Elsevier

Pharmacological Reports

Volume 71, Issue 1, February 2019, Pages 121-129
Pharmacological Reports

Original article
Identification of competitive inhibitors of the human taurine transporter TauT in a human kidney cell line

https://doi.org/10.1016/j.pharep.2018.10.005Get rights and content

Abstract

Background

The osmolyte and antioxidant taurine plays an important role in regulation of cellular volume, oxidative status and Ca2+-homeostasis. Taurine uptake in human cells is regulated by the Na+- and Cl-dependent taurine transporter TauT. In order to gain deeper structural insights about the substrate binding pocket of TauT, a HEK293 cell line producing a GFP-TauT fusion protein was generated.

Methods

Transport activity was validated using cell-based [3H]-taurine transport assays. We determined the Km and IC50 values of taurine, β-alanine and γ-aminobutyrate. Additionally we were able to identify structurally similar compounds as potential new substrates or inhibitors of the TauT transporter. Substrate induced cytotoxicity was analyzed using a cell viability assay.

Results

In this study we show competitive effects of the 3-pyridinesulfonate, 2-aminoethylhydrogen sulfate, 5-aminovalerate, β-aminobutyrate, piperidine-4-sulfonate, 2-aminoethylphosphate and homotaurine. We demonstrate that taurine uptake can be inhibited by a phosphate. Furthermore our studies revealed that piperidine-4-sulfonate interacts with TauT with a higher affinity than γ-aminobutyrate and imidazole-4-acetate.

Conclusion

We propose that piperidine-4-sulfonate may serve as a potential lead structure for the design of novel drug candidates required for specific modulation of the TauT transporter in therapy of neurodegenerative diseases.

Introduction

The transport and homeostasis of bioactive solutes and small compounds is of fundamental importance in human physiology. The conditionally essential β-amino acid taurine (2-aminoethanesulfonate) is one of the most highly accumulated amino acids in the brain and plays a critical role in neural development [1]. The total taurine content in mammals is derived from two sources: mainly by dietary intake and partly also by taurine biosynthesis in liver and kidney [2]. Taurine is involved in fundamental biological processes. For instance, taurine is an indirect antioxidant and plays a key role in osmoregulation, membrane stabilization and modulation of Ca2+-signaling [3]. In brain and retinal tissues, the function of taurine as antioxidant and osmolyte is essential for cell volume homeostasis. In this context it was shown that the regulatory effect of taurine on cell volume leads to neuroprotection [4]. Taurine deficiency in humans causes development of retinal disorders, which in severe cases can lead to blindness [5], [6]. The function of solute carriers (SLC) in drug absorption and disposition has increased their clinical importance in recent years [7].

Taurine supply is regulated by the taurine transporter TauT (SLC6A6) [8] in combination with the gamma-aminobutyric acid (GABA) transporter GAT2 (SLC6A13) [9] and the proton-coupled amino acid transporter PAT1 (SLC36A1) [10]. The transport activity of TauT is sodium- and chloride dependent with a stoichiometry of 2:1:1 (Na+:Cl: taurine) [11]. As a high-affinity and low-specificity transporter TauT has the capability to transport hypotaurine, β-alanine and γ-aminobutyrate (GABA), apart from taurine [12]. In addition there are several drug candidates that interfere with GABA transporters, but so far there is only little knowledge about the interference of structural analogs with TauT.

In 2016, Rasmussen et al. investigated the influence of basic GABA-analogs like nipecotic acid, guvacine, δ-aminolevulinic acid, vigabatrin und gaboxadol on taurine uptake [13].

The anti-epileptic drug vigabatrin increases the intracellular level of GABA by irreversible inhibition of the GABA-aminotransferase [14]. The intestinal absorption of vigabatrin is mainly regulated by PAT1 with a partial involvement of TauT [15]. Gaboxadol, also known as 4,5,6,7-tetrahydroisoxazolo(5,4-c)pyridine-3-ol (THIP), acts as a GABAA receptor type A (GABAAR) agonist. GABAA receptors are Cl-channels, which form the major inhibitory system in the central nervous system. Gaboxadol showed inhibitory properties on the GABAAR as well as on the TauT-mediated uptake of taurine in SKPT cells and ARPE-19 cells [13], [16].

Valembois et al. demonstrated 2017 that various other GABA analogs (imidazole-4-acetat (I4AA), 4,5,6,7-tetrahydropyrazolo[5,4-c]pyridin-3-ol (Aza-THIP), muscimol and thiomuscimol) also act as competitive inhibitors of taurine transport [16]. In particular the histamine metabolite I4AA significantly inhibits taurine uptake in a concentration-dependent manner. Due to its additional interaction with the GABAAR, they defined I4AA as a new lead structure for the development of new compounds.

In the meantime, Suárez et al. reported guanidinoethyl sulfonate [17] to mimic the effects of taurine on long-term synaptic potentiation. GES, a natural guanidine-taurine analog, causes taurine depletion by acting as a competitive inhibitor of TauT, PAT1 and GAT2 as well as interfering with taurine biosynthesis [18], [19], [20]. In order to analyze the involvement of taurine transport by TauT in relation to PAT1- and GAT2-mediated taurine uptake, TauT was recombinantly overproduced in this study.

To gain a deeper knowledge about the structural requirements of ligands for interaction with the taurine transporter (TauT), we investigated the competitive inhibition by various GABA- and TauT-analogs. Therefore we selected structural similar compounds containing one acidic and one amino head group separated by a carbon chain and determined their inhibitory properties on taurine uptake (Fig. 1). The compounds analyzed in this study were chosen mostly due to their known physiological effects on either humans or other eukaryotes. Our taurine uptake assays using HEK293-cells producing TauT-GFP indicated that homotaurine, ethanolamine-O-sulfate (EOS), piperidine-4-sulfonate (P4S) and 3-pyridinesulfonate (PYR) act as taurine analogs; 5-aminovalerate (5AVA) and β-aminobutyrate (BABA) as GABA analogs and 2-aminoethylphosphate (2AEP) as competitive inhibitors for TauT with a phosphate group. Here we report a robust in vitro model for studying TauT and the inhibitory potencies of substrate analogs on taurine uptake.

Section snippets

Chemicals

N-Methyltaurine (PubChem CID: 7882) was purchased from Santa Cruz Biotechnology (Dallas, TX, USA) and sodium dodecyl sulphate (PubChem CID: 3423265) from Carl ROTH (Karlsruhe, Germany). Taurine (PubChem CID: 1123), β-alanine (PubChem CID: 239), γ-aminobutyrate (PubChem CID: 119), hypotaurine (PubChem CID: 107812), 3-pyridinesulfonate (PubChem CID: 23677935), DL-homocysteic acid (PubChem CID: 92117), 2-Aminoethylhydrogen sulfate (PubChem CID: 70223), 5-Aminovalerate (PubChem CID: 138),

Impact of incubation time on taurine uptake

In order to increase the TauT-mediated uptake signals, TauT-GFP was produced in a Flp-In™ T-REx™ 293 cell line. SLC6A6-GFP gene expression was induced with doxycycline hydrochloride, instead of up regulation of SLC6A6 gene expression by hyperosmotic conditions [24], was chosen to avoid effects by altered osmolality. GFP-Fusion was chosen for a rapid evaluation of protein production in stably transfected cell line.

Before investigation of taurine uptake, TauT-GFP production in Flp-In™ T-REx™ 293

Discussion

In the present work we investigated the effect of GABA- and taurine-analogs on 3H-taurine uptake by the taurine transporter TauT, produced in the mammalian kidney cell line Flp-In™ T-REx™ 293. Our key findings reveal that several substrate analogs inhibit taurine transport to a large extent without affecting cell viability.

Conclusion

P4S, 2AEP, 5AVA, EOS, homotaurine, PYR and BABA show affinities for TauT below or in the same range as the affinity of GABA. However, additional experiments are necessary to determine the mechanism of interaction of compounds with TauT as inhibitor or alternative substrates. Improved inhibitors are required for treatment of TauT mediated dysregulation, which is directly connected to the development of retinal blindness [1,6] and renal diseases [3]. Furthermore, TauT mediated anti-oxidation and

Acknowledgements

This work was supported by the Max Planck Society and by the Deutsche Forschungsgemeinschaft (the SFB807 and the cluster of excellence “macromolecular complexes” Frankfurt). We would like to thank Schara Safarian for helpful discussions and proof reading of the manuscript.

References (42)

  • D.L. Martin et al.

    High affinity transport of taurine and beta-alanine and low affinity transport of gamma-aminobutyric acid by a single transport system in cultured glioma cells

    J Biol Chem

    (1979)
  • L. O’Flaherty et al.

    Dexamethasone and lipopolysaccharide regulation of taurine transport in Caco-2 cells

    J Surg Res

    (1997)
  • T. Yahara et al.

    Hypertonicity enhances GABA uptake by cultured rat retinal capillary endothelial cells

    Drug Metab Pharmacokinet

    (2010)
  • S. Kato et al.

    Sensitive and insensitive states of cultured glioma cells to glutamate damage

    Brain Res.

    (1984)
  • A.B. Pramod et al.

    SLC6 transporters: structure, function, regulation, disease association and therapeutics

    Mol Aspects Med

    (2013)
  • R.J. Huxtable

    Physiological actions of taurine

    Physiol Rev

    (1992)
  • K.C. Hayes

    Taurine nutrition

    Nutr Res Rev

    (1988)
  • H.S. Geggel et al.

    Nutritional requirement for taurine in patients receiving long-term parenteral nutrition

    N Engl J Med

    (1985)
  • B. Heller-Stilb et al.

    Disruption of the taurine transporter gene (taut) leads to retinal degeneration in mice

    FASEB J

    (2002)
  • C. International Transporter et al.

    Membrane transporters in drug development

    Nat Rev Drug Discov

    (2010)
  • Q.R. Liu et al.

    Cloning and expression of a cDNA encoding the transporter of taurine and beta-alanine in mouse brain

    Proc Natl Acad Sci U S A

    (1992)
  • Cited by (7)

    • Homotaurine ameliorates the core ASD symptomatology in VPA rats through GABAergic signaling: Role of GAD67

      2022, Brain Research Bulletin
      Citation Excerpt :

      Homotaurine leads to improvements in the biochemical parameters (decrease in oxidative stress; decreased inflammatory cytokines) in the PFC as well as the cerebellum. The anti-inflammatory and antioxidant effect of homotaurine was demonstrated in multiple diseases (Bossù et al., 2018; Tian et al., 2021; Filippelli et al., 2020; Richter et al., 2019; Patel and Lau-Cam, 2017; Adedara et al., 2017). Also, the histopathological findings of the VPA group showed an increase in the neuronal injury score in PFC and Purkinje cell loss in the cerebellum.

    • Identification and pharmacological modification of resistance mechanisms to protoporphyrin-mediated photodynamic therapy in human cutaneous squamous cell carcinoma cell lines

      2022, Photodiagnosis and Photodynamic Therapy
      Citation Excerpt :

      This inconsistency between expression and inhibitory effect could also be observed when cells were inhibited with GAT-3 inhibitor (S)-SNAP-5114. Notably, GABA shows affinity to TauT with Km = 1.46 - 2.4 mM [63,64] and IC50 = 0.72 - 1.014 mM [62,64]. Considering the IC50 of GABA in cSCC cell lines (0.23 - 0.52 mM), this is within the range of the Km/IC50 of GABA to TauT as stated above.

    View all citing articles on Scopus
    View full text