ACS Publications. Most Trusted. Most Cited. Most Read
My Activity
CONTENT TYPES

Figure 1Loading Img

Characterization of Monocarboxylate Transport in Human Kidney HK-2 Cells

View Author Information
Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Amherst, New York 14260, and Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York 14263
Cite this: Mol. Pharm. 2006, 3, 6, 675–685
Publication Date (Web):October 12, 2006
https://doi.org/10.1021/mp060037b
Copyright © 2006 American Chemical Society

    Article Views

    1285

    Altmetric

    -

    Citations

    44
    LEARN ABOUT THESE METRICS
    Other access options

    Abstract

    Abstract Image

    The objectives of this study were to characterize the expression and function of monocarboxylate transporters (MCTs) in human kidney HK-2 cells and to compare the expression of MCTs in HK-2 cells to that found in human kidney. mRNA and protein expression of MCTs were determined by RT-PCR and Western analyses, respectively, while immunofluorescence staining was used to determine the membrane localization of MCT1. The driving force, transport kinetics, and inhibition of two MCT substrates, d-lactate and butyrate, were characterized in HK-2 cells. mRNA of MCT1, -2, -3, -4 isoforms were present in HK-2 cells and in human kidney cortex. MCT1 was present predominantly on the basal membranes of HK-2 cells. The cellular uptake of d-lactate and butyrate exhibited pH- and concentration-dependence (d-lactate, Km of 26.5 ± 2.2 mM and Vmax of 72.0 ± 14.5 nmol mg-1 min-1; butyrate, Km of 0.8 ± 0.3 mM, Vmax of 29.3 ± 2.5 nmol mg-1 min-1, and a diffusional clearance of 2.1 μL mg-1 min-1). The uptake of d-lactate and butyrate by HK-2 cells was inhibited by MCT analogues and the classical MCT inhibitors α-cyano-4-hydroxycinnamate, pCMB, and phloretin. The uptake of d-lactate and butyrate by HK-2 cells significantly decreased after transfection with small-interference RNA for MCT1. In summary, MCTs were present in both HK-2 cells and human kidney cortex, and HK-2 cells exhibited polarized MCT expression and pH-dependent transport of d-lactate and butyrate. Our results also support the usefulness of HK-2 cells as an in vitro model for studying monocarboxylate transport in renal proximal tubule cells.

    Keywords: Monocarboxylate transporter (MCT); HK-2 cells; RNA interference; cellular transport

    Read this article

    To access this article, please review the available access options below.

    Get instant access

    Purchase Access

    Read this article for 48 hours. Check out below using your ACS ID or as a guest.

    Recommended

    Access through Your Institution

    You may have access to this article through your institution.

    Your institution does not have access to this content. You can change your affiliated institution below.

     State University of New York.

     Present address:  Novartis Institutes for BioMedical Research, 250 Mass St., 3C-341-04, Cambridge, MA 02139.

    §

     Roswell Park Cancer Institute.

     Present address:  Cognigen Corporation, 395 Youngs Road, Williamsville, NY 14221.

    *

     Author to whom correspondence should be addressed. Mailing address:  Department of Pharmaceutical Sciences, 517 Hochstetter Hall, University at Buffalo, State University of New York, Amherst, NY 14260-1200. Tel:  (716) 645-2842 ext 230. Fax:  (716) 645-3693. E-mail:  [email protected].

    Cited By

    This article is cited by 44 publications.

    1. R. Seyedi, S.M. Tayebi, D. Zhang, Q. Yiming. The role of monocarboxylate transporter-1 and -4 in exercise and training: A mini-review article. Science & Sports 2024, 39 (2) , 144-152. https://doi.org/10.1016/j.scispo.2022.11.009
    2. Takaharu Takiguchi, Kazuaki Sugio, Masayuki Masuda, Shotaro Sasaki, Seiji Miyauchi. Uptake of Fluorescein via a pH-Dependent Monocarboxylate Transporter by Human Kidney 2 (HK-2) Cells. Biological and Pharmaceutical Bulletin 2024, 47 (1) , 79-87. https://doi.org/10.1248/bpb.b23-00570
    3. Tomas Rousar, Jiri Handl, Jan Capek, Pavlina Nyvltova, Erika Rousarova, Miroslav Kubat, Lenka Smid, Jana Vanova, David Malinak, Kamil Musilek, Petr Cesla. Cysteine conjugates of acetaminophen and p-aminophenol are potent inducers of cellular impairment in human proximal tubular kidney HK-2 cells. Archives of Toxicology 2023, 97 (11) , 2943-2954. https://doi.org/10.1007/s00204-023-03569-2
    4. Yue Wu, Wanqi Ma, Wei Liu, Shuping Zhang. Lactate: a pearl dropped in the ocean—an overlooked signal molecule in physiology and pathology. Cell Biology International 2023, 47 (2) , 295-307. https://doi.org/10.1002/cbin.11975
    5. Klaudia Grochowalska, Piotr Pikul, Agnieszka Piwkowska. Insights into the regulation of podocyte and glomerular function by lactate and its metabolic sensor G‐protein‐coupled receptor 81. Journal of Cellular Physiology 2022, 237 (11) , 4097-4111. https://doi.org/10.1002/jcp.30874
    6. John Quiroga, Pablo Alarcón, Carolina Manosalva, Stefanie Teuber, María Daniella Carretta, Rafael Agustín Burgos. d-lactate-triggered extracellular trap formation in cattle polymorphonuclear leucocytes is glucose metabolism dependent. Developmental & Comparative Immunology 2022, 135 , 104492. https://doi.org/10.1016/j.dci.2022.104492
    7. Chenyu Li, Yaquan Liang, Yuan Qiao. Messengers From the Gut: Gut Microbiota-Derived Metabolites on Host Regulation. Frontiers in Microbiology 2022, 13 https://doi.org/10.3389/fmicb.2022.863407
    8. Marilyn E. Morris, Nisha V. Kwatra. MONOCARBOXYLIC ACID TRANSPORTERS. 2022, 143-164. https://doi.org/10.1002/9781119739883.ch7
    9. Fan Zhang, Li Li, Jia-hui Zhao, Xiu-tong Ge, Hui Gao, Tian-zhu Jia. The Effects of Salt-Water Processing of Phellodendri Chinensis Cortex on the Enhancement of Kidney Absorption of the Main Alkaloids. Natural Product Communications 2022, 17 (2) , 1934578X2210762. https://doi.org/10.1177/1934578X221076218
    10. Patrick D. Bosshart, Roch-Philippe Charles, Rachel-Ann A. Garibsingh, Avner Schlessinger, Dimitrios Fotiadis. SLC16 Family: From Atomic Structure to Human Disease. Trends in Biochemical Sciences 2021, 46 (1) , 28-40. https://doi.org/10.1016/j.tibs.2020.07.005
    11. Cortney E. Heim, Megan E. Bosch, Kelsey J. Yamada, Amy L. Aldrich, Sujata S. Chaudhari, David Klinkebiel, Casey M. Gries, Abdulelah A. Alqarzaee, Yixuan Li, Vinai C. Thomas, Edward Seto, Adam R. Karpf, Tammy Kielian. Lactate production by Staphylococcus aureus biofilm inhibits HDAC11 to reprogramme the host immune response during persistent infection. Nature Microbiology 2020, 5 (10) , 1271-1284. https://doi.org/10.1038/s41564-020-0756-3
    12. J Handl, J Čapek, P Majtnerová, J Báčová, T Roušar. The effect of repeated passaging on the susceptibility of human proximal tubular HK-2 cells to toxic compounds. Physiological Research 2020, , 731-738. https://doi.org/10.33549/physiolres.934491
    13. Bernhard Sattler, Mathias Kranz, Barbara Wenzel, Nalin T. Jain, Rareş-Petru Moldovan, Magali Toussaint, Winnie Deuther-Conrad, Friedrich-Alexander Ludwig, Rodrigo Teodoro, Tatjana Sattler, Masoud Sadeghzadeh, Osama Sabri, Peter Brust. Preclinical Incorporation Dosimetry of [18F]FACH—A Novel 18F-Labeled MCT1/MCT4 Lactate Transporter Inhibitor for Imaging Cancer Metabolism with PET. Molecules 2020, 25 (9) , 2024. https://doi.org/10.3390/molecules25092024
    14. Masoud Sadeghzadeh, Barbara Wenzel, Daniel Gündel, Winnie Deuther-Conrad, Magali Toussaint, Rareş-Petru Moldovan, Steffen Fischer, Friedrich-Alexander Ludwig, Rodrigo Teodoro, Shirisha Jonnalagadda, Sravan K. Jonnalagadda, Gerrit Schüürmann, Venkatram R. Mereddy, Lester R. Drewes, Peter Brust. Development of Novel Analogs of the Monocarboxylate Transporter Ligand FACH and Biological Validation of One Potential Radiotracer for Positron Emission Tomography (PET) Imaging. Molecules 2020, 25 (10) , 2309. https://doi.org/10.3390/molecules25102309
    15. Lindsay B. Baker, Anthony S. Wolfe. Physiological mechanisms determining eccrine sweat composition. European Journal of Applied Physiology 2020, 120 (4) , 719-752. https://doi.org/10.1007/s00421-020-04323-7
    16. Melanie A. Felmlee, Robert S. Jones, Vivian Rodriguez-Cruz, Kristin E. Follman, Marilyn E. Morris, . Monocarboxylate Transporters (SLC16): Function, Regulation, and Role in Health and Disease. Pharmacological Reviews 2020, 72 (2) , 466-485. https://doi.org/10.1124/pr.119.018762
    17. Yuchen Sun, Kosuke Saito, Ryota Iiji, Yoshiro Saito. Application of Ion Chromatography Coupled with Mass Spectrometry for Human Serum and Urine Metabolomics. SLAS Discovery 2019, 24 (7) , 778-786. https://doi.org/10.1177/2472555219850082
    18. Piyush Bajaj, Swapan K. Chowdhury, Robert Yucha, Edward J. Kelly, Guangqing Xiao. Emerging Kidney Models to Investigate Metabolism, Transport, and Toxicity of Drugs and Xenobiotics. Drug Metabolism and Disposition 2018, 46 (11) , 1692-1702. https://doi.org/10.1124/dmd.118.082958
    19. Melanie A. Felmlee, Bridget L. Morse, Kristin E. Follman, Marilyn E. Morris. The Drug of Abuse Gamma-Hydroxybutyric Acid Exhibits Tissue-Specific Nonlinear Distribution. The AAPS Journal 2018, 20 (1) https://doi.org/10.1208/s12248-017-0180-7
    20. Rutwij A. Dave, Kristin E. Follman, Marilyn E. Morris. γ-Hydroxybutyric Acid (GHB) Pharmacokinetics and Pharmacodynamics: Semi-Mechanistic and Physiologically Relevant PK/PD Model. The AAPS Journal 2017, 19 (5) , 1449-1460. https://doi.org/10.1208/s12248-017-0111-7
    21. Hoon Choi, Ting Liu, Kavindra Nath, Rong Zhou, I-Wei Chen. Peptide nanoparticle with pH-sensing cargo solubility enhances cancer drug efficiency. Nano Today 2017, 13 , 15-22. https://doi.org/10.1016/j.nantod.2017.02.008
    22. Mariline Gameiro, Renata Silva, Carolina Rocha-Pereira, Helena Carmo, Félix Carvalho, Maria Bastos, Fernando Remião. Cellular Models and In Vitro Assays for the Screening of modulators of P-gp, MRP1 and BCRP. Molecules 2017, 22 (4) , 600. https://doi.org/10.3390/molecules22040600
    23. M. VRBOVÁ, E. ROUŠAROVÁ, L. BRŮČKOVÁ, P. ČESLA, T. ROUŠAR. Characterization of Acetaminophen Toxicity in Human Kidney HK-2 Cells. Physiological Research 2016, , 627-635. https://doi.org/10.33549/physiolres.933158
    24. Martina Vrbová, Eva Dastychová, Tomáš Roušar. RENAL CELL LINES FOR STUDY OF NEPHROTOXICITY IN VITRO. Military Medical Science Letters 2016, 85 (2) , 69-74. https://doi.org/10.31482/mmsl.2016.013
    25. Saurabh Saxena, Dhananjay Shukla, Anju Bansal. Expression of Monocarboxylate Transporter Isoforms in Rat Skeletal Muscle Under Hypoxic Preconditioning and Endurance Training. High Altitude Medicine & Biology 2016, 17 (1) , 32-42. https://doi.org/10.1089/ham.2015.0048
    26. Renuka Sriram, Mark Van Criekinge, Ailin Hansen, Zhen J. Wang, Daniel B. Vigneron, David M. Wilson, Kayvan R. Keshari, John Kurhanewicz. Real‐time measurement of hyperpolarized lactate production and efflux as a biomarker of tumor aggressiveness in an MR compatible 3D cell culture bioreactor. NMR in Biomedicine 2015, 28 (9) , 1141-1149. https://doi.org/10.1002/nbm.3354
    27. Harindarpal S. Gill, Kun-Young Choi, Lakshmi Kammili, Anastas Popratiloff. Rescue of the temperature-sensitive, autosomal-recessive mutation R298S in the sodium-bicarbonate cotransporter NBCe1-A characterized by a weakened dimer and abnormal aggregation. Biochimica et Biophysica Acta (BBA) - General Subjects 2015, 1850 (6) , 1286-1296. https://doi.org/10.1016/j.bbagen.2015.02.014
    28. Pamela V. Chang, Liming Hao, Stefan Offermanns, Ruslan Medzhitov. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proceedings of the National Academy of Sciences 2014, 111 (6) , 2247-2252. https://doi.org/10.1073/pnas.1322269111
    29. Samuel A. Roiko, Nisha Vijay, Melanie A. Felmlee, Marilyn E. Morris. Brain Extracellular γ-hydroxybutyrate Concentrations are Decreased by L-lactate in Rats: Role in the Treatment of Overdoses. Pharmaceutical Research 2013, 30 (5) , 1338-1348. https://doi.org/10.1007/s11095-013-0973-z
    30. Lidia de Bari, Loredana Moro, Salvatore Passarella. Prostate cancer cells metabolize d ‐lactate inside mitochondria via a d ‐lactate dehydrogenase which is more active and highly expressed than in normal cells. FEBS Letters 2013, 587 (5) , 467-473. https://doi.org/10.1016/j.febslet.2013.01.011
    31. Sarah E. Jenkinson, Git W. Chung, Ellen van Loon, Nur S. Bakar, Abigail M. Dalzell, Colin D. A. Brown. The limitations of renal epithelial cell line HK-2 as a model of drug transporter expression and function in the proximal tubule. Pflügers Archiv - European Journal of Physiology 2012, 464 (6) , 601-611. https://doi.org/10.1007/s00424-012-1163-2
    32. Binbing Ling, Fei Peng, Jane Alcorn, Katharina Lohmann, Brian Bandy, Gordon A Zello. D-Lactate altered mitochondrial energy production in rat brain and heart but not liver. Nutrition & Metabolism 2012, 9 (1) https://doi.org/10.1186/1743-7075-9-6
    33. Mingxiang Liao, Cindy Q. Xia. Applications of iRNA Technologies in Drug Transporters and Drug Metabolizing Enzymes. 2012, 513-543. https://doi.org/10.1002/9781118180778.ch34
    34. Samuel A. Roiko, Melanie A. Felmlee, Marilyn E. Morris. Brain Uptake of the Drug of Abuse γ-Hydroxybutyric Acid in Rats. Drug Metabolism and Disposition 2012, 40 (1) , 212-218. https://doi.org/10.1124/dmd.111.041749
    35. Melanie A. Felmlee, Qi Wang, Dapeng Cui, Samuel A. Roiko, Marilyn E. Morris. Mechanistic Toxicokinetic Model for γ-Hydroxybutyric Acid: Inhibition of Active Renal Reabsorption as a Potential Therapeutic Strategy. The AAPS Journal 2010, 12 (3) , 407-416. https://doi.org/10.1208/s12248-010-9197-x
    36. Helen M. Becker, Nilufar Mohebbi, Angelica Perna, Vadivel Ganapathy, Giovambattista Capasso, Carsten A. Wagner. Localization of members of MCT monocarboxylate transporter family Slc16 in the kidney and regulation during metabolic acidosis. American Journal of Physiology-Renal Physiology 2010, 299 (1) , F141-F154. https://doi.org/10.1152/ajprenal.00488.2009
    37. Carlos Luna-Tortós, Maren Fedrowitz, Wolfgang Löscher. Evaluation of transport of common antiepileptic drugs by human multidrug resistance-associated proteins (MRP1, 2 and 5) that are overexpressed in pharmacoresistant epilepsy. Neuropharmacology 2010, 58 (7) , 1019-1032. https://doi.org/10.1016/j.neuropharm.2010.01.007
    38. Wing Ki Lam, Melanie A. Felmlee, Marilyn E. Morris. Monocarboxylate Transporter-Mediated Transport of γ-Hydroxybutyric Acid in Human Intestinal Caco-2 Cells. Drug Metabolism and Disposition 2010, 38 (3) , 441-447. https://doi.org/10.1124/dmd.109.030775
    39. Keisuke Mitsuoka, Yoshiyuki Shirasaka, Akimasa Fukushi, Masanobu Sato, Toshimichi Nakamura, Takeo Nakanishi, Ikumi Tamai. Transport characteristics of L ‐citrulline in renal apical membrane of proximal tubular cells. Biopharmaceutics & Drug Disposition 2009, 30 (3) , 126-137. https://doi.org/10.1002/bdd.653
    40. Haruko Yanase, Kumiko Takebe, Junko Nio-Kobayashi, Hiromi Takahashi-Iwanaga, Toshihiko Iwanaga. Cellular expression of a sodium-dependent monocarboxylate transporter (Slc5a8) and the MCT family in the mouse kidney. Histochemistry and Cell Biology 2008, 130 (5) , 957-966. https://doi.org/10.1007/s00418-008-0490-z
    41. Marilyn E. Morris, Melanie A. Felmlee. Overview of the Proton-coupled MCT (SLC16A) Family of Transporters: Characterization, Function and Role in the Transport of the Drug of Abuse γ-Hydroxybutyric Acid. The AAPS Journal 2008, 10 (2) https://doi.org/10.1208/s12248-008-9035-6
    42. Emilio Lecona, Nieves Olmo, Javier Turnay, Angélica Santiago-Gómez, Isabel López de Silanes, Myriam Gorospe, M. Antonia Lizarbe. Kinetic analysis of butyrate transport in human colon adenocarcinoma cells reveals two different carrier-mediated mechanisms. Biochemical Journal 2008, 409 (1) , 311-320. https://doi.org/10.1042/BJ20070374
    43. Qi Wang, Marilyn E. Morris. The Role of Monocarboxylate Transporter 2 and 4 in the Transport of γ-Hydroxybutyric Acid in Mammalian Cells. Drug Metabolism and Disposition 2007, 35 (8) , 1393-1399. https://doi.org/10.1124/dmd.107.014852
    44. Qi Wang, Ye Lu, Marilyn E. Morris. Monocarboxylate Transporter (MCT) Mediates the Transport of γ-Hydroxybutyrate in Human Kidney HK-2 cells. Pharmaceutical Research 2007, 24 (6) , 1067-1078. https://doi.org/10.1007/s11095-006-9228-6

    Pair your accounts.

    Export articles to Mendeley

    Get article recommendations from ACS based on references in your Mendeley library.

    Pair your accounts.

    Export articles to Mendeley

    Get article recommendations from ACS based on references in your Mendeley library.

    You’ve supercharged your research process with ACS and Mendeley!

    STEP 1:
    Click to create an ACS ID

    Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

    Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

    Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

    MENDELEY PAIRING EXPIRED
    Your Mendeley pairing has expired. Please reconnect