1
|
Carretero VJ, Álvarez-Merz I, Hernández-Campano J, Kirov SA, Hernández-Guijo JM. Targeting harmful effects of non-excitatory amino acids as an alternative therapeutic strategy to reduce ischemic damage. Neural Regen Res 2025; 20:2454-2463. [PMID: 39314160 PMCID: PMC11801293 DOI: 10.4103/nrr.nrr-d-24-00536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/16/2024] [Accepted: 08/14/2024] [Indexed: 09/25/2024] Open
Abstract
The involvement of the excitatory amino acids glutamate and aspartate in cerebral ischemia and excitotoxicity is well-documented. Nevertheless, the role of non-excitatory amino acids in brain damage following a stroke or brain trauma remains largely understudied. The release of amino acids by necrotic cells in the ischemic core may contribute to the expansion of the penumbra. Our findings indicated that the reversible loss of field excitatory postsynaptic potentials caused by transient hypoxia became irreversible when exposed to a mixture of just four non-excitatory amino acids (L-alanine, glycine, L-glutamine, and L-serine) at their plasma concentrations. These amino acids induce swelling in the somas of neurons and astrocytes during hypoxia, along with permanent dendritic damage mediated by N-methyl-D-aspartate receptors. Blocking N-methyl-D-aspartate receptors prevented neuronal damage in the presence of these amino acids during hypoxia. It is likely that astroglial swelling caused by the accumulation of these amino acids via the alanine-serine-cysteine transporter 2 exchanger and system N transporters activates volume-regulated anion channels, leading to the release of excitotoxins and subsequent neuronal damage through N-methyl-D-aspartate receptor activation. Thus, previously unrecognized mechanisms involving non-excitatory amino acids may contribute to the progression and expansion of brain injury in neurological emergencies such as stroke and traumatic brain injury. Understanding these pathways could highlight new therapeutic targets to mitigate brain injury.
Collapse
Affiliation(s)
| | - Iris Álvarez-Merz
- Department of Pharmacology and Therapeutic, School of Medicine, Univ. Autónoma de Madrid, Madrid, Spain
- Ramón y Cajal Institute for Health Research (IRYCIS), Neurobiology-Research Service, Hospital Ramón y Cajal, Madrid, Spain
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jorge Hernández-Campano
- Department of Pharmacology and Therapeutic, School of Medicine, Univ. Autónoma de Madrid, Madrid, Spain
| | - Sergei A. Kirov
- Department of Neuroscience and Regenerative Medicine & Department of Neurosurgery, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Jesús M. Hernández-Guijo
- Department of Pharmacology and Therapeutic, School of Medicine, Univ. Autónoma de Madrid, Madrid, Spain
- Ramón y Cajal Institute for Health Research (IRYCIS), Neurobiology-Research Service, Hospital Ramón y Cajal, Madrid, Spain
| |
Collapse
|
2
|
Sharma S, Rodems BJ, Baker CD, Kaszuba CM, Franco EI, Smith BR, Ito T, Swovick K, Welle K, Zhang Y, Rock P, Chaves FA, Ghaemmaghami S, Calvi LM, Ganguly A, Burack WR, Becker MW, Liesveld JL, Brookes PS, Munger JC, Jordan CT, Ashton JM, Bajaj J. Taurine from tumour niche drives glycolysis to promote leukaemogenesis. Nature 2025:10.1038/s41586-025-09018-7. [PMID: 40369079 DOI: 10.1038/s41586-025-09018-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 04/14/2025] [Indexed: 05/16/2025]
Abstract
Signals from the microenvironment are known to be critical for development, stem cell self-renewal and oncogenic progression. Although some niche-driven signals that promote cancer progression have been identified1-5, concerted efforts to map disease-relevant microenvironmental ligands of cancer stem cell receptors have been lacking. Here, we use temporal single-cell RNA-sequencing (scRNA-seq) to identify molecular cues from the bone marrow stromal niche that engage leukaemia stem-enriched cells (LSCs) during oncogenic progression. We integrate these data with our human LSC RNA-seq and in vivo CRISPR screen of LSC dependencies6 to identify LSC-niche interactions that are essential for leukaemogenesis. These analyses identify the taurine-taurine transporter (TAUT) axis as a critical dependency of aggressive myeloid leukaemias. We find that cysteine dioxygenase type 1 (CDO1)-driven taurine biosynthesis is restricted to osteolineage cells, and increases during myeloid disease progression. Blocking CDO1 expression in osteolineage cells impairs LSC growth and improves survival outcomes. Using TAUT genetic loss-of-function mouse models and patient-derived acute myeloid leukaemia (AML) cells, we show that TAUT inhibition significantly impairs in vivo myeloid leukaemia progression. Consistent with elevated TAUT expression in venetoclax-resistant AML, TAUT inhibition synergizes with venetoclax to block the growth of primary human AML cells. Mechanistically, our multiomic approaches indicate that the loss of taurine uptake inhibits RAG-GTP dependent mTOR activation and downstream glycolysis. Collectively, our work establishes the temporal landscape of stromal signals during leukaemia progression and identifies taurine as a key regulator of myeloid malignancies.
Collapse
Affiliation(s)
- Sonali Sharma
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Benjamin J Rodems
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Cameron D Baker
- Genomics Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Christina M Kaszuba
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Edgardo I Franco
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Bradley R Smith
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA
| | - Takashi Ito
- Department of Bioscience and Technology, Graduate School of Bioscience and Technology, Fukui Prefectural University, Fukui, Japan
| | - Kyle Swovick
- Mass Spectrometry Resource Laboratory, University of Rochester, Rochester, NY, USA
| | - Kevin Welle
- Mass Spectrometry Resource Laboratory, University of Rochester, Rochester, NY, USA
| | - Yi Zhang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Philip Rock
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Francisco A Chaves
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Sina Ghaemmaghami
- Mass Spectrometry Resource Laboratory, University of Rochester, Rochester, NY, USA
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Laura M Calvi
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Archan Ganguly
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
| | - W Richard Burack
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Michael W Becker
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Jane L Liesveld
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Paul S Brookes
- Department of Anesthesiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Joshua C Munger
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, USA
| | - Craig T Jordan
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - John M Ashton
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Genomics Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Jeevisha Bajaj
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA.
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
3
|
Livengood EJ, Fong RAMV, Pratt AM, Alinskas VO, Van Gorder G, Mezzio M, Mulligan ME, Voura EB. Taurine stimulation of planarian motility: a role for the dopamine receptor pathway. PeerJ 2024; 12:e18671. [PMID: 39655335 PMCID: PMC11627082 DOI: 10.7717/peerj.18671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024] Open
Abstract
Taurine, a normal dietary component that is found in many tissues, is considered important for a number of physiological processes. It is thought to play a particular role in eye development and in the maturation of both the muscular and nervous systems, leading to its suggested use as a therapeutic for Alzheimer's and Parkinson's diseases. Taurine increases metabolism and has also been touted as a weight loss aid. Due to its possible benefits to health and development, taurine is added as a supplement to a wide array of products, including infant formula and energy drinks. Despite its pervasive use as a nutritional additive and implied physiological actions, there is little consensus on how taurine functions. This is likely because, mechanistically, taurine has been demonstrated to affect multiple metabolic pathways. Simple models and straightforward assay systems are required to make headway in understanding this complexity. We chose to begin this work using the planarian because these animals have basic, well-understood muscular and nervous systems and are the subjects of many well-tested assays examining how their physiology is influenced by exposure to various environmental, nutritional, and therapeutic agents. We used a simple behavioral assay, the planarian locomotor velocity test (pLmV), to gain insight into the stimulant properties of taurine. Using this assay, we observed that taurine is a mild stimulant that is not affected by sugars or subject to withdrawal. We also provide evidence that taurine makes use of the dopamine D1 receptor to mediate this stimulant effect. Given the pervasiveness of taurine in many commercial products, our findings using the planarian system provide needed insight into the stimulant properties of taurine that should be considered when adding it to the diet.
Collapse
Affiliation(s)
- Elisa J. Livengood
- Division of Environmental and Renewable Resources, State University of New York (SUNY) at Morrisville, Morrisville, New York, United States
| | - Robyn A. M. V. Fong
- Division of Environmental and Renewable Resources, State University of New York (SUNY) at Morrisville, Morrisville, New York, United States
| | - Angela M. Pratt
- Division of Environmental and Renewable Resources, State University of New York (SUNY) at Morrisville, Morrisville, New York, United States
| | - Veronika O. Alinskas
- Division of Environmental and Renewable Resources, State University of New York (SUNY) at Morrisville, Morrisville, New York, United States
| | - Grace Van Gorder
- Division of Environmental and Renewable Resources, State University of New York (SUNY) at Morrisville, Morrisville, New York, United States
| | - Michael Mezzio
- Department of Math and Science, Dominican University, Orangeburg, New York, United States
| | - Margaret E. Mulligan
- Department of Math and Science, Dominican University, Orangeburg, New York, United States
| | - Evelyn B. Voura
- Crouse Neuroscience Institute, Crouse Health at Crouse Hospital, Crouse Medical Practice, Syracuse, New York, United States
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, United States
| |
Collapse
|
4
|
Álvarez-Merz I, Muñoz MD, Hernández-Guijo JM, Solís JM. Identification of Non-excitatory Amino Acids and Transporters Mediating the Irreversible Synaptic Silencing After Hypoxia. Transl Stroke Res 2024; 15:1070-1087. [PMID: 37755645 DOI: 10.1007/s12975-023-01192-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/03/2023] [Accepted: 09/01/2023] [Indexed: 09/28/2023]
Abstract
The contribution of excitatory amino acids (AA) to ischemic brain injury has been widely described. In addition, we reported that a mixture of non-excitatory AA at plasmatic concentrations turns irreversible the depression of synaptic transmission caused by hypoxia. Here, we describe that the presence of seven non-excitatory AA (L-alanine, L-glutamine, glycine, L-histidine, L-serine, taurine, and L-threonine) during hypoxia provokes an irreversible neuronal membrane depolarization, after an initial phase of hyperpolarization. The collapse of the membrane potential correlates with a great increase in fiber volley amplitude. Nevertheless, we show that the presence of all seven AA is not necessary to cause the irreversible loss of fEPSP after hypoxia and that the minimal combination of AA able to provoke a solid, replicable effect is the mixture of L-alanine, glycine, L-glutamine, and L-serine. Additionally, L-glutamine seems necessary but insufficient to induce these harmful effects. We also prove that the deleterious effects of the AA mixtures on field potentials during hypoxia depend on both the identity and concentration of the individual AA in the mixture. Furthermore, we find that the accumulation of AA in the whole slice does not determine the outcome caused by the AA mixtures on the synaptic transmission during hypoxia. Finally, results obtained using pharmacological inhibitors and specific substrates of AA transporters suggest that system N and the alanine-serine-cysteine transporter 2 (ASCT2) participate in the non-excitatory AA-mediated deleterious effects during hypoxia. Thus, these AA transporters might represent therapeutical targets for the treatment of brain ischemia.
Collapse
Affiliation(s)
- Iris Álvarez-Merz
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Instituto Teófilo Hernando, Universidad Autónoma de Madrid, Madrid, Spain
- Servicio de Neurobiología-Investigación, Hospital Ramón y Cajal, IRYCIS, Madrid, Spain
| | - María-Dolores Muñoz
- Servicio de Neurobiología-Investigación, Hospital Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Jesús M Hernández-Guijo
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Instituto Teófilo Hernando, Universidad Autónoma de Madrid, Madrid, Spain.
- Servicio de Neurobiología-Investigación, Hospital Ramón y Cajal, IRYCIS, Madrid, Spain.
| | - José M Solís
- Servicio de Neurobiología-Investigación, Hospital Ramón y Cajal, IRYCIS, Madrid, Spain
| |
Collapse
|
5
|
Kumrungsee T. Is hepatic GABA transaminase a promising target for obesity and epilepsy treatments? Biosci Biotechnol Biochem 2024; 88:839-849. [PMID: 38749549 DOI: 10.1093/bbb/zbae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 05/05/2024] [Indexed: 07/23/2024]
Abstract
γ-Aminobutyric acid (GABA) transaminase (GABA-T) is a GABA-degrading enzyme that plays an essential role in regulating GABA levels and maintaining supplies of GABA. Although GABA in the mammalian brain was discovered 70 years ago, research on GABA and GABA-T has predominantly focused on the brain. Notwithstanding the high activity and expression of GABA-T in the liver, the exact functions of GABA-T in the liver remain unknown. This article reviews the up-to-date information on GABA-T in the liver. It presents recent findings on the role of liver GABA-T in food intake suppression and appetite regulation. Finally, the potential functions of liver GABA-T in other neurological diseases, natural GABA-T inhibitors, and future perspectives in this research area are discussed.
Collapse
Affiliation(s)
- Thanutchaporn Kumrungsee
- Program of Food and AgriLife Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
- Smart Agriculture, Graduate School of Innovation and Practice for Smart Society, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
6
|
Rodems BJ, Sharma S, Baker CD, Kaszuba CM, Ito T, Liesveld JL, Calvi LM, Becker MW, Jordan CT, Ashton JM, Bajaj J. Temporal Single Cell Analysis of Leukemia Microenvironment Identifies Taurine-Taurine Transporter Axis as a Key Regulator of Myeloid Leukemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.11.593633. [PMID: 38798540 PMCID: PMC11118281 DOI: 10.1101/2024.05.11.593633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Signals from the microenvironment are known to be critical for development, sustaining adult stem cells, and for oncogenic progression. While candidate niche-driven signals that can promote cancer progression have been identified1-6, concerted efforts to comprehensively map microenvironmental ligands for cancer stem cell specific surface receptors have been lacking. Here, we use temporal single cell RNA-sequencing to identify molecular cues from the bone marrow stromal niche that engage leukemia stem cells (LSC) during oncogenic progression. We integrate these data with our RNA-seq analysis of human LSCs from distinct aggressive myeloid cancer subtypes and our CRISPR based in vivo LSC dependency map7 to develop a temporal receptor-ligand interactome essential for disease progression. These analyses identify the taurine transporter (TauT)-taurine axis as a critical dependency of myeloid malignancies. We show that taurine production is restricted to the osteolineage population during cancer initiation and expansion. Inhibiting taurine synthesis in osteolineage cells impairs LSC growth and survival. Our experiments with the TauT genetic loss of function murine model indicate that its loss significantly impairs the progression of aggressive myeloid leukemias in vivo by downregulating glycolysis. Further, TauT inhibition using a small molecule strongly impairs the growth and survival of patient derived myeloid leukemia cells. Finally, we show that TauT inhibition can synergize with the clinically approved oxidative phosphorylation inhibitor venetoclax8, 9 to block the growth of primary human leukemia cells. Given that aggressive myeloid leukemias continue to be refractory to current therapies and have poor prognosis, our work indicates targeting the taurine transporter may be of therapeutic significance. Collectively, our data establishes a temporal landscape of stromal signals during cancer progression and identifies taurine-taurine transporter signaling as an important new regulator of myeloid malignancies.
Collapse
Affiliation(s)
- Benjamin J. Rodems
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Sonali Sharma
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Cameron D. Baker
- Genomics Research Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Christina M. Kaszuba
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA
| | - Takashi Ito
- Department of Bioscience and Technology, Graduate School of Bioscience and Technology, Fukui Prefectural University, Fukui, Japan
| | - Jane L. Liesveld
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Division of Hematology and Oncology, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Laura M. Calvi
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Michael W. Becker
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Division of Hematology and Oncology, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Craig T. Jordan
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - John M. Ashton
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Genomics Research Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jeevisha Bajaj
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
7
|
Nishimura T, Araki H, Higuchi K, Noguchi S, Saito K, Hara K, Yagishita H, Akashi R, Obata S, Tomi M. Involvement of GAT2/Slc6a13 in hypotaurine uptake at fetal-facing plasma membrane of syncytiotrophoblasts at mid-to-late gestation in rats and mice. Placenta 2024; 147:59-67. [PMID: 38325050 DOI: 10.1016/j.placenta.2024.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/11/2024] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Abstract
INTRODUCTION Hypotaurine, a precursor to taurine, is known for its antioxidant properties and is prominently present in fetal plasma and the placenta. Our previous research revealed that ezrin-knockout mice experience fetal growth retardation, coinciding with reduced hypotaurine levels in fetal plasma. This study aims to elucidate the expression and role of hypotaurine transporters within the placenta. METHODS We employed quantitative RT-PCR to measure mRNA expression of GAT transporter family members in the placenta during mid-to-late gestation. LC/MS/MS was used to analyze the distribution of hypotaurine in different placental subregions. Immunohistochemistry was utilized to examine the localization of GAT2 in mice. Placental hypotaurine uptake from fetal circulation was studied via umbilical perfusion in rats. RESULTS Among hypotaurine transporters, GAT2 exhibited increased mRNA and protein expression in murine placenta during mid-to-late gestation. Notably, GAT2/Slc6a13 mRNA and hypotaurine were most concentrated in the labyrinth of murine placenta. In contrast, enzymes responsible for hypotaurine synthesis, such as cysteine dioxygenase, cysteine sulfinic acid decarboxylase, and 2-aminoethanethiol dioxygenase, showed minimal expression in the labyrinth. These findings suggest that GAT2 is a key determinant of hypotaurine levels in the placental labyrinth. Immunohistochemical examination unveiled that GAT2 was predominantly localized on the fetal-facing plasma membrane within syncytiotrophoblasts, which co-localized with ezrin. In rat umbilical perfusion experiments, the GAT2/3 and TauT inhibitor, SNAP-5114, significantly reduced hypotaurine extraction from fetal circulation to the placenta. DISCUSSION The results suggest that GAT2 plays a pivotal role in the concentrative uptake of hypotaurine from fetal plasma within syncytiotrophoblasts of the placenta.
Collapse
Affiliation(s)
- Tomohiro Nishimura
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Hikari Araki
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Kei Higuchi
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Saki Noguchi
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Kei Saito
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Kanako Hara
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Haruya Yagishita
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Risa Akashi
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Sakiko Obata
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Masatoshi Tomi
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan.
| |
Collapse
|
8
|
Turbitt J, Moffett RC, Brennan L, Johnson PRV, Flatt PR, McClenaghan NH, Tarasov AI. Molecular determinants and intracellular targets of taurine signalling in pancreatic islet β-cells. Acta Physiol (Oxf) 2024; 240:e14101. [PMID: 38243723 DOI: 10.1111/apha.14101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 12/26/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024]
Abstract
AIM Despite its abundance in pancreatic islets of Langerhans and proven antihyperglycemic effects, the impact of the essential amino acid, taurine, on islet β-cell biology has not yet received due consideration, which prompted the current studies exploring the molecular selectivity of taurine import into β-cells and its acute and chronic intracellular interactions. METHODS The molecular aspects of taurine transport were probed by exposing the clonal pancreatic BRIN BD11 β-cells and primary mouse and human islets to a range of the homologs of the amino acid (assayed at 2-20 mM), using the hormone release and imaging of intracellular signals as surrogate read-outs. Known secretagogues were employed to profile the interaction of taurine with acute and chronic intracellular signals. RESULTS Taurine transporter TauT was expressed in the islet β-cells, with the transport of taurine and homologs having a weak sulfonate specificity but significant sensitivity to the molecular weight of the transporter. Taurine, hypotaurine, homotaurine, and β-alanine enhanced insulin secretion in a glucose-dependent manner, an action potentiated by cytosolic Ca2+ and cAMP. Acute and chronic β-cell insulinotropic effects of taurine were highly sensitive to co-agonism with GLP-1, forskolin, tolbutamide, and membrane depolarization, with an unanticipated indifference to the activation of PKC and CCK8 receptors. Pre-culturing with GLP-1 or KATP channel inhibitors sensitized or, respectively, desensitized β-cells to the acute taurine stimulus. CONCLUSION Together, these data demonstrate the pathways whereby taurine exhibits a range of beneficial effects on insulin secretion and β-cell function, consistent with the antidiabetic potential of its dietary low-dose supplementation.
Collapse
Affiliation(s)
- Julie Turbitt
- School of Biomedical Sciences, Ulster University, Coleraine, UK
| | | | - Lorraine Brennan
- UCD Institute of Food and Health, UCD School of Agriculture and Food Science, University College Dublin, Dublin 4, Republic of Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Republic of Ireland
| | - Paul R V Johnson
- Nuffield Department of Surgical Sciences, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- Oxford Biomedical Research Centre (OxBRC), Oxford, UK
| | - Peter R Flatt
- School of Biomedical Sciences, Ulster University, Coleraine, UK
| | - Neville H McClenaghan
- School of Biomedical Sciences, Ulster University, Coleraine, UK
- Department of Life Sciences, Atlantic Technological University, Sligo, Republic of Ireland
| | | |
Collapse
|
9
|
Kalbe C, Metzger K, Gariépy C, Palin MF. Effect of muscle fibre types and carnosine levels on the expression of carnosine-related genes in pig skeletal muscle. Histochem Cell Biol 2023; 160:63-77. [PMID: 37171629 PMCID: PMC10313551 DOI: 10.1007/s00418-023-02193-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2023] [Indexed: 05/13/2023]
Abstract
It is generally accepted that carnosine (β-alanyl-L-histidine) content is higher in glycolytic than in oxidative muscle fibres, but the underlying mechanisms responsible for this difference remain to be elucidated. A first study to better understand potential mechanisms involved was undertaken (1) to determine whether differences in the expression of carnosine-related enzymes (CARNS1, CNDP2) and transporters (SLC6A6, SLC15A3, SLC15A4, SLC36A1) exist between oxidative and glycolytic myofibres and (2) to study the effect of carnosine on myoblast proliferative growth and on carnosine-related gene expression in cultured myoblasts isolated from glycolytic and oxidative muscles. Immunohistochemistry analyses were conducted to determine the cellular localization of carnosine-related proteins. Laser-capture microdissection and qPCR analyses were performed to measure the expression of carnosine-related genes in different myofibres isolated from the longissimus dorsi muscle of ten crossbred pigs. Myogenic cells originating from glycolytic and oxidative muscles were cultured to assess the effect of carnosine (0, 10, 25 and 50 mM) on their proliferative growth and on carnosine-related gene expression. The mRNA abundance of CNDP2 and of the studied carnosine transporters was higher in oxidative than in glycolytic myofibres. Since carnosine synthase (CARNS1) mRNA abundance was not affected by either the fibre type or the addition of carnosine to myoblasts, its transcriptional regulation would not be the main process by which carnosine content differences are determined in oxidative and glycolytic muscles. The addition of carnosine to myoblasts leading to a dose-dependent increase in SLC15A3 transcripts, however, suggests a role for this transporter in carnosine uptake and/or efflux to maintain cellular homeostasis.
Collapse
Affiliation(s)
- Claudia Kalbe
- Research Institute for Farm Animal Biology, Institute of Muscle Biology and Growth, Dummerstorf, Germany
| | - Katharina Metzger
- Research Institute for Farm Animal Biology, Institute of Behavioural Physiology, Dummerstorf, Germany
| | - Claude Gariépy
- Agriculture and Agri-Food Canada, St-Hyacinthe Research and Development Centre, St-Hyacinthe, QC, Canada
| | - Marie-France Palin
- Agriculture and Agri-Food Canada, Sherbrooke Research and Development Centre, Sherbrooke, QC, Canada.
| |
Collapse
|
10
|
Sinn M, Stanoppi M, Hauth F, Fleming JR, Funck D, Mayans O, Hartig JS. Guanidino acid hydrolysis by the human enzyme annotated as agmatinase. Sci Rep 2022; 12:22088. [PMID: 36543883 PMCID: PMC9772407 DOI: 10.1038/s41598-022-26655-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Guanidino acids such as taurocyamine, guanidinobutyrate, guanidinopropionate, and guanidinoacetate have been detected in humans. However, except for guanidionacetate, which is a precursor of creatine, their metabolism and potential functions remain poorly understood. Agmatine has received considerable attention as a potential neurotransmitter and the human enzyme so far annotated as agmatinase (AGMAT) has been proposed as an important modulator of agmatine levels. However, conclusive evidence for the assigned enzymatic activity is lacking. Here we show that AGMAT hydrolyzed a range of linear guanidino acids but was virtually inactive with agmatine. Structural modelling and direct biochemical assays indicated that two naturally occurring variants differ in their substrate preferences. A negatively charged group in the substrate at the end opposing the guanidine moiety was essential for efficient catalysis, explaining why agmatine was not hydrolyzed. We suggest to rename AGMAT as guanidino acid hydrolase (GDAH). Additionally, we demonstrate that the GDAH substrates taurocyamine, guanidinobutyrate and guanidinopropionate were produced by human glycine amidinotransferase (GATM). The presented findings show for the first time an enzymatic activity for GDAH/AGMAT. Since agmatine has frequently been proposed as an endogenous neurotransmitter, the current findings clarify important aspects of the metabolism of agmatine and guanidino acid derivatives in humans.
Collapse
Affiliation(s)
- Malte Sinn
- grid.9811.10000 0001 0658 7699Department of Chemistry, University of Konstanz, Konstanz, Germany
| | - Marco Stanoppi
- grid.9811.10000 0001 0658 7699Department of Chemistry, University of Konstanz, Konstanz, Germany
| | - Franziskus Hauth
- grid.9811.10000 0001 0658 7699Department of Chemistry, University of Konstanz, Konstanz, Germany ,grid.9811.10000 0001 0658 7699Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, Konstanz, Germany
| | - Jennifer R. Fleming
- grid.9811.10000 0001 0658 7699Department of Biology, University of Konstanz, Konstanz, Germany
| | - Dietmar Funck
- grid.9811.10000 0001 0658 7699Department of Chemistry, University of Konstanz, Konstanz, Germany
| | - Olga Mayans
- grid.9811.10000 0001 0658 7699Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, Konstanz, Germany ,grid.9811.10000 0001 0658 7699Department of Biology, University of Konstanz, Konstanz, Germany
| | - Jörg S. Hartig
- grid.9811.10000 0001 0658 7699Department of Chemistry, University of Konstanz, Konstanz, Germany ,grid.9811.10000 0001 0658 7699Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, Konstanz, Germany
| |
Collapse
|
11
|
Multi-target action of β-alanine protects cerebellar tissue from ischemic damage. Cell Death Dis 2022; 13:747. [PMID: 36038575 PMCID: PMC9424312 DOI: 10.1038/s41419-022-05159-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/26/2022] [Accepted: 08/03/2022] [Indexed: 01/21/2023]
Abstract
Brain ischemic stroke is among the leading causes of death and long-term disability. New treatments that alleviate brain cell damage until blood supply is restored are urgently required. The emerging focus of anti-stroke strategies has been on blood-brain-barrier permeable drugs that exhibit multiple sites of action. Here, we combine single-cell electrophysiology with live-cell imaging to find that β-Alanine (β-Ala) protects key physiological functions of brain cells that are exposed to acute stroke-mimicking conditions in ex vivo brain preparations. β-Ala exerts its neuroprotective action through several distinct pharmacological mechanisms, none of which alone could reproduce the neuroprotective effect. Since β-Ala crosses the blood-brain barrier and is part of a normal human diet, we suggest that it has a strong potential for acute stroke treatment and facilitation of recovery.
Collapse
|
12
|
Tochitani S. Taurine: A Maternally Derived Nutrient Linking Mother and Offspring. Metabolites 2022; 12:metabo12030228. [PMID: 35323671 PMCID: PMC8954275 DOI: 10.3390/metabo12030228] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 12/11/2022] Open
Abstract
Mammals can obtain taurine from food and synthesize it from sulfur-containing amino acids. Mammalian fetuses and infants have little ability to synthesize taurine. Therefore, they are dependent on taurine given from mothers either via the placenta or via breast milk. Many lines of evidence demonstrate that maternally derived taurine is essential for offspring development, shaping various traits in adults. Various environmental factors, including maternal obesity, preeclampsia, and undernutrition, can affect the efficacy of taurine transfer via either the placenta or breast milk. Thus, maternally derived taurine during the perinatal period can influence the offspring’s development and even determine health and disease later in life. In this review, I will discuss the biological function of taurine during development and the regulatory mechanisms of taurine transport from mother to offspring. I also refer to the possible environmental factors affecting taurine functions in mother-offspring bonding during perinatal periods. The possible functions of taurine as a determinant of gut microbiota and in the context of the Developmental Origins of Health and Disease (DOHaD) hypothesis will also be discussed.
Collapse
Affiliation(s)
- Shiro Tochitani
- Division of Health Science, Graduate School of Health Science, Suzuka University of Medical Science, Suzuka 513-8670, Japan; ; Tel.: +81-59-373-7069
- Department of Radiological Technology, Faculty of Health Science, Suzuka University of Medical Science, Suzuka 513-8670, Japan
- Center for Preventive Medical Sciences, Chiba University, Chiba 263-8522, Japan
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| |
Collapse
|
13
|
Jomura R, Akanuma SI, Tachikawa M, Hosoya KI. SLC6A and SLC16A family of transporters: Contribution to transport of creatine and creatine precursors in creatine biosynthesis and distribution. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:183840. [PMID: 34921896 DOI: 10.1016/j.bbamem.2021.183840] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/06/2021] [Accepted: 12/09/2021] [Indexed: 11/16/2022]
Abstract
Creatine (Cr) is needed to maintain high energy levels in cells. Since Cr plays reportedly a critical role in neurodevelopment and the immune system, Cr dynamics should be strictly regulated to control these physiological events. This review focuses on the role of transporters that recognize Cr and/or Cr precursors. Our previous studies revealed physiological roles of SLC6A and SLC16A family transporters in Cr dynamics. Creatine transporter (CRT/SLC6A8) contributes to the influx transport of Cr in Cr distribution. γ-Aminobutyric acid transporter 2 (GAT2/SLC6A13) mediates incorporation of guanidinoacetate (GAA), a Cr precursor, in the process of Cr biosynthesis. Monocarboxylate transporter 12 (MCT12/SLC16A12) functions as an efflux transporter for Cr and GAA, and contributes to the process of Cr biosynthesis. Accordingly, the SLC6A and SLC16A family of transporters play important roles in the process of Cr biosynthesis and distribution via permeation of Cr and Cr precursors across the plasma membrane.
Collapse
Affiliation(s)
- Ryuta Jomura
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Shin-Ichi Akanuma
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Masanori Tachikawa
- Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Shomachi, Tokushima 770-8505, Japan.
| | - Ken-Ichi Hosoya
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| |
Collapse
|
14
|
Involvement of TauT/SLC6A6 in Taurine Transport at the Blood-Testis Barrier. Metabolites 2022; 12:metabo12010066. [PMID: 35050188 PMCID: PMC8782047 DOI: 10.3390/metabo12010066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 12/30/2022] Open
Abstract
Taurine transport was investigated at the blood–testis barrier (BTB) formed by Sertoli cells. An integration plot analysis of mice showed the apparent influx permeability clearance of [3H]taurine (27.7 μL/(min·g testis)), which was much higher than that of a non-permeable paracellular marker, suggesting blood-to-testis transport of taurine, which may involve a facilitative taurine transport system at the BTB. A mouse Sertoli cell line, TM4 cells, showed temperature- and concentration-dependent [3H]taurine uptake with a Km of 13.5 μM, suggesting that the influx transport of taurine at the BTB involves a carrier-mediated process. [3H]Taurine uptake by TM4 cells was significantly reduced by the substrates of taurine transporter (TauT/SLC6A6), such as β-alanine, hypotaurine, γ-aminobutyric acid (GABA), and guanidinoacetic acid (GAA), with no significant effect shown by L-alanine, probenecid, and L-leucine. In addition, the concentration-dependent inhibition of [3H]taurine uptake revealed an IC50 of 378 μM for GABA. Protein expression of TauT in the testis, seminiferous tubules, and TM4 cells was confirmed by Western blot analysis and immunohistochemistry by means of anti-TauT antibodies, and knockdown of TauT showed significantly decreased [3H]taurine uptake by TM4 cells. These results suggest the involvement of TauT in the transport of taurine at the BTB.
Collapse
|
15
|
Baliou S, Adamaki M, Ioannou P, Pappa A, Panayiotidis MI, Spandidos DA, Christodoulou I, Kyriakopoulos AM, Zoumpourlis V. Protective role of taurine against oxidative stress (Review). Mol Med Rep 2021; 24:605. [PMID: 34184084 PMCID: PMC8240184 DOI: 10.3892/mmr.2021.12242] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/03/2021] [Indexed: 12/14/2022] Open
Abstract
Taurine is a fundamental mediator of homeostasis that exerts multiple roles to confer protection against oxidant stress. The development of hypertension, muscle/neuro‑associated disorders, hepatic cirrhosis, cardiac dysfunction and ischemia/reperfusion are examples of some injuries that are linked with oxidative stress. The present review gives a comprehensive description of all the underlying mechanisms of taurine, with the aim to explain its anti‑oxidant actions. Taurine is regarded as a cytoprotective molecule due to its ability to sustain normal electron transport chain, maintain glutathione stores, upregulate anti‑oxidant responses, increase membrane stability, eliminate inflammation and prevent calcium accumulation. In parallel, the synergistic effect of taurine with other potential therapeutic modalities in multiple disorders are highlighted. Apart from the results derived from research findings, the current review bridges the gap between bench and bedside, providing mechanistic insights into the biological activity of taurine that supports its potential therapeutic efficacy in clinic. In the future, further clinical studies are required to support the ameliorative effect of taurine against oxidative stress.
Collapse
Affiliation(s)
- Stella Baliou
- National Hellenic Research Foundation, 11635 Athens, Greece
| | - Maria Adamaki
- National Hellenic Research Foundation, 11635 Athens, Greece
| | - Petros Ioannou
- Department of Internal Medicine and Infectious Diseases, University Hospital of Heraklion, 71110 Heraklion, Greece
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Mihalis I. Panayiotidis
- Department of Cancer Genetics, Therapeutics and Ultrastructural Pathology, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
- The Cyprus School of Molecular Medicine, 2371 Nicosia, Cyprus
| | - Demetrios A. Spandidos
- Department of Internal Medicine and Infectious Diseases, University Hospital of Heraklion, 71110 Heraklion, Greece
| | | | | | | |
Collapse
|
16
|
Álvarez-Merz I, Luengo JG, Muñoz MD, Hernández-Guijo JM, Solís JM. Hypoxia-induced depression of synaptic transmission becomes irreversible by intracellular accumulation of non-excitatory amino acids. Neuropharmacology 2021; 190:108557. [PMID: 33848510 DOI: 10.1016/j.neuropharm.2021.108557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 03/10/2021] [Accepted: 04/02/2021] [Indexed: 10/21/2022]
Abstract
The intracellular accumulation of some amino acids (AAs), mainly glutamine, can contribute to brain edema observed during liver failure. We recently demonstrated that individual applications of high concentrations (10 mM) of some non-excitatory AAs increase the electrical resistance of hippocampal slices, indicating cell swelling. Therefore, we pondered whether an AA mixture's application might cause cell swelling at a physiological concentration range. In rat hippocampal slices, we carried out extra- and intracellular electrophysiological recordings and AAs analysis to address this question. We applied a mixture of 19 AAs at their plasmatic concentrations (Plasma solution: Ala, Gly, Gln, His, Ser, Tau, Thr, Arg, Leu, Met, Pro, Val, Asn, Cys, Phe, Ile, Lys, Tyr, and Trp). This solution was afterward divided into two according to the individual AAs at 10 mM concentration inducing synaptic potentiation (Plasma1, containing the first seven AAs of Plasma) or not (Plasma2, with the remaining AAs). Plasma application increased evoked field potentials requiring extracellular chloride. This effect was mimicked by the Plasma1 but not the Plasma2 solution. Plasma1-induced potentiation was independent of changes in release probability, basic electrophysiological membrane properties, and NMDAR activation. AAs in Plasma1 act cooperatively to accumulate intracellularly and to induce synaptic potentiation. In the presence of Plasma1, the reversible synaptic depression caused by a 40-min hypoxia period turned into an irreversible disappearance of synaptic potentials through an NMDAR-dependent mechanism. The presence of a system A transport inhibitor did not block Plasma1-mediated effects. These results indicate that cell swelling, induced by the accumulation of non-excitotoxic AAs through unidentified transporters, might foster deleterious effects produced by hypoxia-ischemia episodes.
Collapse
Affiliation(s)
- Iris Álvarez-Merz
- Servicio de Neurobiología-Investigación, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain; Departamento de Farmacología y Terapeútica, ITH, Facultad de Medicina, Universidad Autónoma de Madrid, IRYCIS, Avda. Arzobispo Morcillo 4, 28029, Madrid, Spain
| | - Javier G Luengo
- Servicio de Neurobiología-Investigación, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain; Departamento de Farmacología y Terapeútica, ITH, Facultad de Medicina, Universidad Autónoma de Madrid, IRYCIS, Avda. Arzobispo Morcillo 4, 28029, Madrid, Spain
| | - María-Dolores Muñoz
- Unidad de Neurología Experimental, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, 28034, Spain
| | - Jesús M Hernández-Guijo
- Departamento de Farmacología y Terapeútica, ITH, Facultad de Medicina, Universidad Autónoma de Madrid, IRYCIS, Avda. Arzobispo Morcillo 4, 28029, Madrid, Spain
| | - José M Solís
- Servicio de Neurobiología-Investigación, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain.
| |
Collapse
|
17
|
Fairweather SJ, Shah N, Brӧer S. Heteromeric Solute Carriers: Function, Structure, Pathology and Pharmacology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 21:13-127. [PMID: 33052588 DOI: 10.1007/5584_2020_584] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Solute carriers form one of three major superfamilies of membrane transporters in humans, and include uniporters, exchangers and symporters. Following several decades of molecular characterisation, multiple solute carriers that form obligatory heteromers with unrelated subunits are emerging as a distinctive principle of membrane transporter assembly. Here we comprehensively review experimentally established heteromeric solute carriers: SLC3-SLC7 amino acid exchangers, SLC16 monocarboxylate/H+ symporters and basigin/embigin, SLC4A1 (AE1) and glycophorin A exchanger, SLC51 heteromer Ost α-Ost β uniporter, and SLC6 heteromeric symporters. The review covers the history of the heteromer discovery, transporter physiology, structure, disease associations and pharmacology - all with a focus on the heteromeric assembly. The cellular locations, requirements for complex formation, and the functional role of dimerization are extensively detailed, including analysis of the first complete heteromer structures, the SLC7-SLC3 family transporters LAT1-4F2hc, b0,+AT-rBAT and the SLC6 family heteromer B0AT1-ACE2. We present a systematic analysis of the structural and functional aspects of heteromeric solute carriers and conclude with common principles of their functional roles and structural architecture.
Collapse
Affiliation(s)
- Stephen J Fairweather
- Research School of Biology, Australian National University, Canberra, ACT, Australia. .,Resarch School of Chemistry, Australian National University, Canberra, ACT, Australia.
| | - Nishank Shah
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Stefan Brӧer
- Research School of Biology, Australian National University, Canberra, ACT, Australia.
| |
Collapse
|
18
|
Sears SM, Hewett SJ. Influence of glutamate and GABA transport on brain excitatory/inhibitory balance. Exp Biol Med (Maywood) 2021; 246:1069-1083. [PMID: 33554649 DOI: 10.1177/1535370221989263] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
An optimally functional brain requires both excitatory and inhibitory inputs that are regulated and balanced. A perturbation in the excitatory/inhibitory balance-as is the case in some neurological disorders/diseases (e.g. traumatic brain injury Alzheimer's disease, stroke, epilepsy and substance abuse) and disorders of development (e.g. schizophrenia, Rhett syndrome and autism spectrum disorder)-leads to dysfunctional signaling, which can result in impaired cognitive and motor function, if not frank neuronal injury. At the cellular level, transmission of glutamate and GABA, the principle excitatory and inhibitory neurotransmitters in the central nervous system control excitatory/inhibitory balance. Herein, we review the synthesis, release, and signaling of GABA and glutamate followed by a focused discussion on the importance of their transport systems to the maintenance of excitatory/inhibitory balance.
Collapse
Affiliation(s)
- Sheila Ms Sears
- Department of Biology, Program in Neuroscience, 2029Syracuse University, Syracuse, NY 13244, USA
| | - Sandra J Hewett
- Department of Biology, Program in Neuroscience, 2029Syracuse University, Syracuse, NY 13244, USA
| |
Collapse
|
19
|
Nałęcz KA. Amino Acid Transporter SLC6A14 (ATB 0,+) - A Target in Combined Anti-cancer Therapy. Front Cell Dev Biol 2020; 8:594464. [PMID: 33195271 PMCID: PMC7609839 DOI: 10.3389/fcell.2020.594464] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer cells are characterized by quick growth and proliferation, demanding constant supply of various nutrients. Several plasma membrane transporters delivering such compounds are upregulated in cancer. Solute carrier family 6 member 14 (SLC6A14), known as amino acid transporter B0,+ (ATB0,+) transports all amino acids with exception of the acidic ones: aspartate and glutamate. Its malfunctioning is correlated with several pathological states and it is upregulated in solid tumors. The high expression of SLC6A14 is prognostic and unfavorable in pancreatic cancer, while in breast cancer it is expressed in estrogen receptor positive cells. As many plasma membrane transporters it resides in endoplasmic reticulum (ER) membrane after translation before further trafficking through Golgi to the cell surface. Transporter exit from ER is strictly controlled. The proper folding of SLC6A14 was shown to be controlled from the cytoplasmic side by heat shock proteins, further exit from ER and formation of coatomer II (COPII) coated vesicles depends on specific interaction with COPII cargo-recognizing subunit SEC24C, phosphorylated by kinase AKT. Inhibition of heat shock proteins, known to be upregulated in cancer, directs SLC6A14 to degradation. Targeting proteins regulating SLC6A14 trafficking is proposed as an additional pharmacological treatment of cancer.
Collapse
Affiliation(s)
- Katarzyna A Nałęcz
- Laboratory of Transport Through Biomembranes, Nencki Institute of Experimental Biology, Warsaw, Poland
| |
Collapse
|
20
|
Sub-chronic taurine administration induces behavioral sensitization but does not influence ethanol-induced dopamine release in the nucleus accumbens. Pharmacol Biochem Behav 2019; 188:172831. [PMID: 31770542 DOI: 10.1016/j.pbb.2019.172831] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/20/2019] [Accepted: 11/22/2019] [Indexed: 02/03/2023]
Abstract
Preclinical studies have shown that the amino acid taurine is of importance for the dopamine elevating properties of ethanol. Taurine intake has escalated over the last decade due to increased consumption of taurine-containing energy drinks and dietary supplements. Whether long-term intake of large amounts of taurine induces adaptations affecting ethanol-induced dopamine elevation is not clear. Thus the aim of the present studies was to explore the impact of repeated administration of large amounts of taurine on ethanol-induced behavior and dopamine neurotransmission. Repeated daily systemic administration of taurine increased taurine-induced locomotor activity and rearing. Acute administration of taurine and ethanol in naïve animals produced an additive effect on extracellular taurine but no alteration of the ethanol-induced dopamine elevation, as measured by in vivo microdialysis. Sub-chronic administration of taurine did not modify the taurine- or dopamine-elevating properties of ethanol. Daily taurine treatment also failed to change the mRNA expression of the taurine transporter and GABAA- and glycine-receptor subunits, as measured by qPCR in nucleus accumbens tissue. We conclude that systemic administration of taurine may have long lasting central effects, here displayed as behavioral sensitization. However, repeated daily exposure to taurine does not appear to influence the dopamine elevating properties of ethanol.
Collapse
|
21
|
Luengo JG, Muñoz MD, Álvarez-Merz I, Herranz AS, González JC, Martín del Río R, Hernández-Guijo JM, Solís JM. Intracellular accumulation of amino acids increases synaptic potentials in rat hippocampal slices. Amino Acids 2019; 51:1337-1351. [DOI: 10.1007/s00726-019-02771-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 08/11/2019] [Indexed: 12/22/2022]
|
22
|
Preising MN, Görg B, Friedburg C, Qvartskhava N, Budde BS, Bonus M, Toliat MR, Pfleger C, Altmüller J, Herebian D, Beyer M, Zöllner HJ, Wittsack HJ, Schaper J, Klee D, Zechner U, Nürnberg P, Schipper J, Schnitzler A, Gohlke H, Lorenz B, Häussinger D, Bolz HJ. Biallelic mutation of human SLC6A6 encoding the taurine transporter TAUT is linked to early retinal degeneration. FASEB J 2019; 33:11507-11527. [PMID: 31345061 DOI: 10.1096/fj.201900914rr] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We previously reported that inactivation of the transmembrane taurine transporter (TauT or solute carrier 6a6) causes early retinal degeneration in mice. Compatible with taurine's indispensability for cell volume homeostasis, protein stabilization, cytoprotection, antioxidation, and immuno- and neuromodulation, mice develop multisystemic dysfunctions (hearing loss; liver fibrosis; and behavioral, heart, and skeletal muscle abnormalities) later on. Here, by genetic, cell biologic, in vivo 1H-magnetic resonance spectroscopy and molecular dynamics simulation studies, we conducted in-depth characterization of a novel disorder: human TAUT deficiency. Loss of TAUT function due to a homozygous missense mutation caused panretinal degeneration in 2 brothers. TAUTp.A78E still localized in the plasma membrane but is predicted to impact structural stabilization. 3H-taurine uptake by peripheral blood mononuclear cells was reduced by 95%, and taurine levels were severely reduced in plasma, skeletal muscle, and brain. Extraocular dysfunctions were not yet detected, but significantly increased urinary excretion of 8-oxo-7,8-dihydroguanosine indicated generally enhanced (yet clinically unapparent) oxidative stress and RNA oxidation, warranting continuous broad surveillance.-Preising, M. N., Görg, B., Friedburg, C., Qvartskhava, N., Budde, B. S., Bonus, M., Toliat, M. R., Pfleger, C., Altmüller, J., Herebian, D., Beyer, M., Zöllner, H. J., Wittsack, H.-J., Schaper, J., Klee, D., Zechner, U., Nürnberg, P., Schipper, J., Schnitzler, A., Gohlke, H., Lorenz, B., Häussinger, D., Bolz, H. J. Biallelic mutation of human SLC6A6 encoding the taurine transporter TAUT is linked to early retinal degeneration.
Collapse
Affiliation(s)
- Markus N Preising
- Department of Ophthalmology, Justus-Liebig University Giessen, Giessen, Germany
| | - Boris Görg
- Department of Gastroenterology, Hepatology, and Infectious Diseases, University Hospital of Düsseldorf-Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christoph Friedburg
- Department of Ophthalmology, Justus-Liebig University Giessen, Giessen, Germany
| | - Natalia Qvartskhava
- Department of Gastroenterology, Hepatology, and Infectious Diseases, University Hospital of Düsseldorf-Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Birgit S Budde
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Michele Bonus
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Mohammad R Toliat
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Christopher Pfleger
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Janine Altmüller
- Cologne Center for Genomics, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Diran Herebian
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf-Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Mila Beyer
- Department of Gastroenterology, Hepatology, and Infectious Diseases, University Hospital of Düsseldorf-Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Helge J Zöllner
- Institute of Clinical Neuroscience and Medical Psychology, University Hospital of Düsseldorf-Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Hans-Jörg Wittsack
- Department of Diagnostic and Interventional Radiology, University Hospital of Düsseldorf-Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jörg Schaper
- Department of Diagnostic and Interventional Radiology, University Hospital of Düsseldorf-Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Dirk Klee
- Department of Diagnostic and Interventional Radiology, University Hospital of Düsseldorf-Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ulrich Zechner
- Senckenberg Centre for Human Genetics, Frankfurt on the Main, Germany.,Institute of Human Genetics, Mainz University Medical Center, Mainz, Germany
| | - Peter Nürnberg
- Cologne Center for Genomics, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Jörg Schipper
- Klinik für Hals-Nasen-Ohren Heilkunde, University Hospital of Düsseldorf-Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Alfons Schnitzler
- Institute of Clinical Neuroscience and Medical Psychology, University Hospital of Düsseldorf-Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Holger Gohlke
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,John von Neumann Institute for Computing (NIC)-Jülich Supercomputing Centre (JSC)-Structural Biochemistry, Institute of Complex Systems (ICS 6), Research Centre Jülich, Jülich, Germany
| | - Birgit Lorenz
- Department of Ophthalmology, Justus-Liebig University Giessen, Giessen, Germany
| | - Dieter Häussinger
- Department of Gastroenterology, Hepatology, and Infectious Diseases, University Hospital of Düsseldorf-Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Hanno J Bolz
- Institute of Human Genetics, University of Cologne, Cologne, Germany.,Senckenberg Centre for Human Genetics, Frankfurt on the Main, Germany
| |
Collapse
|
23
|
Nishimura T, Higuchi K, Yoshida Y, Sugita-Fujisawa Y, Kojima K, Sugimoto M, Santo M, Tomi M, Nakashima E. Hypotaurine Is a Substrate of GABA Transporter Family Members GAT2/Slc6a13 and TAUT/Slc6a6. Biol Pharm Bull 2019; 41:1523-1529. [PMID: 30270321 DOI: 10.1248/bpb.b18-00168] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hypotaurine is a precursor of taurine and a physiological antioxidant that circulates in adult and fetal plasma. The purpose of the present study was to clarify whether hypotaurine is a substrate of Slc6a/gamma-aminobutyric acid (GABA) transporter family members. Radiolabeled hypotaurine was synthesized from radiolabeled cysteamine and 2-aminoethanethiol dioxygenase. The uptakes of [3H]GABA, [3H]taurine, and [14C]hypotaurine by HEK293 cells expressing mouse GAT1/Slc6a1, TAUT/Slc6a6, GAT3/Slc6a11, BGT1/Slc6a12, and GAT2/Slc6a13 were measured. TAUT and GAT2 showed strong [14C]hypotaurine uptake activity, while BGT1 showed moderate activity, and GAT1 and GAT3 showed slight but significant activity. Mouse TAUT and GAT2 both showed Michaelis constants of 11 µM for hypotaurine uptake. GAT2-expressing cells pretreated with hypotaurine showed resistance to H2O2-induced oxidative stress. These results suggest that under physiological conditions, TAUT and GAT2 would be major contributors to hypotaurine transfer across the plasma membrane, and that uptake of hypotaurine via GAT2 contributes to the cellular resistance to oxidative stress.
Collapse
Affiliation(s)
| | - Kei Higuchi
- Faculty of Pharmacy, Keio University.,Faculty of Pharma-Sciences, Teikyo University
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Richter M, Moroniak SJ, Michel H. Identification of competitive inhibitors of the human taurine transporter TauT in a human kidney cell line. Pharmacol Rep 2019; 71:121-129. [PMID: 30612046 DOI: 10.1016/j.pharep.2018.10.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 09/24/2018] [Accepted: 10/10/2018] [Indexed: 10/28/2022]
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.
Collapse
Affiliation(s)
- Michelle Richter
- Department of Molecular Membrane Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Selina J Moroniak
- Department of Molecular Membrane Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Hartmut Michel
- Department of Molecular Membrane Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| |
Collapse
|
25
|
Abstract
The small intestine mediates the absorption of amino acids after ingestion of protein and sustains the supply of amino acids to all tissues. The small intestine is an important contributor to plasma amino acid homeostasis, while amino acid transport in the large intestine is more relevant for bacterial metabolites and fluid secretion. A number of rare inherited disorders have contributed to the identification of amino acid transporters in epithelial cells of the small intestine, in particular cystinuria, lysinuric protein intolerance, Hartnup disorder, iminoglycinuria, and dicarboxylic aminoaciduria. These are most readily detected by analysis of urine amino acids, but typically also affect intestinal transport. The genes underlying these disorders have all been identified. The remaining transporters were identified through molecular cloning techniques to the extent that a comprehensive portrait of functional cooperation among transporters of intestinal epithelial cells is now available for both the basolateral and apical membranes. Mouse models of most intestinal transporters illustrate their contribution to amino acid homeostasis and systemic physiology. Intestinal amino acid transport activities can vary between species, but these can now be explained as differences of amino acid transporter distribution along the intestine. © 2019 American Physiological Society. Compr Physiol 9:343-373, 2019.
Collapse
Affiliation(s)
- Stefan Bröer
- Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia
| | - Stephen J Fairweather
- Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia
| |
Collapse
|
26
|
Seidel U, Huebbe P, Rimbach G. Taurine: A Regulator of Cellular Redox Homeostasis and Skeletal Muscle Function. Mol Nutr Food Res 2018; 63:e1800569. [DOI: 10.1002/mnfr.201800569] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/10/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Ulrike Seidel
- Institute of Human Nutrition and Food ScienceUniversity of Kiel Kiel Germany
| | - Patricia Huebbe
- Institute of Human Nutrition and Food ScienceUniversity of Kiel Kiel Germany
| | - Gerald Rimbach
- Institute of Human Nutrition and Food ScienceUniversity of Kiel Kiel Germany
| |
Collapse
|
27
|
Ulenius L, Adermark L, Söderpalm B, Ericson M. Ethanol-Induced Taurine Elevation in the Rat Dorsal Striatum. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 975 Pt 1:173-181. [PMID: 28849453 DOI: 10.1007/978-94-024-1079-2_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
In the search for the primary mechanism underlying the dopamine elevating properties of ethanol we have established that raised levels of taurine in the nucleus accumbens (nAc) is pivotal. In the nAc, the release of taurine appears to be connected to osmoregulation, and neither taurine nor dopamine is increased if ethanol is administered in a hypertonic saline solution. However, even though the nAc is important for drug-reinforcement, manifestation of addiction has been postulated to recruit the more dorsal parts of the striatum (DS). How ethanol influences dopamine and taurine in the DS and their role in addiction is thus far poorly understood. By means of in vivo microdialysis in freely moving rats we concomitantly monitored extracellular levels of dopamine and taurine in the DS following administration of ethanol diluted either in an isotonic or hypertonic saline solution. In a different set of rats, placed in a voluntary ethanol consumption paradigm (intermittent access to 20% ethanol for 2 months), taurine and dopamine were monitored following an acute injection of ethanol. We found that neither administration of ethanol diluted in a hypertonic saline solution, nor 2 months of moderate ethanol consumption, influence the ethanol-induced increase of taurine in the DS. We propose that there may be regional differences in the relationship between taurine, dopamine and ethanol in the nAc and in the DS. It remains to be determined if this subregion-specificity is important for the transition from recreational drug use to a compulsive habit.
Collapse
Affiliation(s)
- Lisa Ulenius
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| | - Louise Adermark
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Beroendekliniken, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mia Ericson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
28
|
Hadj-Saïd W, Fradot V, Ivkovic I, Sahel JA, Picaud S, Froger N. Taurine Promotes Retinal Ganglion Cell Survival Through GABA B Receptor Activation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 975 Pt 2:687-701. [PMID: 28849492 DOI: 10.1007/978-94-024-1079-2_54] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Retinal ganglion cell (RGC) degeneration occurs in numerous retinal diseases, either as a primary process like in glaucoma, or secondary to photoreceptor loss and no efficient compound targeting directly RGC neuroprotection is yet available. We previously described that taurine exerts a direct protective effect on RGCs cultured under serum-deprived conditions. Because taurine was known to have an agonist-like activity for GABA/glycine receptors, we investigated here if the taurine-elicited neuroprotective effect may be mediated through the activation of these receptors using selective antagonist ligands. RGCs were purified, seeded in 96-well plate and maintained in culture during 6 days in vitro. Viable cells were labelled with calcein and densities in full-well area were then automatically counted. Here we show that the protective effect of taurine against RGC loss observed under serum deprivation can be mediated through the GABAB receptor stimulation. Hence, two selective agonists, including baclofen, at this metabotropic GABAB receptor were found to reproduce taurine action by enhancing RGC survival in culture. This study suggests that GABAB receptor stimulation provides direct neuroprotection for RGCs. Accordingly, drugs targeting GABAB receptor may represent a new way for the prevention of RGC degeneration.
Collapse
Affiliation(s)
- Wahiba Hadj-Saïd
- INSERM, U968, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
- UPMC Univ Paris 06, UMR_S968, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
- CNRS, UMR 7210, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
| | - Valérie Fradot
- INSERM, U968, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
- UPMC Univ Paris 06, UMR_S968, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
- CNRS, UMR 7210, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
| | - Ivana Ivkovic
- INSERM, U968, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
- UPMC Univ Paris 06, UMR_S968, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
- CNRS, UMR 7210, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
| | - José-Alain Sahel
- INSERM, U968, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
- UPMC Univ Paris 06, UMR_S968, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
- CNRS, UMR 7210, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Paris, F-75012, France
- Fondation Ophtalmologique Adolphe de Rothschild, Paris, F-75020, France
- Institute of Ophthalmology, University College of London, London, UK
| | - Serge Picaud
- INSERM, U968, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
- UPMC Univ Paris 06, UMR_S968, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
- CNRS, UMR 7210, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France
- Institute of Ophthalmology, University College of London, London, UK
| | - Nicolas Froger
- INSERM, U968, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France.
- UPMC Univ Paris 06, UMR_S968, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France.
- CNRS, UMR 7210, Institut de la Vision, 17 rue Moreau, Paris, F-75012, France.
| |
Collapse
|
29
|
Identification and characterization of the Fasciola hepatica sodium- and chloride-dependent taurine transporter. PLoS Negl Trop Dis 2018; 12:e0006428. [PMID: 29702654 PMCID: PMC5942844 DOI: 10.1371/journal.pntd.0006428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 05/09/2018] [Accepted: 04/03/2018] [Indexed: 01/06/2023] Open
Abstract
The parasitic liver fluke Fasciola hepatica infests mainly ruminants, but it can also cause fasciolosis in people, who ingest the metacercariae encysted on plants. The drug of choice to treat fasciolosis is triclabendazole (TBZ), which has been on the market for several decades. This is also true for the other available drugs. Accordingly, drug-resistant flukes have been emerging at an increasing rate making it desirable to identify alternative drug targets. Here, we focused on the fact that adult F. hepatica persists in the hostile environment of the bile ducts of infected organisms. A common way to render bile acids less toxic is to conjugate them to taurine (2-aminoethanesulfonic acid). We cloned a transporter from the solute carrier-6 (SLC6) family, which was most closely related to the GABA-transporter-2 of other organisms. When heterologously expressed, this F. hepatica transporter supported the high-affinity cellular uptake of taurine (KM = 12.0 ± 0.5 μM) but not of GABA. Substrate uptake was dependent on Na+- and Cl- (calculated stoichiometry 2:1). Consistent with the low chloride concentration in mammalian bile, the F. hepatica transporter had a higher apparent affinity for Cl- (EC50 = 14±3 mM) than the human taurine transporter (EC50 = 55±7 mM). We incubated flukes with unconjugated bile acids in the presence and absence of taurine: taurine promoted survival of flukes; the taurine transporter inhibitor guanidinoethansulfonic acid abolished this protective effect of taurine. Based on these observations, we conclude that the taurine transporter is critical for the survival of liver flukes in the bile. Thus, the taurine transporter represents a candidate drug target.
Collapse
|
30
|
Tachikawa M, Yashiki A, Akanuma SI, Matsukawa H, Ide S, Minami M, Hosoya KI. Astrocytic γ-aminobutyric acid (GABA) transporters mediate guanidinoacetate transport in rat brain. Neurochem Int 2018; 113:1-7. [DOI: 10.1016/j.neuint.2017.11.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 11/11/2017] [Accepted: 11/22/2017] [Indexed: 10/18/2022]
|
31
|
Nielsen CU, Bjerg M, Ulaganathan N, Holm R. Oral and intravenous pharmacokinetics of taurine in sprague-dawley rats: the influence of dose and the possible involvement of the proton-coupled amino acid transporter, PAT1, in oral taurine absorption. Physiol Rep 2017; 5:5/19/e13467. [PMID: 29038364 PMCID: PMC5641942 DOI: 10.14814/phy2.13467] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 08/30/2017] [Accepted: 09/10/2017] [Indexed: 11/24/2022] Open
Abstract
Taurine is involved in various physiological processes, and one of the most abundant amino acids in human. The aim was to investigate the mechanism for intestinal absorption of taurine in vivo using also in vitro mechanistic studies. Taurine absorption was measured in male Sprague‐Dawley rats at 10–997 mg/kg and 1–30 mg/kg for oral and intravenous administration, respectively. Oral absorption was measured in the presence of substrates for the proton‐coupled amino acid transporter, PAT1, that is, 200 mg/kg proline (Pro) and sarcosine (Sar), and in the presence of 2‐Amino‐2‐norbornanecarboxylic acid (BCH) (200 mg/kg). BCH is not an inhibitor of PAT1 or the taurine transporter, TauT, hence it was included as a negative control. In vitro studies investigating the transport mechanism of taurine were conducted in human intestinal Caco‐2 cells. The pharmacokinetic investigations showed that intestinal taurine absorption was not saturable at the investigated doses, but that the time (tmax) to reach the maximal plasma concentration (Cmax) increased with dose. Furthermore, Sar and Pro, but not BCH, decreased taurine Cmax. In vitro it was clearly shown that PAT1 mediated the cellular uptake of taurine and thereby facilitated the transepithelial taurine transport, which could be inhibited by Pro and Sar, but not BCH. In vivo and in vitro results suggest that taurine absorption from the intestine is caused by PAT1.
Collapse
Affiliation(s)
- Carsten Uhd Nielsen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M, Denmark
| | - Maria Bjerg
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M, Denmark
| | - Nithiya Ulaganathan
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M, Denmark
| | - René Holm
- Pharmaceutical Science and CMC Biologics, H. Lundbeck A/S, Valby, Denmark
| |
Collapse
|
32
|
Carnosine content in the porcine longissimus thoracis muscle and its association with meat quality attributes and carnosine-related gene expression. Meat Sci 2017; 124:84-94. [DOI: 10.1016/j.meatsci.2016.11.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/07/2016] [Accepted: 11/08/2016] [Indexed: 11/18/2022]
|
33
|
Kubo Y, Akanuma SI, Hosoya KI. Impact of SLC6A Transporters in Physiological Taurine Transport at the Blood-Retinal Barrier and in the Liver. Biol Pharm Bull 2017; 39:1903-1911. [PMID: 27904033 DOI: 10.1248/bpb.b16-00597] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cumulative studies showed that taurine (2-aminoethanesulfonic acid) contributes to a variety of physiological events. Transport study suggested the cellular taurine transport in an Na+- and Cl--dependent manner, and the several members of SLC6A family have been shown as taurine transporter. At the inner blood-retinal barrier (BRB), taurine transporter (TauT/SLC6A) is involved in the transport of taurine to the retina from the circulating blood. The involvement of TauT is also suggested in γ-aminobutyric acid (GABA) transport at the inner BRB, and its role is assumed in the elimination of GABA from the retinal interstitial fluid. In the retina, taurine is thought to be a major organic osmolyte, and its influx and efflux through TauT and volume-sensitive organic osmolyte and anion channel (VSOAC) in Müller cells regulate the osmolarity in the retinal microenvironment to maintain a healthy retina. In the liver, hepatocytes take up taurine via GABA transporter 2 (GAT2/SLC6A13, the orthologue of mouse GAT3) expressed at the sinusoidal membrane of periportal hepatocytes, contributing to the metabolism of bile acid. Site-directed mutagenesis study suggests amino acid residues that are crucial in the recognition of substrates by GATs and TauT. The evidence suggests the physiological impact of taurine transporters in tissues.
Collapse
Affiliation(s)
- Yoshiyuki Kubo
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | | | | |
Collapse
|
34
|
|
35
|
The taurine transporter substrate guanidinoethyl sulfonate mimics the action of taurine on long-term synaptic potentiation. Amino Acids 2016; 48:2647-2656. [DOI: 10.1007/s00726-016-2298-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/06/2016] [Indexed: 12/20/2022]
|
36
|
Taurine content in different brain structures during ageing: effect on hippocampal synaptic plasticity. Amino Acids 2016; 48:1199-208. [DOI: 10.1007/s00726-015-2155-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/10/2015] [Indexed: 10/22/2022]
|
37
|
Abstract
Histidine-containing dipeptides like carnosine and anserine have protective functions in both health and disease. Animal studies suggest that carnosine can be metabolized within the kidney. The goal of this study was to obtain evidence of carnosine metabolism in the human kidney and to provide insight with regards to diabetic nephropathy. Expression, distribution, and localization of carnosinase-1 (CNDP1), carnosine synthase (CARNS), and taurine transporters (TauT) were measured in human kidneys. CNDP1 and CARNS activities were measured in vitro. CNDP1 and CARNS were located primarily in distal and proximal tubules, respectively. Specifically, CNDP1 levels were high in tubular cells and podocytes (20.3 ± 3.4 and 15 ± 3.2 ng/mg, respectively) and considerably lower in endothelial cells (0.5 ± 0.1 ng/mg). CNDP1 expression was correlated with the degradation of carnosine and anserine (r = 0.88 and 0.81, respectively). Anserine and carnosine were also detectable by HPLC in the renal cortex. Finally, TauT mRNA and protein were found in all renal epithelial cells. In diabetic patients, CNDP1 seemed to be reallocated to proximal tubules. We report compelling evidence that the kidney has an intrinsic capacity to metabolize carnosine. Both CNDP1 and CARNS are expressed in glomeruli and tubular cells. Carnosine-synthesizing and carnosine-hydrolyzing enzymes are localized in distinct compartments in the nephron and increased CNDP1 levels suggest a higher CNDP1 activity in diabetic kidneys.
Collapse
|
38
|
Lambert IH, Kristensen DM, Holm JB, Mortensen OH. Physiological role of taurine--from organism to organelle. Acta Physiol (Oxf) 2015; 213:191-212. [PMID: 25142161 DOI: 10.1111/apha.12365] [Citation(s) in RCA: 241] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 08/01/2014] [Accepted: 08/13/2014] [Indexed: 12/21/2022]
Abstract
Taurine is often referred to as a semi-essential amino acid as newborn mammals have a limited ability to synthesize taurine and have to rely on dietary supply. Taurine is not thought to be incorporated into proteins as no aminoacyl tRNA synthetase has yet been identified and is not oxidized in mammalian cells. However, taurine contributes significantly to the cellular pool of organic osmolytes and has accordingly been acknowledged for its role in cell volume restoration following osmotic perturbation. This review describes taurine homeostasis in cells and organelles with emphasis on taurine biophysics/membrane dynamics, regulation of transport proteins involved in active taurine uptake and passive taurine release as well as physiological processes, for example, development, lung function, mitochondrial function, antioxidative defence and apoptosis which seem to be affected by a shift in the expression of the taurine transporters and/or the cellular taurine content.
Collapse
Affiliation(s)
- I. H. Lambert
- Section of Cellular and Developmental Biology; Department of Biology; University of Copenhagen; Copenhagen Ø Denmark
| | - D. M. Kristensen
- Section of Genomics and Molecular Biomedicine; Department of Biology; University of Copenhagen; Copenhagen Denmark
- Cellular and Metabolic Research Section; Department of Biomedical Sciences; Panum Institute; University of Copenhagen; Copenhagen N Denmark
| | - J. B. Holm
- Section of Genomics and Molecular Biomedicine; Department of Biology; University of Copenhagen; Copenhagen Denmark
| | - O. H. Mortensen
- Cellular and Metabolic Research Section; Department of Biomedical Sciences; Panum Institute; University of Copenhagen; Copenhagen N Denmark
| |
Collapse
|
39
|
Lambert IH, Jensen JV, Pedersen PA. mTOR ensures increased release and reduced uptake of the organic osmolyte taurine under hypoosmotic conditions in mouse fibroblasts. Am J Physiol Cell Physiol 2014; 306:C1028-40. [DOI: 10.1152/ajpcell.00005.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mammalian target of rapamycin (mTOR) is a serine/threonine kinase that modulates translation in response to growth factors and alterations in nutrient availability following hypoxia and DNA damage. Here we demonstrate that mTOR activity in Ehrlich Lettré ascites (ELA) cells is transiently increased within minutes following osmotic cell swelling and that inhibition of phosphatidylinositol-3-phosphatase (PTEN) counteracts the upstream phosphatidylinositol kinase and potentiates mTOR activity. PTEN inhibition concomitantly potentiates swelling-induced taurine release via the volume-sensitive transporter for organic osmolytes and anion channels (VSOAC) and enhances swelling-induced inhibition of taurine uptake via the taurine-specific transporter (TauT). Chronic osmotic stress, i.e., exposure to hypotonic or hypertonic media for 24 h, reduces and increases mTOR activity in ELA cells, respectively. Using rapamycin, we demonstrate that mTOR inhibition is accompanied by reduction in TauT activity and increase in VSOAC activity in cells expressing high (NIH3T3 fibroblasts) or low (ELA) amounts of mTOR protein. The effect of mTOR inhibition on TauT activity reflects reduced TauT mRNA, TauT protein abundance, and an overall reduction in protein synthesis, whereas the effect on VSOAC is mimicked by catalase inhibition and correlates with reduced catalase mRNA abundance. Hence, mTOR activity favors loss of taurine following hypoosmotic cell swelling, i.e., release via VSOAC and uptake via TauT during acute hypotonic exposure is potentiated and reduced, respectively, by phosphorylation involving mTOR and/or the kinases upstream to mTOR. Decrease in TauT activity during chronic hypotonic exposure, on the other hand, involves reduction in expression/activity of TauT and enzymes in antioxidative defense.
Collapse
Affiliation(s)
- Ian Henry Lambert
- Department of Biology, Section of Cellular and Developmental Biology, University of Copenhagen, Copenhagen, Denmark; and
| | - Jane Vendelbo Jensen
- Department of Biology, Section of Cellular and Developmental Biology, University of Copenhagen, Copenhagen, Denmark; and
| | - Per Amstrup Pedersen
- Department of Biology, Section for Molecular Integrative Physiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
40
|
Kempson SA, Zhou Y, Danbolt NC. The betaine/GABA transporter and betaine: roles in brain, kidney, and liver. Front Physiol 2014; 5:159. [PMID: 24795654 PMCID: PMC4006062 DOI: 10.3389/fphys.2014.00159] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 04/04/2014] [Indexed: 12/18/2022] Open
Abstract
The physiological roles of the betaine/GABA transporter (BGT1; slc6a12) are still being debated. BGT1 is a member of the solute carrier family 6 (the neurotransmitter, sodium symporter transporter family) and mediates cellular uptake of betaine and GABA in a sodium- and chloride-dependent process. Most of the studies of BGT1 concern its function and regulation in the kidney medulla where its role is best understood. The conditions here are hostile due to hyperosmolarity and significant concentrations of NH4Cl and urea. To withstand the hyperosmolarity, cells trigger osmotic adaptation, involving concentration of a transcriptional factor TonEBP/NFAT5 in the nucleus, and accumulate betaine and other osmolytes. Data from renal cells in culture, primarily MDCK, revealed that transcriptional regulation of BGT1 by TonEBP/NFAT5 is relatively slow. To allow more acute control of the abundance of BGT1 protein in the plasma membrane, there is also post-translation regulation of BGT1 protein trafficking which is dependent on intracellular calcium and ATP. Further, betaine may be important in liver metabolism as a methyl donor. In fact, in the mouse the liver is the organ with the highest content of BGT1. Hepatocytes express high levels of both BGT1 and the only enzyme that can metabolize betaine, namely betaine:homocysteine –S-methyltransferase (BHMT1). The BHMT1 enzyme removes a methyl group from betaine and transfers it to homocysteine, a potential risk factor for cardiovascular disease. Finally, BGT1 has been proposed to play a role in controlling brain excitability and thereby represents a target for anticonvulsive drug development. The latter hypothesis is controversial due to very low expression levels of BGT1 relative to other GABA transporters in brain, and also the primary location of BGT1 at the surface of the brain in the leptomeninges. These issues are discussed in detail.
Collapse
Affiliation(s)
- Stephen A Kempson
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine Indianapolis, IN, USA
| | - Yun Zhou
- Department of Anatomy, Centre of Molecular Biology and Neuroscience, Institute of Basic Medical Sciences, University of Oslo Oslo, Norway
| | - Niels C Danbolt
- Department of Anatomy, Centre of Molecular Biology and Neuroscience, Institute of Basic Medical Sciences, University of Oslo Oslo, Norway
| |
Collapse
|
41
|
Froger N, Moutsimilli L, Cadetti L, Jammoul F, Wang QP, Fan Y, Gaucher D, Rosolen SG, Neveux N, Cynober L, Sahel JA, Picaud S. Taurine: the comeback of a neutraceutical in the prevention of retinal degenerations. Prog Retin Eye Res 2014; 41:44-63. [PMID: 24721186 DOI: 10.1016/j.preteyeres.2014.03.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 03/11/2014] [Accepted: 03/12/2014] [Indexed: 12/21/2022]
Abstract
Taurine is the most abundant amino acid in the retina. In the 1970s, it was thought to be involved in retinal diseases with photoreceptor degeneration, because cats on a taurine-free diet presented photoreceptor loss. However, with the exception of its introduction into baby milk and parenteral nutrition, taurine has not yet been incorporated into any commercial treatment with the aim of slowing photoreceptor degeneration. Our recent discovery that taurine depletion is involved in the retinal toxicity of the antiepileptic drug vigabatrin has returned taurine to the limelight in the field of neuroprotection. However, although the retinal toxicity of vigabatrin principally involves a deleterious effect on photoreceptors, retinal ganglion cells (RGCs) are also affected. These findings led us to investigate the possible role of taurine depletion in retinal diseases with RGC degeneration, such as glaucoma and diabetic retinopathy. The major antioxidant properties of taurine may influence disease processes. In addition, the efficacy of taurine is dependent on its uptake into retinal cells, microvascular endothelial cells and the retinal pigment epithelium. Disturbances of retinal vascular perfusion in these retinal diseases may therefore affect the retinal uptake of taurine, resulting in local depletion. The low plasma taurine concentrations observed in diabetic patients may further enhance such local decreases in taurine concentration. We here review the evidence for a role of taurine in retinal ganglion cell survival and studies suggesting that this compound may be involved in the pathophysiology of glaucoma or diabetic retinopathy. Along with other antioxidant molecules, taurine should therefore be seriously reconsidered as a potential treatment for such retinal diseases.
Collapse
Affiliation(s)
- Nicolas Froger
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France.
| | - Larissa Moutsimilli
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France
| | - Lucia Cadetti
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France
| | - Firas Jammoul
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France
| | - Qing-Ping Wang
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France
| | - Yichao Fan
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France
| | - David Gaucher
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France; Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Paris, France; Nouvel hôpital civil, hôpitaux universitaires de Strasbourg and Laboratoire de Bactériologie (EA-7290), Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, France
| | - Serge G Rosolen
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France
| | - Nathalie Neveux
- Department of Nutrition, Faculty of Pharmacy, Paris Descartes University, Paris, France; Clinical Chemistry, Hôtel-Dieu-Cochin Hospitals, AP-HP, Paris, France
| | - Luc Cynober
- Department of Nutrition, Faculty of Pharmacy, Paris Descartes University, Paris, France; Clinical Chemistry, Hôtel-Dieu-Cochin Hospitals, AP-HP, Paris, France
| | - José-Alain Sahel
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France; Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Paris, France; Institute of Ophthalmology, University College of London, UK; Fondation Ophtalmologique Adolphe de Rothschild, Paris, France; French Academy of Sciences, Paris, France
| | - Serge Picaud
- INSERM, U968, Institut de la Vision, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie (Paris-6), UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Institut de la Vision, Paris, France; Fondation Ophtalmologique Adolphe de Rothschild, Paris, France.
| |
Collapse
|
42
|
Suárez LM, Bustamante J, Orensanz LM, Martín del Río R, Solís JM. Cooperation of taurine uptake and dopamine D1 receptor activation facilitates the induction of protein synthesis-dependent late LTP. Neuropharmacology 2014; 79:101-11. [DOI: 10.1016/j.neuropharm.2013.10.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 10/28/2013] [Accepted: 10/30/2013] [Indexed: 01/07/2023]
|
43
|
|
44
|
|
45
|
Askwith T. Taurine Treatment for Complications of Diabetes. OXIDATIVE STRESS IN APPLIED BASIC RESEARCH AND CLINICAL PRACTICE 2014. [DOI: 10.1007/978-1-4899-8035-9_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
46
|
Zhou Y, Waanders LF, Holmseth S, Guo C, Berger UV, Li Y, Lehre AC, Lehre KP, Danbolt NC. Proteome analysis and conditional deletion of the EAAT2 glutamate transporter provide evidence against a role of EAAT2 in pancreatic insulin secretion in mice. J Biol Chem 2013; 289:1329-44. [PMID: 24280215 DOI: 10.1074/jbc.m113.529065] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Islet function is incompletely understood in part because key steps in glutamate handling remain undetermined. The glutamate (excitatory amino acid) transporter 2 (EAAT2; Slc1a2) has been hypothesized to (a) provide islet cells with glutamate, (b) protect islet cells against high extracellular glutamate concentrations, (c) mediate glutamate release, or (d) control the pH inside insulin secretory granules. Here we floxed the EAAT2 gene to produce the first conditional EAAT2 knock-out mice. Crossing with Nestin-cyclization recombinase (Cre) eliminated EAAT2 from the brain, resulting in epilepsy and premature death, confirming the importance of EAAT2 for brain function and validating the genetic construction. Crossing with insulin-Cre lines (RIP-Cre and IPF1-Cre) to obtain pancreas-selective deletion did not appear to affect survival, growth, glucose tolerance, or β-cell number. We found (using TaqMan RT-PCR, immunoblotting, immunocytochemistry, and proteome analysis) that the EAAT2 levels were too low to support any of the four hypothesized functions. The proteome analysis detected more than 7,000 islet proteins of which more than 100 were transporters. Although mitochondrial glutamate transporters and transporters for neutral amino acids were present at high levels, all other transporters with known ability to transport glutamate were strikingly absent. Glutamate-metabolizing enzymes were abundant. The level of glutamine synthetase was 2 orders of magnitude higher than that of glutaminase. Taken together this suggests that the uptake of glutamate by islets from the extracellular fluid is insignificant and that glutamate is intracellularly produced. Glutamine synthetase may be more important for islets than assumed previously.
Collapse
Affiliation(s)
- Yun Zhou
- From The Neurotransporter Group, Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Usui T, Kubo Y, Akanuma SI, Hosoya KI. β-Alanine and l-histidine transport across the inner blood-retinal barrier: Potential involvement in l-carnosine supply. Exp Eye Res 2013; 113:135-42. [DOI: 10.1016/j.exer.2013.06.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 05/18/2013] [Accepted: 06/03/2013] [Indexed: 11/25/2022]
|
48
|
Kletke O, Gisselmann G, May A, Hatt H, A. Sergeeva O. Partial agonism of taurine at gamma-containing native and recombinant GABAA receptors. PLoS One 2013; 8:e61733. [PMID: 23637894 PMCID: PMC3640040 DOI: 10.1371/journal.pone.0061733] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 03/13/2013] [Indexed: 11/18/2022] Open
Abstract
Taurine is a semi-essential sulfonic acid found at high concentrations in plasma and mammalian tissues which regulates osmolarity, ion channel activity and glucose homeostasis. The structural requirements of GABAA-receptors (GABAAR) gated by taurine are not yet known. We determined taurine potency and efficacy relative to GABA at different types of recombinant GABAAR occurring in central histaminergic neurons of the mouse hypothalamic tuberomamillary nucleus (TMN) which controls arousal. At binary α1/2β1/3 receptors taurine was as efficient as GABA, whereas incorporation of the γ1/2 subunit reduced taurine efficacy to 60–90% of GABA. The mutation γ2F77I, which abolishes zolpidem potentiation, significantly reduced taurine efficacy at recombinant and native receptors compared to the wild type controls. As taurine was a full- or super- agonist at recombinant αxβ1δ-GABAAR, we generated a chimeric γ2 subunit carrying the δ subunit motif around F77 (MTVFLH). At α1/2β1γ2(MTVFLH) receptors taurine became a super-agonist, similar to δ-containing ternary receptors, but remained a partial agonist at β3-containing receptors. In conclusion, using site-directed mutagenesis we found structural determinants of taurine’s partial agonism at γ-containing GABAA receptors. Our study sheds new light on the β1 subunit conferring the widest range of taurine-efficacies modifying GABAAR function under (patho)physiological conditions.
Collapse
Affiliation(s)
- Olaf Kletke
- Department of Cell Physiology of the Ruhr-University, Bochum, Germany
- Department of Neurophysiology, Medical Faculty of Heinrich-Heine University, Düsseldorf, Germany
| | | | - Andrea May
- Department of Neurophysiology, Medical Faculty of Heinrich-Heine University, Düsseldorf, Germany
| | - Hanns Hatt
- Department of Cell Physiology of the Ruhr-University, Bochum, Germany
| | - Olga A. Sergeeva
- Department of Neurophysiology, Medical Faculty of Heinrich-Heine University, Düsseldorf, Germany
- * E-mail:
| |
Collapse
|
49
|
Wu Y, Janetopoulos C. Systematic analysis of γ-aminobutyric acid (GABA) metabolism and function in the social amoeba Dictyostelium discoideum. J Biol Chem 2013; 288:15280-90. [PMID: 23548898 DOI: 10.1074/jbc.m112.427047] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
While GABA has been suggested to regulate spore encapsulation in the social amoeba Dictyostelium discoideum, the metabolic profile and other potential functions of GABA during development remain unclear. In this study, we investigated the homeostasis of GABA metabolism by disrupting genes related to GABA metabolism and signaling. Extracellular levels of GABA are tightly regulated during early development, and GABA is generated by the glutamate decarboxylase, GadB, during growth and in early development. However, overexpression of the prespore-specific homologue, GadA, in the presence of GadB reduces production of extracellular GABA. Perturbation of extracellular GABA levels delays the process of aggregation. Cytosolic GABA is degraded by the GABA transaminase, GabT, in the mitochondria. Disruption of a putative vesicular GABA transporter (vGAT) homologue DdvGAT reduces secreted GABA. We identified the GABAB receptor-like family member GrlB as the major GABA receptor during early development, and either disruption or overexpression of GrlB delays aggregation. This delay is likely the result of an abolished pre-starvation response and late expression of several "early" developmental genes. Distinct genes are employed for GABA generation during sporulation. During sporulation, GadA alone is required for generating GABA and DdvGAT is likely responsible for GABA secretion. GrlE but not GrlB is the GABA receptor during late development.
Collapse
Affiliation(s)
- Yuantai Wu
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | |
Collapse
|
50
|
Abstract
The solute carrier 6 (SLC6) family of the human genome comprises transporters for neurotransmitters, amino acids, osmolytes and energy metabolites. Members of this family play critical roles in neurotransmission, cellular and whole body homeostasis. Malfunction or altered expression of these transporters is associated with a variety of diseases. Pharmacological inhibition of the neurotransmitter transporters in this family is an important strategy in the management of neurological and psychiatric disorders. This review provides an overview of the biochemical and pharmacological properties of the SLC6 family transporters.
Collapse
Affiliation(s)
- Stefan Bröer
- Research School of Biology, Australian National University, Canberra, ACT, Australia.
| | | |
Collapse
|