Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Biol Pharm Bull ; 43(4): 697-706, 2020.
Article in English | MEDLINE | ID: mdl-32238712

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.


Subject(s)
Amino Acids/pharmacology , Mesalamine/pharmacology , Peptide Transporter 1/physiology , Amino Acids/chemistry , Animals , Biological Transport , Caco-2 Cells , Humans , Mesalamine/chemistry , Oocytes/drug effects , Oocytes/physiology , Peptide Transporter 1/genetics , Xenopus laevis
2.
Biochem Soc Trans ; 48(2): 337-346, 2020 04 29.
Article in English | MEDLINE | ID: mdl-32219385

ABSTRACT

Solute carrier (SLC) transporters play important roles in regulating the movement of small molecules and ions across cellular membranes. In mammals, they play an important role in regulating the uptake of nutrients and vitamins from the diet, and in controlling the distribution of their metabolic intermediates within the cell. Several SLC families also play an important role in drug transport and strategies are being developed to hijack SLC transporters to control and regulate drug transport within the body. Through the addition of amino acid and peptide moieties several novel antiviral and anticancer agents have been developed that hijack the proton-coupled oligopeptide transporters, PepT1 (SCL15A1) and PepT2 (SLC15A2), for improved intestinal absorption and renal retention in the body. A major goal is to understand the rationale behind these successes and expand the library of prodrug molecules that utilise SLC transporters. Recent co-crystal structures of prokaryotic homologues of the human PepT1 and PepT2 transporters have shed important new insights into the mechanism of prodrug recognition. Here, I will review recent developments in our understanding of ligand recognition and binding promiscuity within the SLC15 family, and discuss current models for prodrug recognition.


Subject(s)
Peptide Transporter 1/physiology , Prodrugs/pharmacology , Symporters/physiology , Animals , Biological Transport , Crystallography, X-Ray , Drug Design , Humans , Oligopeptides/chemistry , Peptide Transporter 1/chemistry , Symporters/chemistry , Valacyclovir/pharmacology , Valganciclovir/pharmacology
3.
Compr Physiol ; 8(2): 731-760, 2018 03 25.
Article in English | MEDLINE | ID: mdl-29687900

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.


Subject(s)
Inflammatory Bowel Diseases/physiopathology , Peptide Transporter 1/physiology , Amino Acid Sequence , Animals , Biological Transport/physiology , Humans , Membrane Transport Proteins/physiology , Peptide Transporter 1/genetics , Sequence Alignment , Symporters/genetics , Symporters/physiology
4.
Compr Physiol ; 8(2): 843-869, 2018 03 25.
Article in English | MEDLINE | ID: mdl-29687907

ABSTRACT

As the organ with one of the largest surface areas facing the environment and responsible for nutrient uptake, the small intestine expresses numerous transport proteins in its brush-border membrane for efficient absorption and supply of dietary macro- and micronutrients. The understanding of regulation and functional interplay of these nutrient transporters is of emerging interest in nutrition and medical physiology research in respect to development of diabetes, obesity, and inflammatory bowel disease worldwide. The peptide transporter 1 (PepT1, SLC15A1) is abundantly expressed particularly in the intestinal tract and provides highly effective transport of amino acids in the form of di- and tripeptides and features a substantial acceptance for structurally related compounds and drugs. These characteristics bring PepT1 into focus for nutritional and medical/pharmaceutical approaches, as it is the essential hub responsible for oral bioavailability of dietary protein/peptide supplements and peptide-like drugs in eukaryotic organisms. Detailed analysis of molecular processes regulating PepT1 expression and function achieved in the last two decades has helped to define and use adjusting tools and to better integrate the transporter's role in cell and organ physiology. In this article, we provide an overview of the current knowledge on PepT1 function in health and disease, and on regulatory factors modulating its gene and protein expression as well as transport activity. © 2018 American Physiological Society. Compr Physiol 8:843-869, 2018.


Subject(s)
Intestinal Absorption/physiology , Peptide Transporter 1/physiology , Animals , Biological Transport/physiology , Diabetes Mellitus/metabolism , Gene Expression Regulation/physiology , Homeostasis/physiology , Humans , Inflammatory Bowel Diseases/metabolism , MicroRNAs/genetics , Nutritional Physiological Phenomena/physiology , Obesity/metabolism , Peptide Transporter 1/genetics
5.
Mol Pharm ; 14(12): 4685-4693, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29111754

ABSTRACT

In humans, peptides derived from dietary proteins and peptide-like drugs are transported via the proton-dependent oligopeptide transporter hPepT1 (SLC15A1). hPepT1 is located across the apical membranes of the small intestine and kidney, where it serves as a high-capacity low-affinity transporter of a broad range of di- and tripeptides. hPepT1 is also overexpressed in the colon of inflammatory bowel disease (IBD) patients, where it mediates the transport of harmful peptides of bacterial origin. Therefore, hPepT1 is a drug target for prodrug substrates interacting with intracellular proteins or inhibitors blocking the transport of toxic bacterial products. In this study, we construct multiple structural models of hPepT1 representing different conformational states that occur during transport and inhibition. We then identify and characterize five ligands of hPepT1 using computational methods, such as virtual screening and QM-polarized ligand docking (QPLD), and experimental testing with uptake kinetic measurements and electrophysiological assays. Our results improve our understanding of the substrate and inhibitor specificity of hPepT1. Furthermore, the newly discovered ligands exhibit unique chemotypes, providing a framework for developing tool compounds with optimal intestinal absorption as well as future IBD therapeutics against this emerging drug target.


Subject(s)
Models, Chemical , Oligopeptides/chemistry , Peptide Transporter 1/chemistry , Prodrugs/chemistry , Biological Transport, Active/drug effects , Drug Evaluation, Preclinical/methods , Humans , Inhibitory Concentration 50 , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Kinetics , Ligands , Models, Molecular , Molecular Docking Simulation , Oligopeptides/metabolism , Peptide Transporter 1/antagonists & inhibitors , Peptide Transporter 1/physiology , Prodrugs/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...