HGNC Family | Solute carriers (SLC) |
Name | solute carrier organic anion transporter family, member 1A2 |
Description | This gene encodes a sodium-independent transporter which mediates cellular uptake of organic ions in the liver. Its substrates include bile acids, bromosulphophthalein, and some steroidal compounds. The protein is a member of the SLC21A family of solute carriers. Alternative splicing of this gene results in multiple transcript variants. [provided by RefSeq, Dec 2008] |
Summary |
{"type": "root", "children": [{"type": "p", "children": [{"type": "t", "text": "\nSLCO1A2 encodes the organic anion‐transporting polypeptide 1A2 (OATP1A2), a versatile uptake transporter that is broadly expressed in human tissues—including the brain capillaries, intestinal epithelium, kidney, liver, retina, and even in certain tumor types—and facilitates the cellular entry of a wide range of endogenous molecules and drugs. OATP1A2 mediates the transport of compounds such as estrone 3‐sulfate, imatinib, fexofenadine, various anticancer agents (e.g. doxorubicin and taxanes), triptans for migraine, and even all‐trans‐retinol into specialized tissues. Its activity is often pH‐dependent and can be modulated by post‐translational events, which collectively underscore its key role in absorption, distribution, blood–brain barrier penetration and even in pathological states such as breast cancer and neurodegeneration."}, {"type": "fg", "children": [{"type": "fg_fs", "start_ref": "1", "end_ref": "15"}]}, {"type": "t", "text": "\n"}]}, {"type": "t", "text": "\n\n"}, {"type": "p", "children": [{"type": "t", "text": "\nNumerous studies have demonstrated that genetic variation within SLCO1A2 can have profound effects on transporter function, with single nucleotide polymorphisms sometimes leading to altered transporter kinetics through impaired plasma membrane targeting, changes in glycosylation patterns, or disruption of intracellular regulatory interactions. In vitro investigations have shown that naturally occurring missense variants and polymorphisms within critical domains (including regions such as the sixth transmembrane segment and miRNA‐binding sites in the 3′‐UTR) can reduce the uptake capacity for substrates including estrone 3‐sulfate, imatinib, methotrexate, and fexofenadine. Such alterations in transporter performance have been linked in clinical settings to inter‐individual variability in drug disposition, altered therapeutic efficacy and toxicity, and may even influence disease risk or progression in conditions such as gallstone formation and breast cancer."}, {"type": "fg", "children": [{"type": "fg_fs", "start_ref": "16", "end_ref": "30"}]}, {"type": "t", "text": "\n"}]}, {"type": "t", "text": "\n\n"}, {"type": "p", "children": [{"type": "t", "text": "\nIn addition to genetic variation, SLCO1A2 function is subject to regulation by multiple intracellular signaling pathways and modulatory proteins. Studies have shown that kinase pathways such as those involving AMPK, as well as epigenetic modifications mediated by histone deacetylases (for example, HDAC6) and PDZ-domain adaptor proteins (PDZK1 and NHERF1), can influence OATP1A2 trafficking, membrane stability and recycling. These regulatory mechanisms further modulate substrate uptake and may have clinical implications in diverse settings ranging from altered drug–drug interactions and variable chemotherapeutic responses (e.g. celiprolol and imatinib) to potential roles in neurodegenerative disease and cancer prognosis."}, {"type": "fg", "children": [{"type": "fg_fs", "start_ref": "31", "end_ref": "38"}]}, {"type": "t", "text": "\n"}]}, {"type": "rg", "children": [{"type": "r", "ref": 1, "children": [{"type": "t", "text": "Wooin Lee, Hartmut Glaeser, L Harris Smith, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Polymorphisms in human organic anion-transporting polypeptide 1A2 (OATP1A2): implications for altered drug disposition and central nervous system drug entry."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Biol Chem (2005)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1074/jbc.M411092200"}], "href": "https://doi.org/10.1074/jbc.M411092200"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "15632119"}], "href": "https://pubmed.ncbi.nlm.nih.gov/15632119"}]}, {"type": "r", "ref": 2, "children": [{"type": "t", "text": "Yaming Su, Xiaoping Zhang, Patrick J Sinko "}, {"type": "b", "children": [{"type": "t", "text": "Human organic anion-transporting polypeptide OATP-A (SLC21A3) acts in concert with P-glycoprotein and multidrug resistance protein 2 in the vectorial transport of Saquinavir in Hep G2 cells."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Mol Pharm (2004)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1021/mp0340136"}], "href": "https://doi.org/10.1021/mp0340136"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "15832500"}], "href": "https://pubmed.ncbi.nlm.nih.gov/15832500"}]}, {"type": "r", "ref": 3, "children": [{"type": "t", "text": "Zoulikha M Zaïr, Jyrki J Eloranta, Bruno Stieger, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Pharmacogenetics of OATP (SLC21/SLCO), OAT and OCT (SLC22) and PEPT (SLC15) transporters in the intestine, liver and kidney."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Pharmacogenomics (2008)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.2217/14622416.9.5.597"}], "href": "https://doi.org/10.2217/14622416.9.5.597"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "18466105"}], "href": "https://pubmed.ncbi.nlm.nih.gov/18466105"}]}, {"type": "r", "ref": 4, "children": [{"type": "t", "text": "Masatomo Miura, Hideaki Kagaya, Shigeru Satoh, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Influence of drug transporters and UGT polymorphisms on pharmacokinetics of phenolic glucuronide metabolite of mycophenolic acid in Japanese renal transplant recipients."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Ther Drug Monit (2008)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1097/FTD.0b013e3181838063"}], "href": "https://doi.org/10.1097/FTD.0b013e3181838063"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "18695635"}], "href": "https://pubmed.ncbi.nlm.nih.gov/18695635"}]}, {"type": "r", "ref": 5, "children": [{"type": "t", "text": "Henriette E Meyer zu Schwabedissen, Rommel G Tirona, Cindy S Yip, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Interplay between the nuclear receptor pregnane X receptor and the uptake transporter organic anion transporter polypeptide 1A2 selectively enhances estrogen effects in breast cancer."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Cancer Res (2008)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1158/0008-5472.CAN-08-0265"}], "href": "https://doi.org/10.1158/0008-5472.CAN-08-0265"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "19010908"}], "href": "https://pubmed.ncbi.nlm.nih.gov/19010908"}]}, {"type": "r", "ref": 6, "children": [{"type": "t", "text": "N Picard, S W Yee, J-B Woillard, et al. "}, {"type": "b", "children": [{"type": "t", "text": "The role of organic anion-transporting polypeptides and their common genetic variants in mycophenolic acid pharmacokinetics."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Clin Pharmacol Ther (2010)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/clpt.2009.205"}], "href": "https://doi.org/10.1038/clpt.2009.205"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "19890249"}], "href": "https://pubmed.ncbi.nlm.nih.gov/19890249"}]}, {"type": "r", "ref": 7, "children": [{"type": "t", "text": "Ruben C Hartkoorn, Wai San Kwan, Victoria Shallcross, et al. "}, {"type": "b", "children": [{"type": "t", "text": "HIV protease inhibitors are substrates for OATP1A2, OATP1B1 and OATP1B3 and lopinavir plasma concentrations are influenced by SLCO1B1 polymorphisms."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Pharmacogenet Genomics (2010)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1097/FPC.0b013e328335b02d"}], "href": "https://doi.org/10.1097/FPC.0b013e328335b02d"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "20051929"}], "href": "https://pubmed.ncbi.nlm.nih.gov/20051929"}]}, {"type": "r", "ref": 8, "children": [{"type": "t", "text": "Alli Laitinen, Mikko Niemi "}, {"type": "b", "children": [{"type": "t", "text": "Frequencies of single-nucleotide polymorphisms of SLCO1A2, SLCO1B3 and SLCO2B1 genes in a Finnish population."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Basic Clin Pharmacol Toxicol (2011)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1111/j.1742-7843.2010.00605.x"}], "href": "https://doi.org/10.1111/j.1742-7843.2010.00605.x"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "20560925"}], "href": "https://pubmed.ncbi.nlm.nih.gov/20560925"}]}, {"type": "r", "ref": 9, "children": [{"type": "t", "text": "Stephan Buch, Clemens Schafmayer, Henry Völzke, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Loci from a genome-wide analysis of bilirubin levels are associated with gallstone risk and composition."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Gastroenterology (2010)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1053/j.gastro.2010.09.003"}], "href": "https://doi.org/10.1053/j.gastro.2010.09.003"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "20837016"}], "href": "https://pubmed.ncbi.nlm.nih.gov/20837016"}]}, {"type": "r", "ref": 10, "children": [{"type": "t", "text": "Fanfan Zhou, Andy C Lee, Katja Krafczyk, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Protein kinase C regulates the internalization and function of the human organic anion transporting polypeptide 1A2."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Br J Pharmacol (2011)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1111/j.1476-5381.2010.01144.x"}], "href": "https://doi.org/10.1111/j.1476-5381.2010.01144.x"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "21133891"}], "href": "https://pubmed.ncbi.nlm.nih.gov/21133891"}]}, {"type": "r", "ref": 11, "children": [{"type": "t", "text": "Ziqiang Cheng, Houfu Liu, Na Yu, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Hydrophilic anti-migraine triptans are substrates for OATP1A2, a transporter expressed at human blood-brain barrier."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Xenobiotica (2012)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.3109/00498254.2012.675455"}], "href": "https://doi.org/10.3109/00498254.2012.675455"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "22509823"}], "href": "https://pubmed.ncbi.nlm.nih.gov/22509823"}]}, {"type": "r", "ref": 12, "children": [{"type": "t", "text": "Evita van de Steeg, Anita van Esch, Els Wagenaar, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Influence of human OATP1B1, OATP1B3, and OATP1A2 on the pharmacokinetics of methotrexate and paclitaxel in humanized transgenic mice."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Clin Cancer Res (2013)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1158/1078-0432.CCR-12-2080"}], "href": "https://doi.org/10.1158/1078-0432.CCR-12-2080"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "23243220"}], "href": "https://pubmed.ncbi.nlm.nih.gov/23243220"}]}, {"type": "r", "ref": 13, "children": [{"type": "t", "text": "Fanfan Zhou, Jian Zheng, Ling Zhu, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Functional analysis of novel polymorphisms in the human SLCO1A2 gene that encodes the transporter OATP1A2."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "AAPS J (2013)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1208/s12248-013-9515-1"}], "href": "https://doi.org/10.1208/s12248-013-9515-1"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "23918469"}], "href": "https://pubmed.ncbi.nlm.nih.gov/23918469"}]}, {"type": "r", "ref": 14, "children": [{"type": "t", "text": "Ting Chan, Ling Zhu, Michele C Madigan, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Human organic anion transporting polypeptide 1A2 (OATP1A2) mediates cellular uptake of all-trans-retinol in human retinal pigmented epithelial cells."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Br J Pharmacol (2015)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1111/bph.13060"}], "href": "https://doi.org/10.1111/bph.13060"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "25560245"}], "href": "https://pubmed.ncbi.nlm.nih.gov/25560245"}]}, {"type": "r", "ref": 15, "children": [{"type": "t", "text": "Hannah H Lee, Brenda F Leake, Richard B Kim, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Contribution of Organic Anion-Transporting Polypeptides 1A/1B to Doxorubicin Uptake and Clearance."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Mol Pharmacol (2017)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1124/mol.116.105544"}], "href": "https://doi.org/10.1124/mol.116.105544"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "27777271"}], "href": "https://pubmed.ncbi.nlm.nih.gov/27777271"}]}, {"type": "r", "ref": 16, "children": [{"type": "t", "text": "S-Y Chan, A Martín-Santos, L S Loubière, et al. "}, {"type": "b", "children": [{"type": "t", "text": "The expression of thyroid hormone transporters in the human fetal cerebral cortex during early development and in N-Tera-2 neurodifferentiation."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Physiol (2011)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1113/jphysiol.2011.207290"}], "href": "https://doi.org/10.1113/jphysiol.2011.207290"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "21486766"}], "href": "https://pubmed.ncbi.nlm.nih.gov/21486766"}]}, {"type": "r", "ref": 17, "children": [{"type": "t", "text": "K Eechoute, R M Franke, W J Loos, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Environmental and genetic factors affecting transport of imatinib by OATP1A2."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Clin Pharmacol Ther (2011)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/clpt.2011.42"}], "href": "https://doi.org/10.1038/clpt.2011.42"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "21508937"}], "href": "https://pubmed.ncbi.nlm.nih.gov/21508937"}]}, {"type": "r", "ref": 18, "children": [{"type": "t", "text": "Y Yamakawa, A Hamada, T Shuto, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Pharmacokinetic impact of SLCO1A2 polymorphisms on imatinib disposition in patients with chronic myeloid leukemia."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Clin Pharmacol Ther (2011)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1038/clpt.2011.102"}], "href": "https://doi.org/10.1038/clpt.2011.102"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "21633340"}], "href": "https://pubmed.ncbi.nlm.nih.gov/21633340"}]}, {"type": "r", "ref": 19, "children": [{"type": "t", "text": "Sam Rebello, Sally Zhao, Sam Hariry, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Intestinal OATP1A2 inhibition as a potential mechanism for the effect of grapefruit juice on aliskiren pharmacokinetics in healthy subjects."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Eur J Clin Pharmacol (2012)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1007/s00228-011-1167-4"}], "href": "https://doi.org/10.1007/s00228-011-1167-4"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "22124880"}], "href": "https://pubmed.ncbi.nlm.nih.gov/22124880"}]}, {"type": "r", "ref": 20, "children": [{"type": "t", "text": "Huimin Wang, Ziru Yan, Minyue Dong, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Alteration in placental expression of bile acids transporters OATP1A2, OATP1B1, OATP1B3 in intrahepatic cholestasis of pregnancy."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Arch Gynecol Obstet (2012)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1007/s00404-011-2183-4"}], "href": "https://doi.org/10.1007/s00404-011-2183-4"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "22203093"}], "href": "https://pubmed.ncbi.nlm.nih.gov/22203093"}]}, {"type": "r", "ref": 21, "children": [{"type": "t", "text": "Jyrki J Eloranta, Christian Hiller, Moritz Jüttner, et al. "}, {"type": "b", "children": [{"type": "t", "text": "The SLCO1A2 gene, encoding human organic anion-transporting polypeptide 1A2, is transactivated by the vitamin D receptor."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Mol Pharmacol (2012)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1124/mol.112.077909"}], "href": "https://doi.org/10.1124/mol.112.077909"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "22474172"}], "href": "https://pubmed.ncbi.nlm.nih.gov/22474172"}]}, {"type": "r", "ref": 22, "children": [{"type": "t", "text": "Yumi Hashimoto, Shigenobu Tatsumi, Risa Takeda, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Expression of organic anion-transporting polypeptide 1A2 and organic cation transporter 6 as a predictor of pathologic response to neoadjuvant chemotherapy in triple negative breast cancer."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Breast Cancer Res Treat (2014)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1007/s10549-014-2913-y"}], "href": "https://doi.org/10.1007/s10549-014-2913-y"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "24671357"}], "href": "https://pubmed.ncbi.nlm.nih.gov/24671357"}]}, {"type": "r", "ref": 23, "children": [{"type": "t", "text": "Jian Zheng, Ting Chan, Florence Shin Gee Cheung, et al. "}, {"type": "b", "children": [{"type": "t", "text": "PDZK1 and NHERF1 regulate the function of human organic anion transporting polypeptide 1A2 (OATP1A2) by modulating its subcellular trafficking and stability."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "PLoS One (2014)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1371/journal.pone.0094712"}], "href": "https://doi.org/10.1371/journal.pone.0094712"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "24728453"}], "href": "https://pubmed.ncbi.nlm.nih.gov/24728453"}]}, {"type": "r", "ref": 24, "children": [{"type": "t", "text": "Ting Chan, Jian Zheng, Ling Zhu, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Putative transmembrane domain 6 of the human organic anion transporting polypeptide 1A2 (OATP1A2) influences transporter substrate binding, protein trafficking, and quality control."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Mol Pharm (2015)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1021/mp500459b"}], "href": "https://doi.org/10.1021/mp500459b"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "25387129"}], "href": "https://pubmed.ncbi.nlm.nih.gov/25387129"}]}, {"type": "r", "ref": 25, "children": [{"type": "t", "text": "Yinhui Zhou, Jingjing Yuan, Zhisong Li, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Genetic polymorphisms and function of the organic anion-transporting polypeptide 1A2 and its clinical relevance in drug disposition."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Pharmacology (2015)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1159/000381313"}], "href": "https://doi.org/10.1159/000381313"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "25924632"}], "href": "https://pubmed.ncbi.nlm.nih.gov/25924632"}]}, {"type": "r", "ref": 26, "children": [{"type": "t", "text": "Leila Chaouch, Miniar Kalai1, Imen Darragi, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Genetic link with cholelithiasis among pediatric SCA Tunisian patients: Examples of UGT1A1, SLCO1A2 and SLCO1B1."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Hematology (2016)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1179/1607845415Y.0000000030"}], "href": "https://doi.org/10.1179/1607845415Y.0000000030"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "26146896"}], "href": "https://pubmed.ncbi.nlm.nih.gov/26146896"}]}, {"type": "r", "ref": 27, "children": [{"type": "t", "text": "Xiaozhe Qi, Els Wagenaar, Wentao Xu, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Ochratoxin A transport by the human breast cancer resistance protein (BCRP), multidrug resistance protein 2 (MRP2), and organic anion-transporting polypeptides 1A2, 1B1 and 2B1."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Toxicol Appl Pharmacol (2017)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/j.taap.2017.05.022"}], "href": "https://doi.org/10.1016/j.taap.2017.05.022"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "28532671"}], "href": "https://pubmed.ncbi.nlm.nih.gov/28532671"}]}, {"type": "r", "ref": 28, "children": [{"type": "t", "text": "Vinicius A Sortica, Juliana D Lindenau, Maristela G Cunha, et al. "}, {"type": "b", "children": [{"type": "t", "text": "SLCO1A2, SLCO1B1 and SLCO2B1 polymorphisms influences chloroquine and primaquine treatment in Plasmodium vivax malaria."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Pharmacogenomics (2017)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.2217/pgs-2017-0077"}], "href": "https://doi.org/10.2217/pgs-2017-0077"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "28975866"}], "href": "https://pubmed.ncbi.nlm.nih.gov/28975866"}]}, {"type": "r", "ref": 29, "children": [{"type": "t", "text": "Shu-Mei Wang, Wei-Xin Zeng, Wan-Shui Wu, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Association between a microRNA binding site polymorphism in SLCO1A2 and the risk of delayed methotrexate elimination in Chinese children with acute lymphoblastic leukemia."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Leuk Res (2018)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/j.leukres.2018.01.004"}], "href": "https://doi.org/10.1016/j.leukres.2018.01.004"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "29306656"}], "href": "https://pubmed.ncbi.nlm.nih.gov/29306656"}]}, {"type": "r", "ref": 30, "children": [{"type": "t", "text": "Zhaojian Xiang, Weike Li, Lixue Wang, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Identification of a NF"}, {"type": "a", "children": [{"type": "t", "text": "i"}], "href": "i"}, {"type": "t", "text": "κ"}, {"type": "a", "children": [{"type": "t", "text": "/i"}], "href": "/i"}, {"type": "t", "text": "B Inhibition Site on the Proximal Promoter Region of Human Organic Anion Transporting Polypeptide 1A2 Coding Gene "}, {"type": "a", "children": [{"type": "t", "text": "i"}], "href": "i"}, {"type": "t", "text": "SLCO1A2"}, {"type": "a", "children": [{"type": "t", "text": "/i"}], "href": "/i"}, {"type": "t", "text": "."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Drug Metab Dispos (2018)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1124/dmd.117.078832"}], "href": "https://doi.org/10.1124/dmd.117.078832"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "29549185"}], "href": "https://pubmed.ncbi.nlm.nih.gov/29549185"}]}, {"type": "r", "ref": 31, "children": [{"type": "t", "text": "Monica Y Sanchez-Contreras, Naomi Kouri, Casey N Cook, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Replication of progressive supranuclear palsy genome-wide association study identifies SLCO1A2 and DUSP10 as new susceptibility loci."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Mol Neurodegener (2018)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1186/s13024-018-0267-3"}], "href": "https://doi.org/10.1186/s13024-018-0267-3"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "29986742"}], "href": "https://pubmed.ncbi.nlm.nih.gov/29986742"}]}, {"type": "r", "ref": 32, "children": [{"type": "t", "text": "Xiaoxi Lu, Ting Chan, Zhengqi Cheng, et al. "}, {"type": "b", "children": [{"type": "t", "text": "The 5'-AMP-Activated Protein Kinase Regulates the Function and Expression of Human Organic Anion Transporting Polypeptide 1A2."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Mol Pharmacol (2018)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1124/mol.118.113423"}], "href": "https://doi.org/10.1124/mol.118.113423"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "30348897"}], "href": "https://pubmed.ncbi.nlm.nih.gov/30348897"}]}, {"type": "r", "ref": 33, "children": [{"type": "t", "text": "Jiayi Wang, Jun Yin, Wei Li, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Association between SLCO1A2 genetic variation and methotrexate toxicity in human rheumatoid arthritis treatment."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Biochem Mol Toxicol (2020)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1002/jbt.22513"}], "href": "https://doi.org/10.1002/jbt.22513"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "32304147"}], "href": "https://pubmed.ncbi.nlm.nih.gov/32304147"}]}, {"type": "r", "ref": 34, "children": [{"type": "t", "text": "Jingli Cai, Zhaowen Wang, Guiming Chen, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Reabsorption of bile acids regulated by FXR-OATP1A2 is the main factor for the formation of cholesterol gallstone."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Am J Physiol Gastrointest Liver Physiol (2020)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1152/ajpgi.00385.2019"}], "href": "https://doi.org/10.1152/ajpgi.00385.2019"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "32597704"}], "href": "https://pubmed.ncbi.nlm.nih.gov/32597704"}]}, {"type": "r", "ref": 35, "children": [{"type": "t", "text": "Viktor Hlavac, Radka Vaclavikova, Veronika Brynychova, et al. "}, {"type": "b", "children": [{"type": "t", "text": "SLC46A1 Haplotype with Predicted Functional Impact has Prognostic Value in Breast Carcinoma."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Mol Diagn Ther (2021)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1007/s40291-020-00506-2"}], "href": "https://doi.org/10.1007/s40291-020-00506-2"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "33387348"}], "href": "https://pubmed.ncbi.nlm.nih.gov/33387348"}]}, {"type": "r", "ref": 36, "children": [{"type": "t", "text": "Xiaoli Zheng, Jian V Zhang, Yanfeng Bai, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Upregulation of OATP1A2 in human oesophageal squamous cell carcinoma cells via the HDAC6-GCN5/PCAF-H3K9Ac axis."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Xenobiotica (2021)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1080/00498254.2021.2001076"}], "href": "https://doi.org/10.1080/00498254.2021.2001076"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "34823432"}], "href": "https://pubmed.ncbi.nlm.nih.gov/34823432"}]}, {"type": "r", "ref": 37, "children": [{"type": "t", "text": "Hongye Han, Takeshi Akiyoshi, Tokio Morita, et al. "}, {"type": "b", "children": [{"type": "t", "text": "Comparison of the transport kinetics of fexofenadine and its pH dependency among OATP1A2 genetic variants."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "Drug Metab Pharmacokinet (2022)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/j.dmpk.2022.100470"}], "href": "https://doi.org/10.1016/j.dmpk.2022.100470"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "36116173"}], "href": "https://pubmed.ncbi.nlm.nih.gov/36116173"}]}, {"type": "r", "ref": 38, "children": [{"type": "t", "text": "Hiroki Kataoka, Takeshi Akiyoshi, Yasuo Uchida, et al. "}, {"type": "b", "children": [{"type": "t", "text": "The Effects of N-Glycosylation on the Expression and Transport Activity of OATP1A2 and OATP2B1."}]}, {"type": "t", "text": " "}, {"type": "i", "children": [{"type": "t", "text": "J Pharm Sci (2024)"}]}, {"type": "t", "text": " DOI: "}, {"type": "a", "children": [{"type": "t", "text": "10.1016/j.xphs.2024.02.012"}], "href": "https://doi.org/10.1016/j.xphs.2024.02.012"}, {"type": "t", "text": " PMID: "}, {"type": "a", "children": [{"type": "t", "text": "38432624"}], "href": "https://pubmed.ncbi.nlm.nih.gov/38432624"}]}]}]}
|
Synonyms | OATP, OATP1A2, OATP-A, SLC21A3 |
Proteins | SO1A2_HUMAN |
NCBI Gene ID | 6579 |
API | |
Download Associations | |
Predicted Functions |
![]() |
Co-expressed Genes |
![]() |
Expression in Tissues and Cell Lines |
![]() |
SLCO1A2 has 3,796 functional associations with biological entities spanning 8 categories (molecular profile, organism, chemical, functional term, phrase or reference, disease, phenotype or trait, structural feature, cell line, cell type or tissue, gene, protein or microRNA) extracted from 102 datasets.
Click the + buttons to view associations for SLCO1A2 from the datasets below.
If available, associations are ranked by standardized value
Dataset | Summary | |
---|---|---|
Allen Brain Atlas Adult Human Brain Tissue Gene Expression Profiles | tissues with high or low expression of SLCO1A2 gene relative to other tissues from the Allen Brain Atlas Adult Human Brain Tissue Gene Expression Profiles dataset. | |
Allen Brain Atlas Adult Mouse Brain Tissue Gene Expression Profiles | tissues with high or low expression of SLCO1A2 gene relative to other tissues from the Allen Brain Atlas Adult Mouse Brain Tissue Gene Expression Profiles dataset. | |
Allen Brain Atlas Developing Human Brain Tissue Gene Expression Profiles by Microarray | tissue samples with high or low expression of SLCO1A2 gene relative to other tissue samples from the Allen Brain Atlas Developing Human Brain Tissue Gene Expression Profiles by Microarray dataset. | |
Allen Brain Atlas Developing Human Brain Tissue Gene Expression Profiles by RNA-seq | tissue samples with high or low expression of SLCO1A2 gene relative to other tissue samples from the Allen Brain Atlas Developing Human Brain Tissue Gene Expression Profiles by RNA-seq dataset. | |
Allen Brain Atlas Prenatal Human Brain Tissue Gene Expression Profiles | tissues with high or low expression of SLCO1A2 gene relative to other tissues from the Allen Brain Atlas Prenatal Human Brain Tissue Gene Expression Profiles dataset. | |
BioGPS Cell Line Gene Expression Profiles | cell lines with high or low expression of SLCO1A2 gene relative to other cell lines from the BioGPS Cell Line Gene Expression Profiles dataset. | |
BioGPS Human Cell Type and Tissue Gene Expression Profiles | cell types and tissues with high or low expression of SLCO1A2 gene relative to other cell types and tissues from the BioGPS Human Cell Type and Tissue Gene Expression Profiles dataset. | |
BioGPS Mouse Cell Type and Tissue Gene Expression Profiles | cell types and tissues with high or low expression of SLCO1A2 gene relative to other cell types and tissues from the BioGPS Mouse Cell Type and Tissue Gene Expression Profiles dataset. | |
CCLE Cell Line Gene CNV Profiles | cell lines with high or low copy number of SLCO1A2 gene relative to other cell lines from the CCLE Cell Line Gene CNV Profiles dataset. | |
CCLE Cell Line Gene Expression Profiles | cell lines with high or low expression of SLCO1A2 gene relative to other cell lines from the CCLE Cell Line Gene Expression Profiles dataset. | |
ChEA Transcription Factor Binding Site Profiles | transcription factor binding site profiles with transcription factor binding evidence at the promoter of SLCO1A2 gene from the CHEA Transcription Factor Binding Site Profiles dataset. | |
ChEA Transcription Factor Targets | transcription factors binding the promoter of SLCO1A2 gene in low- or high-throughput transcription factor functional studies from the CHEA Transcription Factor Targets dataset. | |
ChEA Transcription Factor Targets 2022 | transcription factors binding the promoter of SLCO1A2 gene in low- or high-throughput transcription factor functional studies from the CHEA Transcription Factor Targets 2022 dataset. | |
CMAP Signatures of Differentially Expressed Genes for Small Molecules | small molecule perturbations changing expression of SLCO1A2 gene from the CMAP Signatures of Differentially Expressed Genes for Small Molecules dataset. | |
COMPARTMENTS Curated Protein Localization Evidence Scores | cellular components containing SLCO1A2 protein from the COMPARTMENTS Curated Protein Localization Evidence Scores dataset. | |
COMPARTMENTS Curated Protein Localization Evidence Scores 2025 | cellular components containing SLCO1A2 protein from the COMPARTMENTS Curated Protein Localization Evidence Scores 2025 dataset. | |
COMPARTMENTS Text-mining Protein Localization Evidence Scores | cellular components co-occuring with SLCO1A2 protein in abstracts of biomedical publications from the COMPARTMENTS Text-mining Protein Localization Evidence Scores dataset. | |
COMPARTMENTS Text-mining Protein Localization Evidence Scores 2025 | cellular components co-occuring with SLCO1A2 protein in abstracts of biomedical publications from the COMPARTMENTS Text-mining Protein Localization Evidence Scores 2025 dataset. | |
COSMIC Cell Line Gene CNV Profiles | cell lines with high or low copy number of SLCO1A2 gene relative to other cell lines from the COSMIC Cell Line Gene CNV Profiles dataset. | |
COSMIC Cell Line Gene Mutation Profiles | cell lines with SLCO1A2 gene mutations from the COSMIC Cell Line Gene Mutation Profiles dataset. | |
CTD Gene-Chemical Interactions | chemicals interacting with SLCO1A2 gene/protein from the curated CTD Gene-Chemical Interactions dataset. | |
CTD Gene-Disease Associations | diseases associated with SLCO1A2 gene/protein from the curated CTD Gene-Disease Associations dataset. | |
dbGAP Gene-Trait Associations | traits associated with SLCO1A2 gene in GWAS and other genetic association datasets from the dbGAP Gene-Trait Associations dataset. | |
DepMap CRISPR Gene Dependency | cell lines with fitness changed by SLCO1A2 gene knockdown relative to other cell lines from the DepMap CRISPR Gene Dependency dataset. | |
DISEASES Experimental Gene-Disease Association Evidence Scores | diseases associated with SLCO1A2 gene in GWAS datasets from the DISEASES Experimental Gene-Disease Assocation Evidence Scores dataset. | |
DISEASES Experimental Gene-Disease Association Evidence Scores 2025 | diseases associated with SLCO1A2 gene in GWAS datasets from the DISEASES Experimental Gene-Disease Assocation Evidence Scores 2025 dataset. | |
DISEASES Text-mining Gene-Disease Association Evidence Scores | diseases co-occuring with SLCO1A2 gene in abstracts of biomedical publications from the DISEASES Text-mining Gene-Disease Assocation Evidence Scores dataset. | |
DISEASES Text-mining Gene-Disease Association Evidence Scores 2025 | diseases co-occuring with SLCO1A2 gene in abstracts of biomedical publications from the DISEASES Text-mining Gene-Disease Assocation Evidence Scores 2025 dataset. | |
DisGeNET Gene-Disease Associations | diseases associated with SLCO1A2 gene in GWAS and other genetic association datasets from the DisGeNET Gene-Disease Associations dataset. | |
DisGeNET Gene-Phenotype Associations | phenotypes associated with SLCO1A2 gene in GWAS and other genetic association datasets from the DisGeNET Gene-Phenoptype Associations dataset. | |
DrugBank Drug Targets | interacting drugs for SLCO1A2 protein from the curated DrugBank Drug Targets dataset. | |
ENCODE Histone Modification Site Profiles | histone modification site profiles with high histone modification abundance at SLCO1A2 gene from the ENCODE Histone Modification Site Profiles dataset. | |
ENCODE Transcription Factor Binding Site Profiles | transcription factor binding site profiles with transcription factor binding evidence at the promoter of SLCO1A2 gene from the ENCODE Transcription Factor Binding Site Profiles dataset. | |
ENCODE Transcription Factor Targets | transcription factors binding the promoter of SLCO1A2 gene in ChIP-seq datasets from the ENCODE Transcription Factor Targets dataset. | |
ESCAPE Omics Signatures of Genes and Proteins for Stem Cells | PubMedIDs of publications reporting gene signatures containing SLCO1A2 from the ESCAPE Omics Signatures of Genes and Proteins for Stem Cells dataset. | |
GAD Gene-Disease Associations | diseases associated with SLCO1A2 gene in GWAS and other genetic association datasets from the GAD Gene-Disease Associations dataset. | |
GAD High Level Gene-Disease Associations | diseases associated with SLCO1A2 gene in GWAS and other genetic association datasets from the GAD High Level Gene-Disease Associations dataset. | |
GDSC Cell Line Gene Expression Profiles | cell lines with high or low expression of SLCO1A2 gene relative to other cell lines from the GDSC Cell Line Gene Expression Profiles dataset. | |
GeneRIF Biological Term Annotations | biological terms co-occuring with SLCO1A2 gene in literature-supported statements describing functions of genes from the GeneRIF Biological Term Annotations dataset. | |
GeneSigDB Published Gene Signatures | PubMedIDs of publications reporting gene signatures containing SLCO1A2 from the GeneSigDB Published Gene Signatures dataset. | |
GEO Signatures of Differentially Expressed Genes for Diseases | disease perturbations changing expression of SLCO1A2 gene from the GEO Signatures of Differentially Expressed Genes for Diseases dataset. | |
GEO Signatures of Differentially Expressed Genes for Gene Perturbations | gene perturbations changing expression of SLCO1A2 gene from the GEO Signatures of Differentially Expressed Genes for Gene Perturbations dataset. | |
GEO Signatures of Differentially Expressed Genes for Kinase Perturbations | kinase perturbations changing expression of SLCO1A2 gene from the GEO Signatures of Differentially Expressed Genes for Kinase Perturbations dataset. | |
GEO Signatures of Differentially Expressed Genes for Small Molecules | small molecule perturbations changing expression of SLCO1A2 gene from the GEO Signatures of Differentially Expressed Genes for Small Molecules dataset. | |
GEO Signatures of Differentially Expressed Genes for Transcription Factor Perturbations | transcription factor perturbations changing expression of SLCO1A2 gene from the GEO Signatures of Differentially Expressed Genes for Transcription Factor Perturbations dataset. | |
GEO Signatures of Differentially Expressed Genes for Viral Infections | virus perturbations changing expression of SLCO1A2 gene from the GEO Signatures of Differentially Expressed Genes for Viral Infections dataset. | |
GO Biological Process Annotations 2015 | biological processes involving SLCO1A2 gene from the curated GO Biological Process Annotations 2015 dataset. | |
GO Biological Process Annotations 2023 | biological processes involving SLCO1A2 gene from the curated GO Biological Process Annotations 2023 dataset. | |
GO Biological Process Annotations 2025 | biological processes involving SLCO1A2 gene from the curated GO Biological Process Annotations2025 dataset. | |
GO Cellular Component Annotations 2015 | cellular components containing SLCO1A2 protein from the curated GO Cellular Component Annotations 2015 dataset. | |
GO Cellular Component Annotations 2025 | cellular components containing SLCO1A2 protein from the curated GO Cellular Component Annotations 2025 dataset. | |
GO Molecular Function Annotations 2015 | molecular functions performed by SLCO1A2 gene from the curated GO Molecular Function Annotations 2015 dataset. | |
GO Molecular Function Annotations 2023 | molecular functions performed by SLCO1A2 gene from the curated GO Molecular Function Annotations 2023 dataset. | |
GO Molecular Function Annotations 2025 | molecular functions performed by SLCO1A2 gene from the curated GO Molecular Function Annotations 2025 dataset. | |
GTEx Tissue Gene Expression Profiles | tissues with high or low expression of SLCO1A2 gene relative to other tissues from the GTEx Tissue Gene Expression Profiles dataset. | |
GTEx Tissue Gene Expression Profiles 2023 | tissues with high or low expression of SLCO1A2 gene relative to other tissues from the GTEx Tissue Gene Expression Profiles 2023 dataset. | |
GTEx Tissue Sample Gene Expression Profiles | tissue samples with high or low expression of SLCO1A2 gene relative to other tissue samples from the GTEx Tissue Sample Gene Expression Profiles dataset. | |
Guide to Pharmacology Chemical Ligands of Receptors | ligands (chemical) binding SLCO1A2 receptor from the curated Guide to Pharmacology Chemical Ligands of Receptors dataset. | |
GWAS Catalog SNP-Phenotype Associations | phenotypes associated with SLCO1A2 gene in GWAS datasets from the GWAS Catalog SNP-Phenotype Associations dataset. | |
GWASdb SNP-Disease Associations | diseases associated with SLCO1A2 gene in GWAS and other genetic association datasets from the GWASdb SNP-Disease Associations dataset. | |
GWASdb SNP-Phenotype Associations | phenotypes associated with SLCO1A2 gene in GWAS datasets from the GWASdb SNP-Phenotype Associations dataset. | |
Heiser et al., PNAS, 2011 Cell Line Gene Expression Profiles | cell lines with high or low expression of SLCO1A2 gene relative to other cell lines from the Heiser et al., PNAS, 2011 Cell Line Gene Expression Profiles dataset. | |
HMDB Metabolites of Enzymes | interacting metabolites for SLCO1A2 protein from the curated HMDB Metabolites of Enzymes dataset. | |
HPA Cell Line Gene Expression Profiles | cell lines with high or low expression of SLCO1A2 gene relative to other cell lines from the HPA Cell Line Gene Expression Profiles dataset. | |
HPA Tissue Gene Expression Profiles | tissues with high or low expression of SLCO1A2 gene relative to other tissues from the HPA Tissue Gene Expression Profiles dataset. | |
HPA Tissue Protein Expression Profiles | tissues with high or low expression of SLCO1A2 protein relative to other tissues from the HPA Tissue Protein Expression Profiles dataset. | |
HPA Tissue Sample Gene Expression Profiles | tissue samples with high or low expression of SLCO1A2 gene relative to other tissue samples from the HPA Tissue Sample Gene Expression Profiles dataset. | |
HuBMAP Azimuth Cell Type Annotations | cell types associated with SLCO1A2 gene from the HuBMAP Azimuth Cell Type Annotations dataset. | |
HuGE Navigator Gene-Phenotype Associations | phenotypes associated with SLCO1A2 gene by text-mining GWAS publications from the HuGE Navigator Gene-Phenotype Associations dataset. | |
InterPro Predicted Protein Domain Annotations | protein domains predicted for SLCO1A2 protein from the InterPro Predicted Protein Domain Annotations dataset. | |
JASPAR Predicted Transcription Factor Targets | transcription factors regulating expression of SLCO1A2 gene predicted using known transcription factor binding site motifs from the JASPAR Predicted Transcription Factor Targets dataset. | |
Klijn et al., Nat. Biotechnol., 2015 Cell Line Gene CNV Profiles | cell lines with high or low copy number of SLCO1A2 gene relative to other cell lines from the Klijn et al., Nat. Biotechnol., 2015 Cell Line Gene CNV Profiles dataset. | |
Klijn et al., Nat. Biotechnol., 2015 Cell Line Gene Mutation Profiles | cell lines with SLCO1A2 gene mutations from the Klijn et al., Nat. Biotechnol., 2015 Cell Line Gene Mutation Profiles dataset. | |
KnockTF Gene Expression Profiles with Transcription Factor Perturbations | transcription factor perturbations changing expression of SLCO1A2 gene from the KnockTF Gene Expression Profiles with Transcription Factor Perturbations dataset. | |
LINCS L1000 CMAP Chemical Perturbation Consensus Signatures | small molecule perturbations changing expression of SLCO1A2 gene from the LINCS L1000 CMAP Chemical Perturbations Consensus Signatures dataset. | |
LOCATE Curated Protein Localization Annotations | cellular components containing SLCO1A2 protein in low- or high-throughput protein localization assays from the LOCATE Curated Protein Localization Annotations dataset. | |
LOCATE Predicted Protein Localization Annotations | cellular components predicted to contain SLCO1A2 protein from the LOCATE Predicted Protein Localization Annotations dataset. | |
MGI Mouse Phenotype Associations 2023 | phenotypes of transgenic mice caused by SLCO1A2 gene mutations from the MGI Mouse Phenotype Associations 2023 dataset. | |
MotifMap Predicted Transcription Factor Targets | transcription factors regulating expression of SLCO1A2 gene predicted using known transcription factor binding site motifs from the MotifMap Predicted Transcription Factor Targets dataset. | |
MPO Gene-Phenotype Associations | phenotypes of transgenic mice caused by SLCO1A2 gene mutations from the MPO Gene-Phenotype Associations dataset. | |
MSigDB Signatures of Differentially Expressed Genes for Cancer Gene Perturbations | gene perturbations changing expression of SLCO1A2 gene from the MSigDB Signatures of Differentially Expressed Genes for Cancer Gene Perturbations dataset. | |
NURSA Protein Complexes | protein complexs containing SLCO1A2 protein recovered by IP-MS from the NURSA Protein Complexes dataset. | |
Pathway Commons Protein-Protein Interactions | interacting proteins for SLCO1A2 from the Pathway Commons Protein-Protein Interactions dataset. | |
PerturbAtlas Signatures of Differentially Expressed Genes for Gene Perturbations | gene perturbations changing expression of SLCO1A2 gene from the PerturbAtlas Signatures of Differentially Expressed Genes for Gene Perturbations dataset. | |
PFOCR Pathway Figure Associations 2023 | pathways involving SLCO1A2 protein from the PFOCR Pathway Figure Associations 2023 dataset. | |
PFOCR Pathway Figure Associations 2024 | pathways involving SLCO1A2 protein from the Wikipathways PFOCR 2024 dataset. | |
Reactome Pathways 2014 | pathways involving SLCO1A2 protein from the Reactome Pathways dataset. | |
Reactome Pathways 2024 | pathways involving SLCO1A2 protein from the Reactome Pathways 2024 dataset. | |
Roadmap Epigenomics Cell and Tissue DNA Methylation Profiles | cell types and tissues with high or low DNA methylation of SLCO1A2 gene relative to other cell types and tissues from the Roadmap Epigenomics Cell and Tissue DNA Methylation Profiles dataset. | |
Roadmap Epigenomics Histone Modification Site Profiles | histone modification site profiles with high histone modification abundance at SLCO1A2 gene from the Roadmap Epigenomics Histone Modification Site Profiles dataset. | |
RummaGEO Drug Perturbation Signatures | drug perturbations changing expression of SLCO1A2 gene from the RummaGEO Drug Perturbation Signatures dataset. | |
RummaGEO Gene Perturbation Signatures | gene perturbations changing expression of SLCO1A2 gene from the RummaGEO Gene Perturbation Signatures dataset. | |
TargetScan Predicted Conserved microRNA Targets | microRNAs regulating expression of SLCO1A2 gene predicted using conserved miRNA seed sequences from the TargetScan Predicted Conserved microRNA Targets dataset. | |
TargetScan Predicted Nonconserved microRNA Targets | microRNAs regulating expression of SLCO1A2 gene predicted using nonconserved miRNA seed sequences from the TargetScan Predicted Nonconserved microRNA Targets dataset. | |
TCGA Signatures of Differentially Expressed Genes for Tumors | tissue samples with high or low expression of SLCO1A2 gene relative to other tissue samples from the TCGA Signatures of Differentially Expressed Genes for Tumors dataset. | |
TISSUES Curated Tissue Protein Expression Evidence Scores | tissues with high expression of SLCO1A2 protein from the TISSUES Curated Tissue Protein Expression Evidence Scores dataset. | |
TISSUES Curated Tissue Protein Expression Evidence Scores 2025 | tissues with high expression of SLCO1A2 protein from the TISSUES Curated Tissue Protein Expression Evidence Scores 2025 dataset. | |
TISSUES Experimental Tissue Protein Expression Evidence Scores | tissues with high expression of SLCO1A2 protein in proteomics datasets from the TISSUES Experimental Tissue Protein Expression Evidence Scores dataset. | |
TISSUES Experimental Tissue Protein Expression Evidence Scores 2025 | tissues with high expression of SLCO1A2 protein in proteomics datasets from the TISSUES Experimental Tissue Protein Expression Evidence Scores 2025 dataset. | |
TISSUES Text-mining Tissue Protein Expression Evidence Scores | tissues co-occuring with SLCO1A2 protein in abstracts of biomedical publications from the TISSUES Text-mining Tissue Protein Expression Evidence Scores dataset. | |
TISSUES Text-mining Tissue Protein Expression Evidence Scores 2025 | tissues co-occuring with SLCO1A2 protein in abstracts of biomedical publications from the TISSUES Text-mining Tissue Protein Expression Evidence Scores 2025 dataset. | |
WikiPathways Pathways 2024 | pathways involving SLCO1A2 protein from the WikiPathways Pathways 2024 dataset. | |