Dr David Meredith
MA DPhil, Fellow of the HEA
Senior Lecturer in Biochemistry / Biomedical Science
School of Biological and Medical Sciences
Role
I am the Subject co-ordinator for BSc Biological Sciences; I am also Knowledge Exchange Lead for the Faculty of Health & Life Sciences.
Areas of expertise
- Membrane transporter protein structure and function (expression and transport assays), including characterising 'orphan' transporters.
- Design, synthesis and biological testing of novel peptidic prodrug molecules (in collaboration with Prof Pat Bailey, London South Bank University and Dr David Foley, University of Cardiff)
Teaching and supervision
Courses
Modules taught
I teach on a number of modules in the Biomedical and Biological Sciences area, from 1st year onwards, including Scientific Skills, Introduction to Biochemistry A & B, Biochemistry of Cell Function, Molecular Medicine and Evidence Based Medicine & Diagnostics.
I also supervise undergraduate project students.
Supervision
I have supervised six students to PhD completion.
Research
I lead the Membrane Transport Group research group. We investigate the structure-function relationship of membrane transport proteins, to try to elucidate how they bind and transport their substrates. In particular, we study (nutrient) transporters that are naturally present in the intestine with a view to designing small drug molecules that can be taken up through them. These transporters include the peptide transporter (PepT1), amino acid transporters (the PAT family), monocarboxylate transporters (MCTs) and organic anion transporters (OATPs).
The techniques involved range from expression of wild-type and mutated transporter proteins in model systems, cell culture, medicinal chemistry (with Prof Pat Bailey, London South Bank University & Dr David Foley, University of Cardiff) and protein crystallography and molecular modelling (with Dr Newstead and Prof Samson's groups, respectively, University of Oxford).
Groups
Publications
Journal articles
-
Harris SE, Blasio MJ, Zhao X, Ma M, Davies KL, Wooding FBP, Hamilton RS, Blache D, Meredith D, Murray AJ, Fowden AL, Forhead AJ, 'Thyroid deficiency before birth alters the adipose transcriptome to promote overgrowth of white adipose tissue and impair thermogenic capacity'
Thyroid 30 (6) (2020) pp.794-805
ISSN: 1050-7256 eISSN: 1557-9077)AbstractPublished here Open Access on RADARBackground. Development of adipose tissue before birth is essential for energy storage and thermoregulation in the neonate and for cardiometabolic health in later life. Thyroid hormones are important regulators of growth and maturation in fetal tissues. Offspring hypothyroid in utero are poorly adapted to regulate body temperature at birth and are at risk of becoming obese and insulin resistant in childhood. The mechanisms by which thyroid hormones regulate the growth and development of adipose tissue in the fetus, however, are unclear. Methods. This study examined the structure, transcriptome and protein expression of perirenal
adipose tissue (PAT) in a fetal sheep model of thyroid hormone deficiency during late gestation. Proportions of unilocular (white) and multilocular (brown) adipocytes, and unilocular adipocyte size, were assessed by histological and stereological techniques. Changes to the adipose transcriptome were investigated by RNA-sequencing and bioinformatic analysis, and proteins of interest were quantified by Western blotting. Results. Hypothyroidism in utero resulted in elevated plasma insulin and leptin concentrations and overgrowth of PAT in the fetus, specifically due to hyperplasia and hypertrophy of unilocular adipocytes with no change in multilocular adipocyte mass. RNA-sequencing and genomic analyses showed that thyroid deficiency affected 34% of the genes identified in fetal adipose tissue. Enriched KEGG and gene ontology pathways were associated with adipogenic, metabolic and thermoregulatory processes, insulin resistance, and a range of endocrine and adipocytokine signalling pathways. Adipose protein levels of signalling molecules, including phosphorylated S6-kinase (pS6K), glucose transporter isoform 4 (GLUT4) and peroxisome proliferator-activated receptor γ (PPARγ), were increased by fetal hypothyroidism. Fetal thyroid deficiency decreased
uncoupling protein 1 (UCP1) protein and mRNA content, and UCP1 thermogenic capacity without
any change in multilocular adipocyte mass. Conclusions. Growth and development of adipose tissue before birth is sensitive to thyroid hormone status in utero. Changes to the adipose transcriptome and phenotype observed in the hypothyroid fetus may have consequences for neonatal survival and the risk of obesity and metabolic dysfunction in later life. -
Foley DW, Pathak RB, Phillips TR, Wilson GL, Bailey PD, Pieri M, Senan A, Meredith D, 'Thiodipeptides targeting the intestinal oligopeptide transporter as a general approach to improving oral drug delivery'
European Journal of Medicinal Chemistry 156 (2018) pp.180-189
ISSN: 0223-5234 eISSN: 1768-3254AbstractThe broad substrate capacity of the intestinal oligopeptide transporter, PepT1, has made it a key target of research into drug delivery. Whilst the substrate capacity of this transporter is broad, studies have largely been limited to small peptides and peptide-like drugs. Here, we demonstrate for the first time that a diverse range of drugs can be targeted towards transport by PepT1 using a hydrolysis resistant carrier. Eleven prodrugs were synthesized by conjugating modified dipeptides containing a thioamide bond to the approved drugs ibuprofen, gabapentin, propofol, aspirin, acyclovir, nabumetone, atenolol, zanamivir, baclofen and mycophenolate. Except for the aspirin and acyclovir prodrugs, which were unstable in the assay conditions and were not further studied, the prodrugs were tested for affinity and transport by PepT1 expressed in Xenopus laevis oocytes: binding affinities ranged from approximately 0.1 to 2 mM. Compounds which showed robust transport in an oocyte trans-stimulation assay were then tested for transcellular transport in Caco-2 cell monolayers: all five tested prodrugs showed significant PepT1-mediated transcellular uptake. Finally, the ibuprofen and propofol prodrugs were tested for absorption in rats: following oral dosing the intact prodrugs and free ibuprofen were measured in the plasma. This provides proof-of-concept for the idea of targeting poorly bioavailable drugs towards PepT1 transport as a general means of improving oral permeability.Published here Open Access on RADAR -
Harris SE, De Blasio MJ, Davis MA, Kelly A, Davenport HM, Wooding FBP, Blache D, Meredith D, Anderson M, Fowden AL, Limesand SW, Forhead AJ, 'Hypothyroidism in utero stimulates pancreatic beta cell proliferation and hyperinsulinaemia in the ovine fetus during late gestation'
The Journal of Physiology 595 (11) (2017) pp.3331-3343
ISSN: 0022-3751 eISSN: 1469-7793AbstractDevelopment of pancreatic beta cell mass before birth is essential for normal growth of the fetus and for long-term control of carbohydrate metabolism in postnatal life. Thyroid hormones are also important regulators of fetal growth, and the present study tested the hypotheses that thyroid hormones promote beta cell proliferation in the fetal ovine pancreatic islets, and that growth retardation in hypothyroid fetal sheep is associated with reductions in pancreatic beta cell mass and circulating insulin concentration in utero. Organ growth and pancreatic islet cell proliferation and mass were examined in sheep fetuses following removal of the thyroid gland in utero. The effects of T3, insulin and leptin on beta cell proliferation rates were determined in isolated fetal ovine pancreatic islets in vitro. Hypothyroidism in the sheep fetus resulted in an asymmetric pattern of organ growth, pancreatic beta cell hyperplasia, and elevated plasma insulin and leptin concentrations. In pancreatic islets isolated from intact fetal sheep, beta cell proliferation in vitro was reduced by T3 in a dose- dependent manner and increased by insulin at high concentrations only. Leptin induced a bimodal response whereby beta cell proliferation was suppressed at the lowest, and increased at the highest, concentrations. Therefore, proliferation of beta cells isolated from the ovine fetal pancreas is sensitive to physiological concentrations of T3, insulin and leptin. Alterations in these hormones may be responsible for the increased beta cell proliferation and mass observed in the hypothyroid sheep fetus and may have consequences for pancreatic function in later life.Published here Open Access on RADAR -
Foley DW, Bermudez I, Bailey PD, Meredith D, 'A cyclosporine derivative is a substrate of the oligopeptide transporter PepT1'
MedChemComm 2016 (7) (2016) pp.999-1002
ISSN: 2040-2503AbstractThe mammalian proton-coupled oligopeptide transport PepT1 is recognised as an important route of oral drug delivery. Here we describe the successful synthesis and PepT1-mediated transport of a dipeptide-conjugated form of the immunosuppressant cyclosporine A, the largest drug tested to date at 1.2 kDa.Published here -
Beale JH, Parker JL, Samsudin F, Barrett AL, Senan A, Bird LE, Scott D, Owens RJ, Sansom MS, Tucker SJ, Meredith D, Fowler PW, Newstead S, 'Crystal Structures of the Extracellular Domain from PepT1 and PepT2 Provide Novel Insights into Mammalian Peptide Transport.'
Structure 23 (10) (2015) pp.1889-1899
ISSN: 0969-2126AbstractMammals obtain nitrogen via the uptake of di- and tri-peptides in the gastrointestinal tract through the action of PepT1 and PepT2, which are members of the POT family of proton-coupled oligopeptide transporters. PepT1 and PepT2 also play an important role in drug transport in the human body. Recent crystal structures of bacterial homologs revealed a conserved peptide-binding site and mechanism of transport. However, a key structural difference exists between bacterial and mammalian homologs with only the latter containing a large extracellular domain, the function of which is currently unknown. Here, we present the crystal structure of the extracellular domain from both PepT1 and PepT2 that reveal two immunoglobulin-like folds connected in tandem, providing structural insight into mammalian peptide transport. Functional and biophysical studies demonstrate that these domains interact with the intestinal protease trypsin, suggesting a role in clustering proteolytic activity to the site of peptide transport in eukaryotic cells.Published here -
Taylor-Wells J, Meredith D, 'The signature sequence region of the human drug transporter organic anion transporting polypeptide 1B1 Is important for protein surface expression'
Journal of Drug Delivery 2014 (2014)
ISSN: 2090-3014AbstractPublished hereThe organic anion transporting polypeptides (OATPs) encompass a family of membrane transport proteins responsible for the uptake of xenobiotic compounds. Human organic anion transporting polypeptide 1B1 (OATP1B1) mediates the uptake of clinically relevant compounds such as statins and chemotherapeutic agents into hepatocytes, playing an important role in drug delivery and detoxification. The OATPs have a putative 12-transmembrane domain topology and a highly conserved signature sequence (human OATP1B1: DSRWVGAWWLNFL), spanning the extracellular loop 3/TM6 boundary. The presence of three conserved tryptophan residues at the TM interface suggests a structural role for the sequence. This was investigated by site-directed mutagenesis of selected amino acids within the sequence D251E, W254F, W258/259F, and N261A. Transport was measured using the substrate estrone-3-sulfate and surface expression detected by luminometry and confocal microscopy, facilitated by an extracellular FLAG epitope. Uptake of estrone-3-sulfate and the surface expression of D251E, W254F, and W258/259F were both significantly reduced from the wild type OATP1B1-FLAG in transfected HEK293T cells. Confocal microscopy revealed that protein was produced but was retained intracellularly. The uptake and expression of N261A were not significantly different. The reduction in surface expression and intracellular protein retention indicates a structural and/or membrane localization role for these signature sequence residues in the human drug transporter OATP1B1.
-
Meredith D, 'Intestinal physiology amongst the dreaming spires: the 24th EITG meeting, 4-7th September 2011, Oxford, UK'
Genes & Nutrition 6 (2012) pp.3-4
ISSN: 1555-8932Published here -
Taylor-Wells JC, Meredith D, Kelly S, 'Structural Determination of Oatp Transporters Utilising Homology Models and Cell Based Assays'
The FASEB Journal 26 (2012)
ISSN: 0892-6638 eISSN: 1530-6860 -
Suhre K, Shin S, Petersen A, Mohney R, Meredith D, Wagele B, Altmaier E, Deloukas P, Erdmann J, Grundberg E, Hammond C, de Angelis M, Kastenmuller G, Kottgen A, Kronenberg F, Mangino M, Meisinger C, Meitinger T, Mewes H, Milburn M, Prehn C, Raffler J, Ried J, Romisch-Margl W, Samani N, Small K, Wichmann H, Zhai G, Illig T, Spector T, Adamski J, Soranzo N, Gieger C, 'Human metabolic individuality in biomedical and pharmaceutical research'
Nature 477 (7362) (2011) pp.54-60
ISSN: 0028-0836 eISSN: 1476-4687AbstractGenome-wide association studies (GWAS) have identified many risk loci for complex diseases, but effect sizes are typically small and information on the underlying biological processes is often lacking. Associations with metabolic traits as functional intermediates can overcome these problems and potentially inform individualized therapy. Here we report a comprehensive analysis of genotype-dependent metabolic phenotypes using a GWAS with non-targeted metabolomics. We identified 37 genetic loci associated with blood metabolite concentrations, of which 25 show effect sizes that are unusually high for GWAS and account for 10-60% differences in metabolite levels per allele copy. Our associations provide new functional insights for many disease-related associations that have been reported in previous studies, including those for cardiovascular and kidney disorders, type 2 diabetes, cancer, gout, venous thromboembolism and Crohn's disease. The study advances our knowledge of the genetic basis of metabolic individuality in humans and generates many new hypotheses for biomedical and pharmaceutical research.Published here -
Foley D, Rajamanickam J, Bailey P, Meredith D, 'Bioavailability Through PepT1: The Role of Computer Modelling in Intelligent Drug Design'
Current Computer Aided-Drug Design 6 (1) (2010) pp.68-78
ISSN: 1573-4099AbstractPublished hereIn addition to being responsible for the majority of absorption of dietary nitrogen, the mammalian proton-coupled di- and tripeptide transporter PepT1 is also recognised as a major route of drug delivery for several important classes of compound, including lactam antibiotics and angiotensin-converting enzyme inhibitors. Thus there is considerable interest in the PepT1 protein and especially its substrate binding site. In the absence of a crystal structure, computer modelling has been used to try to understand the relationship between PepT1 3D structure and function. Two basic approaches have been taken: modelling the transporter protein, and modelling the substrate. For the former, computer modelling has evolved from early interpretations of the twelve transmembrane domain structure to more recent homology modelling based on recently crystallised bacterial members of the major facilitator superfamily (MFS). Substrate modelling has involved the proposal of a substrate binding template, to which all substrates must conform and from which the affinity of a substrate can be estimated relatively accurately, and identification of points of potential interaction of the substrate with the protein by developing a pharmacophore model of the substrates. Most recently, these two approaches have moved closer together, with the attempted docking of a substrate library onto a homology model of the human PepT1 protein. This article will review these two approaches in which computers have been applied to peptide transport and suggest how such computer modelling could affect drug design and delivery through PepT1.
-
Pillai S, Meredith D, 'SLC36A4 (hPAT4) Is a High Affinity Amino Acid Transporter When Expressed in Xenopus laevis Oocytes'
Journal of Biological Chemistry 286 (4) (2010) pp.2455-2460
ISSN: 0021-9258 eISSN: 1083-351XAbstractPublished hereThe SLC36 family of transporters consists of four genes, two of which, SLC36A1 and SLC36A2, have been demonstrated to code for human proton-coupled amino acid transporters or hPATs. Here we report the characterization of the fourth member of the family, SLC36A4 or hPAT4, which when expressed in Xenopus laevis oocytes also encodes a plasma membrane amino acid transporter, but one that is not proton-coupled and has a very high substrate affinity for the amino acids proline and tryptophan. hPAT4 in Xenopus oocytes mediated sodium-independent, electroneutral uptake of [3H]proline, with the highest rate of uptake when the uptake medium pH was 7.4 and an affinity of 3.13 M. Tryptophan was also an excellently transported substrate with a similarly high affinity (1.72 M). Other amino acids that inhibited [3H]proline were isoleucine (Ki 0.23 mM), glutamine (0.43 mM), methionine (0.44 mM), and alanine (1.48 mM), and with lower affinity, glycine, threonine, and cysteine (Ki >5mM for all). Of the amino acids directly tested for transport, only proline, tryptophan, and alanine showed significant uptake, whereas glycine and cysteine did not. Of the non-proteogenic amino acids and drugs tested, only sarcosine produced inhibition (Ki 1.09 mM), whereas -aminobutyric acid (GABA), -alanine, L-Dopa, D-serine, and -aminolevulinic acid were without effect on [3H]proline uptake. This characterization of hPAT4 as a very high affinity/low capacity non-proton-coupled amino acid transporter raises questions about its physiological role, especially as the transport characteristics of hPAT4 are very similar to the Drosophila orthologue PATH, an amino acid -œtransceptor- that plays a role in nutrient sensing.
-
Taylor-Wells JC, Meredith D, Kelly S, 'Structural Determination of OATP Transporters Utilising Homology Models and Cell Based Assays'
The FASEB Journal 24 (1) (2010)
ISSN: 0892-6638 eISSN: 1530-6860AbstractMembrane transport proteins (transporters) are present in the lipid bilayer of cells and selectively transport compounds across the membrane. The organic anion transporting polypeptides (OATPs) are a family of membrane transporters expressed in a variety of tissues, including absorptive/excretory cells of the liver, kidney and intestine. In addition, OATPs transport an array of endogenous and xenobiotic compounds including bile salts, thyroid hormones, statins and antineoplastics. OATPs play an important role in cell homeostasis and drug absorption/excretion, and have also been implicated in disease and drug-drug interactions. However there is still much to determine regarding the binding properties and exact structure of these proteins. The topology and binding properties of OATP1A2, OATP1B1, OATP1B3 and OATP2B1 have been investigated using a HEK293T cell model.OATP proteins have been expressed in HEK293T cells and kinetic properties determined using the OATP substrate estrone-3-sulfate. Bioinformatics analysis using various topology models has revealed that OATP1B1, OATP1B3 and OATP2B1 have between 11–12 transmembrane domains. However there could be as few as 8 domains, as is the predicted topology of the less well characterised OATP1A2. The topology of these proteins has been experimentally investigated using a FLAG epitope tag recognition system. Epitope tags were cloned into the putative extracellular regions of the protein, which were recognised by a FLAG antibody. The detection of the antibody was quantified by a chemiluminescence method and those detected confirmed as extracellular regions. The findings of this study will be used to further determine drug transport pathways and aid in future drug development.
-
Pieri M, Christian H, Wilkins R, Boyd C, Meredith D, 'The apical (hPepT1) and basolateral peptide transport systems of Caco-2 cells are regulated by AMP-activated protein kinase'
American Journal of Physiology - Gastrointestinal and Liver Physiology 299 (1) (2010) pp.G136-G143
ISSN: 0193-1857 eISSN: 1522-1547AbstractPieri M, Christian HC, Wilkins RJ, Boyd CAR, Meredith D. The apical (hPepT1) and basolateral peptide transport systems of Caco-2 cells are regulated by AMP-activated protein kinase. Am J Physiol Gastrointest Liver Physiol 299: G136-G143, 2010. First published April 29, 2010; doi:10.1152/ajpgi.00014.2010.-The effect of 5-aminoimidazole-4-carboxamide-ribonucleoside (AICAR) activation of the AMP-activated protein kinase (AMPK) on the transport of the model radiolabeled dipeptide [(3)H]-D-Phe-L-Gln was investigated in the human epithelial colon cancer cell line Caco-2. Uptake and transepithelial fluxes of [(3)H]-D-Phe-L-Gln were carried out in differentiated Caco-2 cell monolayers, and hPepT1 and glucose transporter 2 (GLUT2) protein levels were quantified by immunogold electron microscopy. AICAR treatment of Caco-2 cells significantly inhibited apical [(3)H]-D-Phe-L-Gln uptake, matched by a decrease in brush-border membrane hPepT1 protein but with a concomitant increase in the facilitated glucose transporter GLUT2. A restructuring of the apical brush-border membrane was seen by electron microscopy. The hPepT1-mediated transepithelial (A-to-B) peptide flux across the Caco-2 monolayers showed no significant alteration in AICAR-treated cells. The electrical resistance in the AICAR-treated monolayers was significantly higher compared with control cells. Inhibition of the sodium/hydrogen exchanger 3 (NHE3) had an additive effect to AICAR, suggesting that the AMPK effect is not via NHE3. Fluorescence measurement of intracellular pH showed no reduction in the proton gradient driving PepT1-mediated apical uptake. The reduction in apical hPepT1 protein and dipeptide uptake after AICAR treatment in Caco-2 cells demonstrates a regulatory effect of AMPK on hPepT1, along with an influence on both the microvilli and tight junction structures. The absence of an associated reduction in transepithelial peptide movement implies an additional stimulatory effect of AICAR on the basolateral peptide transport system in these cells. These results provide a link between the hPepT1 transporter and the metabolic state of this model enterocyte.Published here -
Wilson MC, Meredith D, Bunnun C, Sessions RB, Halestrap AP, 'Studies on the DIDS-binding Site of Monocarboxylate Transporter 1 Suggest a Homology Model of the Open Conformation and a Plausible Translocation Cycle'
Journal of Biological Chemistry 284 (2009) pp.20011-20021
ISSN: 0021-9258 eISSN: 1083-351XAbstractSite-directed mutagenesis of MCT1 was performed on exofacial lysines Lys(38), Lys(45), Lys(282), and Lys(413). K38Q-MCT1 and K38R-MCT1 were inactive when expressed at the plasma membrane of Xenopus laevis oocytes, whereas K45R/K282R/K413R-MCT1 and K45Q/K282Q/K413Q-MCT1 were active. The former exhibited normal reversible and irreversible inhibition by DIDS, whereas the latter showed less reversible and no irreversible inhibition. K45Q/K413Q-MCT1 retained some irreversible inhibition, whereas K45Q/K282Q-MCT1 and K282Q/K413Q-MCT1 did not. These data suggest that the twoDIDSSO(3)(-) groups interact with positively charged Lys(282) together with Lys(45) and/or Lys(413). This positions one DIDS isothiocyanate group close to Lys(38), leading to its covalent modification and irreversible inhibition. Additional mutagenesis revealed that DIDS cross-links MCT1 to its ancillary protein embigin using either Lys(38) or Lys(290) of MCT1 and Lys(160) or Lys(164) of embigin. We have modeled a possible structure for the outward facing (open) conformation of MCT1 by employing modest rotations of the C-terminal domain of the inner facing conformation modeled previously. The resulting model structure has a DIDS-binding site consistent with experimental data and locates Lys(38) in a hydrophobic environment at the bottom of a substrate-binding channel. Our model suggests a translocation cycle in which Lys(38) accepts a proton before binding lactate. Both the lactate and proton are then passed through the channel via Asp(302-) and Asp(306-), an ion pair already identified as important for transport and located adjacent to Phe(360), which controls channel selectivity. The cross-linking data have also been used to model a structure of MCT1 bound to embigin that is consistent with published data.Published here -
Mace O, Lister N, Morgan E, Shepherd E, Affleck J, Helliwell P, Bronk J, Kellett G, Meredith D, Boyd R, Pieri M, Bailey P, Pettcrew R, Foley D, 'An energy supply network of nutrient absorption coordinated by calcium and T1R taste receptors in rat small intestine.'
The Journal of Physiology 587 (1) (2009) pp.195-210
ISSN: 0022-3751AbstractPublished hereT1R taste receptors are present throughout the gastrointestinal tract. Glucose absorption comprises active absorption via SGLT1 and facilitated absorption via GLUT2 in the apical membrane. Trafficking of apical GLUT2 is rapidly up-regulated by glucose and artificial sweeteners, which act through T1R2 + T1R3/alpha-gustducin to activate PLC beta 2 and PKC beta II. We therefore investigated whether non-sugar nutrients are regulated by taste receptors using perfused rat jejunum in vivo. Under different conditions, we observed a Ca(2+)-dependent reciprocal relationship between the H(+)/oligopeptide transporter PepT1 and apical GLUT2, reflecting the fact that trafficking of PepT1 and GLUT2 to the apical membrane is inhibited and activated by PKC beta II, respectively. Addition of l-glutamate or sucralose to a perfusate containing low glucose (20 mm) each activated PKC beta II and decreased apical PepT1 levels and absorption of the hydrolysis-resistant dipeptide l-Phe(Psi S)-l-Ala (1 mm), while increasing apical GLUT2 and glucose absorption within minutes. Switching perfusion from mannitol to glucose (75 mm) exerted similar effects. l-Glutamate induced rapid GPCR internalization of T1R1, T1R3 and transducin, whereas sucralose internalized T1R2, T1R3 and alpha-gustducin. We conclude that l-glutamate acts via amino acid and glucose via sweet taste receptors to coordinate regulation of PepT1 and apical GLUT2 reciprocally through a common enterocytic pool of PKC beta II. These data suggest the existence of a wider Ca(2+) and taste receptor-coordinated transport network incorporating other nutrients and/or other stimuli capable of activating PKC beta II and additional transporters, such as the aspartate/glutamate transporter, EAAC1, whose level was doubled by l-glutamate. The network may control energy supply.
-
Wilson M, Meredith D, Bunnun C, Sessions R, Halestrap A, 'Studies on the DIDS binding site of monocarboxylate transporter 1 suggest a homology model of the open conformation and a plausible translocation cycle'
Journal of Biological Chemistry 284 (30) (2009) pp.20011-20021
ISSN: 0021-9258AbstractSite-directed mutagenesis of MCT1 was performed on exofacial lysines Lys(38), Lys(45), Lys(282), and Lys(413). K38Q-MCT1 and K38R-MCT1 were inactive when expressed at the plasma membrane of Xenopus laevis oocytes, whereas K45R/K282R/K413R-MCT1 and K45Q/K282Q/K413Q-MCT1 were active. The former exhibited normal reversible and irreversible inhibition by DIDS, whereas the latter showed less reversible and no irreversible inhibition. K45Q/K413Q-MCT1 retained some irreversible inhibition, whereas K45Q/K282Q-MCT1 and K282Q/K413Q-MCT1 did not. These data suggest that the twoDIDSSO(3)(-) groups interact with positively charged Lys(282) together with Lys(45) and/or Lys(413). This positions one DIDS isothiocyanate group close to Lys(38), leading to its covalent modification and irreversible inhibition. Additional mutagenesis revealed that DIDS cross-links MCT1 to its ancillary protein embigin using either Lys(38) or Lys(290) of MCT1 and Lys(160) or Lys(164) of embigin. We have modeled a possible structure for the outward facing (open) conformation of MCT1 by employing modest rotations of the C-terminal domain of the inner facing conformation modeled previously. The resulting model structure has a DIDS-binding site consistent with experimental data and locates Lys(38) in a hydrophobic environment at the bottom of a substrate-binding channel. Our model suggests a translocation cycle in which Lys(38) accepts a proton before binding lactate. Both the lactate and proton are then passed through the channel via Asp(302-) and Asp(306-), an ion pair already identified as important for transport and located adjacent to Phe(360), which controls channel selectivity. The cross-linking data have also been used to model a structure of MCT1 bound to embigin that is consistent with published data.Published here -
Foley D, Bailey P, Pieri M, Meredith D, 'Targeting ketone drugs towards transport by the intestinal peptide transporter, PepT1'
Organic and Biomolecular Chemistry 7 (6) (2009) pp.1064-1067
ISSN: 1477-0520AbstractThiodipeptide prodrugs of the ketone nabumetone are shown to have affinity for, and be transported by, PepT1 in vitro.Published here -
Foley D, Pieri M, Pettecrew R, Price R, Miles S, Lam H, Bailey P, Meredith D, 'The in vitro transport of model thiodipeptide prodrugs designed to target the intestinal oligopeptide transporter, PepT1'
Organic and Biomolecular Chemistry 7 (18) (2009) pp.3652-3656
ISSN: 1477-0520AbstractPublished hereA thiodipeptide carrier system is shown to be effective at enabling a range of covalently bound molecules, including benzyl, benzoyl and ibuprofen conjugates, to be transported via the intestinal peptide transporter PepT1, demonstrating its potential as a rational drug delivery target.
-
Meredith D, 'The mammalian proton-coupled peptide cotransporter PepT1: sitting on the transporter-channel fence?'
Philosophical Transactions of the Royal Society B: Biological Sciences 364 (1514) (2009) pp.203-207
ISSN: 0962-8436 eISSN: 1471-2970AbstractThe proton-coupled di-and tripeptide transporter PepT1 (SLC15a1) is the major route by which dietary nitrogen is taken up from the small intestine, as well as being the route of entry for important therapeutic (pro) drugs such as the beta-lactam antibiotics, angiotensin-converting enzyme inhibitors and antiviral and anti-cancer agents. PepT1 is a member of the major facilitator superfamily of 12 transmembrane domain transporter proteins. Expression studies in Xenopus laevis on rabbit PepT1 that had undergone site-directed mutagenesis of a conserved arginine residue (arginine(282) in transmembrane domain 7) to a glutamate revealed that this residue played a role in the coupling of proton and peptide transport and prevented the movement of non-coupled ions during the transporter cycle. Mutations of arginine(282) to other non-positive residues did not uncouple proton peptide cotransport, but did allow additional ion movements when substrate was added. By contrast, mutations to positive residues appeared to function the same as wild- type. These findings are discussed in relation to the functional role that arginine(282) may play in the way PepT1 operates, together with structural information from the homology model of PepT1 based on the Escherichia coli lactose permease crystal structure.Published here -
Pieri M, Gan C, Bailey P, Meredith D, 'The transmembrane tyrosines Y56, Y91 and Y167 play important roles in determining the affinity and transport rate of the rabbit proton-coupled peptide transporter PepT1'
The International Journal of Biochemistry and Cell Biology 41 (11) (2009) pp.2204-2213
ISSN: 1357-2725AbstractThe mammalian proton-coupled peptide transporter PepT1 is widely accepted as the major route of uptake for dietary nitrogen, as well as being responsible for the oral absorption of a number of classes of drugs, including beta-lactam antibiotics and angiotensin-converting enzyme (ACE) inhibitors. Using site-directed mutagenesis and zero-trans transport assays, we investigated the role of conserved tyrosines in the transmembrane domains (TMDs) of rabbit PepT1 as predicted by hydropathy plots. All the individual TMD tyrosines were substituted with phenylalanine and shown to retain the ability to traffic to the plasma membrane of Xenopus laevis oocytes. These single substitutions of TMD tyrosines by phenylalanine residues did not affect the proton dependence of peptide uptake, with all retaining wildtype PepT1-like pH dependence. Individual mutations of four of the nine TMD residue tyrosines (Y64, Y287, Y345 and Y587) were without measurable effect on PepT1 function, whereas the other five (Y12, Y56, Y91, Y167 and Y345) were shown to result in altered transport function compared to the wild-type PepT1. Intriguingly, the affinity of Y56F-PepT1 was found to be dramatically increased (approximately 100-fold) in comparison to that of the wild-type rabbit PepT1. Y91 mutations also affected the substrate affinity of the transporter, which increased in line with the hydrophilicity of the substituted amino acid (F>Y>Q>R). Y167 was demonstrated to play a pivotal role in rabbit PepT1 function since Y167F, Y167R and Y167Q demonstrated very little transport function. These results are discussed with regard to a proposed mechanism for PepT1 substrate binding.Published here -
Pieri M, Hall D, Price R, Bailey P, Meredith D, 'Site-directed mutagenesis of Arginine282 suggests how protons and peptides are co-transported by rabbit PepT1'
The International Journal of Biochemistry and Cell Biology 40 (4) (2008) pp.721-730
ISSN: 1357-2725AbstractThe mammalian proton-coupled peptide transporter PepT1 is the major route of uptake for dietary nitrogen, as well as the oral absorption of a number of drugs, including beta-lactam. antibiotics and angiotensin-converting enzyme inhibitors. Here we have used site-directed mutagenesis to investigate further the role of conserved charged residues in transmembrane domains. Mutation of rabbit PepT1 arginine282 (R282, transmembrane domain 7) to a positive (R282K) or physiologically titratable residue (R282H), resulted in a transporter with wild-type characteristics when expressed in Xenopus laevis oocytes. Neutral (R282A, R282Q) or negatively charged (R282D, R282E) substitutions gave a transporter that was not stimulated by external acidification (reducing pH(out) from 7.4 to 5.5) but transported at the same rate as the wild-type maximal rate (pH,,ut 5.5); however, only the R282E mutation was unable to concentrate substrate above the extracellular level. All of the R282 mutants showed trans-stimulation of efflux comparable to the wild-type, except R282E-PepT1 which was faster. A conserved negatively charged residue, aspartate341 (D341) in transmembrane domain 8 was implicated in forming a charge pair with R282, as R282E/D341R- and R282D/D341R-PepT1 had wild-type transporter characteristics. Despite their differences in ability to accumulate substrate, both R282E- and R282D-PepT1 showed an increased charge:peptide stoichiometry over the wild-type 1: 1 ratio for the neutral dipeptide Gly-L-Gln, measured using two-electrode voltage clamp. This extra charge movement was linked to substrate transport, as 4-aminobenzoic acid, which binds but is not translocated, did not induce membrane potential depolarisation in R282E-expressing oocytes. A model is proposed for the substrate hinding/translocation process in PepT1.Published here -
Meredith D, Christian H, 'The SLC16 monocaboxylate transporter family'
Xenobiotica: the fate of foreign compounds in biological systems 38 (41128) (2008) pp.1072-1106
ISSN: 0049-8254 eISSN: 1366-5928Abstract1. The monocarboxylate transporter (MCT, SLC16) family comprises 14 members, of which to date only MCT1-4 have been shown to carry monocarboxylates, transporting important metabolic compounds such as lactate, pyruvate and ketone bodies in a proton-coupled manner. The transport of such compounds is fundamental for metabolism, and the tissue locations, properties and regulation of these isoforms is discussed.Published here
2. Of the other members of the MCT family, MCT8 (a thyroid hormone transporter) and TAT1 (an aromatic amino acid transporter) have been characterized more recently, and their physiological roles are reviewed herein. The endogenous substrates and functions of the remaining members of the MCT family await elucidation.
3. The MCT proteins have the typical twelve transmembrane-spanning domain (TMD) topology of membrane transporter proteins, and their structure-function relationship is discussed, especially in relation to the future impact of the single nucleotide polymorphism (SNP) databases and, given their ability to transport pharmacologically relevant compounds, the potential impact for pharmacogenomics. -
Meredith D, Gehl KA, Seymour J, Ellory JC, Wilkins RJ, 'Characterization of sulphate transporters in isolated bovine articular chondrocytes'
Journal of Orthopaedic Research 25 (9) (2007) pp.1145-1153
ISSN: 0736-0266 eISSN: 1554-527XAbstractUptake of SO42- by articular chondrocytes is an essential step in the pathway for sulphation of glycosammoglycans (GAGs), with mutations in So(4)(2-) transport proteins resulting in abnormalities of skeletal growth. In the present study, the transporters mediating SO42- transport in bovine articular chondrocytes have been characterized. Expression of candidate transporters was determined using RT-PCR, while SO42- transport was measured in radioisotope flux experiments. RT-PCR experiments showed that bovine articular chondrocytes express three transporters known to transport SO42- : AE2 (SLC4a2), DTDST (SLC26a2), and SLC26a11. Other transporters-NaS-1 (SLC13a1), SAT-1 (SLC26a1), DRA (SLC26a3), SLC26a6 (PAT1), SLC26a7, SLC26a8 (Tat-1), and SLC26a9-were, however, not detected. In functional experiments, SO42- uptake was temperature sensitive, inhibited by 60% by DIDS (50 mu m) and exhibited saturation kinetics, with a K-m value of 16 mM. Uptake was also inhibited at alkaline extracellular pH. In further experiments, a K-i value for DIDS inhibition of SO42- efflux of 5 mu M was recorded. A DlDS-sensitive component of SO42- efflux persisted in solutions lacking Cl- ions. These data are interpreted as evidence for the preferential operation of carrier-mediated exchange of SO42- - for Cl-, while an alternative SO42- -OH- exchange mode is also possible. (c) 2007 Orthopaedic Research Society.Published here -
Wilson C, Vereshchagina N, Reynolds B, Meredith D, Boyd CAR, Goberdhan DCI, 'Extracellular and subcellular regulation of the PI3K/Akt cassette: new mechanisms for controlling insulin and growth factor signalling'
Biochemical Society Transactions 35 (2) (2007) pp.219-221
ISSN: 0300-5127 eISSN: 1470-8752AbstractThe PI3K (phosphoinositide 3-kinase)/Akt (also called protein kinase B) signalling cassette plays a central role in the response to growth factors, particularly insulin-like molecules, and its misregulation is a characteristic feature of diabetes and many forms of human cancer. Recent molecular genetic studies initiated in the fruitfly, Drosophila melanogaster, have highlighted two new cell-type-specific mechanisms regulating PI3K/Akt signalling and its downstream effects. First, the cellular response to this cassette is modulated by several classes of cell-surface transporters and sensors, suggesting an important role for extracellular nutrients in insulin-sensitivity. Secondly, various cell types show a markedly different subcellular distribution of the activated kinase Akt, influencing the cellular functions of this molecule. These findings reveal new mechanisms by which processes such as growth, lipogenesis and insulin resistance can be differentially regulated and may suggest novel strategies for treating insulin-linked diseases.Published here -
Solomon DH, Wilkins RJ, Meredith D, Browning JA, 'Characterisation of inorganic phosphate transport in bovine articular chondrocytes'
Cellular Physiology and Biochemistry 20 (1-4) (2007) pp.99-108
ISSN: 1015-8987AbstractIn mineralising tissues such as growth plate cartilage extracellular organelles derived from the chondrocyte membrane are present. These matrix vesicles (MV) possess membrane transporters that accumulate Ca2+ and inorganic phosphate (P-i), and initiate the formation of hydroxyapatite crystals. MV are also present in articular cartilage, and hydroxyapatite crystals are believed to promote cartilage degradation in osteoarthritic joints. In the present study, P-i transport pathways in isolated bovine articular chondrocytes have been characterised. P-i uptake was temperature-sensitive and could be resolved into Na+-dependent and Na+-independent components. The Na+-dependent component saturated at high concentrations of extracellular P-i, with a K-m for P-i of 0.17mM. In solutions lacking Na+, uptake did not fully saturate, implying that under these conditions carrier-mediated uptake is supplemented by a diffusive pathway. Both Na+-dependent and Na+-independent components were sensitive to the P-i transport inhibitors phosphonoacetate and arsenate, although a fraction of Na+-independent P-i uptake was resistant to these anions. Total P-i uptake was optimal at pH 7.4, and reduced as pH was made more acidic or more alkaline, an effect that represented reduced Na+-dependent influx. RT-PCR analysis confirmed that two members of the NaPi III family, Pit-1 and Pit-2, are expressed, but that NaPi II transporters are not.Published here -
Meredith D, Price RA, 'Molecular modeling of PepT1- Towards a structure'
Journal of Membrane Biology 213 (2) (2006) pp.79-88
ISSN: 0022-2631 eISSN: 1432-1424AbstractThe proton-coupled uptake of di- and tri-peptides is the major route of dietary nitrogen absorption in the intestine and of reabsorption of filtered protein in the kidney. In addition, the transporters involved, PepT1 (SLC15a1) and PepT2 (SLC15a2), are responsible for the uptake and tissue distribution of a wide range of pharmaceutically important compounds, including beta-lactam antibiotics, angiotensin-converting enzyme inhibitors, anti-cancer and anti-viral drugs. PepT1 and PepT2 are large proteins, with over 700 amino acids, and to date there are no reports of their crystal structures, nor of those of related proteins from lower organisms. Therefore there is virtually no information about the protein 3-D structure, although computer-based approaches have been used to both model the transmembrane domain (TM) layout and to produce a substrate binding template. These models will be discussed, and a new one proposed from homology modeling rabbit PepT1 to the recently crystallized bacterial transporters LacY and GlpT. Understanding the mechanism by which PepT1 and PepT2 bind and transport their substrates is of great interest to researchers, both in academia and in the pharmaceutical industries.Published here -
Omer S, Meredith D, Morris JF, Christian HC, 'Evidence for the role of adenosine 5 '-triphosphate-binding cassette (ABC)-A1 in the externalization of annexin 1 from pituitary folliculostellate cells and ABCA1-transfected cell models'
Endocrinology 147 (7) (2006) pp.3219-3227
ISSN: 0013-7227 eISSN: 1945-7170AbstractAnnexin 1 (ANXA1), a 37-kDa protein, is a member of the superfamily of Ca2+- and phospholipid-binding annexin proteins. In the anterior pituitary, ANXA1 is expressed mainly by folliculostellate (FS) cells and mediates the early delayed feedback inhibition exerted by glucocorticoids on the release of ACTH and other pituitary hormones. It has been previously demonstrated that TtT/GF cells (a FS cell line) express and externalize ANXA1 in response to glucocorticoid treatment. However, ANXA1 lacks a cleavable signal sequence and externalization is not affected by inhibitors of the secretory pathway. We have previously shown that glyburide, an ATP-binding cassette (ABC) transporter inhibitor, inhibits the externalization of ANXA1 from TtT/GF cells and pituitary tissue. Here we investigated whether ABCA1 is involved in ANXA1 externalization. The use of the ABCA1-transporter inhibitors geranyl-geranyl pyrophosphate and sulfobromophthalein significantly inhibited ANXA1 externalization. Partial silencing of ABCA1 expression in TtT/GF cells by siRNA also significantly decreased the amount of cell surface ANXA1. However, anterior pituitary tissue from ABCA1-null mice was found to externalize ANXA1 normally. Because compensation by other ABC family members may occur in vivo, ANXA1 externalization was studied in two transfection models: Xenopus oocytes injected with ABCA1 mRNA and AtT20 D1 corticoctroph cells cotransfected with ABCA1-green fluorescent protein and ANXA1. ABCA1-expressing oocytes, but not water-injected controls, were found to externalize ANXA1. Expression of ABCA1 in AtT20 D1 cells significantly increased the amount of cell surface ANXA1, compared with mock-transfected and ANXA1-only transfected controls. Together these data provide evidence for a role of ABCA1 in ANXA1 export.Published here -
Panitsas KE, Boyd CAR, Meredith D, 'Evidence that the rabbit proton-peptide co-transporter PepT1 is a multimer when expressed in Xenopus laevis oocytes'
Pflügers Archiv European Journal of Physiology 452 (1) (2006) pp.53-63
ISSN: 0031-6768 eISSN: 1432-2013AbstractTo test whether the rabbit proton-coupled peptide transporter PepT1 is a multimer, we have employed a combination of transport assays, luminometry and site-directed mutagenesis. A functional epitope-tagged PepT1 construct (PepT1-FLAG) was co-expressed in Xenopus laevis oocytes with a non-functional but normally trafficked mutant form of the same transporter (W294F-PepT1). The amount of PepT1-FLAG cRNA injected into the oocytes was kept constant, while the amount of W294F-PepT1 cRNA was increased over the mole fraction range of 0 to 1. The uptake of [H-3]-D-Phe-L-Gln into the oocytes was measured at pH(out) 5.5, and the surface expression of PepT I-FLAG was quantified by luminometry. As the mole fraction of injected W294F-PepT1 increased, the uptake of D-Phe-L-Gln decreased. This occurred despite the surface expression of PepT1-FLAG remaining constant, and so we can conclude that PepT1 must be a multimer. Assuming that PepT1 acts as a homomultimer, the best fit for the modelling suggests that PepT1 could be a tetramer, with a minimum requirement of two functional subunits in each protein complex. Western blotting also showed the presence of higher-order complexes of PepT1-FLAG in oocyte membranes. It should be noted that we cannot formally exclude the possibility that PepT1 interacts with unidentified Xenopus protein(s). The finding that PepT1 is a multimer has important implications for the molecular modelling of this protein.Published here -
Bailey PD, Boyd CAR, Collier ID, George JP, Kellett GL, Meredith D, Morgan KM, Pettecrew R, Price RA, 'Affinity prediction for substrates of the peptide transporter PepT1'
Chemical Communications 2006 (3) (2006) pp.323-325
ISSN: 1359-7345 eISSN: 1364-548XAbstractA quantitative method has been developed for determining the affinity of substrates for the peptide transporter PepT1, allowing oral availability of drugs via PepT1 to be estimated.Published here -
Bailey PD, Boyd CAR, Collier ID, Kellett GL, Meredith D, Morgan KM, Pettecrew R, Price RA, 'Probing dipeptide trans/cis stereochemistry using pH control of thiopeptide analogues, and application to the PepT1 transporter'
Organic and Biomolecular Chemistry 3 (2005) pp.4038-4039
ISSN: 1477-0520 eISSN: 1477-0539AbstractThe stereochemistry of thiodipeptides of proline [e.g. Ala-psi[CS-N]-Pro] can be controlled using pH, allowing the trans-preference for substrates of the peptide transporter PepT1 to be confirmed.Published here -
Wilson MC, Meredith D, Fox JEM, Manoharan C, Davies AJ, Halestrap AP, 'Basigin (CD147) is the target for organomercurial inhibition of monocarboxylate transporter isoforms 1 and 4 - The ancillary protein for the insensitive MCT2 is embigin (gp70)'
Journal of Biological Chemistry 280 (2005) pp.27213-27221
ISSN: 0021-9258 eISSN: 1083-351XAbstractTranslocation of monocarboxylate transporters MCT1 and MCT4 to the plasma membrane requires CD147 (basigin) with which they remain tightly associated. However, the importance of CD147 for MCT activity is unclear. MCT1 and MCT4 are both inhibited by the cell-impermeant organomercurial reagent p-chloromercuribenzene sulfonate (pCMBS). Here we demonstrate by site-directed mutagenesis that removal of all accessible cysteine residues on MCT4 does not prevent this inhibition. pCMBS treatment of cells abolished co-immunoprecipitation of MCT1 and MCT4 with CD147 and enhanced labeling of CD147 with a biotinylated-thiol reagent. This suggested that CD147 might be the target of pCMBS, and further evidence for this was obtained by treatment of cells with the bifunctional organomercurial reagent fluorescein dimercury acetate that caused oligomerization of CD147. Site-directed mutagenesis of CD147 implicated the disulfide bridge in the Ig-like C2 domain of CD147 as the target of pCMBS attack. MCT2, which is pCMBS-insensitive, was found to co-immunoprecipitate with gp70 rather than CD147. The interaction between gp70 and MCT2 was confirmed using fluorescence resonance energy transfer between the cyan fluorescent protein- and yellow fluorescent protein-tagged MCT2 and gp70. pCMBS strongly inhibited lactate transport into rabbit erythrocytes, where MCT1 interacts with CD147, but not into rat erythrocytes where it interacts with gp70. These data imply that inhibition of MCT1 and MCT4 activity by pCMBS is mediated through its binding to CD147, whereas MCT2, which associates with gp70, is insensitive to pCMBS. We conclude that ancillary proteins are required to maintain the catalytic activity of MCTs as well as for their translocation to the plasma membrane.Published here -
Goberdhan DCI, Meredith D, Boyd CAR, Wilson C, 'PAT-related amino acid transporters regulate growth via a novel mechanism that does not require bulk transport of amino acids'
Development 132 (2005) pp.2365-2375
ISSN: 0950-1991 eISSN: 1477-9129AbstractGrowth in normal and tumour cells is regulated by evolutionarily conserved extracellular inputs from the endocrine insulin receptor (InR) signalling pathway and by local nutrients. Both signals modulate activity of the intracellular TOR kinase, with nutrients at least partly acting through changes in intracellular amino acid levels mediated by amino acid transporters. We show that in Drosophila, two molecules related to mammalian proton-assisted SLC36 amino acid transporters (PATs), CG3424 and CG1139, are potent mediators of growth. These transporters genetically interact with TOR and other InR signalling components, indicating that they control growth by directly or indirectly modulating the effects of TOR signalling. A mutation in the CG3424 gene, which we have named pathetic (path), reduces growth in the fly. In a heterologous Xenopus oocyte system, PATH also activates the TOR target S6 kinase in an amino acid-dependent way. However, functional analysis reveals that PATH has an extremely low capacity and an exceptionally high affinity compared with characterised human PATs and the CG1139 transporter. PATH and potentially other PAT-related transporters must therefore control growth via a mechanism that does not require bulk transport of amino acids into the cell. As PATH is likely to be saturated in vivo, we propose that one specialised. function of high-affinity PAT-related molecules is to maintain growth as local nutrient levels fluctuate during development.Published here -
Bailey PD, Boyd CAR, Collier ID, George JG, Kellett GL, Meredith D, Morgan KM, Pettecrew R, Price RA, Pritchard RG, 'Conformational and spacial preferences for substrates of PepT1'
Chemical Communications 2005 (42) (2005) pp.5352-5354
ISSN: 1359-7345 eISSN: 1364-548XAbstractThe conformation at the first residue of dipeptide substrates for the peptide transporter PepT1 has been probed using constrained peptide analogues, and the active conformation has been identified.Published here -
Meredith D, 'Site-directed mutation of arginine 282 to glutamate uncouples the movement of peptides and protons by the rabbit proton-peptide cotransporter PepT1'
Journal of Biological Chemistry 279 (2004) pp.15795-15798
ISSN: 0021-9258 eISSN: 1083-351XAbstractA conserved positive residue in the seventh transmembrane domain of the mammalian proton-coupled di- and tripeptide transporter PepT1 has been shown by site-directed mutagenesis to be a key residue for protein function. Substitution of arginine 282 with a glutamate residue (R282E-PepT1) gave a protein at the plasma membrane of Xenopus laevis oocytes that was able to transport the non-hydrolyzable dipeptide [H-3]D-Phe-LGln, although unlike the wild type, the rate of transport by R282E-PepT1 was independent of the extracellular pH level, and the substrate could not be accumulated above equilibrium. The binding affinity of the mutant transport protein was unchanged from the wild type. Thus, R282E-Pept1 appears to have been changed from a proton-driven to a facilitated transporter for peptides. In addition, peptide transport by R282E-PepT1 still induced depolarization as measured by microelectrode recordings of membrane potential. A more detailed study by two-electrode voltage clamping revealed that R282E-PepT1 behaved as a peptide-gated non-selective cation channel with the ion selectivity series lithium > sodium > N-methyl-D-glucamine at pH 7.4. There was also a proton conductance (comparing pH 7.4 and 8.4), and at pH 5.5 the predominant conductance was for potassium ions. Therefore, it can be concluded that changing arginine 282 to a glutamate not only uncouples the cotransport of protons and peptides of the wild-type PepT1 but also creates a peptide-gated cation channel in the protein.Published here -
Halestrap AP, Meredith D, 'The SLC16 gene family - from monocarboxylate transporters (MCTs) to aromatic amino acid transporters and beyond'
Pflügers Archiv European Journal of Physiology 447 (5) (2004) pp.619-628
ISSN: 0031-6768 eISSN: 1432-2013AbstractThe monocarboxylate cotransporter (MCT) family now comprises 14 members, of which only the first four (MCT1-MCT4) have been demonstrated experimentally to catalyse the proton-linked transport of metabolically important monocarboxylates such as lactate, pyruvate and ketone bodies. SLC16A10 (T-type amino-acid transporter-1, TAT1) is an aromatic amino acid transporter whilst the other members await characterization. MCTs have 12 transmembrane domains (TMDs) with intracellular N- and C-termini and a large intracellular loop between TMDs 6 and 7. MCT1 and MCT4 require a monotopic ancillary protein, CD147, for expression of functional protein at the plasma membrane. Lactic acid transport across the plasma membrane is fundamental for the metabolism of and pH regulation of all cells, removing lactic acid produced by glycolysis and allowing uptake by those cells utilizing it for gluconeogenesis (liver and kidney) or as a respiratory fuel (heart and red muscle). The properties of the different MCT isoforms and their tissue distribution and regulation reflect these roles.Published here -
Windhaber RAJ, Wilkins RJ, Meredith D, 'Functional characterisation of glucose transport in bovine articular chondrocytes'
Pflügers Archiv European Journal of Physiology 446 (5) (2003) pp.572-577
ISSN: 0031-6768 eISSN: 1432-2013AbstractThe adequate provision of glucose to articular chondrocytes is essential to sustain their predominantly anaerobic metabolism; glucose is also a precursor for the extracellular matrix macromolecules which these cells synthesise. Impaired glucose uptake would compromise cell function and potentially result in an imbalance of matrix synthesis and degradation, leading to osteoarthritis. We studied the glucose influx pathway into bovine articular chondrocytes using 2-deoxy-d-[H-3]-glucose (DOG). Uptake occurs via an extracellular pH (pH(o))-insensitive, phloretin- and cytochalasin B-sensitive pathway, hallmarks of the GLUT family of facilitative glucose transporters, with a K-m of 0.35+/-0.11 mM. Uptake was affected by a number of physiologically relevant factors: (1) raised hydrostatic pressure (1-30 MPa) inhibited DOG uptake by up to 30%; (2) interleukin-1 (IL-1beta) reduced uptake via an increase in transporter affinity; (3) glucosamine inhibited glucose uptake in a manner consistent with the actions of a competitive inhibitor. Given the involvement of IL-1beta in osteoarthritis and the protective role assigned to glucosamine, these findings implicate an important role for glucose delivery in chondrocyte energy production and matrix metabolism, which, therefore, may potentially affect the maintenance of cartilage integrity.Published here -
Tattersall AL, Meredith D, Furla P, Shen MR, Ellory JC, Wilkins RJ, 'Molecular and functional identification of the Na+/H+ exchange isoforms NHE1 and NHE3 in isolated bovine articular chondrocytes'
Cellular Physiology and Biochemistry 13 (4) (2003) pp.215-222
ISSN: 1015-8987AbstractCartilage matrix turnover is sensitive to changes in intracellular pH (pH) and previous studies have shown that articular chondrocytes regulate pH, predominantly using an amiloride-sensitive Na+/H+ exchanger (NHE) with hallmark properties of the housekeeper isoform NHE1. Immunoblotting studies have, however, demonstrated that bovine chondrocytes express both the NHE1 and NHE3 isoforms of Na+/H+ exchange. In the present study the effect of exposure to serum on acid extrusion from chondrocytes has been studied. The pH-sensitive fluoroprobe BCECF was used to measure pH(i) in isolated bovine articular chondrocytes, and proton equivalent membrane transporters were characterised for cells isolated in the absence and presence of 5% fetal bovine serum (FBS). The contribution of NHE isoforms to acid extrusion, following ammonium-induced acidification, was assessed using a combination of ion substitution and the specific NHE1 inhibitor HOE694. While Na+-dependent acid extrusion was entirely inhibited by HOE694 in FBS untreated cells, the operation of both NHE1 and NHE3 was detected in cells exposed to FBS. In parallel, RT-PCR and immunohistochemistry experiments demonstrated both NHE1 and NHE3 mRNA and expression of both proteins respectively. While serum growth factors are virtually excluded from healthy cartilage they could permeate a damaged matrix. The regulatory characteristics of NHE3 are distinct from NHE1 so that an altered pattern of response to components of mechanical stress such as hyperosmolarity may be associated with increased access of growth factors in diseased tissue.Published here -
Pollard M, Meredith D, McGivan JD, 'Identification of a plasma membrane glutamine transporter from the rat hepatoma cell line H4-IIE-C3'
Biochemical Journal 368 (1) (2002) pp.371-375
ISSN: 0264-6021 eISSN: 1470-8728AbstractGlutamine is taken up into the rat hepatoma cell line H4-IIE-C3 by a Na+-dependent transport system which is specific for glutamine, alanine, serine, cysteine and asparagine and does not tolerate substitution of Na+ by Li+. Glutamine transport was relatively weakly inhibited by a 50-fold excess of leucine and was not inhibited by phenylalanine or N-methyl aminoisobutyrate. These general properties are characteristic of the recently identified ASCT/B-0 family of transporters. Using a reverse transcriptase PCR-based homology cloning approach, we have characterized a cDNA for a novel member of this transporter family (H4-ASCT2) from H4-IIE-C3 cells. The cDNA encodes a 551-amino acid protein which exhibits similarities of between 75 and 85% with ASCT/B-0 transporters previously cloned from other sources. When expressed in Xenopus oocytes, this transporter catalyses Na+-dependent glutamine uptake with characteristics very similar to those of glutamine uptake into the H4-IIE-C3 cells. This newly characterized transporter possesses a number of amino acid sequence differences from ASCT2 clones recently isolated from rat astroglial cells and from normal rat liver. In particular, the loop region between transmembrane helices 3 and 4 from H4-ASCT2 shares less than 66% sequence similarity with ASCT2 from rat liver; furthermore, there are some 25 single amino acid substitutions elsewhere in the H4-ASCT2 sequence compared with that from rat liver. Thus enhanced glutamine uptake in rat hepatoma cells is mediated by the expression of a novel ASCT/B-0 transporter isoform rather than by increased expression of the ASCT2 mRNA found in normal rat liver.Published here -
Meredith D, 'The N-terminal blocked amino acid N-acetyl-L-phenylalanine is a non-translocated substrate for the mammalian peptide transporter PepT1 expressed in Xenopus laevis oocytes'
The Journal of Physiology 544 (S090) (2002) pp.91P-92P
ISSN: 0022-3751 eISSN: 1469-7793AbstractCommunicationsPublished here -
Pollard M, Meredith D, McGivan JD, 'Characterisation and cloning of a Na+-dependent broad-specificity neutral amino acid transporter from NBL-1 cells: a novel member of the ASC/B-0 transporter family'
Biochimica et Biophysica Acta (BBA) - Biomembranes 1561 (2) (2002) pp.202-208
ISSN: 0005-2736AbstractNa-dependent neutral amino acid transport into the bovine renal epithelia] cell line NBL-1 is catalysed by a broad-specificity transporter originally termed System B-0). This transporter is shown to differ in specificity from the 130 transporter cloned from JAR cells [J. Biol. Chem. 271 (1996) 18657] in that it interacts much more strongly with phenylalanine. Using probes designed to conserved transmembrane regions of the ASC/B-0 transporter family we have isolated a cDNA encoding the NBL-1 cell System B0 transporter. When expressed in Xenopus oocytes the clone catalysed Na-dependent alanine uptake which was inhibited by glutamine, leucine and phenylalanine. However, the clone did not catalyse Na+-dependent phenylalanine transport, again as in NBL-1 cells. The clone encoded a protein of 539 amino acids; the predicted transmembrane domains were almost identical in sequence to those of the other members of the B-0/ASC transporter family. Comparison of the sequences of NBL-1 and JAR cell transporters showed some differences near the N-terminus, C-terminus and in the loop between helices 3 and 4. The NBL-1 B-0 transporter is not the same as the renal brush border membrane transporter since it does not transport phenylalanine. Differences in specificity in this protein family arise from relatively small differences in amino acid sequence. (C) 2002 Elsevier Science B.V. All rights reserved.Published here -
Wilson MC, Meredith D, Halestrap AP, 'Fluorescence resonance energy transfer studies on the interaction between the lactate transporter MCT1 and CD147 provide information on the topology and stoichiometry of the complex in situ'
Journal of Biological Chemistry 277 (2002) pp.3666-3672
ISSN: 0021-9258 eISSN: 1083-351XAbstractThe monocarboxylate (lactate) transporters MCTI and MCT4 require the membrane-spanning glycoprotein CD147 for their correct plasma membrane expression and function. We have successfully expressed CD147 and MCTI tagged on their C or N termini with either the cyan (CFP) or yellow (YFP) variants of green fluorescent protein. The tagged proteins were correctly targeted to the plasma membrane of COS-7 cells and were functionally active. Measurements of fluorescence resonance energy transfer (FRET) between all combinations of the tagged proteins were made. FRET was observed when either the C or N terminus of MCT1 (intracellular) is tagged with CFP or YFP and co-expressed with CD147 tagged with YFP or CFP on the C terminus (intracellular) but not the N terminus (extracellular). FRET was also observed between two CD147 molecules when both YFP and CFP were on the C terminus but not when both were on the N terminus or one on either end. No FRET was observed between MCT1-YFP and MCT-CFP in any combination. A wide range of controls including photobleaching were employed to confirm that where FRET was observed, it was not an artifact of direct excitation of YFP by the CFP excitation laser. It was also shown that nonspecific overcrowding of proteins did not induce FRET. Because FRET only occurs between two fluorophores if they are less than 100 A apart and in a suitable orientation, our data provide important information on the topology of CD147 and MCTI within the plasma membrane. The minimum configuration consistent with the data is a dimer of CD147 associating with two MCT1 molecules such that the C terminus of CD147 in the cytosol is close to the C terminus of its partner CD147 and to the C and N termini of an associated MCT1 molecule. FRET may provide a non-invasive technique for measuring changes in these interactions in living cells.Published here -
Meredith D, Pollard M, McGivan JD, 'Functional characterization of the NBL-1 B-0 clone expressed in Xenopus oocytes: a new member of the ASC/B-0 family of amino acid transporters'
The Journal of Physiology 539P (S061) (2002) pp.9P-9P
ISSN: 0022-3751 eISSN: 1469-7793 -
Meredith D, Bell P, McClure B, Wilkins R, 'Functional and molecular characterisation of lactic acid transport in bovine articular chondrocytes'
Cellular Physiology and Biochemistry 12 (4) (2002) pp.227-234
ISSN: 1015-8987AbstractPublished hereChondrocytes, which control the turnover of cartilage, undergo predominantly glycolytic metabolism due to the avascular nature of the tissue. This will result in high levels of lactic acid production, and this lactic acid must leave the cells for their normal intracellular pH to be maintained. However to date the mechanism by which lactic acid is removed from the chondrocyteshas not been elucidated. In the present study lactic acid transport has been characterised using the intracellular pH-sensitive fluorimetric dye BCECF to measure intracellular pH (pH). Addition of extracellular lactic acid-induced an acidification which was sensitive to alpha-cyano-4-hydroxycinnamate (alpha-CHC) and phloretin indicating the involvement of isoform(s) of the monocarboxylate transporter (MCT) family. The results studies of transport kinetics were consistent with the MCT4 isoform (K-m 14.1 mM), common to other glycolytic cells. Western blotting confirmed that MCT4 was the predominantly expressed isoform, although both MCT1 and MCT4 transcripts were present when cells were assayed by RT-PCR. Through effects on pH(i), the activity of this transporter may therefore modify cartilage turnover. Copyright (C) 2002 S. Karger AG, Basel.
-
Meredith D, Halestrap AP, 'OX47 (basigin) may act as a chaperone for expression of the monocarboxylate transporter MCT1 at the plasma membrane of Xenopus laevis oocytes'
The Journal of Physiology 526P (2000) pp.23P-24P
ISSN: 0022-3751 eISSN: 1469-7793 -
Meredith D, Temple CS, Guha N, Sword CJ, Boyd CAR, Collier ID, Morgan KM, Bailey PD, 'Modified amino acids and peptides as substrates for the intestinal peptide transporter PepT1'
European Journal of Biochemistry 267 (12) (2000) pp.3723-3728
ISSN: 0014-2956 eISSN: 1432-1033AbstractThe binding affinities of a number of amino-acid and peptide derivatives by the mammalian intestinal peptide transporter PepT1 were investigated, using the Xenopus laevis expression system. A series of blocked amino acids, namely N-acetyl-Phe (Ac-Phe), phe-amide (Phe-NH2), N-acetyl-Phe-amide (Ac-Phe-NH2) and the parent compound Phe, was compared for efficacy in inhibiting the uptake of the peptide [H-3]-D-Phe-L-Gln. In an equivalent set of experiments, the blocked peptides Ac-Phe-Tyr, Phe-Tyr-NH2 and Ac-Phe-Tyr-NH2 were compared with the parent compound Phe-Tyr. Comparing amino acids and derivatives, only Ac-Phe was an effective inhibitor of peptide uptake (K-i = 1.81 +/- 0.37 mM). Ac-Phe-NH2 had a very weak interaction with PepT1 (K-i = 16.8 +/- 5.64 mM); neither Phe nor Phe-NH2 interacted with PepT1 with measurable affinity. With the dipeptide and derivatives, unsurprisingly the highest affinity interaction was with Phe-Tyr (K-i = 0.10 +/- 0.04 mM). The blocked C-terminal peptide Phe-Tyr-NH2 also interacted with PepT1 with a relatively high affinity (K-i = 0.94 +/- 0.38 mM). Both Ac-Phe-Tyr and Ac-Phe-Tyr-NH2 interacted weakly with PepT1 (K-i = 8.41 +/- 0.11 and 9.97 +/- 4.01 mM, respectively). The results suggest that the N-terminus is the primary binding site for both dipeptides and tripeptides. Additional experiments with four stereoisomers of Ala-Ala-Ala support this conclusion, and lead us to propose that a histidine residue is involved in binding the C-terminus of dipeptides. In addition, a substrate binding model for PepT1 is proposed.Published here -
Meredith D, Boyd CAR, 'Structure and function of eukaryotic peptide transporters'
Cellular and Molecular Life Sciences 57 (5) (2000) pp.754-778
ISSN: 1420-682X eISSN: 1420-9071AbstractThe cotransport of protons and peptides is now recognised as a major route by which dietary nitrogen is absorbed from the intestine, and filtered protein reabsorbed in the kidney. Recently, molecular biology has had a very substantial impact on the study of peptide transport, and here we review the molecular and functional information available within the framework of physiology. To this end we consider not only the mammalian peptide transporters and their tissue distribution and regulation but also those from other species (including Caenorhabditis elegans) which make up the proton-dependent oligopeptide transport superfamily. In addition, understanding the binding requirements for transported substrates may allow future design and targeted tissue delivery of peptide and peptidomimetic drugs. Finally, we aim to highlight some of the less well understood areas of peptide transport, in the hope that it will stimulate further research into this challenging yet exciting topic.Published here -
Bailey PD, Boyd CAR, Bronk JR, Collier ID, Meredith D, Morgan KM, Temple CS, 'How to make drugs orally active: A substrate template for peptide transporter PepT1'
Angewandte Chemie International Edition 39 (3) (2000) pp.505-508
ISSN: 1433-7851 eISSN: 1521-3773AbstractBy building key structural features into hydrophilic drugs, they can be recognized by the PepT1 transporter system of the small intestine and rendered orally active. The model shown provides, for the first time, a 3D template for all known substrates of PepT1.Published here -
Meredith D, Boyd CAR, Bronk JR, Bailey PD, Morgan KM, Collier ID, Temple CS, '4-aminomethylbenzoic acid is a non-translocated competitive inhibitor of the epithelial peptide transporter PepT1'
The Journal of Physiology 512 (3) (1998) pp.629-634
ISSN: 0022-3751 eISSN: 1469-7793AbstractPublished here1 4-Aminomethylbenzoic acid, a molecule which mimics the spatial configuration of a dipeptide, competitively inhibits peptide influx in both Xenopus laevis oocytes expressing rabbit PepT1 and through PepT1 in rat renal brush border membrane vesicles.
2 This molecule is not translocated through PepT1 as measured both by direct HPLC analysis in PepT1-expressing oocytes and indirectly by its failure to trans-stimulate labelled peptide efflux through PepT1 in oocytes and in renal membrane vesicles.
3 However 4-aminomethylbenzoic acid does reverse trans-stimulation through expressed PepT1 of labelled peptide efflux induced by unlabelled peptide. Quantitatively this reversal is compatible with 4-aminomethylbenzoic acid competitively binding to the external surface of PepT1.
4 4-Aminomethylbenzoic acid (the first molecule discovered to be a non-translocated competitive inhibitor of proton-coupled oligopeptide transport) and its derivatives may thus be particularly useful as experimental tools.
-
Temple CS, Stewart AK, Meredith D, Lister NA, Morgan KM, Collier ID, Vaughan-Jones RD, Boyd CAR, Bailey PD, Bronk JR, 'Peptide mimics as substrates for the intestinal peptide transporter'
Journal of Biological Chemistry 273 (1998) pp.20-22
ISSN: 0021-9258 eISSN: 1083-351XAbstractThese results with 4-APAA show for the first time that the presence of a peptide bond is not a requirement for rapid translocation through the proton-linked oligopeptide transporter (PepT1), Further investigation will be needed to determine the minimal structural requirements for a molecule to be a substrate for this transporter. -
Chen S, Meredith D, Boyd CAR, 'Both the H13 gene product and 4F2 antigen are involved in the induction of system y(+) cationic amino-acid transport following activation of human peripheral blood mononuclear cells (PBM)'
Biochimica et Biophysica Acta (BBA) - Biomembranes 1284 (1996) pp.1-3
ISSN: 0005-2736AbstractPrior transfection with antisense oligonucleotides to the H13 and 4F2 hc genes, singly or in combination, was found to inhibit phytohaemagglutinin-induced activation of cationic amino-acid transport system y(+) in human peripheral blood mononuclear cells (mostly circulating lymphocytes). These effects on system y(+) function or expression mean that 4F2 he cannot only be the molecular basis of system y(+)L (Fei, Y.-J., Prasad, P.D., Leibach, F.H. and Ganapathy, V. (1995) Biochemistry 34, 8744-8751). -
Meredith D, Laynes RW, 'Dipeptide transport in brush-border membrane vesicles (BBMV) prepared from human full-term placentae'
Placenta 17 (1996) pp.173-179
ISSN: 0143-4004 eISSN: 1532-3102AbstractThe uptakes of the tritiated, hydrolysis-resistant cationic (D-Phe-L-Lys), neutral (D-Phe-L-Ala) and anionic (D-Phe-L-Glu) peptides into human full-term placental brush-border membrane vesicles (BBMV) were time-dependent and into an osmotically-active space. Uptakes of D-Phe-L-Lys and D-Phe-L-Glu were temperature-dependent. Uptake of D-Phe-L-Lys was electroneutral (either cation exchange or anion co-transport), whereas D-Phe-L-Ala and D-Phe-L-Glu were both stimulated by an increasingly inside-positive membrane potential (explained by either cation exchange or anion co-transport, or translocation alone, respectively). Uptake of D-Phe-L-Ala was stimulated (approximately 50 per cent) by an inwardly-directed proton gradient (pH(in)=7.4, pH(out)=5.5), whereas D-Phe-L-Glu was unaffected, and D-Phe-L-Lys uptake was inhibited (approximately 50 per cent) but was unaffected by the organic cation-eschange inhibitors 1,1-diethyl-2,2-cyanine (decynium22) and 5-(N-methyl-N-isobutyl)amiloride (MIBA). Over the concentration range studied, the peptides did not self-inhibit, and the only cross-inhibition was by D-Phe-L-Glu on D-Phe-L-Lys uptake (estimated K-I 24.2 +/- 1.36 mM), suggesting very low affinity transporter(s). Under conditions favouring its transport by PepT1, D-Phe-L-Glu uptake was unaffected by diethylpyrocarbonate (DEPC); neither D-Phe-L-Ala nor D-Phe-L-Lys was inhibited by DEPC under maximally proton-stimulated conditions of uptake. We conclude that PepT-like transporters are not responsible for peptide uptake into human placental BBMV; while the molecular identity of the transporter(s) involved remains unclear, we hypothesize that they could be similar to the as yet unidentified epithelial basolateral peptide transporter(s). (C) 1996 W. B. Saunders Company LtdPublished here -
MEREDITH D, BOYD CAR, 'DIPEPTIDE TRANSPORT CHARACTERISTICS OF THE APICAL MEMBRANE OF RAT LUNG TYPE-II PNEUMOCYTES'
American Journal of Physiology - Lung Cellular and Molecular Physiology 269 (1995) pp.L137-L143
ISSN: 1040-0605 eISSN: 1522-1504AbstractThe transport of a hydrolysis-resistant dipeptide, D-phenylalanyl-L-alanine (D-Phe-L-Ala), has been studied by high-performance liquid chromatography in rat lung epithelial cells and apical membrane vesicles. Time-dependent uptake of D-Phe-L-Ala into isolated type II pneumocytes was shown. Uptake was saturable, and Michaelis-Menten kinetics were fitted to the data and gave an apparent Michaelis constant (K-m) of 3.4 mM and a maximum velocity (V-max) of 7.0 nmol . mg protein(-1). min(-1). However, known peptide transport inhibitors unexpectedly increased intracellular D-Phe-L-Ala concentration when initial rates of peptide uptake were studied. Apical (brush-border) membrane vesicles prepared from rat lung also showed time- and concentration-dependent influx of D-Phe-L-Ala (apparent K-m 2.0 mM, V-max 0.53 nmol . mg protein(-1). min(-1)). Influx of this neutral dipeptide into the vesicles was shown to be both electrogenic and stimulated by an inwardly directed proton gradient. Influx was inhibitable by mercuric chloride and by the amino acid residue modifying compounds N-acetylimidazole and diethylpyrocarbonate. These findings strongly suggest the presence of a proton-coupled peptide transport protein in the apical surface of the type II cell. This transporter may play a role in lung homeostasis. -
HELLIWELL PA, MEREDITH D, BOYD CAR, BRONK JR, LISTER N, BAILEY PD, 'TRIPEPTIDE TRANSPORT IN RAT LUNG'
Biochimica et Biophysica Acta (BBA) - Biomembranes 1190 (1994) pp.430-434
ISSN: 0005-2736AbstractTransport Of L-alanyl-D-phenylalanyl-L-alanine was investigated with an in situ vascular perfusion preparation of rat lung and brush border membrane vesicles prepared from type II pneumocytes. In the perfused lung 1 mM tripeptide was transported intact from the alveolar lumen to the vascular perfusate at a mean rate of 25.1 +/- 1.29 (3) nmol/min per g dry weight. D-Phenylalanine also appeared in the vascular perfusate at a rate of 21.9 +/- 1.74 (3) nmol/min per g dry weight indicating that 47% of the absorbed tripeptide was split during passage across the epithelial layer. No dipeptide could be detected in the vascular effluent during perfusions with tripeptide. Rapid L-alanyl-D-phenylalanyl-L-alanine uptake occurred with fresh apical membrane vesicles prepared from type II pneumocytes and this was abolished by treatment with 0.1% triton. The related tripeptide, D-alanyl-L-phenylalanyl-D-alanine, was taken up significantly more slowly by the vesicles. D-phenylalanyl-L-alanine and D-phenylalanyl-D-alanine, were also studied with the vascularly perfused preparation; the mixed dipeptide appeared in the vascular perfusate significantly faster than L-alanyl-D-phenylalanyl-L-alanine whereas D-phenylalanyl-D-alanine appeared more slowly and was not hydrolysed. -
Fervenza FC, Meredith D, Ellory JC, Hendry BM, 'ABNORMAL ERYTHROCYTE CHOLINE TRANSPORT IN PATIENTS WITH CHRONIC-RENAL-FAILURE'
Clinical Science 80 (1991) pp.137-141
ISSN: 0143-5221 eISSN: 1470-8736Abstract4. The kinetics of choline transport have been studied in erythrocytes of patients on haemodialysis and control subjects in 'zero-trans' conditions after depletion of intracellular choline. The mean V(max.) in these conditions was 38.4 (SD 4.6) mu-mol h-1 l-1 cells in patients on haemodialysis compared with 14.2 (SD 3.7) mu-mol h-1 l-1 cells in control subjects. The mean K(m) under 'zero-trans' conditions was 19.4 (SD 2.4) mu-mol/l in patients on haemodialysis and 7.4 (SD 1.4) mu-mol/l in control subjects. These differences were significant (P < 0.001).Published here -
FERVENZA FC, MEREDITH D, ELLORY JC, HENDRY BM, 'URIDINE TRANSPORT IN HUMAN ERYTHROCYTES - DATA FROM NORMAL SUBJECTS AND FROM PATIENTS WITH RENAL-FAILURE'
Experimental Physiology 76 (1) (1991) pp.53-58
ISSN: 0958-0670 eISSN: 1469-445XAbstractErythrocyte uridine transport has been studied in eight normal individuals and eight patients on haemodialysis for chronic renal failure. The initial rate of zero-trans uridine influx at 37-degrees-C has been measured as a function of extracellular uridine concentration using [C-14]-labelled uridine. The results are consistent with Michaelis-Menten kinetics. In normal humans the mean V(max) for uridine influx was 32.8 +/- 6.4-mu-mol (1 cells)-1 s-1 (mean +/- S.D.) and the mean K(m) was 190 +/- 12.3-mu-M. The measurements made in renal failure patients were not significantly different (mean V(max) 30.1 +/- 7.1-mu-mol (1 cells)-1 s-1, mean K(m), 185 +/- 13.2-mu-M). These results are discussed with reference to the reported data on uridine transport in human erythrocytes at temperatures between 4 and 35-degrees-C; it is suggested that zero-trans uridine influx shows a decrease in temperature dependence above 25-degrees-C. The V(max) for zero-trans uridine influx at 37-degrees-C in normal erythrocytes represents a turnover number for the nucleoside transporter of 180 uridine molecules per second.Published here -
Fervenza FC, Meredith D, Ellory JC, Hendry BM, 'A Study of the Membrane Transport of Aminoacids in Erythrocytes from Patients on Haemodialysis'
Nephrology Dialysis Transplantation 5 (8) (1990) pp.594-599
ISSN: 0931-0509 eISSN: 1460-2385AbstractThe initial rates of aminoacid influx to erythro cytes from nine patients on maintenence haemodialysis and from nine normal controls have been studied using radiolabelled L-serine, glycine, L-tryptophan and Lleucine. L-serine uptake had an Na-dependent component fitted by Michaelis-Menten kinetics (ASC system) and a non-saturable Na-independent component. In erythrocytes from haemodialysis patients L-serine uptake via the ASC system had a mean (SEM) Vmax of 233 (33) μmol/l cells per h, compared to a mean Vmax in controls of 353 (21) μmol/l cells per h. The mean Km for L-serine uptake was 105 (11) μM in haemodialysis erythrocytes and 160 (12) μM in controls. These reductions in Vmax and Km for ASC in haemodialysis erythrocytes were significant (2P<0.02). Glycine uptake had a Michaelis-Menten-type chloride-dependent component (gly system), and a chloride-independent non-saturable component. The mean Km for glycine influx via the gly system was 26.3 (2.3) μM in haemodialysis erythrocytes, and 34 (3.1) μM in normals (2P<0.02). The Vmax for the uptake of glycine by the gly system did not differ significantly between haemodialysis and control erythrocytes. The initial influx rates of L-tryptophan via the T system and of L-leucine via the L system did not vary between control and haemodialysis erythrocytes. It is suggested that abnormalities of the ASC and gly membrane transport systems may contributeto the altered interorgan transport of aminoacids found in renal disease.Published here
Books
-
Wilkins R, Megson I, Meredith D, (ed.), Oxford Handbook of Medical Sciences, Oxford University Press (2021)
ISBN: 9780198789895AbstractPublished hereThe Oxford Handbook of Medical Sciences is written by biomedical scientists and clinicians to be the definitive guide to the fundamental scientific principles that underpin medicine and the biomedical sciences. It provides a clear and easily digestible account of basic cell physiology, biochemistry, and molecular and medical genetics, followed by chapters integrating the traditional pillars of biomedicine (anatomy, physiology, biochemistry, pathology, and pharmacology) for each of the major systems and processes of the human body: nerve and muscle, musculoskeletal system, respiratory and cardiovascular systems, urinary system, digestive system, endocrine organs, reproductive system, development from fertilization to birth, neuroanatomy and neurophysiology, infection and immunity, and the growth of tissues and organs. Also included are chapters on medicine and society and techniques used in biomedical science research. In its third edition, the Oxford Handbook of Medical Sciences is now fully illustrated in colour, and cross-referenced to the Oxford Handbook of Clinical Medicine, tenth edition, Oxford Handbook of Clinical Specialities, eleventh edition, and Oxford Handbook of Practical Drug Therapy, second edition. Its concise writing style makes it an invaluable source of information for practitioners and students in medicine, biomedical sciences, and the allied health professions. -- Provided by publisher.
-
Wilkins,Robert; Cross, Simon; Megson, Ian, Oxford Handbook of Medical Sciences, Oxford University Press (2011)
ISBN: 978-0199588442
Conference papers
-
Foley DW, Bailey PD, Price RA, Meredith D, Pettecrew R, 'Improving Drug Oral Bioavailability By Targeting Pept1'
236 (2010)
AbstractPepT1 is a proton dependent oligopeptide transporter located principally in the intestinal epithelial membrane. It transports a wide range of substrates including most di- and tripeptides, penicillins, celphalosporins, ACE inhibitors and amino acid esters of nucleoside based drugs. This broad substrate specificity has made it a key target for improving the oral bioavailability of drugs. We have recently patented hydrolysis resistant thiodipeptide PepT1 substrates of type [A], which enable a variety of drugs to be attached via ester or amide links on the C-terminal side-chain. We will present the synthesis and transport in Caco-2 cells and isolated rat loops of our prodrugs (examples [1] and [2]). This will include the first example of targeting ketone containing drugs towards PepT1 absorption [3]. We will also discuss the results of our structure-transport studies.
Other publications
-
Manning Fox JM, Meredith D, Halestrap AP, 'The kinetics, and substrate and inhibitor specificities of human monocarboxylate tranporter 4 expressed in Xenopus laevis oocytes', (2000)
-
Meredith D, Boyd CAR, 'Functional evidence that rabbit PepT1 may act as a multimer when expressed in Xenopus laevis oocytes', (1998)
-
Meredith D, Temple CS, Boyd CAR, 'Kinetic evidence that neutral and anionic peptides bind to different conformations of the recombinant epithelial peptide transporter PepT1', (1998)
-
Temple CS, Meredith D, Boyd CAR, 'Dipeptide transport by rat renal cortex BBMV is mediated by PepT1; functional analysis using ACE inhibitors', (1997)
-
Meredith D, Boyd CAR, 'His(57) is an essential residue for the transport of the anionic dipeptide D-Phe-L-Glu by PepT1 expressed in Xenopus laevis oocytes', (1996)
-
Meredith D, Temple CS, Boyd CAR, 'Chemical evidence for the involvement of histidine residues in proton-peptide co-transport by PepT1 expressed in Xenopus laevis oocytes', (1996)
-
MEREDITH D, 'DIPEPTIDE TRANSPORT BY HUMAN PLACENTAL BRUSH-BORDER MEMBRANE-VESICLES (BBMV)', (1995)
-
MEREDITH D, BOYD CAR, 'OLIGOPEPTIDE TRANSPORT BY EPITHELIAL-CELLS', (1995)
-
MEREDITH D, BOYD CAR, 'PEPTIDE-TRANSPORT IN TYPE-II PNEUMOCYTES FRESHLY ISOLATED FROM RAT LUNG', (1993)
Professional information
Memberships of professional bodies
- Member of The Physiological Society and the European Intestinal Transport Group
- Fellow of the Higher Education Academy
- School of Biological and Medical Sciences - Faculty of Health and Life Sciences