Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Protein Expr Purif ; 190: 105990, 2022 02.
Article in English | MEDLINE | ID: mdl-34637915

ABSTRACT

The human peptide transporter hPEPT1 (SLC15A1) is responsible for uptake of dietary di- and tripeptides and a number of drugs from the small intestine by utilizing the proton electrochemical gradient, and hence an important target for peptide-like drug design and drug delivery. hPEPT1 belongs to the ubiquitous major facilitator superfamily that all contain a 12TM core structure, with global conformational changes occurring during the transport cycle. Several bacterial homologues of these transporters have been characterized, providing valuable insight into the transport mechanism of this family. Here we report the overexpression and purification of recombinant hPEPT1 in a detergent-solubilized state. Thermostability profiling of hPEPT1 at different pH values revealed that hPEPT1 is more stable at pH 6 as compared to pH 7 and 8. Micro-scale thermophoresis (MST) confirmed that the purified hPEPT1 was able to bind di- and tripeptides respectively. To assess the in-solution oligomeric state of hPEPT1, negative stain electron microscopy was performed, demonstrating a predominantly monomeric state.


Subject(s)
Gene Expression , Peptide Transporter 1 , Hot Temperature , Humans , Hydrogen-Ion Concentration , Peptide Transporter 1/biosynthesis , Peptide Transporter 1/chemistry , Peptide Transporter 1/genetics , Peptide Transporter 1/isolation & purification , Protein Stability , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
2.
J Med Chem ; 65(6): 4565-4577, 2022 03 24.
Article in English | MEDLINE | ID: mdl-34842428

ABSTRACT

The naturally occurring linear dipeptide JBP923 (trans-4-l-Hyp-l-Ser, HS-tLL) with anti-inflammatory effects showed potential for the treatment of inflammatory bowel disease (IBD). However, colon-specific delivery after oral administration is still a challenge because its absorption is mediated by oligopeptide transporter 1 (PEPT1) in the upper small intestine and because of its instability in the gastrointestinal tract. Therefore, we aimed to enhance the colon-targeting efficiency by modulating HS-tLL chirality to synthesize eight enantiomers. Among these enantiomers, trans-4-d-Hyp-d-Ser, cis-4-l-Hyp-d-Ser, cis-4-d-Hyp-l-Ser, and cis-4-d-Hyp-d-Ser did not work as substrates of PEPT1 and were stable in the gastrointestinal tract, resulting in enhanced colonic accumulation through the paracellular pathway due to the loose tight junctions in IBD. Interestingly, cis-4-d-Hyp-d-Ser exerted the most potent therapeutic effect on IBD. Our findings revealed the impact of chirality on the colonic accumulation of the linear dipeptide, providing strategies for the colon-targeted delivery of the linear dipeptide for the treatment of IBD.


Subject(s)
Inflammatory Bowel Diseases , Peptide Transporter 1 , Symporters , Colon , Dipeptides/chemistry , Dipeptides/pharmacology , Humans , Inflammatory Bowel Diseases/drug therapy , Peptide Transporter 1/chemistry , Serine/pharmacology , Symporters/metabolism
3.
Sci Adv ; 7(35)2021 08.
Article in English | MEDLINE | ID: mdl-34433568

ABSTRACT

The SLC15 family of proton-coupled solute carriers PepT1 and PepT2 play a central role in human physiology as the principal route for acquiring and retaining dietary nitrogen. A remarkable feature of the SLC15 family is their extreme substrate promiscuity, which has enabled the targeting of these transporters for the improvement of oral bioavailability for several prodrug molecules. Although recent structural and biochemical studies on bacterial homologs have identified conserved sites of proton and peptide binding, the mechanism of peptide capture and ligand promiscuity remains unclear for mammalian family members. Here, we present the cryo-electron microscopy structure of the outward open conformation of the rat peptide transporter PepT2 in complex with an inhibitory nanobody. Our structure, combined with molecular dynamics simulations and biochemical and cell-based assays, establishes a framework for understanding peptide and prodrug recognition within this pharmaceutically important transporter family.


Subject(s)
Prodrugs , Symporters , Animals , Cryoelectron Microscopy , Mammals/metabolism , Membrane Transport Proteins/metabolism , Peptide Transporter 1/chemistry , Peptides/metabolism , Protons , Rats
4.
Biol Pharm Bull ; 44(5): 678-685, 2021.
Article in English | MEDLINE | ID: mdl-33952823

ABSTRACT

To clarify the role of an amino acid residue in the pH-dependent efflux process in Chinese hamster ovary (CHO) cells expressing the human oligopeptide transporter hPEPT1 (CHO/hPEPT1), we determined the effect of extracellular pH on the hPEPT1-mediated efflux process. The efflux of glycylsarcosine (Gly-Sar), a typical substrate for hPEPT1, was determined using an infinite dilution method after cells were preloaded with [3H]-Gly-Sar. The efflux of [3H]-Gly-Sar was stimulated by 5 mM unlabeled hPEPT1 substrates in the medium. This trans-stimulation phenomenon showed that hPEPT1 mediated the efflux of [3H]-Gly-Sar from CHO/hPEPT1 and that hPEPT1 is a bi-directional transporter. We then determined the effect of extracellular pH (varying from 8.0 to 3.5) on the efflux activity. The efflux activity by hPEPT1 decreased with the decrease in extracellular pH. The Henderson-Hasselbälch-type equation, which fitted well to the pH-profile of efflux activity, indicated that a single amino acid residue with a pKa value of approximately 5.7 regulates the efflux activity. The pH-profile of the efflux activity remained almost unchanged irrespective of the proton gradient across the plasma membrane. In addition, the chemical modification of the histidine residue with diethylpyrocarbonate completely abolished the efflux activity from cells, which could be prevented by the presence of 10 mM Gly-Sar. These data indicate that the efflux process of hPEPT1 is also regulated in a pH-dependent manner by the protonation state of a histidine residue located at or near the substrate recognition site facing the extracellular space.


Subject(s)
Histidine/chemistry , Peptide Transporter 1/metabolism , Recombinant Proteins/metabolism , Animals , CHO Cells , Cricetulus , Dipeptides/metabolism , Hydrogen-Ion Concentration , Peptide Transporter 1/chemistry , Peptide Transporter 1/genetics , Protons , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Tritium/chemistry
5.
Food Chem ; 355: 129574, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-33799251

ABSTRACT

Maillard reaction produces advanced glycation end products (AGEs) that endanger human health. This study investigated the protective effect of (+)-catechin (CC) on different types of dietary AGEs absorption and cytotoxicity in Caco-2 cells. Our results showed that CC had higher inhibitory rate on peptide bound-AGEs absorption than free Nɛ-carboxymethyl lysine (CML), which dropped to 36.24% and 32.21% when treated with 20 and 50 µM CC. The reasons might be that CC could repair the loose tight junction (ZO-1) and down-regulation of protein-coupling peptide carrier 1 (PEPT-1) expression in Caco-2 cells which were in accordance with molecular docking results. Additionally, CC could remarkably decreased the protein levels of receptor of AGEs (RAGE), mitogen-activated protein kinases (MAPK) and nuclear factor-kappa B (NF-κB) that detected by western blotting and immunohistochemical staining method. Taken together, these findings demonstrated that CC may inhibit AGEs absorption and protected Caco-2 cells against RAGE-MAPK-NF-κB signaling suppression.


Subject(s)
Catechin/chemistry , Glycation End Products, Advanced/chemistry , Adsorption , Binding Sites , Caco-2 Cells , Catechin/metabolism , Catechin/pharmacology , Cell Survival/drug effects , Down-Regulation/drug effects , Humans , Lysine/analogs & derivatives , Lysine/chemistry , Maillard Reaction , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Molecular Docking Simulation , NF-kappa B/genetics , NF-kappa B/metabolism , Peptide Transporter 1/chemistry , Peptide Transporter 1/genetics , Peptide Transporter 1/metabolism , Signal Transduction/drug effects , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
6.
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
7.
Methods Mol Biol ; 2168: 73-103, 2020.
Article in English | MEDLINE | ID: mdl-33582988

ABSTRACT

Here, we present a protocol for the functional characterization of the H+-coupled human peptide transporter PepT1 and sufficient notes to transfer the protocol to the Na+-coupled sugar transporter SGLT1, the organic cation transporter OCT2, the Na+/Ca2+ exchanger NCX, and the neuronal glutamate transporter EAAT3.The assay was developed for the commercially available SURFE2R N1 instrument (Nanion Technologies GmbH) which applies solid supported membrane (SSM)-based electrophysiology. This technique is widely used for the functional characterization of membrane transporters with more than 100 different transporters characterized so far. The technique is cost-effective, easy to use, and capable of high-throughput measurements.SSM-based electrophysiology utilizes SSM-coated gold sensors to physically adsorb membrane vesicles containing the protein of interest. A fast solution exchange provides the substrate and activates transport. For the measurement of PepT1 activity, we applied a peptide concentration jump to activate H+/peptide symport. Proton influx charges the sensor. A capacitive current is measured reflecting the transport activity of PepT1 . Multiple measurements on the same sensor allow for comparison of transport activity under different conditions. Here, we determine EC50 for PepT1-mediated glycylglycine transport and perform an inhibition experiment using the specific peptide inhibitor Lys[Z(NO2)]-Val.


Subject(s)
Biosensing Techniques/methods , Cell Membrane/metabolism , Electrophysiology/methods , Peptide Transporter 1/metabolism , Sodium-Glucose Transporter 1/metabolism , Biological Transport , Humans , Peptide Transporter 1/chemistry , Sodium-Glucose Transporter 1/chemistry
8.
Am J Physiol Cell Physiol ; 318(1): C191-C204, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31664857

ABSTRACT

Peptide transporter 1 (PepT1) mediates the uptake of dietary di-/tripeptides in vertebrates. However, in teleost fish gut, more than one PepT1-type transporter might operate, because of teleost-specific whole gen(om)e duplication event(s) that occurred during evolution. Here, we describe a novel teleost di-/tripeptide transporter, i.e., the Atlantic salmon (Salmo salar) peptide transporter 1a [PepT1a; or solute carrier family 15 member 1a (Slc15a1a)], which is a paralog (77% similarity and 64% identity at the amino acid level) of the well-described Atlantic salmon peptide transporter 1b [PepT1b, alias PepT1; or solute carrier family 15 member 1b (Slc15a1b)]. Comparative analysis and evolutionary relationships of gene/protein sequences were conducted after ad hoc database mining. Tissue mRNA expression analysis was performed by quantitative real-time PCR, whereas transport function analysis was accomplished by heterologous expression in Xenopus laevis oocytes and two-electrode voltage-clamp measurements. Atlantic salmon pept1a is highly expressed in the proximal intestine (pyloric ceca ≈ anterior midgut > midgut >> posterior midgut), in the same gut regions as pept1b but notably ~5-fold less abundant. Like PepT1b, Atlantic salmon PepT1a is a low-affinity/high-capacity system. Functional analysis showed electrogenic, Na+-independent/pH-dependent transport and apparent substrate affinity (K0.5) values for Gly-Gln of 1.593 mmol/L at pH 7.6 and 0.076 mmol/L at pH 6.5. In summary, we show that a piscine PepT1a-type transporter is functional. Defining the role of Atlantic salmon PepT1a in the gut will help to understand the evolutionary and functional relationships among peptide transporters. Its functional characterization will contribute to elucidate the relevance of peptide transporters in Atlantic salmon nutritional physiology.


Subject(s)
Dipeptides/metabolism , Fish Proteins/metabolism , Intestinal Absorption , Peptide Transporter 1/metabolism , Salmo salar/metabolism , Animals , Evolution, Molecular , Fish Proteins/chemistry , Fish Proteins/genetics , Gene Expression Regulation , Hydrogen-Ion Concentration , Kinetics , Peptide Transporter 1/chemistry , Peptide Transporter 1/genetics , Phylogeny , Salmo salar/genetics , Xenopus laevis
9.
J Med Chem ; 62(17): 7708-7721, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31393124

ABSTRACT

The cyclic dipeptides generally present lower affinity toward intestinal oligopeptide transporter 1 (PEPT1) than the linear dipeptides. JBP485 (cyclo(l-Hyp-l-Ser)) is a low-affinity substrate of PEPT1 with poor oral bioavailability. However, JBP923 (l-Hyp-l-Ser) is a high-affinity substrate of PEPT1 with high oral absorption. We hypothesize that the bioactivatable pseudo-tripeptidization prodrug strategy is promising to increase the affinity of cyclic dipeptides toward PEPT1. To test our hypothesis, we design five amino acid ester prodrugs of JBP485. Compared with JBP485, the optimal prodrug (JBP485-3-CH2-O-valine, J3V) demonstrates improved affinity of PEPT1, oral bioavailability in rats and beagle dogs. Moreover, J3V can dose-dependently protect against liver injury. Additionally, J3V is stable in the gastrointestinal tract, beneficial to the PEPT1-mediated membrane transport, and is bioactivated in the enterocytes and hepatic cells, essential to elicit its bioactivity. In summary, the bioactivatable pseudo-tripeptidization strategy shows potential in increasing affinity of PEPT1 to enhance oral bioavailability of cyclic dipeptides.


Subject(s)
Antiviral Agents/chemistry , Peptide Transporter 1/chemistry , Peptides, Cyclic/chemistry , Prodrugs/chemistry , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Caco-2 Cells , Cell Membrane Permeability/drug effects , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/pathology , Dogs , Dose-Response Relationship, Drug , Humans , Molecular Docking Simulation , Molecular Structure , Peptide Transporter 1/administration & dosage , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/pharmacokinetics , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
10.
Bioorg Chem ; 92: 103163, 2019 11.
Article in English | MEDLINE | ID: mdl-31450166

ABSTRACT

Curcumin (CUR) is a natural yellow pigment from turmeric with extensive bioactivities. However its relatively poor solubility limited its absorption and bioavailability. In this study, a novel series of CUR-peptide conjugates were designed and synthesized as PepT1-mediated transport drugs and their solubility, cellular uptakes and anti-tumor activities were evaluated. Ten compounds showed better water solubility than CUR due to the dipeptide moiety. Compared with CUR, compound 5e exhibited the slightly better activity and 5d showed the similar activity with CUR. Besides, compounds 5d and 5e performed higher cellular uptakes in Caco-2 cell and dose-dependently inhibited by the addition of PepT1 typical substrate glycylsarcosine (Gly-Sar). Compound 5d and 5e have improved the absorption of CUR by PepT1-mediated without affected the activity. These new dipeptide conjugates of CUR may serve as promising lead compounds for future drug development.


Subject(s)
Antineoplastic Agents/chemical synthesis , Curcumin/chemical synthesis , Dipeptides/chemistry , Drug Carriers/chemistry , Peptide Transporter 1/chemistry , Animals , Antineoplastic Agents/pharmacology , Biological Availability , Caco-2 Cells , Cell Membrane Permeability , Curcumin/pharmacology , Dose-Response Relationship, Drug , Drug Liberation , Humans , Microsomes, Liver/drug effects , Molecular Structure , Rats , Solubility
11.
J Am Chem Soc ; 141(6): 2404-2412, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30644743

ABSTRACT

Members of the solute carrier 15 family (SLC15) transport di- and tripeptides as well as peptidomimetic drugs across the cell membrane. Structures of bacterial homologues have provided valuable information on the binding and transport of their natural substrates, but many do not transport medically relevant drugs. In contrast, a homologue from Escherichia coli, DtpA (dipeptide and tripeptide permease), shows a high similarity to human PepT1 (SLC15A1) in terms of ligand selectivity and transports a similar set of drugs. Here, we present the crystal structure of DtpA in ligand-free form (at 3.30 Å resolution) and in complex with the antiviral prodrug valganciclovir (at 2.65 Å resolution) supported by biochemical data. We show that valganciclovir unexpectedly binds with the ganciclovir moiety mimicking the N-terminal residue of a canonical peptide substrate. On the basis of a homology model we argue that this binding mode also applies to the human PepT1 transporter. Our results provide new insights into the binding mode of prodrugs and will assist the rational design of drugs with improved absorption rates.


Subject(s)
Escherichia coli Proteins/metabolism , Membrane Transport Proteins/metabolism , Peptide Transporter 1/metabolism , Valganciclovir/metabolism , Escherichia coli Proteins/chemistry , Humans , Membrane Transport Proteins/chemistry , Models, Molecular , Peptide Transporter 1/chemistry , Protein Binding , Protein Conformation
12.
Proc Natl Acad Sci U S A ; 116(3): 804-809, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30602453

ABSTRACT

A major challenge in drug development is the optimization of intestinal absorption and cellular uptake. A successful strategy has been to develop prodrug molecules, which hijack solute carrier (SLC) transporters for active transport into the body. The proton-coupled oligopeptide transporters, PepT1 and PepT2, have been successfully targeted using this approach. Peptide transporters display a remarkable capacity to recognize a diverse library of di- and tripeptides, making them extremely promiscuous and major contributors to the pharmacokinetic profile of several important drug classes, including beta-lactam antibiotics and antiviral and antineoplastic agents. Of particular interest has been their ability to recognize amino acid and peptide-based prodrug molecules, thereby providing a rational approach to improving drug transport into the body. However, the structural basis for prodrug recognition has remained elusive. Here we present crystal structures of a prokaryotic homolog of the mammalian transporters in complex with the antiviral prodrug valacyclovir and the peptide-based photodynamic therapy agent, 5-aminolevulinic acid. The valacyclovir structure reveals that prodrug recognition is mediated through both the amino acid scaffold and the ester bond, which is commonly used to link drug molecules to the carrier's physiological ligand, whereas 5-aminolevulinic acid makes far fewer interactions compared with physiological peptides. These structures provide a unique insight into how peptide transporters interact with xenobiotic molecules and provide a template for further prodrug development.


Subject(s)
Peptide Transporter 1/chemistry , Prodrugs/chemistry , Staphylococcus hominis/chemistry , Aminolevulinic Acid/administration & dosage , Antiviral Agents/administration & dosage , Photosensitizing Agents/administration & dosage , Valacyclovir/administration & dosage
13.
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
...