/s41467-024-53096-6] [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: 05/17/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024] Open
Abstract
The uptake and elimination of beta-lactam antibiotics in the human body are facilitated by the proton-coupled peptide transporters PepT1 (SLC15A1) and PepT2 (SLC15A2). The mechanism by which SLC15 family transporters recognize and discriminate between different drug classes and dietary peptides remains unclear, hampering efforts to improve antibiotic pharmacokinetics through targeted drug design and delivery. Here, we present cryo-EM structures of the proton-coupled peptide transporter, PepT2 from Rattus norvegicus, in complex with the widely used beta-lactam antibiotics cefadroxil, amoxicillin and cloxacillin. Our structures, combined with pharmacophore mapping, molecular dynamics simulations and biochemical assays, establish the mechanism of beta-lactam antibiotic recognition and the important role of protonation in drug binding and transport.
Collapse
Affiliation(s)
- Joanne L Parker
- Department of Biochemistry, University of Oxford, Oxford, UK.
- The Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK.
| | - Justin C Deme
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute, Frederick, USA
| | | | - Gabriel Kuteyi
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Philip C Biggin
- Department of Biochemistry, University of Oxford, Oxford, UK.
| | - Susan M Lea
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute, Frederick, USA.
| | - Simon Newstead
- Department of Biochemistry, University of Oxford, Oxford, UK.
- The Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK.
| |
Collapse
|
3
|
Li H, Fang C, Hu Y, Xu J, Zhao W, Li L. The Comparative Analysis of Peptides in Enteral Nutrition Products and Foods for Special Medical Purposes. Foods 2024; 13:2557. [PMID: 39200483 PMCID: PMC11353486 DOI: 10.3390/foods13162557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/31/2024] [Accepted: 08/12/2024] [Indexed: 09/02/2024] Open
Abstract
Enteral nutrition (EN) and foods for special medical purposes (FSMPs) can be used to meet the specific nutritional needs of patients. There are multiple types of EN products and nutritionally complete FSMPs on the market. The peptides in these products are important nutritional components, while their presence in different products remains unclear. To provide better clinical guidelines, we analyzed and compared the molecular weight (MW) distribution, types, and quantities of peptides and bioactive peptides of two EN products (liquid products) and two FSMPs with nutritionally complete formulas (powder products). Our results showed that each product had a unique peptide profile. The two liquid products and one powder product (Samples 1-3) had a higher content of peptides. Sample 1 contained 75.60% peptides with an MW less than 375 Da and contained 95.21% peptides with an MW less than 1000 Da, being rich in short peptides. Sample 2 and 3 had high levels of peptides with MW values between 180 Da and 2000 Da. Additionally, Sample 4 contained high levels of proteins, containing 69.18% peptides with MW values larger than 10,000 Da. Further, Sample 1 had more bioactive dipeptides and Sample 2 had more long bioactive peptides. Our results suggest that peptides in different EN and FSMP products are very different and should be evaluated in more detail. This will provide valuable information for clinical medical professionals, help them to guide patients with different physiological conditions better, and ultimately benefit patients.
Collapse
Affiliation(s)
- Hao Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Chenlu Fang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yushan Hu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jing Xu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wei Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Li Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
4
|
Newstead S, Parker J, Deme J, Lichtinger S, Kuteyi G, Biggin P, Lea S. Structural basis for antibiotic transport and inhibition in PepT2, the mammalian proton-coupled peptide transporter. RESEARCH SQUARE 2024:rs.3.rs-4435259. [PMID: 38903084 PMCID: PMC11188089 DOI: 10.21203/rs.3.rs-4435259/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
The uptake and elimination of beta-lactam antibiotics in the human body are facilitated by the proton-coupled peptide transporters PepT1 (SLC15A1) and PepT2 (SLC15A2). The mechanism by which SLC15 family transporters recognize and discriminate between different drug classes and dietary peptides remains unclear, hampering efforts to improve antibiotic pharmacokinetics through targeted drug design and delivery. Here, we present cryo-EM structures of the mammalian proton-coupled peptide transporter, PepT2, in complex with the widely used beta-lactam antibiotics cefadroxil, amoxicillin and cloxacillin. Our structures, combined with pharmacophore mapping, molecular dynamics simulations and biochemical assays, establish the mechanism of antibiotic recognition and the important role of protonation in drug binding and transport.
Collapse
Affiliation(s)
| | | | - Justin Deme
- National Cancer Institute, National Institutes of Health
| | | | | | | | - Susan Lea
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute
| |
Collapse
|
5
|
Benfica LF, Brito LF, do Bem RD, de Oliveira LF, Mulim HA, Braga LG, Cyrillo JNSG, Bonilha SFM, Mercadante MEZ. Detection and characterization of copy number variation in three differentially-selected Nellore cattle populations. Front Genet 2024; 15:1377130. [PMID: 38694873 PMCID: PMC11061390 DOI: 10.3389/fgene.2024.1377130] [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: 01/26/2024] [Accepted: 04/05/2024] [Indexed: 05/04/2024] Open
Abstract
Introduction: Nellore cattle (Bos taurus indicus) is the main beef cattle breed raised in Brazil. This breed is well adapted to tropical conditions and, more recently, has experienced intensive genetic selection for multiple performance traits. Over the past 43 years, an experimental breeding program has been developed in the Institute of Animal Science (IZ, Sertaozinho, SP, Brazil), which resulted in three differentially-selected lines known as Nellore Control (NeC), Nellore Selection (NeS), and Nellore Traditional (NeT). The primary goal of this selection experiment was to determine the response to selection for yearling weight (YW) and residual feed intake (RFI) on Nellore cattle. The main objectives of this study were to: 1) identify copy number variation (CNVs) in Nellore cattle from three selection lines; 2) identify and characterize CNV regions (CNVR) on these three lines; and 3) perform functional enrichment analyses of the CNVR identified. Results: A total of 14,914 unique CNVs and 1,884 CNVRs were identified when considering all lines as a single population. The CNVRs were non-uniformly distributed across the chromosomes of the three selection lines included in the study. The NeT line had the highest number of CNVRs (n = 1,493), followed by the NeS (n = 823) and NeC (n = 482) lines. The CNVRs covered 23,449,890 bp (0.94%), 40,175,556 bp (1.61%), and 63,212,273 bp (2.54%) of the genome of the NeC, NeS, and NeT lines, respectively. Two CNVRs were commonly identified between the three lines, and six, two, and four exclusive regions were identified for NeC, NeS, and NeT, respectively. All the exclusive regions overlap with important genes, such as SMARCD3, SLC15A1, and MAPK1. Key biological processes associated with the candidate genes were identified, including pathways related to growth and metabolism. Conclusion: This study revealed large variability in CNVs and CNVRs across three Nellore lines differentially selected for YW and RFI. Gene annotation and gene ontology analyses of the exclusive CNVRs to each line revealed specific genes and biological processes involved in the expression of growth and feed efficiency traits. These findings contribute to the understanding of the genetic mechanisms underlying the phenotypic differences among the three Nellore selection lines.
Collapse
Affiliation(s)
- Lorena F. Benfica
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
- Department of Animal Science, Faculty of Agricultural and Veterinary Sciences, Sao Paulo State University, Jaboticabal, São Paulo, Brazil
| | - Luiz F. Brito
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Ricardo D. do Bem
- Department of Animal Science, Faculty of Agricultural and Veterinary Sciences, Sao Paulo State University, Jaboticabal, São Paulo, Brazil
| | | | - Henrique A. Mulim
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Larissa G. Braga
- Department of Animal Science, Faculty of Agricultural and Veterinary Sciences, Sao Paulo State University, Jaboticabal, São Paulo, Brazil
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | | | - Sarah F. M. Bonilha
- Beef Cattle Research Center, Institute of Animal Science, Sertaozinho, São Paulo, Brazil
| | - Maria Eugenia Z. Mercadante
- Department of Animal Science, Faculty of Agricultural and Veterinary Sciences, Sao Paulo State University, Jaboticabal, São Paulo, Brazil
- Beef Cattle Research Center, Institute of Animal Science, Sertaozinho, São Paulo, Brazil
| |
Collapse
|
6
|
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
|
7
|
Higuchi K, Kunieda M, Sugiyama K, Tomabechi R, Kishimoto H, Inoue K. Monocarboxylate Transporter 13 (MCT13/SLC16A13) Functions as a Novel Plasma Membrane Oligopeptide Transporter. Nutrients 2023; 15:3527. [PMID: 37630718 PMCID: PMC10458055 DOI: 10.3390/nu15163527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
SLC16A13, which encodes the monocarboxylate transporter 13 (MCT13), is a susceptibility gene for type 2 diabetes and is expressed in the liver and duodenum. Some peptidase-resistant oligopeptides are absorbed in the gastrointestinal tract and affect glycemic control in the body. Their efficient absorption is mediated by oligopeptide transporter(s) at the apical and basolateral membranes of the intestinal epithelia; however, the molecules responsible for basolateral oligopeptide transport have not been identified. In this study, we examined whether MCT13 functions as a novel basolateral oligopeptide transporter. We evaluated the uptake of oligopeptides and peptidomimetics in MCT13-transfected cells. The uptake of cephradine, a probe for peptide transport system(s), significantly increased in MCT13-transfected cells, and this increase was sensitive to membrane potential. The cellular accumulation of bioactive peptides, such as anserine and carnosine, was decreased by MCT13, indicating MCT13-mediated efflux transport activity. In polarized Caco-2 cells, MCT13 was localized at the basolateral membrane. MCT13 induction enhanced cephradine transport in an apical-to-basal direction across Caco-2 cells. These results indicate that MCT13 functions as a novel efflux transporter of oligopeptides and peptidomimetics, driven by electrochemical gradients across the plasma membrane, and it may be involved in the transport of these compounds across the intestinal epithelia.
Collapse
Affiliation(s)
- Kei Higuchi
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Tokyo 192-0392, Japan; (K.H.); (M.K.); (K.S.); (R.T.); (H.K.)
| | - Misato Kunieda
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Tokyo 192-0392, Japan; (K.H.); (M.K.); (K.S.); (R.T.); (H.K.)
| | - Koki Sugiyama
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Tokyo 192-0392, Japan; (K.H.); (M.K.); (K.S.); (R.T.); (H.K.)
| | - Ryuto Tomabechi
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Tokyo 192-0392, Japan; (K.H.); (M.K.); (K.S.); (R.T.); (H.K.)
- Laboratory of Pharmaceutics, Kitasato University School of Pharmacy, 5-9-1 Shirokane, Tokyo 108-8641, Japan
| | - Hisanao Kishimoto
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Tokyo 192-0392, Japan; (K.H.); (M.K.); (K.S.); (R.T.); (H.K.)
| | - Katsuhisa Inoue
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Tokyo 192-0392, Japan; (K.H.); (M.K.); (K.S.); (R.T.); (H.K.)
| |
Collapse
|
8
|
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
|
9
|
Khavinson V, Linkova N, Kozhevnikova E, Dyatlova A, Petukhov M. Transport of Biologically Active Ultrashort Peptides Using POT and LAT Carriers. Int J Mol Sci 2022; 23:ijms23147733. [PMID: 35887081 PMCID: PMC9323678 DOI: 10.3390/ijms23147733] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 01/27/2023] Open
Abstract
Ultrashort peptides (USPs), consisting of 2–7 amino-acid residues, are a group of signaling molecules that regulate gene expression and protein synthesis under normal conditions in various diseases and ageing. USPs serve as a basis for the development of drugs with a targeted mechanism of action. The purpose of this review is to systematize the available data on USP transport involving POT and LAT transporters in various organs and tissues under normal, pathological and ageing conditions. The carriers of the POT family (PEPT1, PEPT2, PHT1, PHT2) transport predominantly di- and tripeptides into the cell. Methods of molecular modeling and physicochemistry have demonstrated the ability of LAT1 to transfer not only amino acids but also some di- and tripeptides into the cell and out of it. LAT1 and 2 are involved in the regulation of the antioxidant, endocrine, immune and nervous systems’ functions. Analysis of the above data allows us to conclude that, depending on their structure, di- and tripeptides can be transported into the cells of various tissues by POT and LAT transporters. This mechanism is likely to underlie the tissue specificity of peptides, their geroprotective action and effectiveness in the case of neuroimmunoendocrine system disorders.
Collapse
Affiliation(s)
- Vladimir Khavinson
- Department of Biogerontology, Saint Petersburg Institute of Bioregulation and Gerontology, 197110 Saint Petersburg, Russia; (N.L.); (E.K.); (A.D.)
- Group of Peptide Regulation of Aging, Pavlov Institute of Physiology of Russian Academy of Sciences, 199034 Saint Petersburg, Russia
- Correspondence: or ; Tel.: +7-(921)-9110800
| | - Natalia Linkova
- Department of Biogerontology, Saint Petersburg Institute of Bioregulation and Gerontology, 197110 Saint Petersburg, Russia; (N.L.); (E.K.); (A.D.)
- The Laboratory “Problems of Aging”, Belgorod National Research University, 308015 Belgorod, Russia
| | - Ekaterina Kozhevnikova
- Department of Biogerontology, Saint Petersburg Institute of Bioregulation and Gerontology, 197110 Saint Petersburg, Russia; (N.L.); (E.K.); (A.D.)
| | - Anastasiia Dyatlova
- Department of Biogerontology, Saint Petersburg Institute of Bioregulation and Gerontology, 197110 Saint Petersburg, Russia; (N.L.); (E.K.); (A.D.)
| | - Mikhael Petukhov
- Petersburg Nuclear Physics Institute Named after B.P. Konstantinov, NRC “Kurchatov Institute”, 188300 Gatchina, Russia;
- Peter the Great St. Petersburg Group of Biophysics, Higher Engineering and Technical School, Peter the Great St. Petersburg Polytechnic University, 195251 Saint Petersburg, Russia
| |
Collapse
|
10
|
Kaulich E, Carroll T, Ackley BD, Tang YQ, Hardege I, Nehrke K, Schafer WR, Walker DS. Distinct roles for two Caenorhabditis elegans acid-sensing ion channels in an ultradian clock. eLife 2022; 11:e75837. [PMID: 35666106 PMCID: PMC9374441 DOI: 10.7554/elife.75837] [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: 11/24/2021] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
Biological clocks are fundamental to an organism's health, controlling periodicity of behaviour and metabolism. Here, we identify two acid-sensing ion channels, with very different proton sensing properties, and describe their role in an ultradian clock, the defecation motor program (DMP) of the nematode Caenorhabditis elegans. An ACD-5-containing channel, on the apical membrane of the intestinal epithelium, is essential for maintenance of luminal acidity, and thus the rhythmic oscillations in lumen pH. In contrast, the second channel, composed of FLR-1, ACD-3 and/or DEL-5, located on the basolateral membrane, controls the intracellular Ca2+ wave and forms a core component of the master oscillator that controls the timing and rhythmicity of the DMP. flr-1 and acd-3/del-5 mutants show severe developmental and metabolic defects. We thus directly link the proton-sensing properties of these channels to their physiological roles in pH regulation and Ca2+ signalling, the generation of an ultradian oscillator, and its metabolic consequences.
Collapse
Affiliation(s)
- Eva Kaulich
- Neurobiology Division, MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
| | - Trae Carroll
- Department of Pathology and Lab Medicine, University of Rochester Medical CenterRochesterUnited States
| | - Brian D Ackley
- Department of Molecular Biosciences, University of KansasLawrenceUnited States
| | - Yi-Quan Tang
- Neurobiology Division, MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Iris Hardege
- Neurobiology Division, MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
| | - Keith Nehrke
- Department of Medicine, Nephrology Division, University of Rochester Medical CenterRochesterUnited States
| | - William R Schafer
- Neurobiology Division, MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
- Department of Biology, KU LeuvenLeuvenBelgium
| | - Denise S Walker
- Neurobiology Division, MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
| |
Collapse
|
11
|
Jala A, Ponneganti S, Vishnubhatla DS, Bhuvanam G, Mekala PR, Varghese B, Radhakrishnanand P, Adela R, Murty US, Borkar RM. Transporter-mediated drug-drug interactions: advancement in models, analytical tools, and regulatory perspective. Drug Metab Rev 2021; 53:285-320. [PMID: 33980079 DOI: 10.1080/03602532.2021.1928687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 05/05/2021] [Indexed: 02/08/2023]
Abstract
Drug-drug interactions mediated by transporters are a serious clinical concern hence a tremendous amount of work has been done on the characterization of the transporter-mediated proteins in humans and animals. The underlying mechanism for the transporter-mediated drug-drug interaction is the induction or inhibition of the transporter which is involved in the cellular uptake and efflux of drugs. Transporter of the brain, liver, kidney, and intestine are major determinants that alter the absorption, distribution, metabolism, excretion profile of drugs, and considerably influence the pharmacokinetic profile of drugs. As a consequence, transporter proteins may affect the therapeutic activity and safety of drugs. However, mounting evidence suggests that many drugs change the activity and/or expression of the transporter protein. Accordingly, evaluation of drug interaction during the drug development process is an integral part of risk assessment and regulatory requirements. Therefore, this review will highlight the clinical significance of the transporter, their role in disease, possible cause underlying the drug-drug interactions using analytical tools, and update on the regulatory requirement. The recent in-silico approaches which emphasize the advancement in the discovery of drug-drug interactions are also highlighted in this review. Besides, we discuss several endogenous biomarkers that have shown to act as substrates for many transporters, which could be potent determinants to find the drug-drug interactions mediated by transporters. Transporter-mediated drug-drug interactions are taken into consideration in the drug approval process therefore we also provided the extrapolated decision trees from in-vitro to in-vivo, which may trigger the follow-up to clinical studies.
Collapse
Affiliation(s)
- Aishwarya Jala
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Srikanth Ponneganti
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Devi Swetha Vishnubhatla
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Gayathri Bhuvanam
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Prithvi Raju Mekala
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Bincy Varghese
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Pullapanthula Radhakrishnanand
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Ramu Adela
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | | | - Roshan M Borkar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| |
Collapse
|
12
|
Liu X, Lyu W, Liu L, Lv K, Zheng F, Wang Y, Chen J, Dai B, Yang H, Xiao Y. Comparison of Digestive Enzyme Activities and Expression of Small Intestinal Transporter Genes in Jinhua and Landrace Pigs. Front Physiol 2021; 12:669238. [PMID: 34194337 PMCID: PMC8236719 DOI: 10.3389/fphys.2021.669238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/07/2021] [Indexed: 11/24/2022] Open
Abstract
Digestive enzyme activity is involved in the regulation of growth performance because digestive enzymes function to improve the feed efficiency by digestion and in turn to modulate the process of nutrient metabolism. The objective of this study was to investigate the differences of the digestive enzyme activities and expression of nutrient transporters in the intestinal tract between Jinhua and Landrace pigs and to explore the potential breed-specificity in digestion and absorption. The pancreas segments and the digesta and mucosa of the duodenum, jejunum, and ileum were collected from 10 Jinhua pigs and Landrace pigs, respectively. The activities of trypsin, chymotrypsin, amylase, maltase, sucrase, and lipase were measured and the expression levels of PepT1, GLUT2, SGLT1, FABP1, FABP2, and FABP4 were examined. Results showed that the trypsin activity in the pancreas of Jinhua pigs was higher than that in Landrace pigs, but was lower in the small intestine, except for in the jejunal mucosa. The chymotrypsin activity in the small intestine of Jinhua pigs was higher than that in Landrace pigs, except for in jejunal mucosa and contents. Compared with Landrace pigs, the amylase and maltase activity in the small intestine of Jinhua pigs was lower, except for in ileal mucosa. The sucrase activity in the small intestine of Jinhua pigs was also lower than Landrace pigs, except for in jejunal mucosa. Furthermore, the lipase activity in the small intestine of Jinhua pigs was higher than that in Landrace pigs. The mRNA levels of PepT1 and GLUT2 in duodenal, jejunal and ileal mucosa showed no difference between Jinhua and Landrace pigs, whereas SGLT1 in ileal mucosa was lower in Jinhua pigs. The mRNA levels of FABP1, FABP2 and FABP4 in the small intestinal mucosa of Jinhua pigs were higher than in Landrace pigs. These findings indicate that there is a certain difference in the digestibility and absorption of nutrients in small intestine of Jinhua and Landrace pigs, partially resulting in their differences in growth development and fat deposition.
Collapse
Affiliation(s)
- Xiuting Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Wentao Lyu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Lei Liu
- Zhejiang Goshine Test Technologies Co., Ltd., Hangzhou, China
| | - Kaikai Lv
- Zhejiang Goshine Test Technologies Co., Ltd., Hangzhou, China
| | - Fen Zheng
- Agricultural and Rural Bureau of Kaihua County, Kaihua, China
| | - Yuanxia Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Jinggang Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Bing Dai
- Zhejiang Goshine Test Technologies Co., Ltd., Hangzhou, China
| | - Hua Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Yingping Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| |
Collapse
|
13
|
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: 0.5] [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
|
14
|
Labarta-Bajo L, Nilsen SP, Humphrey G, Schwartz T, Sanders K, Swafford A, Knight R, Turner JR, Zúñiga EI. Type I IFNs and CD8 T cells increase intestinal barrier permeability after chronic viral infection. J Exp Med 2021; 217:152069. [PMID: 32880630 PMCID: PMC7953738 DOI: 10.1084/jem.20192276] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 06/29/2020] [Accepted: 08/06/2020] [Indexed: 12/16/2022] Open
Abstract
Intestinal barrier leakage constitutes a potential therapeutic target for many inflammatory diseases and represents a disease progression marker during chronic viral infections. However, the causes of altered gut barrier remain mostly unknown. Using murine infection with lymphocytic choriomeningitis virus, we demonstrate that, in contrast to an acute viral strain, a persistent viral isolate leads to long-term viral replication in hematopoietic and mesenchymal cells, but not epithelial cells (IECs), in the intestine. Viral persistence drove sustained intestinal epithelial barrier leakage, which was characterized by increased paracellular flux of small molecules and was associated with enhanced colitis susceptibility. Type I IFN signaling caused tight junction dysregulation in IECs, promoted gut microbiome shifts and enhanced intestinal CD8 T cell responses. Notably, both type I IFN receptor blockade and CD8 T cell depletion prevented infection-induced barrier leakage. Our study demonstrates that infection with a virus that persistently replicates in the intestinal mucosa increases epithelial barrier permeability and reveals type I IFNs and CD8 T cells as causative factors of intestinal leakage during chronic infections.
Collapse
Affiliation(s)
- Lara Labarta-Bajo
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA
| | - Steven P Nilsen
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Gregory Humphrey
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Tara Schwartz
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Karenina Sanders
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Austin Swafford
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA
| | - Rob Knight
- Department of Pediatrics, University of California, San Diego, La Jolla, CA.,Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA.,Department of Bioengineering, University of California, San Diego, La Jolla, CA.,Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Elina I Zúñiga
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA
| |
Collapse
|
15
|
Ran T, Li H, Liu Y, Zhou C, He Z, Tan Z, Yang W, Beauchemin KA. Cloning, phylogenetic analysis, and postnatal expression of oligopeptide transporter PepT1 in gastrointestinal tract of kid goats receiving supplemental feed or pasture. CANADIAN JOURNAL OF ANIMAL SCIENCE 2020. [DOI: 10.1139/cjas-2019-0155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study aimed to clone the cDNA of PepT1, an H+-dependent oligopeptide transporter, from kid goats and examine effects of physiological development (suckling, weaning, and post-weaning) of the animal and feeding system (supplemental feeding vs. grazing) on peptide transport capability. A 2395 bp cDNA sequence of pept1 (GenBank: MH308024) was cloned and phylogenetic analysis revealed a high homology and structure similarity with PepT1 of sheep and cattle. The pept1 was expressed throughout the gastrointestinal tract of kid goats immediately after birth and during development. Relative abundance of pept1 decreased in all segments except the middle-jejunum during suckling, whereas its expression in most segments of small intestine increased with age after weaning and remained stable thereafter. Middle-jejunum was the predominant expression site and probably the main peptide absorption site. Supplemental feeding enhanced pept1 expression because it increased protein intake compared with grazing. No feeding system × age interaction was observed in most segments; the expression was age related during suckling and diet related during weaning and post-weaning, indicating that feeding system and age had independent effects on pept1 expression. These results indicate that PepT1 plays an important role for protein nutrition in neonatal goats, and its expression can be affected by feeding system.
Collapse
Affiliation(s)
- Tao Ran
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, and National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, and South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan 410125, People’s Republic of China
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB T1J 4B1, Canada
- Faculty of Veterinary Medicine, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Hengzhi Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People’s Republic of China
| | - Yong Liu
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, and National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, and South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan 410125, People’s Republic of China
| | - Chuanshe Zhou
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, and National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, and South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan 410125, People’s Republic of China
- Hunan Co-Innovation Centre of Animal Production Safety, CICAPS, Changsha, Hunan 410128, People’s Republic of China
| | - Zhixiong He
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, and National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, and South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan 410125, People’s Republic of China
| | - Zhiliang Tan
- CAS Key Laboratory for Agro-Ecological Processes in Subtropical Region, and National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, and South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, the Chinese Academy of Sciences, Changsha, Hunan 410125, People’s Republic of China
- Hunan Co-Innovation Centre of Animal Production Safety, CICAPS, Changsha, Hunan 410128, People’s Republic of China
| | - Wenzhu Yang
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB T1J 4B1, Canada
| | - Karen A. Beauchemin
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB T1J 4B1, Canada
| |
Collapse
|
16
|
Yuri T, Kono Y, Okada T, Terada T, Miyauchi S, Fujita T. Transport Characteristics of 5-Aminosalicylic Acid Derivatives Conjugated with Amino Acids via Human H +-Coupled Oligopeptide Transporter PEPT1. Biol Pharm Bull 2020; 43:697-706. [PMID: 32238712 DOI: 10.1248/bpb.b19-01048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
5-Aminosalicylic acid (5-ASA) is used as first line therapy for symptom remission and maintenance of inflammatory bowel disease (IBD). Because 5-ASA is well absorbed from the small intestine when orally administered, several 5-ASA formulations for selective delivery to the colon have been developed and used in clinical practice. However, its delivery efficiency to local inflamed colonic sites remains low. Intestinal H+-coupled oligopeptide transporter 1 (PEPT1) expression in the colon is low, whereas its expression is induced in the colon under chronic inflammation conditions, such as IBD. Therefore, we considered that PEPT1 would be a target transporter to improve 5-ASA delivery efficiency to local colonic lesions. We evaluated the transport characteristics of dipeptide-like 5-ASA derivatives, which were coupling glycine (Gly), lysine, glutamic acid (Glu), valine (Val) and tyrosine to amino or carboxyl group of 5-ASA, in Caco-2 cells. [3H]Glycylsarcosine (Gly-Sar) uptake into Caco-2 cells was inhibited by all 5-ASA derivatives. In addition, 5-ASA derivatives (Gly-ASA, Glu-ASA and Val-ASA), which were coupled by glycine, glutamic acid and valine to amino group of 5-ASA, were taken up in a pH- and concentration-dependent manner and their uptake was inhibited by excess Gly-Sar. Two-electrode voltage-clamp experiment using human PEPT1 expressing Xenopus oocytes showed that Gly-ASA, Glu-ASA and Val-ASA induced marked currents at pH 6.0. Taken together, these results showed that these 5-ASA derivatives are transportable substrates for PEPT1.
Collapse
Affiliation(s)
- Tatsushi Yuri
- Graduate School of Pharmaceutical Sciences, Ritsumeikan University
| | - Yusuke Kono
- Ritsumeikan Global-Innovation Research Organization (R-GIRO), Ritsumeikan University.,College of Pharmaceutical Sciences, Ritsumeikan University
| | - Tomofumi Okada
- College of Pharmaceutical Sciences, Ritsumeikan University
| | - Tomohiro Terada
- Department of Pharmacy, Shiga University of Medical Science Hospital
| | | | - Takuya Fujita
- Graduate School of Pharmaceutical Sciences, Ritsumeikan University.,Ritsumeikan Global-Innovation Research Organization (R-GIRO), Ritsumeikan University.,College of Pharmaceutical Sciences, Ritsumeikan University
| |
Collapse
|
17
|
Li Y, Yuan S, Yong X, zhao T, Liu J. Research progress on small peptides in Chinese Baijiu. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104081] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
18
|
Maes M, Anderson G, Betancort Medina SR, Seo M, Ojala JO. Integrating Autism Spectrum Disorder Pathophysiology: Mitochondria, Vitamin A, CD38, Oxytocin, Serotonin and Melatonergic Alterations in the Placenta and Gut. Curr Pharm Des 2020; 25:4405-4420. [PMID: 31682209 DOI: 10.2174/1381612825666191102165459] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 10/31/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND A diverse array of data has been associated with autism spectrum disorder (ASD), reflecting the complexity of its pathophysiology as well as its heterogeneity. Two important hubs have emerged, the placenta/prenatal period and the postnatal gut, with alterations in mitochondria functioning crucial in both. METHODS Factors acting to regulate mitochondria functioning in ASD across development are reviewed in this article. RESULTS Decreased vitamin A, and its retinoic acid metabolites, lead to a decrease in CD38 and associated changes that underpin a wide array of data on the biological underpinnings of ASD, including decreased oxytocin, with relevance both prenatally and in the gut. Decreased sirtuins, poly-ADP ribose polymerase-driven decreases in nicotinamide adenine dinucleotide (NAD+), hyperserotonemia, decreased monoamine oxidase, alterations in 14-3-3 proteins, microRNA alterations, dysregulated aryl hydrocarbon receptor activity, suboptimal mitochondria functioning, and decreases in the melatonergic pathways are intimately linked to this. Many of the above processes may be modulating, or mediated by, alterations in mitochondria functioning. Other bodies of data associated with ASD may also be incorporated within these basic processes, including how ASD risk factors such as maternal obesity and preeclampsia, as well as more general prenatal stressors, modulate the likelihood of offspring ASD. CONCLUSION Such a mitochondria-focussed integrated model of the pathophysiology of ASD has important preventative and treatment implications.
Collapse
Affiliation(s)
- Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - George Anderson
- CRC Scotland & London, Eccleston Square, London, United Kingdom
| | | | - Moonsang Seo
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Johanna O Ojala
- Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
19
|
The Radiolabeling of a Gly-Sar Dipeptide Derivative with Flourine-18 and Its Use as a Potential Peptide Transporter PET Imaging Agent. Molecules 2020; 25:molecules25030643. [PMID: 32024310 PMCID: PMC7037988 DOI: 10.3390/molecules25030643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/27/2020] [Accepted: 01/30/2020] [Indexed: 11/23/2022] Open
Abstract
We have developed a novel fluorine-18 radiotracer, dipeptide 1, radiolabeled in two steps from mesylate 3. The initial radiolabeling is achieved in a short reaction time (10 min) and purified through solid-phase extraction (SPE) with modest radiochemical yields (rcy = 10 ± 2%, n = 5) in excellent radiochemical purity (rcp > 99%, n = 5). The de-protection of the tert-butyloxycarbonyl (Boc) and trityl group was achieved with mild heating under acidic conditions to provide 18F-tagged dipeptide 1. Preliminary analysis of 18F-dipeptide 1 was performed to confirm uptake by peptide transporters (PepTs) in human pancreatic carcinoma cell lines Panc1, BxPC3, and ASpc1, which are reported to express the peptide transporter 1 (PepT1). Furthermore, we confirmed in vivo uptake of 18F-dipeptide tracer 1 using microPET/CT in mice harboring subcutaneous flank Panc1, BxPC3, and Aspc1 tumors. In conclusion, we have established the radiolabeling of dipeptide 1 with fluoride-18, and demonstrated its potential as an imaging agent which may have clinical applications for the diagnosis of pancreatic carcinomas.
Collapse
|
20
|
Kudo M, Kobayashi-Nakamura K, Kitajima N, Tsuji-Naito K. Alternate expression of PEPT1 and PEPT2 in epidermal differentiation is required for NOD2 immune responses by bacteria-derived muramyl dipeptide. Biochem Biophys Res Commun 2020; 522:151-156. [PMID: 31757425 DOI: 10.1016/j.bbrc.2019.11.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 11/05/2019] [Indexed: 11/30/2022]
Abstract
Peptide transporters 1 and 2 (PEPT1 and PEPT2) are proton-coupled oligopeptide transporter members of the solute carrier 15 family and play a role in the cellular uptake of di/tri-peptides and peptidomimetics. Our previous work showed that PEPT2 is predominantly expressed within undifferentiated keratinocytes. Here we show that PEPT2 expression decreases as keratinocyte differentiation progresses and that PEPT1 alternately is expressed at later stages. Absolute quantification using quantitative polymerase chain reaction revealed that the expression level of PEPT1 is about 17 times greater than that of PEPT2. Immunohistochemical study of human skin provided evidence of PEPT1 in the epidermis. The uptake of glycylsarcosine into keratinocytes was significantly blocked by PEPT inhibitors, including nateglinide and glibenclamide. Moreover, we found that PEPT1 knockdown in differentiated keratinocytes significantly suppressed the influence of a bacterial-derived peptide, muramyl dipeptide (MDP), on the production of proinflammatory cytokine interleukin-8, implying that bacteria-derived oligopeptides can be transported by PEPT1 in advanced differentiated keratinocytes. Taken together, PEPT1 and PEPT2 may concertedly play an important role in MDP-NOD2 signaling in the epidermis, which provides new insight into the mechanisms of skin homeostasis against microbial pathogens.
Collapse
Affiliation(s)
- Michiko Kudo
- DHC Corporation, Fundamental Research Laboratory, Division 2, 2-42 Hamada, Mihama-ku, Chiba, 261-0025, Japan
| | - Kumiko Kobayashi-Nakamura
- DHC Corporation, Fundamental Research Laboratory, Division 2, 2-42 Hamada, Mihama-ku, Chiba, 261-0025, Japan
| | - Natsuko Kitajima
- DHC Corporation, Fundamental Research Laboratory, Division 2, 2-42 Hamada, Mihama-ku, Chiba, 261-0025, Japan
| | - Kentaro Tsuji-Naito
- DHC Corporation, Fundamental Research Laboratory, Division 2, 2-42 Hamada, Mihama-ku, Chiba, 261-0025, Japan.
| |
Collapse
|
21
|
Karimian Pour N, Piquette-Miller M. Dysregulation of renal transporters in a rodent model of viral Infection. Int Immunopharmacol 2020; 80:106135. [PMID: 31951958 DOI: 10.1016/j.intimp.2019.106135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/11/2019] [Accepted: 12/16/2019] [Indexed: 01/01/2023]
Abstract
Inflammation elicited by viral mimetic poly I:C has been shown to impose changes in the expression of drug transporters in the placenta and maternal liver in rats at term pregnancy. This was associated with altered drug disposition in the mother and fetus. Renal transporters play an important role in the elimination of several drugs taken by pregnant women. We examined the impact of poly I:C on the expression of renal transporters in pregnant rats at term. Pregnant Sprague-Dawley rats received single intraperitoneal dose of either poly I:C (5 mg/kg) or saline at gestation day 18 (n = 8/group). Animals were euthanized 24 h after the injection. The mRNA and protein expression of pro-inflammatory cytokines and transporters were measured by qRT-PCR and western blot. Poly I:C caused a fourfold increase in the mRNA of IL-6 in the kidney. As compared to saline controls, the mRNA expression of Mrp2, Bcrp, Octn1, Oat1, Oat2, Oat3, Urat1, Oatp4c1, and Pept2 was downregulated, whereas the Ent1 mRNA was increased. Protein expression of Bcrp, Urat1 and Pept2 were significantly decreased. While there was a trend towards reduced Mrp2, Oat2 and Oat3 protein expression, this did not reach significance. Poly I:C did not impact mRNA levels of Mdr1a, Mdr1b, Mrp4, Oct1, Oct2, Oct3, Octn2, Mate1, Ent2 or Pept1. Viral-induced inflammation mediates significant changes in the expression of several key drug transporters in the kidney of pregnant rats. Many clinically important drugs are substrates for these transporters. Therefore, inflammation-mediated alterations in transporter expression could affect their maternal and fetal disposition.
Collapse
Affiliation(s)
- Navaz Karimian Pour
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.
| | | |
Collapse
|
22
|
Petrick JS, Bell E, Koch MS. Weight of the evidence: independent research projects confirm industry conclusions on the safety of insect-protected maize MON 810. GM CROPS & FOOD 2019; 11:30-46. [PMID: 31651217 PMCID: PMC7064210 DOI: 10.1080/21645698.2019.1680242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 01/16/2023]
Abstract
The cumulative weight of the evidence demonstrates the safety and equivalence of genetically engineered (GE) crops compared to the conventional varieties from which they have been derived. Confirmatory toxicology and animal nutrition studies have nevertheless become an expected/mandated component of GE crop safety assessments, despite the lack of additional value these studies provide for product safety assessment. Characterization and safety data (e.g. trait protein safety; molecular, compositional, and agronomic/phenotypic assessments), and animal feeding studies form a weight of the evidence supporting the safety of insect-protected maize MON 810. Independent animal testing has recently confirmed the lack of MON 810 toxicity in subchronic and chronic toxicity studies. These results could have been predicted from the available safety data. Animal testing of GE crops should be supported by testable scientific hypotheses and testing should be consistent with ethical obligations to reduce, refine, and replace (3Rs) animal testing when possible.
Collapse
Affiliation(s)
- Jay S. Petrick
- Product Safety Center, Bayer Crop Science, Chesterfield, Missouri, USA
| | - Erin Bell
- Product Safety Center, Monsanto Company, Chesterfield, Missouri, USA
| | - Michael S. Koch
- Product Safety Center, Bayer Crop Science, Chesterfield, Missouri, USA
| |
Collapse
|
23
|
Xu Q, Hong H, Wu J, Yan X. Bioavailability of bioactive peptides derived from food proteins across the intestinal epithelial membrane: A review. Trends Food Sci Technol 2019. [DOI: 10.1016/j.tifs.2019.02.050] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
24
|
Amino acid transporters in the regulation of insulin secretion and signalling. Biochem Soc Trans 2019; 47:571-590. [PMID: 30936244 DOI: 10.1042/bst20180250] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 02/24/2019] [Accepted: 02/25/2019] [Indexed: 01/02/2023]
Abstract
Amino acids are increasingly recognised as modulators of nutrient disposal, including their role in regulating blood glucose through interactions with insulin signalling. More recently, cellular membrane transporters of amino acids have been shown to form a pivotal part of this regulation as they are primarily responsible for controlling cellular and circulating amino acid concentrations. The availability of amino acids regulated by transporters can amplify insulin secretion and modulate insulin signalling in various tissues. In addition, insulin itself can regulate the expression of numerous amino acid transporters. This review focuses on amino acid transporters linked to the regulation of insulin secretion and signalling with a focus on those of the small intestine, pancreatic β-islet cells and insulin-responsive tissues, liver and skeletal muscle. We summarise the role of the amino acid transporter B0AT1 (SLC6A19) and peptide transporter PEPT1 (SLC15A1) in the modulation of global insulin signalling via the liver-secreted hormone fibroblast growth factor 21 (FGF21). The role of vesicular vGLUT (SLC17) and mitochondrial SLC25 transporters in providing glutamate for the potentiation of insulin secretion is covered. We also survey the roles SNAT (SLC38) family and LAT1 (SLC7A5) amino acid transporters play in the regulation of and by insulin in numerous affective tissues. We hypothesise the small intestine amino acid transporter B0AT1 represents a crucial nexus between insulin, FGF21 and incretin hormone signalling pathways. The aim is to give an integrated overview of the important role amino acid transporters have been found to play in insulin-regulated nutrient signalling.
Collapse
|
25
|
Shen H, Scialis RJ, Lehman-McKeeman L. Xenobiotic Transporters in the Kidney: Function and Role in Toxicity. Semin Nephrol 2019; 39:159-175. [DOI: 10.1016/j.semnephrol.2018.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
26
|
Schäfer N, Rikkala PR, Veyhl-Wichmann M, Keller T, Jurowich CF, Geiger D, Koepsell H. A Modified Tripeptide Motif of RS1 ( RSC1A1) Down-Regulates Exocytotic Pathways of Human Na +-d-glucose Cotransporters SGLT1, SGLT2, and Glucose Sensor SGLT3 in the Presence of Glucose. Mol Pharmacol 2019; 95:82-96. [PMID: 30355744 DOI: 10.1124/mol.118.113514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/19/2018] [Indexed: 12/11/2022] Open
Abstract
A domain of protein RS1 (RSC1A1) called RS1-Reg down-regulates the plasma membrane abundance of Na+-d-glucose cotransporter SGLT1 by blocking the exocytotic pathway at the trans-Golgi. This effect is blunted by intracellular glucose but prevails when serine in a QSP (Gln-Ser-Pro) motif is replaced by glutamate [RS1-Reg(S20E)]. RS1-Reg binds to ornithine decarboxylase (ODC) and inhibits ODC in a glucose-dependent manner. Because the ODC inhibitor difluoromethylornithine (DFMO) acts like RS1-Reg(S20E), and DFMO and RS1-Reg(S20E) are not cumulative, we raised the hypothesis that RS1-Reg(S20E) down-regulates the exocytotic pathway of SGLT1 at the trans-Golgi by inhibiting ODC. We investigated whether QEP down-regulates human SGLT1 (hSGLT1) like hRS1-Reg(S20E) and whether human Na+-d-glucose cotransporter hSGLT2 and the human glucose sensor hSGLT3 are also addressed. We expressed hSGLT1, hSGLT1 linked to yellow fluorescent protein (hSGLT1-YFP), hSGLT2-YFP and hSGLT3-YFP in oocytes of Xenopus laevis, injected hRS1-Reg(S20E), QEP, DFMO, and/or α-methyl-d-glucopyranoside (AMG), and measured AMG uptake, glucose-induced currents, and plasma membrane-associated fluorescence after 1 hour. We also performed in vitro AMG uptake measurements into small intestinal mucosa of mice and human. The data indicate that QEP down-regulates the exocytotic pathway of SGLT1 similar to hRS1-Reg(S20E). Our results suggests that both peptides also down-regulate hSGLT2 and hSGLT3 via the same pathway. Thirty minutes after application of 5 mM QEP in the presence of 5 mM d-glucose, hSGLT1-mediated AMG uptake into small intestinal mucosa was decreased by 40% to 50%. Thus oral application of QEP in a formulation that optimizes uptake into enterocytes but prevents entry into the blood is proposed as novel antidiabetic therapy.
Collapse
Affiliation(s)
- Nadine Schäfer
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute (N.S., T.K., D.G., H.K.) and Institute of Anatomy and Cell Biology (P.R.R., M.V.-W., H.K.), University of Würzburg, Würzburg, Germany; and Department of General, Visceral, Vascular, and Paedriatic Surgery, University Hospital of Würzburg, Würzburg, Germany (C.F.J.)
| | - Prashanth Reddy Rikkala
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute (N.S., T.K., D.G., H.K.) and Institute of Anatomy and Cell Biology (P.R.R., M.V.-W., H.K.), University of Würzburg, Würzburg, Germany; and Department of General, Visceral, Vascular, and Paedriatic Surgery, University Hospital of Würzburg, Würzburg, Germany (C.F.J.)
| | - Maike Veyhl-Wichmann
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute (N.S., T.K., D.G., H.K.) and Institute of Anatomy and Cell Biology (P.R.R., M.V.-W., H.K.), University of Würzburg, Würzburg, Germany; and Department of General, Visceral, Vascular, and Paedriatic Surgery, University Hospital of Würzburg, Würzburg, Germany (C.F.J.)
| | - Thorsten Keller
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute (N.S., T.K., D.G., H.K.) and Institute of Anatomy and Cell Biology (P.R.R., M.V.-W., H.K.), University of Würzburg, Würzburg, Germany; and Department of General, Visceral, Vascular, and Paedriatic Surgery, University Hospital of Würzburg, Würzburg, Germany (C.F.J.)
| | - Christian Ferdinand Jurowich
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute (N.S., T.K., D.G., H.K.) and Institute of Anatomy and Cell Biology (P.R.R., M.V.-W., H.K.), University of Würzburg, Würzburg, Germany; and Department of General, Visceral, Vascular, and Paedriatic Surgery, University Hospital of Würzburg, Würzburg, Germany (C.F.J.)
| | - Dietmar Geiger
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute (N.S., T.K., D.G., H.K.) and Institute of Anatomy and Cell Biology (P.R.R., M.V.-W., H.K.), University of Würzburg, Würzburg, Germany; and Department of General, Visceral, Vascular, and Paedriatic Surgery, University Hospital of Würzburg, Würzburg, Germany (C.F.J.)
| | - Hermann Koepsell
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute (N.S., T.K., D.G., H.K.) and Institute of Anatomy and Cell Biology (P.R.R., M.V.-W., H.K.), University of Würzburg, Würzburg, Germany; and Department of General, Visceral, Vascular, and Paedriatic Surgery, University Hospital of Würzburg, Würzburg, Germany (C.F.J.)
| |
Collapse
|
27
|
Sun Y, Gan W, Lei M, Jiang W, Cheng M, He J, Sun Q, Liu W, Hu L, Jin Y. PEPT1-mediated prodrug strategy for oral delivery of peramivir. Asian J Pharm Sci 2018; 13:555-565. [PMID: 32104429 PMCID: PMC7032255 DOI: 10.1016/j.ajps.2018.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 05/20/2018] [Accepted: 05/28/2018] [Indexed: 12/11/2022] Open
Abstract
Peramivir was a novel and highly potent neuraminidase (NA) inhibitor for the treatment of influenza A and B. However, it exhibited a very low oral bioavailability (only 3%) due to the high polarity (log P of -1.4) and the low membrane permeability across the intestine. To utilize the PEPT1-mediated prodrug strategy to improve the oral absorption and develop the oral alternative, seven amino acid ester prodrugs and seven amino acid amide prodrugs have been synthesized. The permeability of these prodrugs across Caco-2 cells were screened. Peramivr-(CH2)2-l-Val and Peramivir-l-Ile were of the highest permeability in ester prodrugs and amide prodrugs, respectively, and then they were selected for further studies. Glycylsarcosine (gly-sar) uptake by Caco-2 could be inbihited by Peramivir-(CH2)2-l-Val and Peramivir-l-Ile in a concentration-dependent manner, and the IC50 was 1.34 ± 0.31 mM and 1.78 ± 0.48 mM, respectively. The direct uptake of Peramivir-(CH2)2-l-Val and Peramivir-l-Ile in MDCK-PEPT1 cells were significantly higher than in MDCK mock cells, and could be markedly inhibited by gly-sar. The uptake of Peramivir-(CH2)2-l-Val and Peramivir-l-Ile (0.01 to 50 mM) in MDCK-hPEPT1 cells conformed to Michaelis-Menten Equation. The oral bioavailability of peramivir was 65.3% and 37.3% after the oral administration of Peramivir-(CH2)2-l-Val and Peramivir-l-Ile to rats, respectively. The oral absorption and bioactivation of Peramivir-(CH2)2-l-Val was rapid and extensive, and no Peramivir-(CH2)2-l-Val was found in plasma. Because the amide bond was relatively stable, Peramivir-l-Ile could not be totally converted to the parent drug in vivo. Peramivir-(CH2)2-l-Val with good oral profiles and rapid bioactivation might be a promising prodrug for the further clinic development. The present study also corroborated the idea that the PEPT1-mediated prodrug approach has enormous promise for improving the oral absorption of poorly absorbed drug.
Collapse
Affiliation(s)
- Yongbing Sun
- Division of Pharmaceutics, National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, 56 Yangming Road, Nanchang 330006, China
| | - Wei Gan
- Division of Pharmaceutics, National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, 56 Yangming Road, Nanchang 330006, China
| | - Mingdao Lei
- Department of Pharmacy, Jiangxi Maternal and Child Health Hospital, NO 318 Bayi Road, Nanchang 330001, China
| | - Wei Jiang
- Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang 330004, China
| | - Meng Cheng
- Division of Pharmaceutics, National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, 56 Yangming Road, Nanchang 330006, China
| | - Junwei He
- Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang 330004, China
| | - Qi Sun
- Division of Pharmaceutics, National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, 56 Yangming Road, Nanchang 330006, China
| | - Wan Liu
- Division of Pharmaceutics, National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, 56 Yangming Road, Nanchang 330006, China
| | - Lvjiang Hu
- Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, NO 445 Bayi Road, Nanchang 330006, China
| | - Yi Jin
- Division of Pharmaceutics, National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, 56 Yangming Road, Nanchang 330006, China
| |
Collapse
|
28
|
P-glycoprotein expression in the gastrointestinal tract of male and female rats is influenced differently by food. Eur J Pharm Sci 2018; 123:569-575. [DOI: 10.1016/j.ejps.2018.08.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 12/17/2022]
|
29
|
Tailoring acyclovir prodrugs with enhanced antiviral activity: rational design, synthesis, human plasma stability and in vitro evaluation. Amino Acids 2018; 50:1131-1143. [DOI: 10.1007/s00726-018-2590-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/12/2018] [Indexed: 12/14/2022]
|
30
|
Spanier B, Rohm F. Proton Coupled Oligopeptide Transporter 1 (PepT1) Function, Regulation, and Influence on the Intestinal Homeostasis. Compr Physiol 2018; 8:843-869. [PMID: 29687907 DOI: 10.1002/cphy.c170038] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
31
|
Viennois E, Pujada A, Zen J, Merlin D. Function, Regulation, and Pathophysiological Relevance of the POT Superfamily, Specifically PepT1 in Inflammatory Bowel Disease. Compr Physiol 2018; 8:731-760. [PMID: 29687900 DOI: 10.1002/cphy.c170032] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mammalian members of the proton-coupled oligopeptide transporter family are integral membrane proteins that mediate the cellular uptake of di/tripeptides and peptide-like drugs and couple substrate translocation to the movement of H+ , with the transmembrane electrochemical proton gradient providing the driving force. Peptide transporters are responsible for the (re)absorption of dietary and/or bacterial di- and tripeptides in the intestine and kidney and maintaining homeostasis of neuropeptides in the brain. These proteins additionally contribute to absorption of a number of pharmacologically important compounds. In this overview article, we have provided updated information on the structure, function, expression, localization, and activities of PepT1 (SLC15A1), PepT2 (SLC15A2), PhT1 (SLC15A4), and PhT2 (SLC15A3). Peptide transporters, in particular, PepT1 are discussed as drug-delivery systems in addition to their implications in health and disease. Particular emphasis has been placed on the involvement of PepT1 in the physiopathology of the gastrointestinal tract, specifically, its role in inflammatory bowel diseases. © 2018 American Physiological Society. Compr Physiol 8:731-760, 2018.
Collapse
Affiliation(s)
- Emilie Viennois
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Adani Pujada
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Jane Zen
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Didier Merlin
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA.,Veterans Affairs Medical Center, Decatur, Georgia, USA
| |
Collapse
|
32
|
Abstract
With the global population rising, the need for sustainable and resource-efficiently produced proteins with nutritional and health promoting qualities has become urgent. Proteins are important macronutrients and are involved in most, if not all, biological processes in the human body. This review discusses these absorption mechanisms in the small intestine. To study intestinal transport and predict bioavailability, cell lines are widely applied as screening models and often concern Caco-2, HT-29, HT-29/MTX and T84 cells. Here, we provide an overview of the presence and activities of peptide- and amino acid transporters in these cell models. Further, inter-laboratory differences are discussed as well as the culture micro-environment, both of which may influence cell culture phenotype and performance. Finally, the value of new developments in the field, including culturing cells in 3-dimensional systems under shear stress (i.e., gut-on-chips), is highlighted. In particular, their suitability in screening novel food proteins and prediction of the nutritional quality needed for inclusion in the human diet of the future is addressed.
Collapse
|
33
|
Xu Q, Liu Z, Liu H, Zhao F, Huang X, Wu Y, Liu J. Functional characterization of oligopeptide transporter 1 of dairy cows. J Anim Sci Biotechnol 2018; 9:7. [PMID: 29387385 PMCID: PMC5778758 DOI: 10.1186/s40104-017-0219-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 12/17/2017] [Indexed: 12/03/2022] Open
Abstract
Background It is well known that peptides play a vital role in the nutrition and health of dairy cows. Bovine oligopeptide transporter 1 (bPepT1) is involved in the peptide transport process in the gastrointestinal tracts of dairy cows. However, little information is known in the characteristics of bPepT1. Therefore, the purpose of this study was to characterize bPepT1 functionally using a mammalian cell expression system. The uptake of radiolabeled dipeptide glycyl-sarcosine ([3H]-Gly-Sar) into the bPepT1-transfected Chinese hamster ovary cells was measured at various pH and substrate concentrations and with or without 15 other small peptides that contained Met or Lys. Results Western blot results showed that the abundance of bPepT1 protein in the jejunum and ileum are the highest in the gastrointestinal tract of dairy cows. The uptake of [3H]-Gly-Sar by bPepT1-Chinese hamster ovary cells was dependent on time, pH, and substrate concentration, with a low Km value of 0.94 ± 0.06 mmol/L and a maximum velocity of 20.80 ± 1.74 nmol/(mg protein • 5 min). Most of the di- and tripeptides were the substrates of bPepT1, based on substrate-competitive studies. However, bPepT1 has a higher affinity to the peptides with shorter chains, greater hydrophobicity, and negative or neutral charges. Conclusions These results demonstrated for the first time the functional characteristics of bPepT1, and they provide a new insight and better understanding into its vital role in absorbing a wide range of peptides from the digestive tract of dairy cows.
Collapse
Affiliation(s)
- Qingbiao Xu
- 1Institute of Dairy Science, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, 310058 People's Republic of China.,2College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070 People's Republic of China
| | - Zhixuan Liu
- 1Institute of Dairy Science, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, 310058 People's Republic of China
| | - Hongyun Liu
- 1Institute of Dairy Science, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, 310058 People's Republic of China
| | - Fengqi Zhao
- 1Institute of Dairy Science, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, 310058 People's Republic of China.,3Laboratory of Lactation and Metabolic Physiology, Department of Animal and Veterinary Sciences, University of Vermont, Burlington, VT 05405 USA
| | - Xinbei Huang
- 1Institute of Dairy Science, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, 310058 People's Republic of China
| | - Yueming Wu
- 1Institute of Dairy Science, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, 310058 People's Republic of China
| | - Jianxin Liu
- 1Institute of Dairy Science, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, 310058 People's Republic of China
| |
Collapse
|
34
|
Jafary Omid N, Bahari Javan N, Dehpour AR, Partoazar A, Rafiee Tehrani M, Dorkoosh F. In-vitro and in-vivo cytotoxicity and efficacy evaluation of novel glycyl-glycine and alanyl-alanine conjugates of chitosan and trimethyl chitosan nano-particles as carriers for oral insulin delivery. Int J Pharm 2017; 535:293-307. [PMID: 29138048 DOI: 10.1016/j.ijpharm.2017.11.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 11/09/2017] [Accepted: 11/10/2017] [Indexed: 10/18/2022]
Abstract
PURPOSE The aim of this research work was to explore the possibility of providing multifunctional oral insulin delivery system by conjugating several types of dipeptides on chitosan and trimethyl chitosan to be used as drug carriers. METHOD Conjugates of Glycyl-glycine and alanyl-alanine of chitosan and trimethyl chitosan (on primary alcohol group of polymer located on carbon 6) were synthesized and nanoparticles containing insulin were prepared for oral delivery. Preparation conditions of nanoparticles were optimized and their performance to enhance the permeability of insulin as well as cytotoxicity of nanoparticles in Caco-2 cell line was evaluated. To evaluate the efficacy of orally administered nanoparticles, nanoparticles with the most permeability enhancing ability were studied in male Wistar rats as animal model by measuring insulin and glucose Serum levels. RESULT Structural study of all the conjugates by infrared spectroscopy and nuclear magnetic resonance confirmed the successful formation of the conjugates with the desirable substitution degree. By optimizing preparation conditions, nanoparticles with expected size (157.3-197.7 nm), Zeta potential (24.35-34.37 mV), polydispersity index (0.365-0.512), entrapment efficiency (70.60-86.52%) and loading capacity (30.92-56.81%), proper morphology and desirable release pattern were obtained. Glycyl-glycine and alanyl-alanine conjugate nanoparticles of trimethyl chitosan showed 2.5-3.3 folds more effective insulin permeability in Caco-2 cell line than their chitosan counterparts. In animal model, oral administration of glycyl-glycine and alanyl-alanine conjugate nanoparticles of trimethyl chitosan demonstrated reasonable increase in Serum insulin level with relative bioavailability of 17.19% and 15.46% for glycyl-glycine and alanyl-alanine conjugate nanoparticles, respectively, and reduction in Serum glucose level compared with trimethyl chitosan nanoparticles (p < 0.05). CONCLUSION It seems that glycyl-glycine and alanyl-alanine conjugate nanoparticles of trimethyl chitosan have met the aim of this research work and have been able to orally deliver insulin with more than one mechanism in animal model. Hence, they are promising candidates for further research studies.
Collapse
Affiliation(s)
- Nersi Jafary Omid
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nika Bahari Javan
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad-Reza Dehpour
- Department of Pharmacology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Partoazar
- Department of Pharmacology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Rafiee Tehrani
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Dorkoosh
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Medical Biomaterial Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
35
|
Kawakami K, Moritani C, Uraji M, Fujita A, Kawakami K, Hatanaka T, Suzaki E, Tsuboi S. Sake lees hydrolysate protects against acetaminophen-induced hepatotoxicity via activation of the Nrf2 antioxidant pathway. J Clin Biochem Nutr 2017; 61:203-209. [PMID: 29203962 PMCID: PMC5703781 DOI: 10.3164/jcbn.17-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 07/03/2017] [Indexed: 11/22/2022] Open
Abstract
Acetaminophen is a commonly used analgesic. However, an overdose of acetaminophen causes severe hepatotoxicity via depletion of hepatic glutathione. Here, we investigated the protective effects of sake lees hydrolysate against acetaminophen-induced hepatotoxicity in mice. Sake lees hydrolysate was administered orally to ICR mice for seven days. Six hours after acetaminophen treatment, the mice were sacrificed, and blood and liver samples were collected for analysis. Treatment with acetaminophen markedly increased the levels of serum alanine aminotransferase, aspartate aminotransferase, lactate dehydrogenase, and alkaline phosphatase. Pretreatment with sake lees hydrolysate significantly prevented the increases in the serum levels of these enzymes and inhibited acetaminophen-mediated glutathione depletion. In addition, histopathological evaluation of the livers also revealed that sake lees hydrolysate prevented acetaminophen-induced centrilobular necrosis. The expression of γ-glutamylcysteine synthetase (γ-GCS), hemeoxygenase-1 (HO-1) and nuclear factor erythroid 2-related factor 2 (Nrf2) in the liver were decreased after acetaminophen treatment, whereas pretreatment with sake lees hydrolysate led to an increased expression of all three proteins. Furthermore, sake lees hydrolysate induced the expression of these proteins in HepG2. These results suggested that sake lees hydrolysate could induces HO-1 and γ-GCS expression via activation of the Nrf2 antioxidant pathway, and protects against acetaminophen-induced hepatotoxicity in mice.
Collapse
Affiliation(s)
- Kayoko Kawakami
- School of Pharmacy, Shujitsu University, 1-6-1 Nishigawara, Naka-ku, Okayama 703-8516, Japan
| | - Chie Moritani
- School of Pharmacy, Shujitsu University, 1-6-1 Nishigawara, Naka-ku, Okayama 703-8516, Japan
| | - Misugi Uraji
- Okayama Prefectural Technology Center for Agriculture, Forestry and Fisheries, Research Institute for Biological Sciences (RIBS), Okayama, 7549-1 Kibichuo-cho, Kaga-gun, Okayama 716-1241, Japan
| | - Akiko Fujita
- SATAKE Corporation, 2-30 Saijo Nishihonmachi, Higashi-Hiroshima-shi, Hiroshima 739-8602, Japan
| | - Koji Kawakami
- SATAKE Corporation, 2-30 Saijo Nishihonmachi, Higashi-Hiroshima-shi, Hiroshima 739-8602, Japan
| | - Tadashi Hatanaka
- Okayama Prefectural Technology Center for Agriculture, Forestry and Fisheries, Research Institute for Biological Sciences (RIBS), Okayama, 7549-1 Kibichuo-cho, Kaga-gun, Okayama 716-1241, Japan
| | - Etsuko Suzaki
- School of Pharmacy, Shujitsu University, 1-6-1 Nishigawara, Naka-ku, Okayama 703-8516, Japan
| | - Seiji Tsuboi
- School of Pharmacy, Shujitsu University, 1-6-1 Nishigawara, Naka-ku, Okayama 703-8516, Japan
| |
Collapse
|
36
|
Mai Y, Afonso-Pereira F, Murdan S, Basit AW. Excipient-mediated alteration in drug bioavailability in the rat depends on the sex of the animal. Eur J Pharm Sci 2017; 107:249-255. [DOI: 10.1016/j.ejps.2017.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/17/2017] [Accepted: 07/10/2017] [Indexed: 12/28/2022]
|
37
|
Tereshchenkova VF, Goptar IA, Zhuzhikov DP, Belozersky MA, Dunaevsky YE, Oppert B, Filippova IY, Elpidina EN. Prolidase is a critical enzyme for complete gliadin digestion in Tenebrio molitor larvae. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2017; 95:e21395. [PMID: 28660745 DOI: 10.1002/arch.21395] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Prolidase is a proline-specific metallopeptidase that cleaves imidodipeptides with C-terminal Pro residue. Prolidase was purified and characterized from the Tenebrio molitor larval midgut. The enzyme was localized in the soluble fraction of posterior midgut tissues, corresponding to a predicted cytoplasmic localization of prolidase according to the structure of the mRNA transcript. Expression of genes encoding prolidase and the major digestive proline-specific peptidase (PSP)-dipeptidyl peptidase 4-were similar. The pH optimum of T. molitor prolidase was 7.5, and the enzyme was inhibited by Z-Pro, indicating that it belongs to type I prolidases. In mammals, prolidase is particularly important in the catabolism of a proline-rich protein-collagen. We propose that T. molitor larval prolidase is a critical enzyme for the final stages of digestion of dietary proline-rich gliadins, providing hydrolysis of imidodipeptides in the cytoplasm of midgut epithelial cells. We propose that the products of hydrolysis are absorbed from the luminal contents by peptide transporters, which we have annotated in the T. molitor larval gut transcriptome. The origin of prolidase substrates in the insect midgut is discussed in the context of overall success of grain feeding insects.
Collapse
Affiliation(s)
| | - Irina A Goptar
- Chemical Faculty, Lomonosov Moscow State University, Moscow, Russia
| | | | - Mikhail A Belozersky
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Yakov E Dunaevsky
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Brenda Oppert
- USDA Agricultural Research Service, Center for Grain and Animal Health Research, Manhattan, KS, USA
| | | | - Elena N Elpidina
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
38
|
Shared hemocyte- and intestine-dominant expression profiles of intelectin genes in ascidian Ciona intestinalis: insight into the evolution of the innate immune system in chordates. Cell Tissue Res 2017; 370:129-142. [DOI: 10.1007/s00441-017-2647-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 03/27/2017] [Indexed: 10/19/2022]
|
39
|
Stelzl T, Geillinger-Kästle KE, Stolz J, Daniel H. Glycans in the intestinal peptide transporter PEPT1 contribute to function and protect from proteolysis. Am J Physiol Gastrointest Liver Physiol 2017; 312:G580-G591. [PMID: 28336547 DOI: 10.1152/ajpgi.00343.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 03/16/2017] [Accepted: 03/17/2017] [Indexed: 01/31/2023]
Abstract
Despite the fact that many membrane proteins carry extracellular glycans, little is known about whether the glycan chains also affect protein function. We recently demonstrated that the proton-coupled oligopeptide transporter 1 (PEPT1) in the intestine is glycosylated at six asparagine residues (N50, N406, N439, N510, N515, and N532). Mutagenesis-induced disruption of the individual N-glycosylation site N50, which is highly conserved among mammals, was detected to significantly enhance the PEPT1-mediated inward transport of peptides. Here, we show that for the murine protein the inhibition of glycosylation at sequon N50 by substituting N50 with glutamine, lysine, or cysteine or by replacing S52 with alanine equally altered PEPT1 transport kinetics in oocytes. Furthermore, we provide evidence that the uptake of [14C]glycyl-sarcosine in immortalized murine small intestinal (MODE-K) or colonic epithelial (PTK-6) cells stably expressing the PEPT1 transporter N50Q is also significantly increased relative to the wild-type protein. By using electrophysiological recordings and tracer flux studies, we further demonstrate that the rise in transport velocity observed for PEPT1 N50Q is bidirectional. In line with these findings, we show that attachment of biotin derivatives, comparable in weight with two to four monosaccharides, to the PEPT1 N50C transporter slows down the transport velocity. In addition, our experiments provide strong evidence that glycosylation of PEPT1 confers resistance against proteolytic cleavage by proteinase K, whereas a remarkable intrinsic stability against trypsin, even in the absence of N-linked glycans, was detected.NEW & NOTEWORTHY This study highlights the role of N50-linked glycans in modulating the bidirectional transport activity of the murine peptide transporter PEPT1. Electrophysiological and tracer flux measurements in Xenopus oocytes have shown that removal of the N50 glycans increases the maximal peptide transport rate in the inward and outward directions. This effect could be largely reversed by replacement of N50 glycans with structurally dissimilar biotin derivatives. In addition, N-glycans were detected to stabilize PEPT1 against proteolytic cleavage.
Collapse
Affiliation(s)
- Tamara Stelzl
- Nutritional Physiology, Technische Universität München, Freising, Germany
| | | | - Jürgen Stolz
- Nutritional Physiology, Technische Universität München, Freising, Germany
| | - Hannelore Daniel
- Nutritional Physiology, Technische Universität München, Freising, Germany
| |
Collapse
|
40
|
Wang HC, Shi FY, Hou MJ, Fu XY, Long RJ. Cloning of oligopeptide transport carrier PepT1 and comparative analysis of PepT1 messenger ribonucleic acid expression in response to dietary nitrogen levels in yak () and indigenous cattle () on the Qinghai-Tibetan plateau. J Anim Sci 2017; 94:3431-3340. [PMID: 27695788 DOI: 10.2527/jas.2016-0501] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The gastrointestinal lumen can directly absorb all di- and tripeptide protein degradation products, and oligopeptide absorption depends on the specific peptide transport carriers, which are located in gastrointestinal epithelial cells on the brush border membrane. Yak () use N more efficiently than cattle do, which implies that yak have a specific mechanism of nonprotein utilization including a peptide absorption mechanism. However, this mechanism has not been clarified. Our objective was to explore whether yak possess any adaptive mechanisms of peptide absorption to survive in the harsh foraging environment of the Qinghai-Tibetan plateau. Twelve castrated males of each of 2 genotypes, yak () and indigenous cattle (), were fed diets of various N levels. The yak PepT1 (yPepT1) cDNA was cloned in omasum epithelial tissue. Our results showed that the full-length yPepT1 cDNA contains 2,805 bp, and a 2,121-bp open reading frame encodes a putative protein of 707 AA residues. The yPepT1 AA sequence identified 5 putative extracellular N-glycosylation sites (Asn, Asn, Asn, Asn, and Asn), 2 putative intracellular protein kinase A sites (Ser and Thr), and 3 intracellular putative protein kinase C sites (Ser, Ser, and Ser). The yPepT1 AA sequence was 99, 95, 86, and 83% identical to PepT1 from cattle (), sheep (), pigs (), and humans (), respectively. The relative PepT1 mRNA expression for indigenous cattle was greater than yak in the rumen, omasum, duodenum, ileum, and liver ( < 0.001); however, it was lower in jejunum tissue ( < 0.01). The relative PepT1 mRNA expression in response to increasing dietary N for both genotypes were linear in the rumen and jejunum ( < 0.10); quadratic or cubic in the reticulum ( < 0.01); linear or quadratic in the duodenum, ileum, and liver ( ≤ 0.01); and linear, quadratic, or cubic in the omasum ( < 0.001). Moreover, there were significant interactions between genotype and dietary N in rumen, reticulum, omasum, duodenum, jejunum, ileum, and liver tissues. In conclusion, the PepT1 profile and expression in gastrointestinal epithelial cells of yak varied from those of cattle, implying that yak have evolved a peptide transport mechanism to adapt the environment of the Qinghai-Tibetan plateau.
Collapse
|
41
|
Quistgaard EM, Martinez Molledo M, Löw C. Structure determination of a major facilitator peptide transporter: Inward facing PepTSt from Streptococcus thermophilus crystallized in space group P3121. PLoS One 2017; 12:e0173126. [PMID: 28264013 PMCID: PMC5338821 DOI: 10.1371/journal.pone.0173126] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/15/2017] [Indexed: 12/03/2022] Open
Abstract
Major facilitator superfamily (MFS) peptide transporters (typically referred to as PepT, POT or PTR transporters) mediate the uptake of di- and tripeptides, and so play an important dietary role in many organisms. In recent years, a better understanding of the molecular basis for this process has emerged, which is in large part due to a steep increase in structural information. Yet, the conformational transitions underlying the transport mechanism are still not fully understood, and additional data is therefore needed. Here we report in detail the detergent screening, crystallization, experimental MIRAS phasing, and refinement of the peptide transporter PepTSt from Streptococcus thermophilus. The space group is P3121, and the protein is crystallized in a monomeric inward facing form. The binding site is likely to be somewhat occluded, as the lobe encompassing transmembrane helices 10 and 11 is markedly bent towards the central pore of the protein, but the extent of this potential occlusion could not be determined due to disorder at the apex of the lobe. Based on structural comparisons with the seven previously determined P212121 and C2221 structures of inward facing PepTSt, the structural flexibility as well as the conformational changes mediating transition between the inward open and inward facing occluded states are discussed. In conclusion, this report improves our understanding of the structure and conformational cycle of PepTSt, and can furthermore serve as a case study, which may aid in supporting future structure determinations of additional MFS transporters or other integral membrane proteins.
Collapse
Affiliation(s)
- Esben M. Quistgaard
- Centre for Structural Systems Biology (CSSB), DESY and European Molecular Biology Laboratory Hamburg, Hamburg, Germany
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Maria Martinez Molledo
- Centre for Structural Systems Biology (CSSB), DESY and European Molecular Biology Laboratory Hamburg, Hamburg, Germany
| | - Christian Löw
- Centre for Structural Systems Biology (CSSB), DESY and European Molecular Biology Laboratory Hamburg, Hamburg, Germany
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
42
|
The effect of cephalexin in influencing the pharmacokinetics of a novel drug - 5'-valyl-cytarabine hydrochloride. Asian J Pharm Sci 2017; 12:143-148. [PMID: 32104323 PMCID: PMC7032087 DOI: 10.1016/j.ajps.2016.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/13/2016] [Accepted: 08/22/2016] [Indexed: 11/22/2022] Open
Abstract
The aim of this study is to investigate the pharmacokinetics of 5′-valyl-cytarabine hydrochloride (OPC) when co-administered with cephalexin, which are both the substrates of PepT1. The drugs were administered orally by gavage. Blood samples were collected from the orbital plexus of the rats after oral administration of drug solutions. A new high-performance liquid chromatographic method was validated and used for determination of the two drugs. Pharmacokinetic parameters were calculated using DAS 2.1.1 software with noncompartmental analysis. After oral administration of OPC and co-administration of OPC and cephalexin, there were significant differences in the main pharmacokinetic parameters. The main pharmacokinetic parameters for the OPC group and the co-administrative group were as follows: AUC0-10 (18,168.7 ± 2561.4) ng⋅h/ml and (13,448.5 ± 2544.73) ng⋅h/ml, AUC0-∞ (18,683.1 ± 3066.5) ng⋅h/ml and (13,721.1 ± 2683.0) ng⋅h/ml, Cmax (6654.8 ± 481.3) ng/ml and (4765.1 ± 928.9) ng/ml, respectively. The results showed that the bioavailability of OPC could be reduced when co-administered with cephalexin, suggesting that the efficacy of a novel drug might be reduced when it came to combination use of β-lactam antibiotics.
Collapse
|
43
|
Pak YA, Long AJ, Annes WF, Witcher JW, Knadler MP, Ayan-Oshodi MA, Mitchell MI, Leese P, Hillgren KM. In Vitro and Clinical Evaluations of the Drug-Drug Interaction Potential of a Metabotropic Glutamate 2/3 Receptor Agonist Prodrug with Intestinal Peptide Transporter 1. Drug Metab Dispos 2017; 45:137-144. [PMID: 27895114 PMCID: PMC5267515 DOI: 10.1124/dmd.116.071118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/21/2016] [Indexed: 01/15/2023] Open
Abstract
Despite peptide transporter 1 (PEPT1) being responsible for the bioavailability for a variety of drugs, there has been little study of its potential involvement in drug-drug interactions. Pomaglumetad methionil, a metabotropic glutamate 2/3 receptor agonist prodrug, utilizes PEPT1 to enhance absorption and bioavailability. In vitro studies were conducted to guide the decision to conduct a clinical drug interaction study and to inform the clinical study design. In vitro investigations determined the prodrug (LY2140023 monohydrate) is a substrate of PEPT1 with Km value of approximately 30 µM, whereas the active moiety (LY404039) is not a PEPT1 substrate. In addition, among the eight known PEPT1 substrates evaluated in vitro, valacyclovir was the most potent inhibitor (IC50 = 0.46 mM) of PEPT1-mediated uptake of the prodrug. Therefore, a clinical drug interaction study was conducted to evaluate the potential interaction between the prodrug and valacyclovir in healthy subjects. No effect of coadministration was observed on the pharmacokinetics of the prodrug, valacyclovir, or either of their active moieties. Although in vitro studies showed potential for the prodrug and valacyclovir interaction via PEPT1, an in vivo study showed no interaction between these two drugs. PEPT1 does not appear to easily saturate because of its high capacity and expression in the intestine. Thus, a clinical interaction at PEPT1 is unlikely even with a compound with high affinity for the transporter.
Collapse
Affiliation(s)
- Y Anne Pak
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (Y.A.P., A.J.L.,W.F.A., J.W.W., M.P.K., M.A.A.-O., M.I.M., K.M.H.); and Quintiles Transnational, Clinical Pharmacology, Overland Park, Kansas (P.L.)
| | - Amanda J Long
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (Y.A.P., A.J.L.,W.F.A., J.W.W., M.P.K., M.A.A.-O., M.I.M., K.M.H.); and Quintiles Transnational, Clinical Pharmacology, Overland Park, Kansas (P.L.)
| | - William F Annes
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (Y.A.P., A.J.L.,W.F.A., J.W.W., M.P.K., M.A.A.-O., M.I.M., K.M.H.); and Quintiles Transnational, Clinical Pharmacology, Overland Park, Kansas (P.L.)
| | - Jennifer W Witcher
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (Y.A.P., A.J.L.,W.F.A., J.W.W., M.P.K., M.A.A.-O., M.I.M., K.M.H.); and Quintiles Transnational, Clinical Pharmacology, Overland Park, Kansas (P.L.)
| | - Mary Pat Knadler
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (Y.A.P., A.J.L.,W.F.A., J.W.W., M.P.K., M.A.A.-O., M.I.M., K.M.H.); and Quintiles Transnational, Clinical Pharmacology, Overland Park, Kansas (P.L.)
| | - Mosun A Ayan-Oshodi
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (Y.A.P., A.J.L.,W.F.A., J.W.W., M.P.K., M.A.A.-O., M.I.M., K.M.H.); and Quintiles Transnational, Clinical Pharmacology, Overland Park, Kansas (P.L.)
| | - Malcolm I Mitchell
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (Y.A.P., A.J.L.,W.F.A., J.W.W., M.P.K., M.A.A.-O., M.I.M., K.M.H.); and Quintiles Transnational, Clinical Pharmacology, Overland Park, Kansas (P.L.)
| | - Phillip Leese
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (Y.A.P., A.J.L.,W.F.A., J.W.W., M.P.K., M.A.A.-O., M.I.M., K.M.H.); and Quintiles Transnational, Clinical Pharmacology, Overland Park, Kansas (P.L.)
| | - Kathleen M Hillgren
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana (Y.A.P., A.J.L.,W.F.A., J.W.W., M.P.K., M.A.A.-O., M.I.M., K.M.H.); and Quintiles Transnational, Clinical Pharmacology, Overland Park, Kansas (P.L.)
| |
Collapse
|
44
|
Kodama N, Iwao T, Katano T, Ohta K, Yuasa H, Matsunaga T. Characteristic Analysis of Intestinal Transport in Enterocyte-Like Cells Differentiated from Human Induced Pluripotent Stem Cells. Drug Metab Dispos 2016; 44:0. [PMID: 27417181 DOI: 10.1124/dmd.116.069336] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 07/13/2016] [Indexed: 12/25/2022] Open
Abstract
We previously demonstrated that differentiated enterocytes from human induced pluripotent stem (iPS) cells exhibited drug-metabolizing activities and cytochrome P450 CYP3A4 inducibility. The aim of this study was to apply human iPS cell-derived enterocytes in pharmacokinetic studies by investigating the characteristics of drug transport into enterocyte-like cells. Human iPS cells cultured on feeder cells were differentiated into endodermal cells using activin A. These endodermal-like cells were then differentiated into intestinal stem cells by fibroblast growth factor 2. Finally, epidermal growth factor and small-molecule compounds induced the maturation of the intestinal stem cell-like cells. After differentiation, we performed transepithelial electrical resistance (TEER) measurements, immunofluorescence staining, and transport studies. TEER values increased in a time-dependent manner and reached approximately 100 Ω × cm(2) Efflux transport of Hoechst 33342, a substrate of breast cancer resistance protein (BCRP), was observed and inhibited by the BCRP inhibitor Ko143. The uptake of peptide transporter 1 substrate glycylsarcosine was also confirmed and suppressed when the temperature was lowered to 4°C. Using immunofluorescence staining, villin and Na(+)-K(+) ATPase were expressed. These results suggest that human iPS cell-derived enterocytes had loose tight junctions, polarity, as well as uptake and efflux transport functions. In addition, the rank order of apparent membrane permeability coefficient (Papp) values of these test compounds across the enterocyte-like cell membrane corresponded to the fraction absorbance (Fa) values. Therefore, differentiated enterocytes from human iPS cells may provide a useful comprehensive evaluation model of drug transport and metabolism in the small intestine.
Collapse
Affiliation(s)
- Nao Kodama
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (N.K., T.I., T.M.), Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (T.K., K.O., H.Y.)
| | - Takahiro Iwao
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (N.K., T.I., T.M.), Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (T.K., K.O., H.Y.)
| | - Takahiro Katano
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (N.K., T.I., T.M.), Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (T.K., K.O., H.Y.)
| | - Kinya Ohta
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (N.K., T.I., T.M.), Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (T.K., K.O., H.Y.)
| | - Hiroaki Yuasa
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (N.K., T.I., T.M.), Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (T.K., K.O., H.Y.)
| | - Tamihide Matsunaga
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (N.K., T.I., T.M.), Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (T.K., K.O., H.Y.)
| |
Collapse
|
45
|
Chan T, Lu X, Shams T, Zhu L, Murray M, Zhou F. The Role of N-Glycosylation in Maintaining the Transporter Activity and Expression of Human Oligopeptide Transporter 1. Mol Pharm 2016; 13:3449-3456. [DOI: 10.1021/acs.molpharmaceut.6b00462] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Ting Chan
- Faculty
of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia
| | - Xiaoxi Lu
- Faculty
of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia
| | - Tahiatul Shams
- Faculty
of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia
| | - Ling Zhu
- Retinal
Therapeutics Research Group, Save Sight Institute, The University of Sydney, Sydney, NSW 2000, Australia
| | - Michael Murray
- Pharmacogenomics
and Drug Development Group, Discipline of Pharmacology, School of
Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Fanfan Zhou
- Faculty
of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
46
|
Zhang Y, Viennois E, Zhang M, Xiao B, Han MK, Walter L, Garg P, Merlin D. PepT1 Expression Helps Maintain Intestinal Homeostasis by Mediating the Differential Expression of miRNAs along the Crypt-Villus Axis. Sci Rep 2016; 6:27119. [PMID: 27250880 PMCID: PMC4890533 DOI: 10.1038/srep27119] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/12/2016] [Indexed: 01/25/2023] Open
Abstract
In the jejunum, PepT1 is particularly enriched in the well-differentiated absorptive epithelial cells in the villi. Studies of expression and function of PepT1 along the crypt-villus axis demonstrated that this protein is crucial to the process of di/tripeptide absorption. We recently exhibited that PepT1 plays an important role in multiple biological functions, including the ability to regulate the expression/secretion of specific microRNAs (miRNAs) and the expression levels of multiple proteins. In this study, we observed that PepT1 knockout (KO) mice exhibited reduced body weight and shorten intestinal microvilli. We then examined the expression levels of various miRNAs and their target proteins along the crypt-villi axis in the jejunum of PepT1 KO mice. We found that PepT1 KO altered the distribution of miRNAs along the crypt-villus axis and changed the miRNA profiles of both villi and crypts. Using miRNA-target prediction and 2D-DIGE/mass spectrometry on villi and crypts samples, we found that ablation of PepT1 further directly or indirectly altered expression levels of certain protein targets. Collectively, our results suggest that PepT1 contributes to maintain balance of homeostasis and proper functions in the small intestine, and dysregulated miRNAs and proteins along the crypt-villus axis are highly related to this process.
Collapse
Affiliation(s)
- Yuchen Zhang
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, 30302, USA
| | - Emilie Viennois
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, 30302, USA
| | - Mingzhen Zhang
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, 30302, USA
| | - Bo Xiao
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, 30302, USA.,Institute for Clean Energy and Advanced Materials, Faculty for Materials and Energy, Southwest University, Chongqing, 400715, P. R. China
| | - Moon Kwon Han
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, 30302, USA
| | - Lewins Walter
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, 30302, USA
| | - Pallavi Garg
- Department of Biology, Georgia State University, Atlanta, Georgia, 30302, USA
| | - Didier Merlin
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, 30302, USA.,Atlanta Veterans Affairs Medical Center, Decatur, Georgia, 30033, USA
| |
Collapse
|
47
|
Kodama N, Iwao T, Kabeya T, Horikawa T, Niwa T, Kondo Y, Nakamura K, Matsunaga T. Inhibition of mitogen-activated protein kinase kinase, DNA methyltransferase, and transforming growth factor-β promotes differentiation of human induced pluripotent stem cells into enterocytes. Drug Metab Pharmacokinet 2016; 31:193-200. [DOI: 10.1016/j.dmpk.2016.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 01/21/2016] [Accepted: 02/09/2016] [Indexed: 12/13/2022]
|
48
|
H(+)/peptide transporter (PEPT2) is expressed in human epidermal keratinocytes and is involved in skin oligopeptide transport. Biochem Biophys Res Commun 2016; 475:335-41. [PMID: 27216463 DOI: 10.1016/j.bbrc.2016.05.093] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 05/19/2016] [Indexed: 01/17/2023]
Abstract
Peptide transporter 2 (PEPT2) is a member of the proton-coupled oligopeptide transporter family, which mediates the cellular uptake of oligopeptides and peptide-like drugs. Although PEPT2 is expressed in many tissues, its expression in epidermal keratinocytes remains unclear. We investigated PEPT2 expression profile and functional activity in keratinocytes. We confirmed PEPT2 mRNA expression in three keratinocyte lines (normal human epidermal keratinocytes (NHEKs), immortalized keratinocytes, and malignant keratinocytes) by reverse transcription-polymerase chain reaction (RT-PCR) and quantitative real-time RT-PCR. In contrast to PEPT1, PEPT2 expression in the three keratinocytes was similar or higher than that in HepG2 cells, used as PEPT2-positive cells. Immunolocalization analysis using human skin showed epidermal PEPT2 localization. We studied keratinocyte transport function by measuring the oligopeptide content using liquid chromatography/tandem mass spectrometry. Glycylsarcosine uptake in NHEKs was pH-dependent, suggesting that keratinocytes could absorb small peptides in the presence of an inward H(+) gradient. We also performed a skin-permeability test of several oligopeptides using skin substitute, suggesting that di- and tripeptides pass actively through the epidermis. In conclusion, PEPT2 is expressed in keratinocytes and involved in skin oligopeptide uptake.
Collapse
|
49
|
Moronta J, Smaldini PL, Docena GH, Añón MC. Peptides of amaranth were targeted as containing sequences with potential anti-inflammatory properties. J Funct Foods 2016. [DOI: 10.1016/j.jff.2015.12.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
50
|
Stelzl T, Baranov T, Geillinger KE, Kottra G, Daniel H. Effect of N-glycosylation on the transport activity of the peptide transporter PEPT1. Am J Physiol Gastrointest Liver Physiol 2016; 310:G128-41. [PMID: 26585416 DOI: 10.1152/ajpgi.00350.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 11/10/2015] [Indexed: 01/31/2023]
Abstract
The intestinal peptide transporter PEPT1 provides bulk quantities of amino acids to epithelial cells. PEPT1 is a high-capacity and low-affinity solute carrier of the SLC15 family found in apical membranes of enterocytes in small intestine and distal colon. Surprisingly, murine PEPT1 (mPEPT1) has an apparent molecular mass of ∼95 kDa in the small intestine but ∼105 kDa in the large intestine. Here we describe studies on mPEPT1 protein glycosylation and how glycans affect transport function. Putative N-glycosylation sites of mPEPT1 were altered by site-directed mutagenesis followed by expression in Xenopus laevis oocytes. Replacement of six asparagine residues (N) at positions N50, N406, N439, N510, N515, and N532 by glutamine (Q) resulted in a decrease of the mPEPT1 mass by around 35 kDa. Electrophysiology revealed all glycosylation-deficient transporters to be functional with comparable expression levels in oocyte membranes. Strikingly, the mutant protein with N50Q exhibited a twofold decreased affinity for Gly-Sar but a 2.5-fold rise in the maximal inward currents compared with the wild-type protein. Elevated maximal transport currents were also recorded for cefadroxil and tri-l-alanine. Tracer flux studies performed with [(14)C]-Gly-Sar confirmed the reduction in substrate affinity and showed twofold increased maximal transport rates for the N50Q transporter. Elimination of individual N-glycosylation sites did not alter membrane expression in oocytes or overall transport characteristics except for the mutant protein N50Q. Because transporter surface density was not altered in N50Q, removal of the glycan at this location appears to accelerate the substrate turnover rate.
Collapse
Affiliation(s)
- Tamara Stelzl
- Chair of Nutritional Physiology, Technische Universität München, Freising, Germany; ZIEL, Institute for Food and Health, Freising, Germany
| | - Tatjana Baranov
- Chair of Nutritional Physiology, Technische Universität München, Freising, Germany; ZIEL, Institute for Food and Health, Freising, Germany
| | - Kerstin E Geillinger
- Chair of Nutritional Physiology, Technische Universität München, Freising, Germany; ZIEL, Institute for Food and Health, Freising, Germany
| | - Gabor Kottra
- Chair of Nutritional Physiology, Technische Universität München, Freising, Germany; ZIEL, Institute for Food and Health, Freising, Germany
| | - Hannelore Daniel
- Chair of Nutritional Physiology, Technische Universität München, Freising, Germany; ZIEL, Institute for Food and Health, Freising, Germany
| |
Collapse
|