1
|
Lewis JH, Zhou Y, Lu Z. Examination of conformational dynamics of AdiC transporter with fluorescence-polarization microscopy. J Gen Physiol 2025; 157:e202413709. [PMID: 39976549 PMCID: PMC11841622 DOI: 10.1085/jgp.202413709] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/19/2024] [Accepted: 01/19/2025] [Indexed: 02/23/2025] Open
Abstract
To understand the mechanism underlying the ability of individual AdiC molecules to transport arginine and agmatine, we used a recently developed high-resolution single-molecule fluorescence-polarization microscopy method to investigate conformation-specific changes in the emission polarization of a bifunctional fluorophore attached to an AdiC molecule. With this capability, we resolved AdiC's four conformations characterized by distinct spatial orientations in the absence or presence of the two substrates, and furthermore, each conformation's two energetic states, totaling 24 states. From the lifetimes of individual states and state-to-state transition probabilities, we determined 60 rate constants characterizing the transitions and 4 KD values characterizing the interactions of AdiC's two sides with arginine and agmatine, quantitatively defining a 24-state model. This model satisfactorily predicts the observed Michaelis-Menten behaviors of AdiC. With the acquired temporal information and existing structural information, we illustrated how to build an experiment-based integrative 4D model to capture and exhibit the complex spatiotemporal mechanisms underlying facilitated transport of substrates. However, inconsistent with what is expected from the prevailing hypothesis that AdiC is a 1:1 exchanger, all observed conformations transitioned among themselves with or without the presence of substrates. To corroborate this unexpected finding, we performed radioactive flux assays and found that the results are also incompatible with the hypothesis. As a technical advance, we showed that a monofunctional and the standard bifunctional fluorophore labels report comparable spatial orientation information defined in a local frame of reference. Here, the successful determination of the complex conformation-kinetic mechanism of AdiC demonstrates the unprecedented resolving power of the present microscopy method.
Collapse
Affiliation(s)
- John H. Lewis
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yufeng Zhou
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhe Lu
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
2
|
Dai L, Zeng Q, Zhang T, Zhang Y, Shi Y, Li Y, Xu K, Huang J, Wang Z, Zhou Q, Yan R. Structural basis for the substrate recognition and transport mechanism of the human y +LAT1-4F2hc transporter complex. SCIENCE ADVANCES 2025; 11:eadq0558. [PMID: 40106545 PMCID: PMC11922002 DOI: 10.1126/sciadv.adq0558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 02/12/2025] [Indexed: 03/22/2025]
Abstract
Heteromeric amino acid transporters (HATs), including y+LAT1-4F2hc complex, are responsible for transporting amino acids across membranes, and mutations in y+LAT1 cause lysinuric protein intolerance (LPI), a hereditary disorder characterized by defective cationic amino acid transport. The relationship between LPI and specific mutations in y+LAT1 has yet to be fully understood. In this study, we characterized the function of y+LAT1-4F2hc complex in mammalian cells and determined the cryo-EM structures of the human y+LAT1-4F2hc complex in two distinct conformations: the apo state in an inward-open conformation and the native substrate-bound state in an outward-open conformation. Structural analysis suggests that Asp243 in y+LAT1 plays a crucial role in coordination with sodium ion and substrate selectivity. Molecular dynamic (MD) simulations further revealed the different transport mechanism of cationic amino acids and neutral amino acids. These results provide important insights into the mechanisms of the substrate binding and working cycle of HATs.
Collapse
Affiliation(s)
- Lu Dai
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
- Key University Laboratory of Metabolism and Health of Guangdong, Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Qian Zeng
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
- Key University Laboratory of Metabolism and Health of Guangdong, Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Ting Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
- Key University Laboratory of Metabolism and Health of Guangdong, Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Yuanyuan Zhang
- Research Center for Industries of the Future, Zhejiang Key Laboratory of Structural Biology, School of Life Sciences, Westlake University, Institute of Biology, Westlake Institute for Advanced Study, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China
| | - Yi Shi
- Research Center for Industries of the Future, Zhejiang Key Laboratory of Structural Biology, School of Life Sciences, Westlake University, Institute of Biology, Westlake Institute for Advanced Study, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China
| | - Yaning Li
- Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Kangtai Xu
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
- Key University Laboratory of Metabolism and Health of Guangdong, Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Jing Huang
- Research Center for Industries of the Future, Zhejiang Key Laboratory of Structural Biology, School of Life Sciences, Westlake University, Institute of Biology, Westlake Institute for Advanced Study, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China
| | - Zilong Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
- Key University Laboratory of Metabolism and Health of Guangdong, Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Qiang Zhou
- Research Center for Industries of the Future, Zhejiang Key Laboratory of Structural Biology, School of Life Sciences, Westlake University, Institute of Biology, Westlake Institute for Advanced Study, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China
| | - Renhong Yan
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
- Key University Laboratory of Metabolism and Health of Guangdong, Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| |
Collapse
|
3
|
Scalise M, Scanga R, Console L, Galluccio M, Pochini L, Indiveri C. Lysine 204 is crucial for the antiport function of the human LAT1 transporter. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2025; 1866:149520. [PMID: 39428051 DOI: 10.1016/j.bbabio.2024.149520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/11/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
LAT1 (SLC7A5) catalyzes an antiport reaction of amino acids with specificity towards the essential ones. It is mainly expressed at the Blood Brain Barrier and placenta barriers, but it becomes over-expressed in virtually all human cancers even if originating from tissues with lower expression levels. The antiport reaction of LAT1 is crucial at the BBB since its inherited loss causes Autism Spectrum Disorder. We have investigated the possible molecular determinant of the antiport by site-directed mutagenesis, in vitro transport assay and computational analysis. Previous data indicated that mutation of K204 impairs, but does not knock-out LAT1 functionality. We have investigated the activity changes in the K204Q mutant by following the transport of [3H]-histidine, one of the major substrates, in proteoliposomes harbouring the WT or K204Q. In the mutant, the [3H]-histidine uptake and efflux are not more stimulated by the counter-substrate as they occur in the WT. Moreover, the mutation strongly decreases the substrate affinity and alters the pH dependence of K204Q. Molecular Dynamics analysis correlates well with the experimental data since it shows that substrate prematurely escapes the binding site. In addition, the K204Q shows a strongly increased mobility in those regions, transmembrane domains and random coils, involved in the transport cycle. The identified Lys residue could be responsible of the same phenomenon in those members of the SLC7 family, described as antiporters, in which it is conserved.
Collapse
Affiliation(s)
- Mariafrancesca Scalise
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy
| | - Raffaella Scanga
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy
| | - Lara Console
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy
| | - Michele Galluccio
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy
| | - Lorena Pochini
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy; CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, via Amendola 165/A, 70126 Bari, Italy.
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy; CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, via Amendola 165/A, 70126 Bari, Italy.
| |
Collapse
|
4
|
Fort J, Nicolàs-Aragó A, Maggi L, Martinez-Molledo M, Kapiki D, González-Novoa P, Gómez-Gejo P, Zijlstra N, Bodoy S, Pardon E, Steyaert J, Llorca O, Orozco M, Cordes T, Palacín M. The conserved lysine residue in transmembrane helix 5 is pivotal for the cytoplasmic gating of the L-amino acid transporters. PNAS NEXUS 2025; 4:pgae584. [PMID: 39822574 PMCID: PMC11736713 DOI: 10.1093/pnasnexus/pgae584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 12/18/2024] [Indexed: 01/19/2025]
Abstract
L-Amino acid transporters (LATs) play a key role in a wide range of physiological processes. Defects in LATs can lead to neurological disorders and aminoacidurias, while the overexpression of these transporters is related to cancer. BasC is a bacterial LAT transporter with an APC fold. In this study, to monitor the cytoplasmic motion of BasC, we developed a single-molecule Förster resonance energy transfer assay that can characterize the conformational states of the intracellular gate in solution at room temperature. Based on combined biochemical and biophysical data and molecular dynamics simulations, we propose a model in which the conserved lysine residue in TM5 supports TM1a to explore both open and closed states within the cytoplasmic gate under apo conditions. This equilibrium can be altered by substrates, mutation of conserved lysine 154 in TM5, or a transport-blocking nanobody interacting with TM1a. Overall, these findings provide insights into the transport mechanism of BasC and highlight the significance of the lysine residue in TM5 in the cytoplasmic gating of LATs.
Collapse
Affiliation(s)
- Joana Fort
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Universitat de Barcelona (UB), Diagonal 643, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Adrià Nicolàs-Aragó
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Luca Maggi
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain
| | - Maria Martinez-Molledo
- Structural Biology Programme, Spanish National Cancer Research Centre, 28029 Centro Nacional de Investigaciones Oncológicas (CNIO), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Despoina Kapiki
- Physical and Synthetic Biology, Faculty of Biology, Ludwig-Maximilians-Universität München, Großhadernerstr. 2-4, 82152 Planegg-Martinsried, Germany
| | - Paula González-Novoa
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Universitat de Barcelona (UB), Diagonal 643, 08028 Barcelona, Spain
| | - Patricia Gómez-Gejo
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain
| | - Niels Zijlstra
- Physical and Synthetic Biology, Faculty of Biology, Ludwig-Maximilians-Universität München, Großhadernerstr. 2-4, 82152 Planegg-Martinsried, Germany
| | - Susanna Bodoy
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Biosciences, Universitat de Vic—Universitat Central de Catalunya, de la Laura 13, 08500 Vic, Spain
| | - Els Pardon
- VIB-VUB Center for Structural Biology, VIB, Pleinaan 2, 1050 Brussel, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinaan 2, 1050 Brussel, Belgium
| | - Jan Steyaert
- VIB-VUB Center for Structural Biology, VIB, Pleinaan 2, 1050 Brussel, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinaan 2, 1050 Brussel, Belgium
| | - Oscar Llorca
- Structural Biology Programme, Spanish National Cancer Research Centre, 28029 Centro Nacional de Investigaciones Oncológicas (CNIO), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Modesto Orozco
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Universitat de Barcelona (UB), Diagonal 643, 08028 Barcelona, Spain
| | - Thorben Cordes
- Physical and Synthetic Biology, Faculty of Biology, Ludwig-Maximilians-Universität München, Großhadernerstr. 2-4, 82152 Planegg-Martinsried, Germany
- Biophysical Chemistry, Department of Chemistry and Chemical Biology, Technische Universität Dortmund, Otto-Hahn-Str. 4a, 44227 Dortmund, Germany
| | - Manuel Palacín
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, 08028 Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Universitat de Barcelona (UB), Diagonal 643, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
5
|
Hossen MS, Islam MSU, Yasin M, Ibrahim M, Das A. A Review on the Role of Human Solute Carriers Transporters in Cancer. Health Sci Rep 2025; 8:e70343. [PMID: 39807482 PMCID: PMC11725534 DOI: 10.1002/hsr2.70343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/03/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Background and Aim The high rate of tumor growth results in an increased need for amino acids. As solute carriers (SLC) transporters are capable of transporting different amino acids, cancer may develop as a result of these transporters' over-expression due to their complex formation with other biological molecules. Therefore, this review investigated the role of SLC transporters in the progression of cancer. Methods We retrieved data from Google Scholar, Web of Science, PubMed, Cochrane Library, and EMBASE regarding the influence of human SLCs on the development of cancer. Articles published in English before August 2024 were included in the study. Results The overexpression of SLCs is strongly related to tumor cell proliferation and angiogenesis in a number of cancer types including thyroid, pancreatic, lung, hepatocellular, and colon cancers. They are crucial for the stimulation of several biological signaling pathways, particularly mTOR kinase activity, which starts a signaling cascade, protein synthesis, cell growth, and proliferation, and inhibits apoptosis of cancerous cells. Furthermore, they contribute to the activation of PI3K/AKT signaling, which has an impact on the growth, invasion, and death of cancer cells. Thus, SLC transporters become a potential therapeutic target that plays a crucial role in drug resistance, tumor microenvironment regulation, and modulation of immune response. Conclusion The review recognized the crucial role of SLC transporters in different types of cancer progression. Therefore, to confirm our findings, a case-control study is required to investigate the role of amino acid transporters in cancer development.
Collapse
Affiliation(s)
- Md. Shafiul Hossen
- Department of PharmacyState University of BangladeshDhakaBangladesh
- Department of PharmacyNoakhali Science and Technology UniversitySonapurBangladesh
| | | | - Mohammad Yasin
- Department of PharmacySouthern University BangladeshChittagongBangladesh
| | - Mohammed Ibrahim
- Department of PharmacyState University of BangladeshDhakaBangladesh
| | - Abhijit Das
- Department of PharmacyNoakhali Science and Technology UniversitySonapurBangladesh
| |
Collapse
|
6
|
Zhang Y, Cai J, Hosmane NS, Zhu Y. In silico assessments of the small molecular boron agents to pave the way for artificial intelligence-based boron neutron capture therapy. Eur J Med Chem 2024; 279:116841. [PMID: 39244862 DOI: 10.1016/j.ejmech.2024.116841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/28/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024]
Abstract
Boron neutron capture therapy (BNCT) is a highly targeted, selective and effective technique to cure various types of cancers, with less harm to the healthy cells. In principle, BNCT treatment needs to distribute the 10boron (10B) atoms inside the tumor tissues, selectively and homogeneously, as well as to initiate a nuclear fission reaction by capturing sufficient neutrons which releases high linear energy particles to kill the tumor cells. In BNCT, it is crucial to have high quality boron agents with acceptable bio-selectivity, homogeneous distribution and deliver in required quantity, similar to chemotherapy and other radiotherapy for tumor treatment. Nevertheless, boron drugs currently used in clinical trials yet to meet the full requirements. On the other hand, BNCT processing has opened up the era of renaissance due to the advanced development of the high-quality neutron source and the global construction of new BNCT centers. Consequently, there is an urgent need to use boron agents that have increased biocapacity. Artificial intelligence (AI) tools such as molecular docking and molecular dynamic simulation technologies have been utilized to develop new medicines. In this work, the in silico assessments including bioinformatics assessments of BNCT related tumoral receptor proteins, computational assessments of optimized small molecules of boron agents, are employed to speed up the screening process for boron drugs. The outcomes will be applicable to pave the way for future BNCT that utilizes artificial intelligence. The in silico molecular docking and dynamic simulation results of the optimized small boron agents, such as 4-borono-l-phenylalanine (BPA) with optimized proteins like the L-type amino acid transporter 1 (LTA1, also known as SLC7A5) will be examined. The in silico assessments results will certainly be helpful to researchers in optimizing druggable boron agents for the BNCT application. The clinical status of the optimized proteins, which are highly relevant to cancers that may be treated with BNCT, has been assessed using bioinformatics technology and discussed accordingly. Furthermore, the evaluations of cytotoxicity (IC50), boron uptake and tissue distribution of the optimized ligands 1 and 7 have been presented.
Collapse
Affiliation(s)
- Yingjun Zhang
- Sunshine Lake Pharma Co. Ltd., Dongguan, 523871, China
| | - Jianghong Cai
- School of Pharmacy, Macau University of Science and Technology, Macau, 999078, China
| | - Narayan S Hosmane
- Department of Chemistry and Biochemistry, Northern Illinois University, DeKalb, IL, USA
| | - Yinghuai Zhu
- Sunshine Lake Pharma Co. Ltd., Dongguan, 523871, China.
| |
Collapse
|
7
|
Cimadamore-Werthein C, King MS, Lacabanne D, Pyrihová E, Jaiquel Baron S, Kunji ER. Human mitochondrial carriers of the SLC25 family function as monomers exchanging substrates with a ping-pong kinetic mechanism. EMBO J 2024; 43:3450-3465. [PMID: 38937634 PMCID: PMC11329753 DOI: 10.1038/s44318-024-00150-0] [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: 03/14/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/29/2024] Open
Abstract
Members of the SLC25 mitochondrial carrier family link cytosolic and mitochondrial metabolism and support cellular maintenance and growth by transporting compounds across the mitochondrial inner membrane. Their monomeric or dimeric state and kinetic mechanism have been a matter of long-standing debate. It is believed by some that they exist as homodimers and transport substrates with a sequential kinetic mechanism, forming a ternary complex where both exchanged substrates are bound simultaneously. Some studies, in contrast, have provided evidence indicating that the mitochondrial ADP/ATP carrier (SLC25A4) functions as a monomer, has a single substrate binding site, and operates with a ping-pong kinetic mechanism, whereby ADP is imported before ATP is exported. Here we reanalyze the oligomeric state and kinetic properties of the human mitochondrial citrate carrier (SLC25A1), dicarboxylate carrier (SLC25A10), oxoglutarate carrier (SLC25A11), and aspartate/glutamate carrier (SLC25A13), all previously reported to be dimers with a sequential kinetic mechanism. We demonstrate that they are monomers, except for dimeric SLC25A13, and operate with a ping-pong kinetic mechanism in which the substrate import and export steps occur consecutively. These observations are consistent with a common transport mechanism, based on a functional monomer, in which a single central substrate-binding site is alternately accessible.
Collapse
Affiliation(s)
- Camila Cimadamore-Werthein
- Medical Research Council Mitochondrial Biology Unit, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, United Kingdom
| | - Martin S King
- Medical Research Council Mitochondrial Biology Unit, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, United Kingdom
| | - Denis Lacabanne
- Medical Research Council Mitochondrial Biology Unit, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, United Kingdom
| | - Eva Pyrihová
- Medical Research Council Mitochondrial Biology Unit, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, United Kingdom
| | - Stephany Jaiquel Baron
- Medical Research Council Mitochondrial Biology Unit, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, United Kingdom
| | - Edmund Rs Kunji
- Medical Research Council Mitochondrial Biology Unit, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, United Kingdom.
| |
Collapse
|
8
|
Rullo-Tubau J, Martinez-Molledo M, Bartoccioni P, Puch-Giner I, Arias Á, Saen-Oon S, Stephan-Otto Attolini C, Artuch R, Díaz L, Guallar V, Errasti-Murugarren E, Palacín M, Llorca O. Structure and mechanisms of transport of human Asc1/CD98hc amino acid transporter. Nat Commun 2024; 15:2986. [PMID: 38582862 PMCID: PMC10998858 DOI: 10.1038/s41467-024-47385-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 03/29/2024] [Indexed: 04/08/2024] Open
Abstract
Recent cryoEM studies elucidated details of the structural basis for the substrate selectivity and translocation of heteromeric amino acid transporters. However, Asc1/CD98hc is the only neutral heteromeric amino acid transporter that can function through facilitated diffusion, and the only one that efficiently transports glycine and D-serine, and thus has a regulatory role in the central nervous system. Here we use cryoEM, ligand-binding simulations, mutagenesis, transport assays, and molecular dynamics to define human Asc1/CD98hc determinants for substrate specificity and gain insights into the mechanisms that govern substrate translocation by exchange and facilitated diffusion. The cryoEM structure of Asc1/CD98hc is determined at 3.4-3.8 Å resolution, revealing an inward-facing semi-occluded conformation. We find that Ser 246 and Tyr 333 are essential for Asc1/CD98hc substrate selectivity and for the exchange and facilitated diffusion modes of transport. Taken together, these results reveal the structural bases for ligand binding and transport features specific to human Asc1.
Collapse
Affiliation(s)
- Josep Rullo-Tubau
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, E-08028, Barcelona, Spain
| | - Maria Martinez-Molledo
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro, 3, E-28029, Madrid, Spain
| | - Paola Bartoccioni
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, E-08028, Barcelona, Spain
- The Spanish Center of Rare Diseases (CIBERER U-731), Baldiri Reixac 10, E-08028, Barcelona, Spain
| | - Ignasi Puch-Giner
- Electronic and atomic protein modelling group, Barcelona Supercomputing Center, Plaça d'Eusebi Güell, 1-3, E-08034, Barcelona, Spain
| | - Ángela Arias
- Clinical Biochemistry Department, Sant Joan de Déu Research Institute, Pg. de Sant Joan de Déu, 2, E-08950, Esplugues de Llobregat, Spain
| | - Suwipa Saen-Oon
- Nostrum Biodiscovery, Av. de Josep Tarradellas, 8-10, E-08029, Barcelona, Spain
| | - Camille Stephan-Otto Attolini
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, E-08028, Barcelona, Spain
| | - Rafael Artuch
- The Spanish Center of Rare Diseases (CIBERER U-731), Baldiri Reixac 10, E-08028, Barcelona, Spain
- Clinical Biochemistry Department, Sant Joan de Déu Research Institute, Pg. de Sant Joan de Déu, 2, E-08950, Esplugues de Llobregat, Spain
| | - Lucía Díaz
- Nostrum Biodiscovery, Av. de Josep Tarradellas, 8-10, E-08029, Barcelona, Spain
| | - Víctor Guallar
- Electronic and atomic protein modelling group, Barcelona Supercomputing Center, Plaça d'Eusebi Güell, 1-3, E-08034, Barcelona, Spain
- Nostrum Biodiscovery, Av. de Josep Tarradellas, 8-10, E-08029, Barcelona, Spain
| | - Ekaitz Errasti-Murugarren
- The Spanish Center of Rare Diseases (CIBERER U-731), Baldiri Reixac 10, E-08028, Barcelona, Spain.
- Physiological Sciences Department, Genetics Area, School of Medicine and Health Sciences, University of Barcelona, Bellvitge Campus. Feixa Llarga s/n, E-08907, L'Hospitalet de Llobregat, Spain.
- Human Molecular Genetics Laboratory, Gene, Disease and Therapy Program, IDIBELL, Hospital Duran i Reynals, Avd. Gran Via de L'Hospitalet 199, E-08908, L'Hospitalet de Llobregat, Spain.
| | - Manuel Palacín
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, E-08028, Barcelona, Spain.
- The Spanish Center of Rare Diseases (CIBERER U-731), Baldiri Reixac 10, E-08028, Barcelona, Spain.
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Av. Diagonal, 643, E-08028, Barcelona, Spain.
| | - Oscar Llorca
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro, 3, E-28029, Madrid, Spain.
| |
Collapse
|
9
|
Fordham TM, Morelli NS, Garcia-Reyes Y, Ware MA, Rahat H, Sundararajan D, Fuller KNZ, Severn C, Pyle L, Malloy CR, Jin ES, Parks EJ, Wolfe RR, Cree MG. Metabolic effects of an essential amino acid supplement in adolescents with PCOS and obesity. Obesity (Silver Spring) 2024; 32:678-690. [PMID: 38439205 DOI: 10.1002/oby.23988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 03/06/2024]
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS) is characterized by hyperandrogenism, insulin resistance, and hepatic steatosis (HS). Because dietary essential amino acid (EAA) supplementation has been shown to decrease HS in various populations, this study's objective was to determine whether supplementation would decrease HS in PCOS. METHODS A randomized, double-blind, crossover, placebo-controlled trial was conducted in 21 adolescents with PCOS (BMI 37.3 ± 6.5 kg/m2, age 15.6 ± 1.3 years). Liver fat, very low-density lipoprotein (VLDL) lipogenesis, and triacylglycerol (TG) metabolism were measured following each 28-day phase of placebo or EAA. RESULTS Compared to placebo, EAA was associated with no difference in body weight (p = 0.673). Two markers of liver health improved: HS was lower (-0.8% absolute, -7.5% relative reduction, p = 0.013), as was plasma aspartate aminotransferase (AST) (-8%, p = 0.004). Plasma TG (-9%, p = 0.015) and VLDL-TG (-21%, p = 0.031) were reduced as well. VLDL-TG palmitate derived from lipogenesis was not different between the phases, nor was insulin sensitivity (p > 0.400 for both). Surprisingly, during the EAA phase, participants reported consuming fewer carbohydrates (p = 0.038) and total sugars (p = 0.046). CONCLUSIONS Similar to studies in older adults, short-term EAA supplementation in adolescents resulted in significantly lower liver fat, AST, and plasma lipids and thus may prove to be an effective treatment in this population. Additional research is needed to elucidate the mechanisms for these effects.
Collapse
Affiliation(s)
- Talyia M Fordham
- Department of Nutrition and Exercise Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Nazeen S Morelli
- Department of Pediatrics, Section on Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Yesenia Garcia-Reyes
- Department of Pediatrics, Section on Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Meredith A Ware
- Department of Pediatrics, Section on Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Haseeb Rahat
- Department of Pediatrics, Section on Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Divya Sundararajan
- Department of Pediatrics, Section on Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kelly N Z Fuller
- Department of Pediatrics, Section on Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Cameron Severn
- Child Health Biostatistics Core, Department of Pediatrics, Section of Endocrinology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laura Pyle
- Child Health Biostatistics Core, Department of Pediatrics, Section of Endocrinology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Craig R Malloy
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- VA North Texas Health Care System, Dallas, Texas, USA
| | - Eunsook S Jin
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Elizabeth J Parks
- Department of Nutrition and Exercise Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Robert R Wolfe
- Department of Geriatrics, Donald W. Reynolds Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Melanie G Cree
- Department of Pediatrics, Section on Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Center for Women's Health Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
10
|
Pranav, Bajpai A, Dwivedi PK, Sivakumar S. Chiral nanomaterial-based approaches for diagnosis and treatment of protein-aggregated neurodiseases: current status and future opportunities. J Mater Chem B 2024; 12:1991-2005. [PMID: 38333942 DOI: 10.1039/d3tb02381h] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Protein misfolding and its aggregation, known as amyloid aggregates (Aβ), are some of the major causes of more than 20 diseases such as Parkinson's disease, Alzheimer's disease, and type 2 diabetes. The process of Aβ formation involves an energy-driven oligomerization of Aβ monomers, leading to polymerization and eventual aggregation into fibrils. Aβ fibrils exhibit multilevel chirality arising from its amino acid residues and the arrangement of folded polypeptide chains; thus, a chirality-driven approach can be utilized for the detection and inhibition of Aβ fibrils. In this regard, chiral nanomaterials have recently opened new possibilities for various biomedical applications owing to their stereoselective interaction with biological systems. Leveraging this chirality-driven approach with chiral nanomaterials against protein-aggregated diseases could yield promising results, particularly in the early detection of Aβ forms and the inhibition of Aβ aggregate formation via specific and strong "chiral-chiral interaction." Despite the advantages, the development of advanced theranostic systems using chiral nanomaterials against protein-aggregated diseases has received limited attention so far because of considerably limited formulations for chiral nanomaterials and lack of information of their chiroptical behavior. This review aims to present the current status of chiral nanomaterials explored for detecting and inhibiting Aβ forms. This review covers the origin of chirality in amyloid fibrils and nanomaterials and different chiral detection methods; furthermore, different chiral nanosystems such as chiral plasmonic nanomaterials, chiral carbon-based nanomaterials, and chiral nanosurfaces, which have been used so far for different therapeutic applications against protein-aggregated diseases, are discussed in detail. The findings from this review may pave the way for the development of novel approaches using chiral nanomaterials to combat diseases resulting from protein misfolding and can further be extended to other disease forms.
Collapse
Affiliation(s)
- Pranav
- Centre for Nanosciences, Indian Institute of Technology, Kanpur 208016, India.
| | - Abhishek Bajpai
- Centre for Nanosciences, Indian Institute of Technology, Kanpur 208016, India.
| | - Prabhat K Dwivedi
- Centre for Nanosciences, Indian Institute of Technology, Kanpur 208016, India.
| | - Sri Sivakumar
- Centre for Nanosciences, Indian Institute of Technology, Kanpur 208016, India.
- Department of Chemical Engineering, Indian Institute of Technology, Kanpur 208016, India
- Materials Science Program, Indian Institute of Technology, Kanpur 208016, India
- Centre for Environmental Science and Engineering, India
| |
Collapse
|
11
|
Galluccio M, Mazza T, Scalise M, Tripicchio M, Scarpelli M, Tolomeo M, Pochini L, Indiveri C. Over-Production of the Human SLC7A10 in E. coli and Functional Assay in Proteoliposomes. Int J Mol Sci 2023; 25:536. [PMID: 38203703 PMCID: PMC10779382 DOI: 10.3390/ijms25010536] [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: 11/30/2023] [Revised: 12/28/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
The human SLC7A10 transporter, also known as ASC-1, catalyzes the transport of some neutral amino acids. It is expressed in astrocytes, neurons, and adipose tissues, playing roles in learning, memory processes, and lipid metabolism, thus being involved in neurological and metabolic pathologies. Structure/function studies on this transporter are still in their infancy. In this study, we present a methodology for producing the recombinant human transporter in E. coli. Its transport function was assayed in proteoliposomes following the uptake of radiolabeled L-serine. After the testing of several growth conditions, the hASC-1 transporter was successfully expressed in BL21(DE3) codon plus RIL in the presence of 0.5% glucose and induced with 0.05 mM IPTG. After solubilization with C12E8 and cholesteryl hemisuccinate and purification by Ni-chelating chromatography, hASC-1 was reconstituted in proteoliposomes. In this experimental system it was able to catalyze an Na+-independent homologous antiport of L-serine. A Km for L-serine transport of 0.24 mM was measured. The experimental model developed in this work represents a reproducible system for the transport assay of hASC-1 in the absence of interferences. This tool will be useful to unveil unknown transport properties of hASC-1 and for testing ligands with possible application in human pharmacology.
Collapse
Affiliation(s)
- Michele Galluccio
- Laboratory of Biochemistry, Molecular Biotechnology, and Molecular Biology, Department DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (T.M.); (M.S.); (M.T.); (M.S.); (M.T.); (L.P.)
| | - Tiziano Mazza
- Laboratory of Biochemistry, Molecular Biotechnology, and Molecular Biology, Department DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (T.M.); (M.S.); (M.T.); (M.S.); (M.T.); (L.P.)
| | - Mariafrancesca Scalise
- Laboratory of Biochemistry, Molecular Biotechnology, and Molecular Biology, Department DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (T.M.); (M.S.); (M.T.); (M.S.); (M.T.); (L.P.)
| | - Martina Tripicchio
- Laboratory of Biochemistry, Molecular Biotechnology, and Molecular Biology, Department DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (T.M.); (M.S.); (M.T.); (M.S.); (M.T.); (L.P.)
| | - Martina Scarpelli
- Laboratory of Biochemistry, Molecular Biotechnology, and Molecular Biology, Department DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (T.M.); (M.S.); (M.T.); (M.S.); (M.T.); (L.P.)
| | - Maria Tolomeo
- Laboratory of Biochemistry, Molecular Biotechnology, and Molecular Biology, Department DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (T.M.); (M.S.); (M.T.); (M.S.); (M.T.); (L.P.)
| | - Lorena Pochini
- Laboratory of Biochemistry, Molecular Biotechnology, and Molecular Biology, Department DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (T.M.); (M.S.); (M.T.); (M.S.); (M.T.); (L.P.)
- National Research Council (CNR), Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Via Amendola 122/O, 70126 Bari, Italy
| | - Cesare Indiveri
- Laboratory of Biochemistry, Molecular Biotechnology, and Molecular Biology, Department DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (T.M.); (M.S.); (M.T.); (M.S.); (M.T.); (L.P.)
- National Research Council (CNR), Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Via Amendola 122/O, 70126 Bari, Italy
| |
Collapse
|
12
|
Drehmann P, Milanos S, Schaefer N, Kasaragod VB, Herterich S, Holzbach-Eberle U, Harvey RJ, Villmann C. Dual Role of Dysfunctional Asc-1 Transporter in Distinct Human Pathologies, Human Startle Disease, and Developmental Delay. eNeuro 2023; 10:ENEURO.0263-23.2023. [PMID: 37903619 PMCID: PMC10668224 DOI: 10.1523/eneuro.0263-23.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/11/2023] [Indexed: 11/01/2023] Open
Abstract
Human startle disease is associated with mutations in distinct genes encoding glycine receptors, transporters or interacting proteins at glycinergic synapses in spinal cord and brainstem. However, a significant number of diagnosed patients does not carry a mutation in the common genes GLRA1, GLRB, and SLC6A5 Recently, studies on solute carrier 7 subfamily 10 (SLC7A10; Asc-1, alanine-serine-cysteine transporter) knock-out (KO) mice displaying a startle disease-like phenotype hypothesized that this transporter might represent a novel candidate for human startle disease. Here, we screened 51 patients from our patient cohort negative for the common genes and found three exonic (one missense, two synonymous), seven intronic, and single nucleotide changes in the 5' and 3' untranslated regions (UTRs) in Asc-1. The identified missense mutation Asc-1G307R from a patient with startle disease and developmental delay was investigated in functional studies. At the molecular level, the mutation Asc-1G307R did not interfere with cell-surface expression, but disrupted glycine uptake. Substitution of glycine at position 307 to other amino acids, e.g., to alanine or tryptophan did not affect trafficking or glycine transport. By contrast, G307K disrupted glycine transport similar to the G307R mutation found in the patient. Structurally, the disrupted function in variants carrying positively charged residues can be explained by local structural rearrangements because of the large positively charged side chain. Thus, our data suggest that SLC7A10 may represent a rare but novel gene associated with human startle disease and developmental delay.
Collapse
Affiliation(s)
- Paul Drehmann
- Institute for Clinical Neurobiology, Julius Maximilians University of Würzburg, 97078 Würzburg, Germany
| | - Sinem Milanos
- Institute for Clinical Neurobiology, Julius Maximilians University of Würzburg, 97078 Würzburg, Germany
| | - Natascha Schaefer
- Institute for Clinical Neurobiology, Julius Maximilians University of Würzburg, 97078 Würzburg, Germany
| | - Vikram Babu Kasaragod
- Neurobiology Division, Medical Reserach Council Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Sarah Herterich
- Institute for Clinical Neurobiology, Julius Maximilians University of Würzburg, 97078 Würzburg, Germany
| | - Ulrike Holzbach-Eberle
- Center for Pediatrics and Adolescent Medicine, Pediatric Neurology, Social Pediatrics and Epileptology, University Hospital Gießen, 35392 Giessen, Germany
| | - Robert J Harvey
- School of Health, University of the Sunshine Coast, Sippy Downs, QLD 4558, Australia
- Sunshine Coast Health Institute, Birtinya, QLD 4575, Australia
| | - Carmen Villmann
- Institute for Clinical Neurobiology, Julius Maximilians University of Würzburg, 97078 Würzburg, Germany
| |
Collapse
|
13
|
Raitano A, Martin T, Zhang C, Malinao MC, Capo L, Ikeura M, Carroll R, Quintana JC, Dlamini S, Kulenovic L, Jahanshir E, Kang S, Morrison K, Torgov M, Morrison K. Boronotyrosine, a Borylated Amino Acid Mimetic with Enhanced Solubility, Tumor Boron Delivery, and Retention for the Re-emerging Boron Neutron Capture Therapy Field. J Med Chem 2023; 66:13809-13820. [PMID: 37729617 DOI: 10.1021/acs.jmedchem.3c01265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Boron neutron capture therapy (BNCT) is a re-emerging binary cellular level cancer intervention that occurs through the interaction of a cancer-specific 10boron (10B) drug and neutrons. We created a new 10B drug, 3-borono-l-tyrosine (BTS), that improves on the characteristics of the main historical BNCT drug 4-borono-l-phenylalanine (BPA). BTS has up to 4 times greater uptake in vitro than BPA and increased cellular retention. Like BPA, BTS uptake is mediated by the l-type amino acid transporter-1 (LAT1) but is less sensitive to natural amino acid competition. BTS can be formulated and bolus dosed at much higher levels than BPA, resulting in 2-3 times greater boron delivery in vivo. Fast blood clearance and greater tumor boron delivery result in superior tumor-to-blood ratios. BTS boron delivery appears to correlate with LAT1 expression. BTS is a promising boron delivery drug that has the potential to improve modern BNCT interventions.
Collapse
Affiliation(s)
- Arthur Raitano
- Drug Development Division, TAE Life Sciences, 1756 Cloverfield Boulevard, Santa Monica, California 90404, United States
| | - Tioga Martin
- Drug Development Division, TAE Life Sciences, 1756 Cloverfield Boulevard, Santa Monica, California 90404, United States
| | - Chunying Zhang
- Drug Development Division, TAE Life Sciences, 1756 Cloverfield Boulevard, Santa Monica, California 90404, United States
| | - Maria-Christina Malinao
- Drug Development Division, TAE Life Sciences, 1756 Cloverfield Boulevard, Santa Monica, California 90404, United States
| | - Linnette Capo
- Drug Development Division, TAE Life Sciences, 1756 Cloverfield Boulevard, Santa Monica, California 90404, United States
| | - Maki Ikeura
- Drug Development Division, TAE Life Sciences, 1756 Cloverfield Boulevard, Santa Monica, California 90404, United States
| | - Rebecca Carroll
- Drug Development Division, TAE Life Sciences, 1756 Cloverfield Boulevard, Santa Monica, California 90404, United States
| | - Jason C Quintana
- Drug Development Division, TAE Life Sciences, 1756 Cloverfield Boulevard, Santa Monica, California 90404, United States
| | - Samkeliso Dlamini
- Drug Development Division, TAE Life Sciences, 1756 Cloverfield Boulevard, Santa Monica, California 90404, United States
| | - Leila Kulenovic
- Drug Development Division, TAE Life Sciences, 1756 Cloverfield Boulevard, Santa Monica, California 90404, United States
| | - Eva Jahanshir
- Drug Development Division, TAE Life Sciences, 1756 Cloverfield Boulevard, Santa Monica, California 90404, United States
| | - Sohye Kang
- Drug Development Division, TAE Life Sciences, 1756 Cloverfield Boulevard, Santa Monica, California 90404, United States
| | - Karen Morrison
- Drug Development Division, TAE Life Sciences, 1756 Cloverfield Boulevard, Santa Monica, California 90404, United States
| | - Michael Torgov
- Drug Development Division, TAE Life Sciences, 1756 Cloverfield Boulevard, Santa Monica, California 90404, United States
| | - Kendall Morrison
- Drug Development Division, TAE Life Sciences, 1756 Cloverfield Boulevard, Santa Monica, California 90404, United States
| |
Collapse
|
14
|
Afshinpour M, Parsi P, Mahdiuni H. Investigation of molecular details of a bacterial cationic amino acid transporter (GkApcT) during arginine transportation using molecular dynamics simulation and umbrella sampling techniques. J Mol Model 2023; 29:260. [PMID: 37479900 DOI: 10.1007/s00894-023-05670-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/17/2023] [Indexed: 07/23/2023]
Abstract
CONTEXT Cationic amino acid transporters (CATs) facilitate arginine transport across membranes and maintain its levels in various tissues and organs, but their overexpression has been associated with severe cancers. A recent study identified the alternating access mechanism and critical residues involved in arginine transportation in a cationic amino acid transporter from Geobacillus kaustophilus (GkApcT). Here, we used molecular dynamics (MD) simulation methods to investigate the transportation mechanism of arginine (Arg) through GkApcT. The results revealed that arginine strongly interacts with specific binding site residues (Thr43, Asp111, Glu115, Lys191, Phe231, Ile234, and Asp237). Based on the umbrella sampling, the main driving force for arginine transport is the polar interactions of the arginine with channel-lining residues. An in-depth description of the dissociation mechanism and binding energy analysis brings valuable insight into the interactions between arginine and transporter residues, facilitating the design of effective CAT inhibitors in cancer cells. METHODS The membrane-protein system was constructed by uploading the prokaryotic CAT (PDB ID: 6F34) to the CHARMM-GUI web server. Molecular dynamics simulations were done using the GROMACS package, version 5.1.4, with the CHARMM36 force field and TIP3P water model. The MM-PBSA approach was performed for determining the arginine binding free energy. Furthermore, the hotspot residues were identified through per-residue decomposition analysis. The characteristics of the channel such as bottleneck radius and channel length were analyzed using the CaverWeb 1.1 web server. The proton wire inside the transporter was investigated based on the classic Grotthuss mechanism. We also investigated the atomistic details of arginine transportation using the path-based free energy umbrella sampling technique (US).
Collapse
Affiliation(s)
- Maral Afshinpour
- Bioinformatics Lab, Department of Biology, School of Sciences, Razi University, P.O. Box, Kermanshah, 67149-67346, Iran
- Department of Chemistry and Biochemistry, South Dakota State University (SDSU), Brookings, SD, USA
| | - Parinaz Parsi
- Bioinformatics Lab, Department of Biology, School of Sciences, Razi University, P.O. Box, Kermanshah, 67149-67346, Iran
| | - Hamid Mahdiuni
- Bioinformatics Lab, Department of Biology, School of Sciences, Razi University, P.O. Box, Kermanshah, 67149-67346, Iran.
| |
Collapse
|
15
|
Fuss MF, Wieferig JP, Corey RA, Hellmich Y, Tascón I, Sousa JS, Stansfeld PJ, Vonck J, Hänelt I. Cyclic di-AMP traps proton-coupled K + transporters of the KUP family in an inward-occluded conformation. Nat Commun 2023; 14:3683. [PMID: 37344476 DOI: 10.1038/s41467-023-38944-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 05/22/2023] [Indexed: 06/23/2023] Open
Abstract
Cyclic di-AMP is the only known essential second messenger in bacteria and archaea, regulating different proteins indispensable for numerous physiological processes. In particular, it controls various potassium and osmolyte transporters involved in osmoregulation. In Bacillus subtilis, the K+/H+ symporter KimA of the KUP family is inactivated by c-di-AMP. KimA sustains survival at potassium limitation at low external pH by mediating potassium ion uptake. However, at elevated intracellular K+ concentrations, further K+ accumulation would be toxic. In this study, we reveal the molecular basis of how c-di-AMP binding inhibits KimA. We report cryo-EM structures of KimA with bound c-di-AMP in detergent solution and reconstituted in amphipols. By combining structural data with functional assays and molecular dynamics simulations we reveal how c-di-AMP modulates transport. We show that an intracellular loop in the transmembrane domain interacts with c-di-AMP bound to the adjacent cytosolic domain. This reduces the mobility of transmembrane helices at the cytosolic side of the K+ binding site and therefore traps KimA in an inward-occluded conformation.
Collapse
Affiliation(s)
- Michael F Fuss
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Jan-Philip Wieferig
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Robin A Corey
- Department of Biochemistry, University of Oxford, Oxford, UK
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Yvonne Hellmich
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Igor Tascón
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, Leioa, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Joana S Sousa
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
- UCB Pharma, UCB Biopharma UK, Slough, SL1 3WE, UK
| | - Phillip J Stansfeld
- School of Life Sciences & Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| | - Janet Vonck
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany.
| | - Inga Hänelt
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
16
|
Afshinpour M, Mahdiuni H. Arginine transportation mechanism through cationic amino acid transporter 1: insights from molecular dynamics studies. J Biomol Struct Dyn 2023; 41:13580-13594. [PMID: 36762692 DOI: 10.1080/07391102.2023.2175374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 01/28/2023] [Indexed: 02/11/2023]
Abstract
Metabolic and signaling mechanisms in mammalian cells are facilitated by the transportation of L-arginine (Arg) across the plasma membrane through cationic amino acid transporter (CAT) proteins. Due to a lack of argininosuccinate synthase (ASS) activity in various tumor cells such as acute myeloid leukemia, acute lymphocytic leukemia, and chronic lymphocytic leukemia, these tumor entities are arginine-auxotrophic and therefore depend on the uptake of the amino acid arginine. Cationic amino acid transporter-1 (CAT-1) is the leading arginine importer expressed in the aforementioned tumor entities. Hence, in the present study, to investigate the transportation mechanism of arginine in CAT-1, we performed molecular dynamics (MD) simulation methods on the modeled human CAT-1. The MM-PBSA approach was conducted to determine the critical residues interacting with arginine within the corresponding binding site of CAT-1. In addition, we found out that the water molecules have the leading role in forming the transportation channel within CAT-1. The conductive structure of CAT-1 was formed only when the water molecules were continuously distributed across the channel. Steered molecular dynamics (SMD) simulation approach showed various energy barriers against arginine transportation through CAT-1, especially while crossing the bottlenecks of the related channel. These findings at the molecular level might shed light on identifying the crucial amino acids in the binding of arginine to eukaryotic CATs and also provide fundamental insights into the arginine transportation mechanisms through CAT-1. Understanding the transportation mechanism of arginine is essential to developing CAT-1 blockers, which can be potential medications for some types of cancers.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Maral Afshinpour
- Bioinformatics Lab., Department of Biology, School of Sciences, Razi University, Kermanshah, Iran
| | - Hamid Mahdiuni
- Bioinformatics Lab., Department of Biology, School of Sciences, Razi University, Kermanshah, Iran
| |
Collapse
|
17
|
Gauthier-Coles G, Fairweather SJ, Bröer A, Bröer S. Do Amino Acid Antiporters Have Asymmetric Substrate Specificity? Biomolecules 2023; 13:biom13020301. [PMID: 36830670 PMCID: PMC9953452 DOI: 10.3390/biom13020301] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Amino acid antiporters mediate the 1:1 exchange of groups of amino acids. Whether substrate specificity can be different for the inward and outward facing conformation has not been investigated systematically, although examples of asymmetric transport have been reported. Here we used LC-MS to detect the movement of 12C- and 13C-labelled amino acid mixtures across the plasma membrane of Xenopus laevis oocytes expressing a variety of amino acid antiporters. Differences of substrate specificity between transporter paralogs were readily observed using this method. Our results suggest that antiporters are largely symmetric, equalizing the pools of their substrate amino acids. Exceptions are the antiporters y+LAT1 and y+LAT2 where neutral amino acids are co-transported with Na+ ions, favouring their import. For the antiporters ASCT1 and ASCT2 glycine acted as a selective influx substrate, while proline was a selective influx substrate of ASCT1. These data show that antiporters can display non-canonical modes of transport.
Collapse
|
18
|
Kantipudi S, Harder D, Fotiadis D. Characterization of substrates and inhibitors of the human heterodimeric transporter 4F2hc-LAT1 using purified protein and the scintillation proximity radioligand binding assay. Front Physiol 2023; 14:1148055. [PMID: 36895635 PMCID: PMC9989278 DOI: 10.3389/fphys.2023.1148055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Amino acids have diverse and essential roles in many cellular functions such as in protein synthesis, metabolism and as precursors of different hormones. Translocation of amino acids and derivatives thereof across biological membranes is mediated by amino acid transporters. 4F2hc-LAT1 is a heterodimeric amino acid transporter that is composed of two subunits belonging to the SLC3 (4F2hc) and SLC7 (LAT1) solute carrier families. The ancillary protein 4F2hc is responsible for the correct trafficking and regulation of the transporter LAT1. Preclinical studies have identified 4F2hc-LAT1 as a valid anticancer target due to its importance in tumor progression. The scintillation proximity assay (SPA) is a valuable radioligand binding assay that allows the identification and characterization of ligands of membrane proteins. Here, we present a SPA ligand binding study using purified recombinant human 4F2hc-LAT1 protein and the radioligand [3H]L-leucine as tracer. Binding affinities of different 4F2hc-LAT1 substrates and inhibitors determined by SPA are comparable with previously reported K m and IC 50 values from 4F2hc-LAT1 cell-based uptake assays. In summary, the SPA is a valuable method for the identification and characterization of ligands of membrane transporters including inhibitors. In contrast to cell-based assays, where the potential interference with other proteins such as endogenous transporters persists, the SPA uses purified protein making target engagement and characterization of ligands highly reliable.
Collapse
Affiliation(s)
- Satish Kantipudi
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Daniel Harder
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Dimitrios Fotiadis
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
19
|
Yao H, Cai H, Li D. Fluorescence-Detection Size-Exclusion Chromatography-Based Thermostability Assay for Membrane Proteins. Methods Mol Biol 2023; 2564:299-315. [PMID: 36107350 DOI: 10.1007/978-1-0716-2667-2_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Green fluorescent proteins (GFPs) have lightened up almost every aspect of biological research including protein sciences. In the field of membrane protein structural biology, GFPs have been used widely to monitor membrane protein localization, expression level, the purification process and yield, and the stability inside the cells and in the test tube. Of particular interest is the fluorescence-detector size-exclusion chromatography-based thermostability assay (FSEC-TS). By simple heating and FSEC, the generally applicable method allows rapid assessment of the thermostability of GFP-fused membrane proteins without purification. Here we describe the experimental details and some typical results for the FSEC-TS method.
Collapse
Affiliation(s)
| | | | - Dianfan Li
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
20
|
Zhou Y, Lewis JH, Lu Z. Tracking multiple conformations occurring on angstrom-and-millisecond scales in single amino-acid-transporter molecules. eLife 2023; 12:82175. [PMID: 36800214 PMCID: PMC9937647 DOI: 10.7554/elife.82175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 01/15/2023] [Indexed: 02/18/2023] Open
Abstract
Most membrane protein molecules undergo conformational changes as they transition from one functional state to another one. An understanding of the mechanism underlying these changes requires the ability to resolve individual conformational states, whose changes often occur on millisecond and angstrom scales. Tracking such changes and acquiring a sufficiently large amount of data remain challenging. Here, we use the amino-acid transporter AdiC as an example to demonstrate the application of a high-resolution fluorescence-polarization-microscopy method in tracking multistate conformational changes of a membrane protein. We have successfully resolved four conformations of AdiC by monitoring the emission-polarization changes of a fluorophore label and quantified their probabilities in the presence of a series of concentrations of its substrate arginine. The acquired data are sufficient for determining all equilibrium constants that fully establish the energetic relations among the four states. The KD values determined for arginine in four individual conformations are statistically comparable to the previously reported overall KD determined using isothermal titration calorimetry. This demonstrated strong resolving power of the present polarization-microscopy method will enable an acquisition of the quantitative information required for understanding the expected complex conformational mechanism underlying the transporter's function, as well as those of other membrane proteins.
Collapse
Affiliation(s)
- Yufeng Zhou
- Department of Physiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - John H Lewis
- Department of Physiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Zhe Lu
- Department of Physiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
21
|
Hang TD, Hung HM, Beckers P, Desmet N, Lamrani M, Massie A, Hermans E, Vanommeslaeghe K. Structural investigation of human cystine/glutamate antiporter system xc− (Sxc−) using homology modeling and molecular dynamics. Front Mol Biosci 2022; 9:1064199. [DOI: 10.3389/fmolb.2022.1064199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/15/2022] [Indexed: 12/04/2022] Open
Abstract
The cystine/glutamate antiporter system xc− (Sxc−) belongs to the SLC7 family of plasma membrane transporters. It exports intracellular glutamate along the latter’s concentration gradient as a driving force for cellular uptake of cystine. Once imported, cystine is mainly used for the production of glutathione, a tripeptide thiol crucial in maintenance of redox homeostasis and protection of cells against oxidative stress. Overexpression of Sxc− has been found in several cancer cells, where it is thought to counteract the increased oxidative stress. In addition, Sxc− is important in the central nervous system, playing a complex role in regulating glutamatergic neurotransmission and glutamate toxicity. Accordingly, this transporter is considered a potential target for the treatment of cancer as well as neurodegenerative diseases. Till now, no specific inhibitors are available. We herein present four conformations of Sxc− along its transport pathway, obtained using multi-template homology modeling and refined by means of Molecular Dynamics. Comparison with a very recently released cryo-EM structure revealed an excellent agreement with our inward-open conformation. Intriguingly, our models contain a structured N-terminal domain that is unresolved in the experimental structures and is thought to play a gating role in the transport mechanism of other SLC7 family members. In contrast to the inward-open model, there is no direct experimental counterpart for the other three conformations we obtained, although they are in fair agreement with the other stages of the transport mechanism seen in other SLC7 transporters. Therefore, our models open the prospect for targeting alternative Sxc− conformations in structure-based drug design efforts.
Collapse
|
22
|
Nanobodies targeting LexA autocleavage disclose a novel suppression strategy of SOS-response pathway. Structure 2022; 30:1479-1493.e9. [DOI: 10.1016/j.str.2022.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/29/2022] [Accepted: 09/18/2022] [Indexed: 11/05/2022]
|
23
|
Neumann C, Rosenbæk LL, Flygaard RK, Habeck M, Karlsen JL, Wang Y, Lindorff‐Larsen K, Gad HH, Hartmann R, Lyons JA, Fenton RA, Nissen P. Cryo-EM structure of the human NKCC1 transporter reveals mechanisms of ion coupling and specificity. EMBO J 2022; 41:e110169. [PMID: 36239040 PMCID: PMC9713717 DOI: 10.15252/embj.2021110169] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 12/03/2022] Open
Abstract
The sodium-potassium-chloride transporter NKCC1 of the SLC12 family performs Na+ -dependent Cl- - and K+ -ion uptake across plasma membranes. NKCC1 is important for regulating cell volume, hearing, blood pressure, and regulation of hyperpolarizing GABAergic and glycinergic signaling in the central nervous system. Here, we present a 2.6 Å resolution cryo-electron microscopy structure of human NKCC1 in the substrate-loaded (Na+ , K+ , and 2 Cl- ) and occluded, inward-facing state that has also been observed for the SLC6-type transporters MhsT and LeuT. Cl- binding at the Cl1 site together with the nearby K+ ion provides a crucial bridge between the LeuT-fold scaffold and bundle domains. Cl- -ion binding at the Cl2 site seems to undertake a structural role similar to conserved glutamate of SLC6 transporters and may allow for Cl- -sensitive regulation of transport. Supported by functional studies in mammalian cells and computational simulations, we describe a putative Na+ release pathway along transmembrane helix 5 coupled to the Cl2 site. The results provide insight into the structure-function relationship of NKCC1 with broader implications for other SLC12 family members.
Collapse
Affiliation(s)
- Caroline Neumann
- Danish Research Institute of Translational Neuroscience—DANDRITENordic EMBL Partnership for Molecular MedicineAarhusDenmark,Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| | | | - Rasmus Kock Flygaard
- Danish Research Institute of Translational Neuroscience—DANDRITENordic EMBL Partnership for Molecular MedicineAarhusDenmark,Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| | - Michael Habeck
- Danish Research Institute of Translational Neuroscience—DANDRITENordic EMBL Partnership for Molecular MedicineAarhusDenmark,Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| | | | - Yong Wang
- Linderstrøm‐Lang Centre for Protein Science, Department of BiologyUniversity of CopenhagenCopenhagenDenmark,Shanghai Institute for Advanced Study, Institute of Quantitative Biology, College of Life SciencesZhejiang UniversityHangzhouChina
| | - Kresten Lindorff‐Larsen
- Linderstrøm‐Lang Centre for Protein Science, Department of BiologyUniversity of CopenhagenCopenhagenDenmark
| | - Hans Henrik Gad
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| | - Rune Hartmann
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| | - Joseph Anthony Lyons
- Danish Research Institute of Translational Neuroscience—DANDRITENordic EMBL Partnership for Molecular MedicineAarhusDenmark,Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark,Interdisciplinary Nanoscience Center (iNANO)Aarhus UniversityAarhusDenmark
| | | | - Poul Nissen
- Danish Research Institute of Translational Neuroscience—DANDRITENordic EMBL Partnership for Molecular MedicineAarhusDenmark,Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| |
Collapse
|
24
|
Peter MF, Ruland JA, Depping P, Schneberger N, Severi E, Moecking J, Gatterdam K, Tindall S, Durand A, Heinz V, Siebrasse JP, Koenig PA, Geyer M, Ziegler C, Kubitscheck U, Thomas GH, Hagelueken G. Structural and mechanistic analysis of a tripartite ATP-independent periplasmic TRAP transporter. Nat Commun 2022; 13:4471. [PMID: 35927235 PMCID: PMC9352664 DOI: 10.1038/s41467-022-31907-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 07/08/2022] [Indexed: 11/27/2022] Open
Abstract
Tripartite ATP-independent periplasmic (TRAP) transporters are found widely in bacteria and archaea and consist of three structural domains, a soluble substrate-binding protein (P-domain), and two transmembrane domains (Q- and M-domains). HiSiaPQM and its homologs are TRAP transporters for sialic acid and are essential for host colonization by pathogenic bacteria. Here, we reconstitute HiSiaQM into lipid nanodiscs and use cryo-EM to reveal the structure of a TRAP transporter. It is composed of 16 transmembrane helices that are unexpectedly structurally related to multimeric elevator-type transporters. The idiosyncratic Q-domain of TRAP transporters enables the formation of a monomeric elevator architecture. A model of the tripartite PQM complex is experimentally validated and reveals the coupling of the substrate-binding protein to the transporter domains. We use single-molecule total internal reflection fluorescence (TIRF) microscopy in solid-supported lipid bilayers and surface plasmon resonance to study the formation of the tripartite complex and to investigate the impact of interface mutants. Furthermore, we characterize high-affinity single variable domains on heavy chain (VHH) antibodies that bind to the periplasmic side of HiSiaQM and inhibit sialic acid uptake, providing insight into how TRAP transporter function might be inhibited in vivo.
Collapse
Affiliation(s)
- Martin F Peter
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Jan A Ruland
- Institute for Physical und Theoretical Chemistry, University of Bonn, Wegelerstr. 12, 53127, Bonn, Germany
| | - Peer Depping
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- Aston Centre for Membrane Proteins and Lipids Research, Aston St., B4 7ET, Birmingham, UK
| | - Niels Schneberger
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Emmanuele Severi
- Department of Biology (Area 10), University of York, York, YO10 5YW, UK
- Biosciences Institute, Newcastle University, Newcastle, NE2 4HH, UK
| | - Jonas Moecking
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Karl Gatterdam
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Sarah Tindall
- Department of Biology (Area 10), University of York, York, YO10 5YW, UK
| | - Alexandre Durand
- Institut de Génétique et de Biologie Molecule et Cellulaire, 1 Rue Laurent Fries, 67404, Illkirch Cedex, France
| | - Veronika Heinz
- Institute of Biophysics and Biophysical Chemistry, University of Regensburg, Universitätsstr. 31, 93053, Regensburg, Germany
| | - Jan Peter Siebrasse
- Institute for Physical und Theoretical Chemistry, University of Bonn, Wegelerstr. 12, 53127, Bonn, Germany
| | - Paul-Albert Koenig
- Core Facility Nanobodies, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Matthias Geyer
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Christine Ziegler
- Institute of Biophysics and Biophysical Chemistry, University of Regensburg, Universitätsstr. 31, 93053, Regensburg, Germany
| | - Ulrich Kubitscheck
- Institute for Physical und Theoretical Chemistry, University of Bonn, Wegelerstr. 12, 53127, Bonn, Germany
| | - Gavin H Thomas
- Department of Biology (Area 10), University of York, York, YO10 5YW, UK
| | - Gregor Hagelueken
- Institute of Structural Biology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
25
|
Huang Y, Zhang L, Wang M, Li C, Zheng W, Chen H, Liang Y, Wu Z. Optimization of Precursor Synthesis Conditions of (2S,4S)4–[18F]FPArg and Its Application in Glioma Imaging. Pharmaceuticals (Basel) 2022; 15:ph15080946. [PMID: 36015094 PMCID: PMC9416586 DOI: 10.3390/ph15080946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 01/27/2023] Open
Abstract
Although the tracer (2S,4S)4–[18F]FPArg is expected to provide a powerful imaging method for the diagnosis and treatment of clinical tumors, it has not been realized due to the low yield of chemical synthesis and radiolabeling. A simple synthetic method for the radiolabeled precursor of (2S,4S)4–[18F]FPArg in stable yield was obtained by adjusting the sequence of the synthetic steps. Furthermore, the biodistribution experiments confirmed that (2S,4S)4–[18F]FPArg could be cleared out quickly in wild type mouse. Cell uptake experiments and U87MG tumor mouse microPET–CT imaging experiments showed that the tumor had high uptake of (2S,4S)4–[18F]FPArg and the clearance was slow, but (2S,4S)4–[18F]FPArg was rapidly cleared in normal brain tissue. MicroPET–CT imaging of nude mice bearing orthotopic HS683–Luc showed that (2S,4S)4–[18F]FPArg can penetrate blood–brain barrier and image gliomas with a high contrast. Therefore, (2S,4S)4–[18F]FPArg is expected to be further applied in the diagnosis and efficacy evaluation of clinical glioma.
Collapse
Affiliation(s)
- Yong Huang
- Department of Nuclear Medicine, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China; (Y.H.); (C.L.)
| | - Lu Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China; (L.Z.); (W.Z.); (H.C.)
| | - Meng Wang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China;
| | - Chengze Li
- Department of Nuclear Medicine, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China; (Y.H.); (C.L.)
| | - Wei Zheng
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China; (L.Z.); (W.Z.); (H.C.)
| | - Hualong Chen
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China; (L.Z.); (W.Z.); (H.C.)
| | - Ying Liang
- Department of Nuclear Medicine, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China; (Y.H.); (C.L.)
- Correspondence: (Y.L.); (Z.W.)
| | - Zehui Wu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China; (L.Z.); (W.Z.); (H.C.)
- Correspondence: (Y.L.); (Z.W.)
| |
Collapse
|
26
|
Del Alamo D, Meiler J, Mchaourab HS. Principles of Alternating Access in LeuT-fold Transporters: Commonalities and Divergences. J Mol Biol 2022; 434:167746. [PMID: 35843285 DOI: 10.1016/j.jmb.2022.167746] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 11/15/2022]
Abstract
Found in all domains of life, transporters belonging to the LeuT-fold class mediate the import and exchange of hydrophilic and charged compounds such as amino acids, metals, and sugar molecules. Nearly two decades of investigations on the eponymous bacterial transporter LeuT have yielded a library of high-resolution snapshots of its conformational cycle linked by solution-state experimental data obtained from multiple techniques. In parallel, its topology has been observed in symporters and antiporters characterized by a spectrum of substrate specificities and coupled to gradients of distinct ions. Here we review and compare mechanistic models of transport for LeuT, its well-studied homologs, as well as functionally distant members of the fold, emphasizing the commonalities and divergences in alternating access and the corresponding energy landscapes. Our integrated summary illustrates how fold conservation, a hallmark of the LeuT fold, coincides with divergent choreographies of alternating access that nevertheless capitalize on recurrent structural motifs. In addition, it highlights the knowledge gap that hinders the leveraging of the current body of research into detailed mechanisms of transport for this important class of membrane proteins.
Collapse
Affiliation(s)
- Diego Del Alamo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Department of Chemistry, Vanderbilt University, Nashville, TN, USA. https://twitter.com/DdelAlamo
| | - Jens Meiler
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA; Institute for Drug Discovery, Leipzig University, Leipzig, DE, USA. https://twitter.com/MeilerLab
| | - Hassane S Mchaourab
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
27
|
Rullo-Tubau J, Bartoccioni P, Llorca O, Errasti-Murugarren E, Palacín M. HATs meet structural biology. Curr Opin Struct Biol 2022; 74:102389. [DOI: 10.1016/j.sbi.2022.102389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/31/2022] [Accepted: 04/10/2022] [Indexed: 11/26/2022]
|
28
|
Ca 2+-mediated higher-order assembly of heterodimers in amino acid transport system b 0,+ biogenesis and cystinuria. Nat Commun 2022; 13:2708. [PMID: 35577790 PMCID: PMC9110406 DOI: 10.1038/s41467-022-30293-9] [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: 05/18/2021] [Accepted: 04/22/2022] [Indexed: 02/06/2023] Open
Abstract
Cystinuria is a genetic disorder characterized by overexcretion of dibasic amino acids and cystine, causing recurrent kidney stones and kidney failure. Mutations of the regulatory glycoprotein rBAT and the amino acid transporter b0,+AT, which constitute system b0,+, are linked to type I and non-type I cystinuria respectively and they exhibit distinct phenotypes due to protein trafficking defects or catalytic inactivation. Here, using electron cryo-microscopy and biochemistry, we discover that Ca2+ mediates higher-order assembly of system b0,+. Ca2+ stabilizes the interface between two rBAT molecules, leading to super-dimerization of b0,+AT-rBAT, which in turn facilitates N-glycan maturation and protein trafficking. A cystinuria mutant T216M and mutations of the Ca2+ site of rBAT cause the loss of higher-order assemblies, resulting in protein trapping at the ER and the loss of function. These results provide the molecular basis of system b0,+ biogenesis and type I cystinuria and serve as a guide to develop new therapeutic strategies against it. More broadly, our findings reveal an unprecedented link between transporter oligomeric assembly and protein-trafficking diseases.
Collapse
|
29
|
Yan L, He J, Liao X, Liang T, Zhu J, Wei W, He Y, Zhou X, Peng T. A comprehensive analysis of the diagnostic and prognostic value associated with the SLC7A family members in breast cancer. Gland Surg 2022; 11:389-411. [PMID: 35284318 PMCID: PMC8899434 DOI: 10.21037/gs-21-909] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/30/2022] [Indexed: 07/21/2023]
Abstract
BACKGROUND The solute carrier (SLC) 7 family genes play central roles in cancer cell metabolism as glucose and glutamate transporters. However, their expression and prognostic value in breast cancer (BC) remains to be elucidated. METHODS Clinical data from BC patients were downloaded from The Cancer Genome Atlas (TCGA) and the Kaplan-Meier (KM) plotter database. The mechanisms underlying the association between SLC7A expression and overall survival (OS) were explored using Cox regression and log-rank tests. ESTIMATE gives a measure of the immune-cell infiltrates. Single-sample (ss) Gene Set Enrichment Analysis (GSEA) was conducted to quantify immune cell infiltration. RESULTS High SLC7A5 expression was associated with a poorer survival time in BC patients according to the TCGA and KM plotter data. SLC7A4 was associated with good progression-free interval (PFI) and disease-specific survival (DSS) according to the TCGA data. Furthermore, SLC7A4 was correlated with good prognosis of OS, distant metastasis-free survival (DMFS), relapse-free survival (RFS), and post-progression survival (PPS) according to the KM plotter data. SLC7A3 expression was positively associated with OS, but was not strongly associated with PFI nor DSS in the TCGA data. However, SLC7A3 was positively correlated with DMFS and RFS in the KM database analysis. SLC7A had excellent diagnostic value in BC patients and was strongly correlated with tumor infiltration. T helper 2 (Th2) cells, CD56 bright natural killer (NK) cells, and NK cells were the most strongly correlated with the SLC7A family genes, suggesting that these genes play a crucial role in BC partly by modulating immune infiltration. CONCLUSIONS SLC7A4 and SLC7A5 expression levels may be sensitive biomarkers for predicting BC outcomes. SLC7A3 may be a potential diagnostic and prognostic biomarker in BC, but further studies are warranted to verify these results.
Collapse
Affiliation(s)
- Liping Yan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment Guangxi Medical University, Nanning, China
| | - Jianxin He
- Department of Ultrasound Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Tianyi Liang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jia Zhu
- Department of Breast Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wensong Wei
- Department of Breast Surgery, The Third Hospital of Nanchang, Nanchang, China
| | - Yongfei He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xin Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment Guangxi Medical University, Nanning, China
| |
Collapse
|
30
|
Cysteine 467 of the ASCT2 Amino Acid Transporter Is a Molecular Determinant of the Antiport Mechanism. Int J Mol Sci 2022; 23:ijms23031127. [PMID: 35163050 PMCID: PMC8835248 DOI: 10.3390/ijms23031127] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 02/01/2023] Open
Abstract
The plasma membrane transporter ASCT2 is a well-known Na+-dependent obligatory antiporter of neutral amino acids. The crucial role of the residue C467 in the recognition and binding of the ASCT2 substrate glutamine, has been highlighted by structure/function relationship studies. The reconstitution in proteoliposomes of the human ASCT2 produced in P. pastoris is here employed to unveil another role of the C467 residue in the transport reaction. Indeed, the site-directed mutant C467A displayed a novel property of the transporter, i.e., the ability of mediating a low but measurable unidirectional transport of [3H]-glutamine. This reaction conforms to the main features of the ASCT2-mediated transport, namely the Na+-dependence, the pH dependence, the stimulation by cholesterol included in the proteoliposome membrane, and the specific inhibition by other common substrates of the reconstituted human ASCT2. Interestingly, the WT protein cannot catalyze the unidirectional transport of [3H]-glutamine, demonstrating an unspecific phenomenon. This difference is in favor of a structural conformational change between a WT and C467A mutant that triggers the appearance of the unidirectional flux; this feature has been investigated by comparing the available 3D structures in two different conformations, and two homology models built on the basis of hEAAT1 and GLTPh.
Collapse
|
31
|
Parker JL, Deme JC, Kolokouris D, Kuteyi G, Biggin PC, Lea SM, Newstead S. Molecular basis for redox control by the human cystine/glutamate antiporter system xc . Nat Commun 2021; 12:7147. [PMID: 34880232 PMCID: PMC8654953 DOI: 10.1038/s41467-021-27414-1] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022] Open
Abstract
Cysteine plays an essential role in cellular redox homoeostasis as a key constituent of the tripeptide glutathione (GSH). A rate limiting step in cellular GSH synthesis is the availability of cysteine. However, circulating cysteine exists in the blood as the oxidised di-peptide cystine, requiring specialised transport systems for its import into the cell. System xc- is a dedicated cystine transporter, importing cystine in exchange for intracellular glutamate. To counteract elevated levels of reactive oxygen species in cancerous cells system xc- is frequently upregulated, making it an attractive target for anticancer therapies. However, the molecular basis for ligand recognition remains elusive, hampering efforts to specifically target this transport system. Here we present the cryo-EM structure of system xc- in both the apo and glutamate bound states. Structural comparisons reveal an allosteric mechanism for ligand discrimination, supported by molecular dynamics and cell-based assays, establishing a mechanism for cystine transport in human cells.
Collapse
Affiliation(s)
- Joanne L Parker
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK.
| | - Justin C Deme
- Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
- Central Oxford Structural Molecular Imaging Centre, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | | | - Gabriel Kuteyi
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
| | - Philip C Biggin
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
| | - Susan M Lea
- Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK.
- Central Oxford Structural Molecular Imaging Centre, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK.
- Center for Structural Biology, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA.
| | - Simon Newstead
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK.
- The Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK.
| |
Collapse
|
32
|
Structural basis for substrate specificity of heteromeric transporters of neutral amino acids. Proc Natl Acad Sci U S A 2021; 118:2113573118. [PMID: 34848541 DOI: 10.1073/pnas.2113573118] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 11/18/2022] Open
Abstract
Despite having similar structures, each member of the heteromeric amino acid transporter (HAT) family shows exquisite preference for the exchange of certain amino acids. Substrate specificity determines the physiological function of each HAT and their role in human diseases. However, HAT transport preference for some amino acids over others is not yet fully understood. Using cryo-electron microscopy of apo human LAT2/CD98hc and a multidisciplinary approach, we elucidate key molecular determinants governing neutral amino acid specificity in HATs. A few residues in the substrate-binding pocket determine substrate preference. Here, we describe mutations that interconvert the substrate profiles of LAT2/CD98hc, LAT1/CD98hc, and Asc1/CD98hc. In addition, a region far from the substrate-binding pocket critically influences the conformation of the substrate-binding site and substrate preference. This region accumulates mutations that alter substrate specificity and cause hearing loss and cataracts. Here, we uncover molecular mechanisms governing substrate specificity within the HAT family of neutral amino acid transporters and provide the structural bases for mutations in LAT2/CD98hc that alter substrate specificity and that are associated with several pathologies.
Collapse
|
33
|
Gauthier-Coles G, Vennitti J, Zhang Z, Comb WC, Xing S, Javed K, Bröer A, Bröer S. Quantitative modelling of amino acid transport and homeostasis in mammalian cells. Nat Commun 2021; 12:5282. [PMID: 34489418 PMCID: PMC8421413 DOI: 10.1038/s41467-021-25563-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 08/13/2021] [Indexed: 12/20/2022] Open
Abstract
Homeostasis is one of the fundamental concepts in physiology. Despite remarkable progress in our molecular understanding of amino acid transport, metabolism and signaling, it remains unclear by what mechanisms cytosolic amino acid concentrations are maintained. We propose that amino acid transporters are the primary determinants of intracellular amino acid levels. We show that a cell’s endowment with amino acid transporters can be deconvoluted experimentally and used this data to computationally simulate amino acid translocation across the plasma membrane. Transport simulation generates cytosolic amino acid concentrations that are close to those observed in vitro. Perturbations of the system are replicated in silico and can be applied to systems where only transcriptomic data are available. This work explains amino acid homeostasis at the systems-level, through a combination of secondary active transporters, functionally acting as loaders, harmonizers and controller transporters to generate a stable equilibrium of all amino acid concentrations. Cytosolic amino acid concentrations are carefully maintained, but how homeostasis occurs is unclear. Here, the authors show that amino acid transporters primarily determine intracellular amino acid levels and develop a model that predicts a perturbation response similar to experimental data.
Collapse
Affiliation(s)
| | - Jade Vennitti
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Zhiduo Zhang
- Division of Genome Science and Cancer, ACRF INCITe Centre - ANU Node, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | | | | | - Kiran Javed
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Angelika Bröer
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Stefan Bröer
- Research School of Biology, Australian National University, Canberra, ACT, Australia.
| |
Collapse
|
34
|
Nicolàs-Aragó A, Fort J, Palacín M, Errasti-Murugarren E. Rush Hour of LATs towards Their Transport Cycle. MEMBRANES 2021; 11:602. [PMID: 34436365 PMCID: PMC8399266 DOI: 10.3390/membranes11080602] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 12/29/2022]
Abstract
The mammalian SLC7 family comprises the L-amino acid transporters (LATs) and the cationic amino acid transporters (CATs). The relevance of these transporters is highlighted by their involvement in several human pathologies, including inherited rare diseases and acquired diseases, such as cancer. In the last four years, several crystal or cryo-EM structures of LATs and CATs have been solved. These structures have started to fill our knowledge gap that previously was based on the structural biology of remote homologs of the amino acid-polyamine-organocation (APC) transporters. This review recovers this structural and functional information to start generating the molecular bases of the transport cycle of LATs. Special attention is given to the known transporter conformations within the transport cycle and the molecular bases for substrate interaction and translocation, including the asymmetric interaction of substrates at both sides of the plasma membrane.
Collapse
Affiliation(s)
- Adrià Nicolàs-Aragó
- Laboratory of Amino Acid Transporters and Disease, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain; (A.N.-A.); (J.F.)
| | - Joana Fort
- Laboratory of Amino Acid Transporters and Disease, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain; (A.N.-A.); (J.F.)
- CIBERER (Centro Español en Red de Biomedicina de Enfermedades Raras), 08028 Barcelona, Spain
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Manuel Palacín
- Laboratory of Amino Acid Transporters and Disease, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain; (A.N.-A.); (J.F.)
- CIBERER (Centro Español en Red de Biomedicina de Enfermedades Raras), 08028 Barcelona, Spain
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Ekaitz Errasti-Murugarren
- Laboratory of Amino Acid Transporters and Disease, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain; (A.N.-A.); (J.F.)
- CIBERER (Centro Español en Red de Biomedicina de Enfermedades Raras), 08028 Barcelona, Spain
| |
Collapse
|
35
|
Elorza-Vidal X, Xicoy-Espaulella E, Pla-Casillanis A, Alonso-Gardón M, Gaitán-Peñas H, Engel-Pizcueta C, Fernández-Recio J, Estévez R. Structural basis for the dominant or recessive character of GLIALCAM mutations found in leukodystrophies. Hum Mol Genet 2021; 29:1107-1120. [PMID: 31960914 PMCID: PMC7206653 DOI: 10.1093/hmg/ddaa009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/10/2020] [Accepted: 01/15/2020] [Indexed: 12/27/2022] Open
Abstract
Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a type of leukodystrophy characterized by white matter edema, and it is caused mainly by recessive mutations in MLC1 and GLIALCAM genes. These variants are called MLC1 and MLC2A with both types of patients sharing the same clinical phenotype. In addition, dominant mutations in GLIALCAM have also been identified in a subtype of MLC patients with a remitting phenotype. This variant has been named MLC2B. GLIALCAM encodes for an adhesion protein containing two immunoglobulin (Ig) domains and it is needed for MLC1 targeting to astrocyte–astrocyte junctions. Most mutations identified in GLIALCAM abolish GlialCAM targeting to junctions. However, it is unclear why some mutations behave as recessive or dominant. Here, we used a combination of biochemistry methods with a new developed anti-GlialCAM nanobody, double-mutants and cysteine cross-links experiments, together with computer docking, to create a structural model of GlialCAM homo-interactions. Using this model, we suggest that dominant mutations affect different GlialCAM–GlialCAM interacting surfaces in the first Ig domain, which can occur between GlialCAM molecules present in the same cell (cis) or present in neighbouring cells (trans). Our results provide a framework that can be used to understand the molecular basis of pathogenesis of all identified GLIALCAM mutations.
Collapse
Affiliation(s)
- Xabier Elorza-Vidal
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain.,Centro de Investigación en red de enfermedades raras (CIBERER), ISCIII, Madrid, Spain
| | - Efren Xicoy-Espaulella
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain
| | - Adrià Pla-Casillanis
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain
| | - Marta Alonso-Gardón
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain
| | - Héctor Gaitán-Peñas
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain.,Centro de Investigación en red de enfermedades raras (CIBERER), ISCIII, Madrid, Spain
| | - Carolyn Engel-Pizcueta
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain
| | - Juan Fernández-Recio
- Barcelona Supercomputing Center (BSC), Barcelona, Spain.,Institut de Biologia Molecular de Barcelona, CSIC, Barcelona, Spain.,Instituto de Ciencias de la Vid y del Vino (ICVV), CSIC- Universidad de La Rioja- Gobierno de la Rioja, Logroño, Spain
| | - Raúl Estévez
- Unitat de Fisiologia, Departament de Ciències Fisiològiques, Genes Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain.,Centro de Investigación en red de enfermedades raras (CIBERER), ISCIII, Madrid, Spain
| |
Collapse
|
36
|
Cecchetti C, Scull NJ, Mohan TC, Alguel Y, Jones AMC, Cameron AD, Byrne B. Transfer of stabilising mutations between different secondary active transporter families. FEBS Open Bio 2021; 11:1685-1694. [PMID: 33932145 PMCID: PMC8167854 DOI: 10.1002/2211-5463.13168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/25/2021] [Accepted: 04/15/2021] [Indexed: 11/24/2022] Open
Abstract
Integral membrane transporters play essential roles in the movement of substrates across biological membranes. One approach to produce transporters suitable for structural studies is to introduce mutations that reduce conformational flexibility and increase stability. However, it can be difficult to predict which mutations will result in a more stable protein. Previously, we stabilised the uric acid‐xanthine transporter, UapA, a member of the SLC23 family, through introduction of a single‐point mutation, G411V, trapping the protein in the inward‐facing conformation. Here, we attempted to stabilise the structurally related BOR1 transporter from Arabidopsis thaliana, a member of the SLC4 family, by introducing the equivalent substitution. We identified possible residues, P362 and M363, in AtBOR1, likely to be equivalent to the G411 of UapA, and generated four mutants, P362V or L and M363F or Y. Stability analysis using heated Fluorescent Size Exclusion Chromatography indicated that the M363F/Y mutants were more stable than the WT AtBOR1 and P362V/L mutants. Furthermore, functional complementation analysis revealed that the M363F/Y mutants exhibited reduced transport activity compared to the P362V/L and WT proteins. Purification and crystallisation of the M363F/Y proteins yielded crystals that diffracted better than WT (5.5 vs 7 Å). We hypothesise that the increased bulk of the F and Y substitutions limits the ability of the protein to undergo the conformational rearrangements associated with transport. These proteins represent a basis for future studies on AtBOR1.
Collapse
Affiliation(s)
| | - Nicola J Scull
- Department of Life Sciences, Imperial College London, UK
| | | | - Yilmaz Alguel
- Department of Life Sciences, Imperial College London, UK
| | | | | | | |
Collapse
|
37
|
Morio H, Reien Y, Hirayama Y, Hashimoto H, Anzai N. Protein kinase C activation upregulates human L-type amino acid transporter 2 function. J Physiol Sci 2021; 71:11. [PMID: 33789576 PMCID: PMC10716992 DOI: 10.1186/s12576-021-00795-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 03/05/2021] [Indexed: 11/10/2022]
Abstract
L-type amino acid transporter 2 (LAT2) is a Na+-independent neutral amino acid transporter, whose function regulation system remains unclarified. Since protein kinase C (PKC) is known to regulate the functions of various transporters, we investigated whether human LAT2 (hLAT2) function is regulated by PKC. In mouse proximal tubule S2 cells, hLAT2 transport activity was upregulated by PKC activation. However, we found that the mRNA and protein expression of hLAT2 was not affected by PKC activation and that the upregulation was independent of the three potential PKC consensus sites in the hLAT2 amino acid sequence. Moreover, we found that PKC activation upregulated the Vmax value for hLAT2-mediated alanine transport, which was not accompanied by the induction of hLAT2 membrane insertion. In conclusion, we showed that hLAT2 function is upregulated by PKC activation, which is not related to either the de novo synthesis, the phosphorylation or the membrane insertion of hLAT2.
Collapse
Affiliation(s)
- Hanae Morio
- Department of Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba, 260-8670, Japan
| | - Yoshie Reien
- Department of Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba, 260-8670, Japan
| | - Yuri Hirayama
- Department of Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba, 260-8670, Japan
| | - Hirofumi Hashimoto
- Department of Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba, 260-8670, Japan
| | - Naohiko Anzai
- Department of Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuou-ku, Chiba, 260-8670, Japan.
- Department of Pharmacology and Toxicology, School of Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu-cho, Shimotsuga-gun, Tochigi, 321-0293, Japan.
| |
Collapse
|
38
|
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
|
39
|
Errasti-Murugarren E, Bartoccioni P, Palacín M. Membrane Protein Stabilization Strategies for Structural and Functional Studies. MEMBRANES 2021; 11:membranes11020155. [PMID: 33671740 PMCID: PMC7926488 DOI: 10.3390/membranes11020155] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 02/07/2023]
Abstract
Accounting for nearly two-thirds of known druggable targets, membrane proteins are highly relevant for cell physiology and pharmacology. In this regard, the structural determination of pharmacologically relevant targets would facilitate the intelligent design of new drugs. The structural biology of membrane proteins is a field experiencing significant growth as a result of the development of new strategies for structure determination. However, membrane protein preparation for structural studies continues to be a limiting step in many cases due to the inherent instability of these molecules in non-native membrane environments. This review describes the approaches that have been developed to improve membrane protein stability. Membrane protein mutagenesis, detergent selection, lipid membrane mimics, antibodies, and ligands are described in this review as approaches to facilitate the production of purified and stable membrane proteins of interest for structural and functional studies.
Collapse
Affiliation(s)
- Ekaitz Errasti-Murugarren
- Laboratory of Amino acid Transporters and Disease, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain;
- CIBERER (Centro Español en Red de Biomedicina de Enfermedades Raras), 28029 Barcelona, Spain
- Correspondence: (E.E.-M.); (M.P.)
| | - Paola Bartoccioni
- Laboratory of Amino acid Transporters and Disease, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain;
- CIBERER (Centro Español en Red de Biomedicina de Enfermedades Raras), 28029 Barcelona, Spain
| | - Manuel Palacín
- Laboratory of Amino acid Transporters and Disease, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain;
- CIBERER (Centro Español en Red de Biomedicina de Enfermedades Raras), 28029 Barcelona, Spain
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, 08028 Barcelona, Spain
- Correspondence: (E.E.-M.); (M.P.)
| |
Collapse
|
40
|
Errasti-Murugarren E, Palacín M. Heteromeric Amino Acid Transporters in Brain: from Physiology to Pathology. Neurochem Res 2021; 47:23-36. [PMID: 33606172 DOI: 10.1007/s11064-021-03261-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/27/2021] [Accepted: 01/30/2021] [Indexed: 12/12/2022]
Abstract
In humans, more than 50 transporters are responsible for the traffic and balance of amino acids within and between cells and tissues, and half of them have been associated with disease [1]. Covering all common amino acids, Heteromeric Amino acid Transporters (HATs) are one class of such transporters. This review first highlights structural and functional studies that solved the atomic structure of HATs and revealed molecular clues on substrate interaction. Moreover, this review focuses on HATs that have a role in the central nervous system (CNS) and that are related to neurological diseases, including: (i) LAT1/CD98hc and its role in the uptake of branched chain amino acids trough the blood brain barrier and autism. (ii) LAT2/CD98hc and its potential role in the transport of glutamine between plasma and cerebrospinal fluid. (iii) y+LAT2/CD98hc that is emerging as a key player in hepatic encephalopathy. xCT/CD98hc as a potential therapeutic target in glioblastoma, and (iv) Asc-1/CD98hc as a potential therapeutic target in pathologies with alterations in NMDA glutamate receptors.
Collapse
Affiliation(s)
- Ekaitz Errasti-Murugarren
- Institute for Research in Biomedicine. Institute of Science and Technology (BIST), 08028, Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 08028, Barcelona, Spain.
| | - Manuel Palacín
- Institute for Research in Biomedicine. Institute of Science and Technology (BIST), 08028, Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 08028, Barcelona, Spain. .,Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, 08028, Barcelona, Spain.
| |
Collapse
|
41
|
Zhang S, Zhou J, Zhang Y, Liu T, Friedel P, Zhuo W, Somasekharan S, Roy K, Zhang L, Liu Y, Meng X, Deng H, Zeng W, Li G, Forbush B, Yang M. The structural basis of function and regulation of neuronal cotransporters NKCC1 and KCC2. Commun Biol 2021; 4:226. [PMID: 33597714 PMCID: PMC7889885 DOI: 10.1038/s42003-021-01750-w] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 01/22/2021] [Indexed: 11/08/2022] Open
Abstract
NKCC and KCC transporters mediate coupled transport of Na++K++Cl- and K++Cl- across the plasma membrane, thus regulating cell Cl- concentration and cell volume and playing critical roles in transepithelial salt and water transport and in neuronal excitability. The function of these transporters has been intensively studied, but a mechanistic understanding has awaited structural studies of the transporters. Here, we present the cryo-electron microscopy (cryo-EM) structures of the two neuronal cation-chloride cotransporters human NKCC1 (SLC12A2) and mouse KCC2 (SLC12A5), along with computational analysis and functional characterization. These structures highlight essential residues in ion transport and allow us to propose mechanisms by which phosphorylation regulates transport activity.
Collapse
Affiliation(s)
- Sensen Zhang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jun Zhou
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yuebin Zhang
- State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Tianya Liu
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Perrine Friedel
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Wei Zhuo
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Suma Somasekharan
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Kasturi Roy
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Laixing Zhang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yang Liu
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xianbin Meng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, China
| | - Wenwen Zeng
- Center for Life Sciences, Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Guohui Li
- State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China.
| | - Biff Forbush
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Maojun Yang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China.
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
42
|
Abstract
Drug transporters are integral membrane proteins that play a critical role in drug disposition by affecting absorption, distribution, and excretion. They translocate drugs, as well as endogenous molecules and toxins, across membranes using ATP hydrolysis, or ion/concentration gradients. In general, drug transporters are expressed ubiquitously, but they function in drug disposition by being concentrated in tissues such as the intestine, the kidneys, the liver, and the brain. Based on their primary sequence and their mechanism, transporters can be divided into the ATP-binding cassette (ABC), solute-linked carrier (SLC), and the solute carrier organic anion (SLCO) superfamilies. Many X-ray crystallography and cryo-electron microscopy (cryo-EM) structures have been solved in the ABC and SLC transporter superfamilies or of their bacterial homologs. The structures have provided valuable insight into the structural basis of transport. This chapter will provide particular focus on the promiscuous drug transporters because of their effect on drug disposition and the challenges associated with them.
Collapse
Affiliation(s)
- Arthur G Roberts
- Pharmaceutical and Biomedical Sciences Department, University of Georgia, Athens, GA, USA.
| |
Collapse
|
43
|
Cosco J, Scalise M, Colas C, Galluccio M, Martini R, Rovella F, Mazza T, Ecker GF, Indiveri C. ATP modulates SLC7A5 (LAT1) synergistically with cholesterol. Sci Rep 2020; 10:16738. [PMID: 33028978 PMCID: PMC7541457 DOI: 10.1038/s41598-020-73757-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/17/2020] [Indexed: 01/07/2023] Open
Abstract
The plasma membrane transporter hLAT1 is responsible for providing cells with essential amino acids. hLAT1 is over-expressed in virtually all human cancers making the protein a hot-spot in the fields of cancer and pharmacology research. However, regulatory aspects of hLAT1 biology are still poorly understood. A remarkable stimulation of transport activity was observed in the presence of physiological levels of cholesterol together with a selective increase of the affinity for the substrate on the internal site, suggesting a stabilization of the inward open conformation of hLAT1. A synergistic effect by ATP was also observed only in the presence of cholesterol. The same phenomenon was detected with the native protein. Altogether, the biochemical assays suggested that cholesterol and ATP binding sites are close to each other. The computational analysis identified two neighboring regions, one hydrophobic and one hydrophilic, to which cholesterol and ATP were docked, respectively. The computational data predicted interaction of the ϒ-phosphate of ATP with Lys 204, which was confirmed by site-directed mutagenesis. The hLAT1-K204Q mutant showed an impaired function and response to ATP. Interestingly, this residue is conserved in several members of the SLC7 family.
Collapse
Affiliation(s)
- Jessica Cosco
- Department of DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, via Bucci 4C, 87036, Arcavacata di Rende, Italy
| | - Mariafrancesca Scalise
- Department of DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, via Bucci 4C, 87036, Arcavacata di Rende, Italy
| | - Claire Colas
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090, Wien, Austria
| | - Michele Galluccio
- Department of DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, via Bucci 4C, 87036, Arcavacata di Rende, Italy
| | - Riccardo Martini
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090, Wien, Austria
| | - Filomena Rovella
- Department of DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, via Bucci 4C, 87036, Arcavacata di Rende, Italy
| | - Tiziano Mazza
- Department of DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, via Bucci 4C, 87036, Arcavacata di Rende, Italy
| | - Gerhard F Ecker
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090, Wien, Austria
| | - Cesare Indiveri
- Department of DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, via Bucci 4C, 87036, Arcavacata di Rende, Italy. .,CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), via Amendola 122/O, 70126, Bari, Italy.
| |
Collapse
|
44
|
Cormerais Y, Vucetic M, Pouysségur J. Targeting amino acids transporters (SLCs) to starve cancer cells to death. Biochem Biophys Res Commun 2020; 520:691-693. [PMID: 31761081 DOI: 10.1016/j.bbrc.2019.10.173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 10/24/2019] [Indexed: 10/25/2022]
Affiliation(s)
- Yann Cormerais
- Department of Medical Biology, Centre Scientifique de Monaco (CSM), Monaco; Department of Genetics and Complex Diseases, Harvard Medical School, Boston, USA
| | - Milica Vucetic
- Department of Medical Biology, Centre Scientifique de Monaco (CSM), Monaco
| | - Jacques Pouysségur
- Department of Medical Biology, Centre Scientifique de Monaco (CSM), Monaco; University Côte d'Azur, (IRCAN), CNRS, INSERM, Centre A. Lacassagne, Nice, France.
| |
Collapse
|
45
|
Pandey B, Aarthy M, Sharma M, Singh SK, Kumar V. Computational analysis identifies druggable mutations in human rBAT mediated Cystinuria. J Biomol Struct Dyn 2020; 39:5058-5067. [PMID: 32602810 DOI: 10.1080/07391102.2020.1784792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Culex quinquefasciatus Cqm1 protein acts as the receptor for Lysinibacillus sphaericus mosquito-larvicidal binary (BinAB) toxin that is used worldwide for mosquito control. We found amino acid transporter protein, rBAT, as phylogenetically closest Cqm1 homolog in humans. The present study reveals large evolutionary distance between Cqm1 and rBAT, and rBAT ectodomain lacks the sequence motif which serves as binding-site for the BinAB toxin. Thus, BinAB toxin can be expected to remain safe for humans. rBAT (heavy subunit; SLC3A1) and catalytic b0,+AT (light subunit; SLC7A9), linked by single disulfide bond, mediate renal reabsorption of cystine and dibasic amino acids in Na+ independent manner. Mutations in rBAT cause type I Cystinuria disease which shows global prevalence, and rBAT can be thought as an important pharmacological target. However, 3D structures of rBAT and b0,+AT, the two components of b0,+ heteromeric amino acid transporter systems, are not available. We constructed a reliable homology model of rBAT using Cqm1 coordinates and that of transmembrane b0,+AT subunit using LAT1 coordinates. Mapping of pathogenic mutations onto rBAT ectodomain revealed their scattered distribution throughout the rBAT protein. Further, our computational simulations-based scoring of several known deleterious mutations of rBAT revealed that mutations those do not compromise the protein fold and stability, are localized on the same face of the molecule. These residues are expected to interact with the b0,+AT transporter. The present study thus identifies druggable sites on rBAT that could be targeted for the treatment of type I Cystinuria.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Bharati Pandey
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Murali Aarthy
- Computer-aided drug design Lab, Department of Bioinformatics, Alagappa University, Karaikudi, India
| | - Mahima Sharma
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Sanjeev Kumar Singh
- Computer-aided drug design Lab, Department of Bioinformatics, Alagappa University, Karaikudi, India
| | - Vinay Kumar
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
46
|
Yan R, Li Y, Shi Y, Zhou J, Lei J, Huang J, Zhou Q. Cryo-EM structure of the human heteromeric amino acid transporter b 0,+AT-rBAT. SCIENCE ADVANCES 2020; 6:eaay6379. [PMID: 32494597 PMCID: PMC7159911 DOI: 10.1126/sciadv.aay6379] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 01/22/2020] [Indexed: 05/16/2023]
Abstract
Heteromeric amino acid transporters (HATs) catalyze the transmembrane movement of amino acids, comprising two subunits, a heavy chain and a light chain, linked by a disulfide bridge. The b0,+AT (SLC7A9) is a representative light chain of HATs, forming heterodimer with rBAT, a heavy chain which mediates the membrane trafficking of b0,+AT. The b0,+AT-rBAT complex is an obligatory exchanger, which mediates the influx of cystine and cationic amino acids and the efflux of neutral amino acids in kidney and small intestine. Here, we report the cryo-EM structure of the human b0,+AT-rBAT complex alone and in complex with arginine substrate at resolution of 2.7 and 2.3 Å, respectively. The overall structure of b0,+AT-rBAT exists as a dimer of heterodimer consistent with the previous study. A ligand molecule is bound to the substrate binding pocket, near which an occluded pocket is identified, to which we found that it is important for substrate transport.
Collapse
Affiliation(s)
- Renhong Yan
- Key Laboratory of Structural Biology of Zhejiang Province, Institute of Biology, Westlake Institute for Advanced Study, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Yaning Li
- Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yi Shi
- Key Laboratory of Structural Biology of Zhejiang Province, Institute of Biology, Westlake Institute for Advanced Study, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Jiayao Zhou
- Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jianlin Lei
- Technology Center for Protein Sciences, Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jing Huang
- Key Laboratory of Structural Biology of Zhejiang Province, Institute of Biology, Westlake Institute for Advanced Study, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| | - Qiang Zhou
- Key Laboratory of Structural Biology of Zhejiang Province, Institute of Biology, Westlake Institute for Advanced Study, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou 310024, Zhejiang Province, China
| |
Collapse
|
47
|
Haffke M, Duckely M, Bergsdorf C, Jaakola VP, Shrestha B. Development of a biochemical and biophysical suite for integral membrane protein targets: A review. Protein Expr Purif 2020; 167:105545. [DOI: 10.1016/j.pep.2019.105545] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/18/2019] [Accepted: 11/24/2019] [Indexed: 12/11/2022]
|
48
|
Asc-1 Transporter (SLC7A10): Homology Models And Molecular Dynamics Insights Into The First Steps Of The Transport Mechanism. Sci Rep 2020; 10:3731. [PMID: 32111919 PMCID: PMC7048771 DOI: 10.1038/s41598-020-60617-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 02/14/2020] [Indexed: 12/02/2022] Open
Abstract
The alanine-serine-cysteine transporter Asc-1 regulates the synaptic availability of d-serine and glycine (the two co-agonists of the NMDA receptor) and is regarded as an important drug target. To shuttle the substrate from the extracellular space to the cytoplasm, this transporter undergoes multiple distinct conformational states. In this work, homology modeling, substrate docking and molecular dynamics simulations were carried out to learn more about the transition between the “outward-open” and “outward-open occluded” states. We identified a transition state involving the highly-conserved unwound TM6 region in which the Phe243 flips close to the d-serine substrate without major movements of TM6. This feature and those of other key residues are proposed to control the binding site and substrate translocation. Competitive inhibitors ACPP, LuAE00527 and SMLC were docked and their binding modes at the substrate binding site corroborated the key role played by Phe243 of TM6. For ACPP and LuAE00527, strong hydrophobic interactions with this residue hinder its mobility and prevent the uptake and the efflux of substrates. As for SMLC, the weaker interactions maintain the flexibility of Phe243 and the efflux process. Overall, we propose a molecular basis for the inhibition of substrate translocation of the Asc-1 transporter that should be valuable for rational drug design.
Collapse
|
49
|
Uchański T, Pardon E, Steyaert J. Nanobodies to study protein conformational states. Curr Opin Struct Biol 2020; 60:117-123. [DOI: 10.1016/j.sbi.2020.01.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 01/07/2023]
|
50
|
Qiu XM, Sun YY, Ye XY, Li ZG. Signaling Role of Glutamate in Plants. FRONTIERS IN PLANT SCIENCE 2020; 10:1743. [PMID: 32063909 PMCID: PMC6999156 DOI: 10.3389/fpls.2019.01743] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 12/11/2019] [Indexed: 05/11/2023]
Abstract
It is well known that glutamate (Glu), a neurotransmitter in human body, is a protein amino acid. It plays a very important role in plant growth and development. Nowadays, Glu has been found to emerge as signaling role. Under normal conditions, Glu takes part in seed germination, root architecture, pollen germination, and pollen tube growth. Under stress conditions, Glu participates in wound response, pathogen resistance, response and adaptation to abiotic stress (such as salt, cold, heat, and drought), and local stimulation (abiotic or biotic stress)-triggered long distance signaling transduction. In this review, in the light of the current opinion on Glu signaling in plants, the following knowledge was updated and discussed. 1) Glu metabolism; 2) signaling role of Glu in plant growth, development, and response and adaptation to environmental stress; as well as 3) the underlying research directions in the future. The purpose of this review was to look forward to inspiring the rapid development of Glu signaling research in plant biology, particularly in the field of stress biology of plants.
Collapse
Affiliation(s)
- Xue-Mei Qiu
- School of Life Sciences, Yunnan Normal University, Kunming, China
- Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Ministry of Education, Kunming, China
- Key Laboratory of Biomass Energy and Environmental Biotechnology, Yunnan Province, Yunnan Normal University, Kunming, China
| | - Yu-Ying Sun
- School of Life Sciences, Yunnan Normal University, Kunming, China
- Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Ministry of Education, Kunming, China
- Key Laboratory of Biomass Energy and Environmental Biotechnology, Yunnan Province, Yunnan Normal University, Kunming, China
| | - Xin-Yu Ye
- School of Life Sciences, Yunnan Normal University, Kunming, China
- Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Ministry of Education, Kunming, China
- Key Laboratory of Biomass Energy and Environmental Biotechnology, Yunnan Province, Yunnan Normal University, Kunming, China
| | - Zhong-Guang Li
- School of Life Sciences, Yunnan Normal University, Kunming, China
- Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Ministry of Education, Kunming, China
- Key Laboratory of Biomass Energy and Environmental Biotechnology, Yunnan Province, Yunnan Normal University, Kunming, China
| |
Collapse
|