Anti-SLC22A13 (Aa 38-139) Polyclonal Antibody (DPAB-DC3801) This Product Is for Research Use Only and Is Not Intended for Diagnostic Use

Total Page:16

File Type:pdf, Size:1020Kb

Anti-SLC22A13 (Aa 38-139) Polyclonal Antibody (DPAB-DC3801) This Product Is for Research Use Only and Is Not Intended for Diagnostic Use Anti-SLC22A13 (aa 38-139) polyclonal antibody (DPAB-DC3801) This product is for research use only and is not intended for diagnostic use. PRODUCT INFORMATION Antigen Description This gene encodes a member of the organic-cation transporter family. It is located in a gene cluster with another member of the family, organic cation transporter like 4. The encoded protein is a transmembrane protein involved in the transport of small molecules. This protein can function to mediate urate uptake and is a high affinity nicotinate exchanger in the kidneys and the intestine. Immunogen SLC22A13 (NP_004247, 38 a.a. ~ 139 a.a) partial recombinant protein with GST tag. The sequence is AHVFMVLDEPHHCAVAWVKNHTFNLSAAEQLVLSVPLDTAGHPEPCLMFRPPPANASLQDILSH RFNETQPCDMGWEYPENRLPSLKNEFNLVCDRKHLKDT Source/Host Mouse Species Reactivity Human Conjugate Unconjugated Applications WB (Recombinant protein), ELISA, Size 50 μl Buffer 50 % glycerol Preservative None Storage Store at -20°C or lower. Aliquot to avoid repeated freezing and thawing. GENE INFORMATION Gene Name SLC22A13 solute carrier family 22 (organic anion/urate transporter), member 13 [ Homo sapiens (human) ] Official Symbol SLC22A13 Synonyms SLC22A13; solute carrier family 22 (organic anion/urate transporter), member 13; OAT10; 45-1 Ramsey Road, Shirley, NY 11967, USA Email: [email protected] Tel: 1-631-624-4882 Fax: 1-631-938-8221 1 © Creative Diagnostics All Rights Reserved OCTL1; OCTL3; ORCTL3; ORCTL-3; solute carrier family 22 member 13; organic cation transporter-like 3; organic-cation transporter like 3; organic cationic transporter-like 3; solute carrier family 22, member 13; solute carrier family 22 (organic anion transporter), member 13; Entrez Gene ID 9390 Protein Refseq NP_004247 UniProt ID Q9Y226 Chromosome Location 3p21.3 Function nicotinate transporter activity; organic cation transmembrane transporter activity; 45-1 Ramsey Road, Shirley, NY 11967, USA Email: [email protected] Tel: 1-631-624-4882 Fax: 1-631-938-8221 2 © Creative Diagnostics All Rights Reserved.
Recommended publications
  • Differential Expression of Hydroxyurea Transporters in Normal and Polycythemia Vera Hematopoietic Stem and Progenitor Cell Subpopulations
    Zurich Open Repository and Archive University of Zurich Main Library Strickhofstrasse 39 CH-8057 Zurich www.zora.uzh.ch Year: 2021 Differential expression of hydroxyurea transporters in normal and polycythemia vera hematopoietic stem and progenitor cell subpopulations Tan, Ge ; Meier-Abt, Fabienne Abstract: Polycythemia vera (PV) is a myeloproliferative neoplasm marked by hyperproliferation of the myeloid lineages and the presence of an activating JAK2 mutation. Hydroxyurea (HU) is a standard treat- ment for high-risk patients with PV. Because disease-driving mechanisms are thought to arise in PV stem cells, effective treatments should target primarily the stem cell compartment. We tested for theantipro- liferative effect of patient treatment with HU in fluorescence-activated cell sorting-isolated hematopoietic stem/multipotent progenitor cells (HSC/MPPs) and more committed erythroid progenitors (common myeloid/megakaryocyte-erythrocyte progenitors [CMP/MEPs]) in PV using RNA-sequencing and gene set enrichment analysis. HU treatment led to significant downregulation of gene sets associated with cell proliferation in PV HSCs/MPPs, but not in PV CMP/MEPs. To explore the mechanism underlying this finding, we assessed for expression of solute carrier membrane transporters, which mediate trans- membrane movement of drugs such as HU into target cells. The active HU uptake transporter OCTN1 was upregulated in HSC/MPPs compared with CMP/MEPs of untreated patients with PV, and the HU diffusion facilitator urea transporter B (UTB) was downregulated in HSC/MPPs compared withCM- P/MEPs in all patient and control groups tested. These findings indicate a higher accumulation ofHU within PV HSC/MPPs compared with PV CMP/MEPs and provide an explanation for the differential effects of HU in HSC/MPPs and CMP/MEPs of patients with PV.
    [Show full text]
  • Cellular and Molecular Signatures in the Disease Tissue of Early
    Cellular and Molecular Signatures in the Disease Tissue of Early Rheumatoid Arthritis Stratify Clinical Response to csDMARD-Therapy and Predict Radiographic Progression Frances Humby1,* Myles Lewis1,* Nandhini Ramamoorthi2, Jason Hackney3, Michael Barnes1, Michele Bombardieri1, Francesca Setiadi2, Stephen Kelly1, Fabiola Bene1, Maria di Cicco1, Sudeh Riahi1, Vidalba Rocher-Ros1, Nora Ng1, Ilias Lazorou1, Rebecca E. Hands1, Desiree van der Heijde4, Robert Landewé5, Annette van der Helm-van Mil4, Alberto Cauli6, Iain B. McInnes7, Christopher D. Buckley8, Ernest Choy9, Peter Taylor10, Michael J. Townsend2 & Costantino Pitzalis1 1Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK. Departments of 2Biomarker Discovery OMNI, 3Bioinformatics and Computational Biology, Genentech Research and Early Development, South San Francisco, California 94080 USA 4Department of Rheumatology, Leiden University Medical Center, The Netherlands 5Department of Clinical Immunology & Rheumatology, Amsterdam Rheumatology & Immunology Center, Amsterdam, The Netherlands 6Rheumatology Unit, Department of Medical Sciences, Policlinico of the University of Cagliari, Cagliari, Italy 7Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK 8Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham B15 2WB, UK 9Institute of
    [Show full text]
  • Disease-Induced Modulation of Drug Transporters at the Blood–Brain Barrier Level
    International Journal of Molecular Sciences Review Disease-Induced Modulation of Drug Transporters at the Blood–Brain Barrier Level Sweilem B. Al Rihani 1 , Lucy I. Darakjian 1, Malavika Deodhar 1 , Pamela Dow 1 , Jacques Turgeon 1,2 and Veronique Michaud 1,2,* 1 Tabula Rasa HealthCare, Precision Pharmacotherapy Research and Development Institute, Orlando, FL 32827, USA; [email protected] (S.B.A.R.); [email protected] (L.I.D.); [email protected] (M.D.); [email protected] (P.D.); [email protected] (J.T.) 2 Faculty of Pharmacy, Université de Montréal, Montreal, QC H3C 3J7, Canada * Correspondence: [email protected]; Tel.: +1-856-938-8697 Abstract: The blood–brain barrier (BBB) is a highly selective and restrictive semipermeable network of cells and blood vessel constituents. All components of the neurovascular unit give to the BBB its crucial and protective function, i.e., to regulate homeostasis in the central nervous system (CNS) by removing substances from the endothelial compartment and supplying the brain with nutrients and other endogenous compounds. Many transporters have been identified that play a role in maintaining BBB integrity and homeostasis. As such, the restrictive nature of the BBB provides an obstacle for drug delivery to the CNS. Nevertheless, according to their physicochemical or pharmacological properties, drugs may reach the CNS by passive diffusion or be subjected to putative influx and/or efflux through BBB membrane transporters, allowing or limiting their distribution to the CNS. Drug transporters functionally expressed on various compartments of the BBB involve numerous proteins from either the ATP-binding cassette (ABC) or the solute carrier (SLC) superfamilies.
    [Show full text]
  • Identification of Key Pathways and Genes in Dementia Via Integrated Bioinformatics Analysis
    bioRxiv preprint doi: https://doi.org/10.1101/2021.04.18.440371; this version posted July 19, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Identification of Key Pathways and Genes in Dementia via Integrated Bioinformatics Analysis Basavaraj Vastrad1, Chanabasayya Vastrad*2 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karnataka, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2021.04.18.440371; this version posted July 19, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract To provide a better understanding of dementia at the molecular level, this study aimed to identify the genes and key pathways associated with dementia by using integrated bioinformatics analysis. Based on the expression profiling by high throughput sequencing dataset GSE153960 derived from the Gene Expression Omnibus (GEO), the differentially expressed genes (DEGs) between patients with dementia and healthy controls were identified. With DEGs, we performed a series of functional enrichment analyses. Then, a protein–protein interaction (PPI) network, modules, miRNA-hub gene regulatory network and TF-hub gene regulatory network was constructed, analyzed and visualized, with which the hub genes miRNAs and TFs nodes were screened out. Finally, validation of hub genes was performed by using receiver operating characteristic curve (ROC) analysis.
    [Show full text]
  • Pflugers Final
    CORE Metadata, citation and similar papers at core.ac.uk Provided by Serveur académique lausannois A comprehensive analysis of gene expression profiles in distal parts of the mouse renal tubule. Sylvain Pradervand2, Annie Mercier Zuber1, Gabriel Centeno1, Olivier Bonny1,3,4 and Dmitri Firsov1,4 1 - Department of Pharmacology and Toxicology, University of Lausanne, 1005 Lausanne, Switzerland 2 - DNA Array Facility, University of Lausanne, 1015 Lausanne, Switzerland 3 - Service of Nephrology, Lausanne University Hospital, 1005 Lausanne, Switzerland 4 – these two authors have equally contributed to the study to whom correspondence should be addressed: Dmitri FIRSOV Department of Pharmacology and Toxicology, University of Lausanne, 27 rue du Bugnon, 1005 Lausanne, Switzerland Phone: ++ 41-216925406 Fax: ++ 41-216925355 e-mail: [email protected] and Olivier BONNY Department of Pharmacology and Toxicology, University of Lausanne, 27 rue du Bugnon, 1005 Lausanne, Switzerland Phone: ++ 41-216925417 Fax: ++ 41-216925355 e-mail: [email protected] 1 Abstract The distal parts of the renal tubule play a critical role in maintaining homeostasis of extracellular fluids. In this review, we present an in-depth analysis of microarray-based gene expression profiles available for microdissected mouse distal nephron segments, i.e., the distal convoluted tubule (DCT) and the connecting tubule (CNT), and for the cortical portion of the collecting duct (CCD) (Zuber et al., 2009). Classification of expressed transcripts in 14 major functional gene categories demonstrated that all principal proteins involved in maintaining of salt and water balance are represented by highly abundant transcripts. However, a significant number of transcripts belonging, for instance, to categories of G protein-coupled receptors (GPCR) or serine-threonine kinases exhibit high expression levels but remain unassigned to a specific renal function.
    [Show full text]
  • Modulation of Urate Transport by Drugs
    pharmaceutics Review Modulation of Urate Transport by Drugs Péter Tátrai 1, Franciska Erd˝o 2, Gabriella Dörnyei 3 and Péter Krajcsi 1,2,3,* 1 Solvo Biotechnology, Science Park, Building B2, 4-20 Irinyi József utca, H-1117 Budapest, Hungary; [email protected] 2 Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Hungary; [email protected] 3 Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, H-1088 Budapest, Hungary; [email protected] * Correspondence: [email protected] Abstract: Background: Serum urate (SU) levels in primates are extraordinarily high among mammals. Urate is a Janus-faced molecule that acts physiologically as a protective antioxidant but provokes inflammation and gout when it precipitates at high concentrations. Transporters play crucial roles in urate disposition, and drugs that interact with urate transporters either by intention or by accident may modulate SU levels. We examined whether in vitro transporter interaction studies may clarify and predict such effects. Methods: Transporter interaction profiles of clinically proven urate-lowering (uricosuric) and hyperuricemic drugs were compiled from the literature, and the predictive value of in vitro-derived cut-offs like Cmax/IC50 on the in vivo outcome (clinically relevant decrease or increase of SU) was assessed. Results: Interaction with the major reabsorptive urate transporter URAT1 appears to be dominant over interactions with secretory transporters in determining the net effect of a drug on SU levels. In vitro inhibition interpreted using the recommended cut-offs is useful at predicting the clinical outcome. Conclusions: In vitro safety assessments regarding urate transport should be done early in drug development to identify candidates at risk of causing major imbalances.
    [Show full text]
  • Phylogenetic, Syntenic, and Tissue Expression Analysis of Slc22 Genes in Zebrafish (Danio Rerio)
    Mihaljevic et al. BMC Genomics (2016) 17:626 DOI 10.1186/s12864-016-2981-y RESEARCH ARTICLE Open Access Phylogenetic, syntenic, and tissue expression analysis of slc22 genes in zebrafish (Danio rerio) Ivan Mihaljevic1†, Marta Popovic1,2†, Roko Zaja1,3 and Tvrtko Smital1* Abstract Background: SLC22 protein family is a member of the SLC (Solute carriers) superfamily of polyspecific membrane transporters responsible for uptake of a wide range of organic anions and cations, including numerous endo- and xenobiotics. Due to the lack of knowledge on zebrafish Slc22 family, we performed initial characterization of these transporters using a detailed phylogenetic and conserved synteny analysis followed by the tissue specific expression profiling of slc22 transcripts. Results: We identified 20 zebrafish slc22 genes which are organized in the same functional subgroups as human SLC22 members. Orthologies and syntenic relations between zebrafish and other vertebrates revealed consequences of the teleost-specific whole genome duplication as shown through one-to-many orthologies for certain zebrafish slc22 genes. Tissue expression profiles of slc22 transcripts were analyzed using qRT-PCR determinations in nine zebrafish tissues: liver, kidney, intestine, gills, brain, skeletal muscle, eye, heart, and gonads. Our analysis revealed high expression of oct1 in kidney, especially in females, followed by oat3 and oat2c in females, oat2e in males and orctl4 in females. oct1 was also dominant in male liver. oat2d showed the highest expression in intestine with less noticeable gender differences. All slc22 genes showed low expression in gills, and moderate expression in heart and skeletal muscle. Dominant genes in brain were oat1 in females and oct1 in males, while the highest gender differences were determined in gonads, with dominant expression of almost all slc22 genes in testes and the highest expression of oat2a.
    [Show full text]
  • Organic Anion Transporter 2 (OAT2): an Enigmatic Human Solute Carrier
    DMD Fast Forward. Published on November 21, 2016 as DOI: 10.1124/dmd.116.072264 This article has not been copyedited and formatted. The final version may differ from this version. DMD #072264 Organic Anion Transporter 2 (OAT2): An Enigmatic Human Solute Carrier Hong Shen, Yurong Lai, and A. David Rodrigues Department of Metabolism and Pharmacokinetics (H.S and Y.L), Bristol-Myers Squibb Downloaded from Research and Development, Princeton, New Jersey, and Department of Pharmacokinetics, Dynamics, and Metabolism (A.D. R), Pfizer World Wide Research dmd.aspetjournals.org and Development, Groton, Connecticut, United States at ASPET Journals on September 24, 2021 1 DMD Fast Forward. Published on November 21, 2016 as DOI: 10.1124/dmd.116.072264 This article has not been copyedited and formatted. The final version may differ from this version. DMD #072264 Running title: OAT2 as a membrane transporter of emerging importance Address correspondence to: Dr. Hong Shen, Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Company, Route 206 & Province Line Road, Princeton, NJ 08543. Telephone: (609) 252-4509 E-mail: [email protected] Downloaded from Number of text pages: 28 dmd.aspetjournals.org Number of tables: 2 Number of figures: 3 Number of references: 91 at ASPET Journals on September 24, 2021 Number of words: Abstract: 192 Introduction: 561 Abbreviations: CMD, cimetidine; cGMP, guanosine 3,5-cyclic monophosphate; Cmax, total maximum plasma concentration; Cmax,u, unbound maximum plasma concentration; DHEA, dehydroepiandrosterone; E3S, estrone-3sulfate; GFR, glomerular filtration rate; IC50, inhibitor concentration causing 50% inhibition; IMC, indomethacin; Ki, inhibition constant; LC-MS/MS, liquid chromatography–tandem mass spectrometry; MATE, multidrug and toxin extrusion protein; MPP+, 1-methyl-4-phenylpyridinium; MRP, multi 2 DMD Fast Forward.
    [Show full text]
  • Table SI. Enriched Genes in the Upregulated Genes of the Recovery Group According to the GO Molecular Function Terms. A, Downreg
    Table SI. Enriched genes in the upregulated genes of the recovery group according to the GO Molecular Function terms. A, Downregulated genes Adjusted Total Molecular Rank P‑value genes (n) Function Genes 1 <0.001 266 GO:0019899 Raf1 Timp1 Tbc1d8 Ube2g2 Ube2z enzyme binding Lonrf3 Tbc1d15 Rnf144a Ube2g1 Shc3 Rgcc Rnf19a Ube2j2 Rnf138 Atg13 Cks1b Ube2j1 Rnf19b Trib1 Trib3 Abtb2 Rnf125 Cdc42ep3 Nploc4 Cdc42ep4 Cdc42ep2 Rab11fip5 Arih2 Brms1 Tmem189 Mef2d Hspb1 Cdk9 Ksr1 Tnfaip3 Net1 Rnf180 Fgr Bhlhe41 Irs2 Ppp1r15a Asb4 Trim72 Zfp36 Sfn Xpo6 Fap Sox9 Mapk7 Itga3 Tubb5 Daxx Klf4 Stat3 Gab2 Myo9b Cstb Hmox1 Por Bcl2l1 Plin5 Chp1 Ube2i Sash1 Sqstm1 Rxra Slpi Sdc4 Tnfaip1 Cd40 Slc12a4 Map2k3 Ywhah Ppp1r12a Cry1 Plek Egfr Tnip1 Npc1l1 Rock2 Map2k6 Per1 Nfkbia Bdkrb2 Prkch Hif1a Golga5 Ripk1 Map3k1 Glud1 Nufip1 Clu Spry2 Hcls1 Ifnar2 Tuba1b Cdkn1a Sik1 Tmem173 Map3k2 Tnf Riok3 Ptpn2 Cep192 Smad2 Fas Jak2 Ankrd1 Rela Rps6ka4 Ankrd2 Rabgef1 Prkar1b Nop58 Casp8 Cflar Hdac4 Sele Nek2 Optn Nek6 Lcn2 Stom Traf6 Spred1 Nop56 Src Ccnl1 Ptpn22 Il6ra Pip5k1a F3 Bcl10 3110043O21Rik Tnfrsf1b Slc2a1 Sfpq Rpa2 Errfi1 Mad2l2 Tbc1d14 Uchl1 Glmn Scarb2 Ulk1 Ung Rad18 Mef2a Ctsc Ipo5 Mvp Kctd13 Msn Eif4ebp1 Casp3 Smad1 Ubash3b Ets1 Tirap Smad3 Tgfbr2 Ptgs2 Prr5l Micall1 Cnppd1 Map2k4 Tnks1bp1 Ppp1r32 Prdm4 Midn Ibtk Rusc2 Fmnl2 Ptpn23 Sh3bp4 Nop14 Kdm1a Serpine1 Gch1 Inf2 Csf3 Snx10 Txnip Egr1 Ranbp9 Akap12 Rab3gap2 Ddx58 Bcor Rabggta Pik3r1 Pkp2 Usp22 Shc1 Ptpn11 Fzd5 Cxcr4 Plaur Bag5 Maml1 Camk2n2 Taf7 Ywhag Ezr Jun Camk2d Parp4 Nod2 Ptafr Hmga2 Zfp746 Ptk2b Flot1
    [Show full text]
  • Sex Differences in Urate Handling
    International Journal of Molecular Sciences Review Sex Differences in Urate Handling Victoria L. Halperin Kuhns * and Owen M. Woodward * Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA * Correspondence: [email protected] (V.L.H.K.); [email protected] (O.M.W.); Tel.: +1-410-706-1754 (V.L.H.K.); +1-410-706-1760 (O.M.W.) Received: 27 May 2020; Accepted: 12 June 2020; Published: 16 June 2020 Abstract: Hyperuricemia, or elevated serum urate, causes urate kidney stones and gout and also increases the incidence of many other conditions including renal disease, cardiovascular disease, and metabolic syndrome. As we gain mechanistic insight into how urate contributes to human disease, a clear sex difference has emerged in the physiological regulation of urate homeostasis. This review summarizes our current understanding of urate as a disease risk factor and how being of the female sex appears protective. Further, we review the mechanisms of renal handling of urate and the significant contributions from powerful genome-wide association studies of serum urate. We also explore the role of sex in the regulation of specific renal urate transporters and the power of new animal models of hyperuricemia to inform on the role of sex and hyperuricemia in disease pathogenesis. Finally, we advocate the use of sex differences in urate handling as a potent tool in gaining a further understanding of physiological regulation of urate homeostasis and for presenting new avenues for treating the constellation of urate related pathologies. Keywords: sex differences; gout; uric acid; serum urate; ABCG2; SLC2A9; URAT1 1.
    [Show full text]
  • Lupus Nephritis Supp Table 5
    Supplementary Table 5 : Transcripts and DAVID pathways correlating with the expression of CD4 in lupus kidney biopsies Positive correlation Negative correlation Transcripts Pathways Transcripts Pathways Identifier Gene Symbol Correlation coefficient with CD4 Annotation Cluster 1 Enrichment Score: 26.47 Count P_Value Benjamini Identifier Gene Symbol Correlation coefficient with CD4 Annotation Cluster 1 Enrichment Score: 3.16 Count P_Value Benjamini ILMN_1727284 CD4 1 GOTERM_BP_FAT translational elongation 74 2.50E-42 1.00E-38 ILMN_1681389 C2H2 zinc finger protein-0.40001984 INTERPRO Ubiquitin-conjugating enzyme/RWD-like 17 2.00E-05 4.20E-02 ILMN_1772218 HLA-DPA1 0.934229063 SP_PIR_KEYWORDS ribosome 60 2.00E-41 4.60E-39 ILMN_1768954 RIBC1 -0.400186083 SMART UBCc 14 1.00E-04 3.50E-02 ILMN_1778977 TYROBP 0.933302249 KEGG_PATHWAY Ribosome 65 3.80E-35 6.60E-33 ILMN_1699190 SORCS1 -0.400223681 SP_PIR_KEYWORDS ubl conjugation pathway 81 1.30E-04 2.30E-02 ILMN_1689655 HLA-DRA 0.915891173 SP_PIR_KEYWORDS protein biosynthesis 91 4.10E-34 7.20E-32 ILMN_3249088 LOC93432 -0.400285215 GOTERM_MF_FAT small conjugating protein ligase activity 35 1.40E-04 4.40E-02 ILMN_3228688 HLA-DRB1 0.906190291 SP_PIR_KEYWORDS ribonucleoprotein 114 4.80E-34 6.70E-32 ILMN_1680436 CSH2 -0.400299744 SP_PIR_KEYWORDS ligase 54 1.50E-04 2.00E-02 ILMN_2157441 HLA-DRA 0.902996561 GOTERM_CC_FAT cytosolic ribosome 59 3.20E-33 2.30E-30 ILMN_1722755 KRTAP6-2 -0.400334007 GOTERM_MF_FAT acid-amino acid ligase activity 40 1.60E-04 4.00E-02 ILMN_2066066 HLA-DRB6 0.901531942 SP_PIR_KEYWORDS
    [Show full text]
  • The Interactions of Herbal Compounds with Human Organic Anion/Cation Transporters
    omics & en P Lu et al., J Pharmacogenomics Pharmacoproteomics 2014, 5:5 g h o a c r a m DOI: 10.4172/2153-0645.1000142 m a r c a o Journal of h p P r o f t o e l ISSN: 2153-0645o a m n r i c u s o J Pharmacogenomics & Pharmacoproteomics ReviewResearch Article Article OpenOpen Access Access The Interactions of Herbal Compounds with Human Organic Anion/Cation Transporters Xiaoxi Lu1, Ting Chan1, Chenghao Xu1, Wee Vean Ng1, Ling Zhu2 and Fanfan Zhou2* 1Faculty of Pharmacy, The University of Sydney, NSW, Australia 2Retinal Therapeutics Research Group, Save Sight Institute, The University of Sydney, Sydney, NSW, Australia Abstract The Solute Carrier transporters (SLCs) are a superfamily of membrane proteins responsible for cellular influx of various molecules. The Organic anion transporting polypeptides (OATPs), Organic anion transporters (OATs) and Organic cation transporters (OCTs) are essential SLC subfamilies that largely impact on drug performance. These Organic anion/cation transporters are widely expressed in epithelium throughout the body, where they play essential roles in cellular uptake of many endogenous substances like hormones and a wide range of exogenous molecules including clinically important drugs such as statins, anti-cancer agents and antibiotics. Herbal medicines that derived from plants, fruits and vegetables have been long presumed to be safe by the general public. The growing use of herbal products together with other agents has raised many concerns due to unexpected adverse effects. Drug- drug/herb interactions through SLC transporters often result in unsatisfied therapeutic outcomes and/or unexpected toxicities. This review will give an update on the interactions of herbal compounds with human Organic anion/cation transporters in order to form the basis of better and safer drug therapies.
    [Show full text]