1
|
Hou L, Chen Z, Chen F, Sheng L, Ye W, Dai Y, Guo X, Dong C, Li G, Liao K, Li Y, Ma J, Wei H, Ran W, Shang J, Ling X, Patel JS, Liang SH, Xu H, Wang L. Synthesis, preclinical assessment, and first-in-human study of [ 18F]d 4-FET for brain tumor imaging. Eur J Nucl Med Mol Imaging 2024:10.1007/s00259-024-06964-8. [PMID: 39482500 DOI: 10.1007/s00259-024-06964-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024]
Abstract
PURPOSE Tumor-to-background ratio (TBR) is a critical metric in oncologic PET imaging. This study aims to enhance the TBR of [18F]FET in brain tumor imaging by substituting deuterium ("D") for hydrogen ("H"), thereby improving the diagnostic sensitivity and accuracy. METHODS [18F]d4-FET was synthesised by two automated radiochemistry modules. Biodistribution studies and imaging efficacy were evaluated in vivo and ex vivo in rodent models, while metabolic stability and radiation dosimetry were assessed in non-human primates. Additionally, preliminary imaging evaluations were carried out in five brain tumor patients: three glioma patients underwent imaging with both [18F]d4-FET and [18F]FET, and two patients with brain metastases were imaged using [18F]d4-FET and [18F]FDG. RESULTS [18F]d4-FET demonstrated high radiochemical purity and yield. PET/MRI in rodent models demonstrated superior TBR for [18F]d4-FET compared to [18F]FET, and autoradiography showed tumor margins that correlated well with pathological extents. Studies in cynomolgus monkeys indicated comparable in vivo stability and effective dose with [18F]FET. In glioma patients, [18F]d4-FET showed enhanced TBR, while in patients with brain metastases, [18F]d4-FET displayed superior lesion delineation compared to [18F]FDG, especially in smaller metastatic sites. CONCLUSION We successfully synthesized the novel PET radiotracer [18F]d4-FET, which retains the advantageous properties of [18F]FET while potentially enhancing TBR for glioma imaging. Preliminary studies indicate excellent stability, efficacy, and sensitivity of [18F]d4-FET, suggesting its potential in clinical evaluations of brain tumors. TRIAL REGISTRATION ChiCTR2400081576, registration date: 2024-03-05, https://www.chictr.org.cn/bin/project/edit?pid=206162.
Collapse
Affiliation(s)
- Lu Hou
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Zhiyong Chen
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Fanfan Chen
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518025, China
| | - Lianghe Sheng
- Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Weijian Ye
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yingchu Dai
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Xiaoyu Guo
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Chenchen Dong
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Guocong Li
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Kai Liao
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yinlong Li
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA, 30322, USA
| | - Jie Ma
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Huiyi Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Wenqing Ran
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jingjie Shang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Xueying Ling
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Jimmy S Patel
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA, 30322, USA
| | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Rd, Atlanta, GA, 30322, USA.
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, & Key Laboratory of Basic and Translational Research On Radiopharmaceuticals, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
| |
Collapse
|
2
|
Järvinen J, Pulkkinen H, Rautio J, Timonen JM. Amino Acid-Based Boron Carriers in Boron Neutron Capture Therapy (BNCT). Pharmaceutics 2023; 15:2663. [PMID: 38140004 PMCID: PMC10748186 DOI: 10.3390/pharmaceutics15122663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/18/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Interest in the design of boronated amino acids has emerged, partly due to the utilization of boronophenylalanine (BPA), one of the two agents employed in clinical Boron Neutron Capture Therapy (BNCT). The boronated amino acids synthesized thus far for BNCT investigations can be classified into two categories based on the source of boron: boronic acids or carboranes. Amino acid-based boron carriers, employed in the context of BNCT treatment, demonstrate significant potential in the treatment of challenging tumors, such as those located in the brain. This review aims to shed light on the developmental journey and challenges encountered over the years in the field of amino acid-based boron delivery compound development. The primary focus centers on the utilization of the large amino acid transporter 1 (LAT1) as a target for boron carriers in BNCT. The development of efficient carriers remains a critical objective, addressing challenges related to tumor specificity, effective boron delivery, and rapid clearance from normal tissue and blood. LAT1 presents an intriguing and promising target for boron delivery, given its numerous characteristics that make it well suited for drug delivery into tumor tissues, particularly in the case of brain tumors.
Collapse
Affiliation(s)
- Juulia Järvinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Herkko Pulkkinen
- Department of Technical Physics, Faculty of Science, Forestry and Technology, University of Eastern Finland, 70211 Kuopio, Finland
| | - Jarkko Rautio
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Juri M. Timonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70211 Kuopio, Finland
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, P.O. Box 56, 00014 Helsinki, Finland
| |
Collapse
|
3
|
Qiu H, Shao N, Liu J, Zhao J, Chen C, Li Q, He Z, Zhao X, Xu L. Amino acid metabolism in tumor: New shine in the fog? Clin Nutr 2023:S0261-5614(23)00184-X. [PMID: 37321900 DOI: 10.1016/j.clnu.2023.06.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 05/10/2023] [Accepted: 06/03/2023] [Indexed: 06/17/2023]
Abstract
Alterations in amino acid metabolism is closely related to the occurrence of clinical diseases. The mechanism of tumorigenesis is complex, involving the complicated relationship between tumor cells and immune cells in local tumor microenvironment. A series of recent studies have shown that metabolic remodeling is intimately related to tumorigenesis. And amino acid metabolic reprogramming is one of the important characteristics of tumor metabolic remodeling, which participates in tumor cells growth, survival as well as the immune cell activation and function in the local tumor microenvironment, thereby affecting tumor immune escape. Recent studies have further shown that controlling the intake of specific amino acids can significantly improve the effect of clinical intervention in tumors, suggesting that amino acid metabolism is gradually becoming one of the new promising targets of clinical intervention in tumors. Therefore, developing new intervention strategies based on amino acid metabolism has broad prospects. In this article, we review the abnormal changes in the metabolism of some typical amino acids, including glutamine, serine, glycine, asparagine and so on in tumor cells and summarize the relationship among amino acid metabolism, tumor microenvironment and the function of T cells. In particular, we discuss the current issues that need to be addressed in the related fields of tumor amino acid metabolism, aiming to provide a theoretical basis for the development of new strategies for clinical interventions in tumors based on amino acid metabolism reprogramming.
Collapse
Affiliation(s)
- Hui Qiu
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi Guizhou 563000, China
| | - Nan Shao
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi Guizhou 563000, China
| | - Jing Liu
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi Guizhou 563000, China
| | - Juanjuan Zhao
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi Guizhou 563000, China
| | - Chao Chen
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi Guizhou 563000, China
| | - Qihong Li
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi Guizhou 563000, China
| | - Zhixu He
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi Guizhou 563000, China; Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi Guizhou 563000, China
| | - Xu Zhao
- School of Medicine, Guizhou University, Guizhou Guiyang, 550025 China; Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi Guizhou 563000, China.
| | - Lin Xu
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi Guizhou 563000, China.
| |
Collapse
|
4
|
Krishnan KS, Billups B. ASC Transporters Mediate D-Serine Transport into Astrocytes Adjacent to Synapses in the Mouse Brain. Biomolecules 2023; 13:biom13050819. [PMID: 37238689 DOI: 10.3390/biom13050819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
D-serine is an important signalling molecule, which activates N-methyl D-aspartate receptors (NMDARs) in conjunction with its fellow co-agonist, the neurotransmitter glutamate. Despite its involvement in plasticity and memory related to excitatory synapses, its cellular source and sink remain a question. We hypothesise that astrocytes, a type of glial cell that surrounds synapses, are likely candidates to control the extracellular concentration of D-Serine by removing it from the synaptic space. Using in situ patch clamp recordings and pharmacological manipulation of astrocytes in the CA1 region of the mouse hippocampal brain slices, we investigated the transport of D-serine across the plasma membrane. We observed the D-serine-induced transport-associated currents upon puff-application of 10 mM D-serine on astrocytes. Further, O-benzyl-L-serine and trans-4-hydroxy-proline, known substrate inhibitors of the alanine serine cysteine transporters (ASCT), reduced D-serine uptake. These results indicate that ASCT is a central mediator of astrocytic D-serine transport and plays a role in regulating its synaptic concentration by sequestration into astrocytes. Similar results were observed in astrocytes of the somatosensory cortex and Bergmann glia in the cerebellum, indicative of a general mechanism expressed across a range of brain areas. This removal of synaptic D-serine and its subsequent metabolic degradation are expected to reduce its extracellular availability, influencing NMDAR activation and NMDAR-dependent synaptic plasticity.
Collapse
Affiliation(s)
- Karthik Subramanian Krishnan
- Eccles Institute of Neuroscience, The John Curtin School of Medical Research, The Australian National University, 131 Garran Road, Canberra, ACT 2601, Australia
| | - Brian Billups
- Eccles Institute of Neuroscience, The John Curtin School of Medical Research, The Australian National University, 131 Garran Road, Canberra, ACT 2601, Australia
| |
Collapse
|
5
|
Wang X, Chen J, Liu XH, Zeng XY, Long QY, Liu YH, Mao Q. Evaluation of CD98 light chain-LAT1 as a potential marker of cancer stem-like cells in glioblastoma. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119303. [PMID: 35659617 DOI: 10.1016/j.bbamcr.2022.119303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 05/23/2022] [Accepted: 05/27/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE Glioma stem cells (GSCs) are a minority population of glioma cells that regarded as the cause of tumor formation and recurrence. Identifying new molecular strategies targeting GSCs must be urgently developed to treat glioblastoma. In this study, one of CD98 light chain-L type amino acid transporter 1 (LAT1) was found as a potential GSC marker. LAT1 served as EAA transporter has been shown to be closely related with tumor invasion, metastasis, angiogenesis, and radiosensitivity. METHODS LAT1+ and LAT1- glioma cells were sorted by flow cytometry. Cellular immunofluorescence, sphere-formation arrays, and in vitro limiting dilution experiments were used to identify cell stemness. Differentiated glioma stem cells were cultured, and the expressions of β-tubulinIII, GFAP, and LAT1 were detected by Western blot. Nude mouse models were constructed to observe tumor formation and metastasis in nude mice. RESULTS LAT1+ glioma cells were testified a small percentage of all cells and selected as the subsequent sorting marker. LAT1+ cells were separated from U87 and U251 cells could express high level of stem cell markers, and possessed GSC properties including self-renewal ability and multi-directional differentiation potential. But LAT1- cells did not have these characteristics. In addition, LAT1+ cells were able to generate tumors in vivo, tumor size of LAT1+ cells formed were much bigger than that of LAT1- cells. CONCLUSION Our study, including molecular, cell, vitro and vivo experiments, has shown that LAT1+ cells possess GSC properties, and present for the first time that LAT1 can be used as a new marker for GSCs screening.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Neurosurgery, West China Hospital of Sichuan University, China.
| | - Jinxiu Chen
- Department of Radiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Sciences and Technology of China, China
| | - Xiang-Hao Liu
- Department of Neurosurgery, West China Hospital of Sichuan University, China
| | - Xiang-Yi Zeng
- Department of Neurosurgery, West China Hospital of Sichuan University, China
| | - Qiang-You Long
- Department of Neurosurgery, West China Hospital of Sichuan University, China
| | - Yan-Hui Liu
- Department of Neurosurgery, West China Hospital of Sichuan University, China
| | - Qing Mao
- Department of Neurosurgery, West China Hospital of Sichuan University, China
| |
Collapse
|
6
|
Leiske MN, Mazrad ZAI, Zelcak A, Wahi K, Davis TP, McCarroll JA, Holst J, Kempe K. Zwitterionic Amino Acid-Derived Polyacrylates as Smart Materials Exhibiting Cellular Specificity and Therapeutic Activity. Biomacromolecules 2022; 23:2374-2387. [PMID: 35508075 DOI: 10.1021/acs.biomac.2c00143] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The synthesis of new amino acid-containing, cell-specific, therapeutically active polymers is presented. Amino acids served as starting material for the preparation of tailored polymers with different amino acids in the side chain. The reversible addition-fragmentation chain-transfer (RAFT) polymerization of acrylate monomers yielded polymers of narrow size distribution (Đ ≤ 1.3). In particular, glutamate (Glu)-functionalized, zwitterionic polymers revealed a high degree of cytocompatibility and cellular specificity, i.e., showing association to different cancer cell lines, but not with nontumor fibroblasts. Energy-dependent uptake mechanisms were confirmed by means of temperature-dependent cellular uptake experiments as well as localization of the polymers in cellular lysosomes determined by confocal laser scanning microscopy (CLSM). The amino acid receptor antagonist O-benzyl-l-serine (BzlSer) was chosen as an active ingredient for the design of therapeutic copolymers. RAFT copolymerization of Glu acrylate and BzlSer acrylate resulted in tailored macromolecules with distinct monomer ratios. The targeted, cytotoxic activity of copolymers was demonstrated by means of multiday in vitro cell viability assays. To this end, polymers with 25 mol % BzlSer content showed cytotoxicity against cancer cells, while leaving fibroblasts unaffected over a period of 3 days. Our results emphasize the importance of biologically derived materials to be included in synthetic polymers and the potential of zwitterionic, amino acid-derived materials for cellular targeting. Furthermore, it highlights that the fine balance between cellular specificity and unspecific cytotoxicity can be tailored by monomer ratios within a copolymer.
Collapse
Affiliation(s)
- Meike N Leiske
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Zihnil A I Mazrad
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Aykut Zelcak
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Kanu Wahi
- School of Medical Sciences and Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Thomas P Davis
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Joshua A McCarroll
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW 2052, Australia.,School of Women's and Children's Health, UNSW Sydney, Sydney, NSW 2052, Australia.,Australian Centre for NanoMedicine, UNSW Sydney, Sydney, NSW 2052, Australia.,UNSW RNA Institute, Sydney, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Jeff Holst
- School of Medical Sciences and Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Kristian Kempe
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia.,Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
7
|
Bay C, Bajraktari-Sylejmani G, Haefeli WE, Burhenne J, Weiss J, Sauter M. Functional Characterization of the Solute Carrier LAT-1 (SLC7A5/SLC2A3) in Human Brain Capillary Endothelial Cells with Rapid UPLC-MS/MS Quantification of Intracellular Isotopically Labelled L-Leucine. Int J Mol Sci 2022; 23:ijms23073637. [PMID: 35408997 PMCID: PMC8998838 DOI: 10.3390/ijms23073637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/22/2022] [Accepted: 03/25/2022] [Indexed: 11/16/2022] Open
Abstract
The solute carrier L-type amino acid transporter 1 (LAT-1/SLC7A5) is a viable target for drug delivery to the central nervous system (CNS) and tumors due to its high abundance at the blood-brain barrier and in tumor tissue. LAT-1 is only localized on the cell surface as a heterodimer with CD98, which is not required for transporter function. To support future CNS drug-delivery development based on LAT-1 targeting, we established an ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) assay for stable isotopically labeled leucine ([13C6, 15N]-L-leucine), with a dynamic range of 0.1-1000 ng/mL that can be applied for the functional testing of LAT-1 activity when combined with specific inhibitors and, consequently, the LAT-1 inhibition capacity of new compounds. The assay was established in a 96-well format, facilitating high-throughput experiments, and, hence, can support the screening for novel inhibitors. Applicable recommendations of the US Food and Drug Administration and European Medicines Agency for bioanalytical method validation were followed to validate the assay. The assay was applied to investigate the IC50 of two well-known LAT-1 inhibitors on hCMEC/D3 cells: the highly specific LAT-1 inhibitor JPH203, which was also used to demonstrate LAT-1 specific uptake, and the general system L inhibitor BCH. In addition, the [13C6, 15N]-L-leucine uptake was determined on two human brain capillary endothelial cell lines (NKIM-6 and hCMEC/D3), which were characterized for their expressional differences of LAT-1 at the protein and mRNA level and the surface amount of CD98. The IC50 values of the inhibitors were in concordance with previously reported values. Furthermore, the [13C6, 15N]-L-leucine uptake was significantly higher in hCMEC/D3 cells compared to NKIM-6 cells, which correlated with higher expression of LAT-1 and a higher surface amount of CD98. Therefore, the UPLC-MS/MS quantification of ([13C6, 15N]-L-leucine is a feasible strategy for the functional characterization of LAT-1 activity in cells or tissue.
Collapse
Affiliation(s)
| | | | | | | | | | - Max Sauter
- Correspondence: ; Tel.: +49-6221-56-32899
| |
Collapse
|
8
|
Sexually dimorphic and brain region-specific transporter adaptations in system x c- null mice. Neurochem Int 2020; 141:104888. [PMID: 33199267 DOI: 10.1016/j.neuint.2020.104888] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023]
Abstract
System xc- is a heterodimeric amino acid antiporter that, in the central nervous system, is best known for linking the import of L-cystine (CySS) with the export of L-glutamate for the production and maintenance of cellular glutathione (GSH) and extracellular glutamate levels, respectively. Yet, mice that are null for system xc- are healthy, fertile, and, morphologically, their brains are grossly normal. This suggests other glutamate and/or cyst(e)ine transport mechanisms may be upregulated in compensation. To test this, we measured the plasma membrane expression of Excitatory Amino Acid Transporters (EAATs) 1-3, the Alanine-Serine-Cysteine-Transporter (ASCT) 1, the sodium-coupled neutral amino acid transporter (SNAT) 3 and the L Amino Acid Transporter (LAT) 2 in striatum, hippocampus and cortex of male and female mice using Western Blot analysis. Present results demonstrate brain region and transporter-specific changes occurs in female system xc- null mice with increased expression of EAAT1 and ASCT1 occurring in the striatum and cortex, respectively, and decreased SNAT 3 expression in cortex. In male system xc- null brain, only SNAT3 was altered significantly - increasing in the cortex, but decreasing in the striatum. Total levels of GSH and CyS were similar to that found in age and sex-matched littermate control mice, however, reductions in the ratio of reduced to oxidized GSH (GSH/GSSG) - a hallmark of oxidative stress - were found in all three brain regions in female system xc- null mice, whereas this occurred exclusively in the striatum of males. Protein levels of Superoxide dismutase (SOD) 1 were reduced, whereas SOD2 was enhanced in the hippocampus of male xc- null mice only. Finally, striatal vulnerability to 3-nitropropionic acid (3-NP)-mediated oxidative stress in either sex showed no genotype difference, although 3-NP was more toxic to female mice of either genotype, as evidenced by an increase in moribundity as compared to males.
Collapse
|
9
|
Peng H, Wang Y, Luo W. Multifaceted role of branched-chain amino acid metabolism in cancer. Oncogene 2020; 39:6747-6756. [PMID: 32978521 PMCID: PMC7606751 DOI: 10.1038/s41388-020-01480-z] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/09/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023]
Abstract
Metabolic reprogramming fulfils increased nutrient demands and regulates
numerous oncogenic processes in tumors, leading to tumor malignancy.
Branched-chain amino acids (BCAAs, i.e., valine, leucine, and isoleucine)
function as nitrogen donors to generate macromolecules such as nucleotides and
are indispensable for human cancer cell growth. The cell-autonomous and
non-autonomous roles of altered BCAA metabolism have been implicated in cancer
progression and the key proteins in the BCAA metabolic pathway serve as possible
prognostic and diagnostic biomarkers in human cancers. Here we summarize how
BCAA metabolic reprogramming is regulated in cancer cells and how it influences
cancer progression.
Collapse
Affiliation(s)
- Hui Peng
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Yingfei Wang
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA. .,Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| | - Weibo Luo
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA. .,Department of Pharmacology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| |
Collapse
|
10
|
Satou M, Wang J, Nakano-Tateno T, Teramachi M, Suzuki T, Hayashi K, Lamothe S, Hao Y, Kurata H, Sugimoto H, Chik C, Tateno T. L-type amino acid transporter 1, LAT1, in growth hormone-producing pituitary tumor cells. Mol Cell Endocrinol 2020; 515:110868. [PMID: 32579901 DOI: 10.1016/j.mce.2020.110868] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 05/03/2020] [Accepted: 05/11/2020] [Indexed: 11/22/2022]
Abstract
Pituitary tumors (PTs) can cause significant mortality and morbidity due to limited therapeutic options. L-type amino acid transporters (LATs), in particular, the LAT1 isoform, is expressed in a variety of tumor cells. Pharmacological inhibition or genetic ablation of LAT1 can suppress leucine transport into cancer cells, resulting in suppression of cancer cell growth. However, roles of LAT1 in PTs have not been elucidated. Therefore, we assessed LAT1 expression in PTs and evaluated a LAT1-specific inhibitor, JPH203, on rat somatomammotroph tumor cells, GH4 cells. GH4 cells dominantly express LAT1 mRNA rather than other LAT isoforms, whereas LAT2 transcripts were most abundant in normal rat pituitary tissues. JPH203 inhibited leucine uptake and cell growth in GH4 cells in a concentration-dependent manner, and appeared to be independent of the mechanistic target, the rapamycin pathway. Although JPH203 did not induce apoptosis, it suppressed growth hormone production in GH4 cells. Also, genetic downregulation of LAT1 showed similar effects on cell growth and hormone production. These results indicated that restriction of LAT1 substrates by JPH203 modulated both cell growth and hormone production. In conclusion, LAT1 may be a new therapeutic target for PTs because its inhibition leads to suppression of cell growth as well as hormone production. JPH203 may represent a promising drug for clinical use in patients with PTs, with the potential of hormonal control and tumor suppression.
Collapse
Affiliation(s)
- Motoyasu Satou
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Department of Biochemistry, Dokkyo Medical University School of Medicine, Mibu, Tochigi, Japan
| | - Jason Wang
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Tae Nakano-Tateno
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Mariko Teramachi
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | | - Keitaro Hayashi
- Department of Pharmacology and Toxicology, Dokkyo Medical University School of Medicine, Mibu, Tochigi, Japan
| | - Shawn Lamothe
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Yubin Hao
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Harley Kurata
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Hiroyuki Sugimoto
- Department of Biochemistry, Dokkyo Medical University School of Medicine, Mibu, Tochigi, Japan
| | - Constance Chik
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Toru Tateno
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
11
|
Rani N, Singh B, Kumar N, Singh P, Hazari PP, Jaswal A, Gupta SK, Chhabra R, Radotra BD, Mishra AK. The diagnostic performance of 99mTc-methionine single-photon emission tomography in grading glioma preoperatively: a comparison with histopathology and Ki-67 indices. Nucl Med Commun 2020; 41:848-857. [PMID: 32796472 DOI: 10.1097/mnm.0000000000001230] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To characterize glioma preoperatively using quantitative 99mTc-methionine SPECT and comparison with MR-perfusion/spectroscopy and histopatholgical/Ki-67 scoring. METHODS Twenty-nine patients (21M: 8F; mean age 42.3 ± 10.5 years) with clinical and radiological suspicion of glioma assessed by 99mTc-MDM/SPECT and ceMRI. Additionally, 12/29 patients underwent dynamic susceptibility contrast-enhanced (DSCE) MRI and magnetic resonance spectroscopy (MRS) examination. Three patients with benign pathologies were recruited as controls. Histopathological tumor analysis was done in all (n = 29) the patients, and the Ki-67 index was evaluated in 20/29 patients. The target-to-nontarget (T/NT) methionine tumor uptake ratios, normalized cerebral blood volume (nCBV) and metabolites [choline/N-acetyl aspartate (Cho/NAA), Cho/creatine (Cr), Cr/NAA and Cr/Cho) ratios were measured in tumor areas. RESULTS On histopathological analysis, 26/29 patients had glioma (G IV-13; G III-04; G II-09). The mean T/NT ratio in G-II was significantly lower (2.46 ± 2.3) than in G-III (7.13 ± 2.2) and G-IV (5.16 ± 1.2). However, the mean ratio was highest (15.9 ± 6.8) in meningioma (n=3). The T/NT cutoff ratio of 3.08 provided 100% sensitivity, 87.5% specificity for discriminating high-grade glioma (HGG) from low-grade glioma (LGG) disease. Likewise, the nCBV cutoff of 2.43 offered 100% sensitivity and 80% specificity. Only the Cho/NAA cutoff value of greater than 3.34 provided reasonable sensitivity and specificity of 85.7% and 80.0% respectively for this differentiation. T/NT ratio correlated significantly with nCBV and Cho/NAA, Cho/Cr ratios but not with Ki-67. CONCLUSION Quantitative 99mTc-MDM -SPECT provided high sensitivity and specificity to differentiate HGG versus LGG preoperatively and demonstrated a potential role for the differential diagnosis of glial versus nonglial tumors.
Collapse
Affiliation(s)
| | | | | | | | - Puja P Hazari
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Science, DRDO, New Delhi
| | - Ambika Jaswal
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Science, DRDO, New Delhi
| | | | | | | | - Anil K Mishra
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Science, DRDO, New Delhi
| |
Collapse
|
12
|
Sakata T, Hana K, Mikami T, Yoshida T, Endou H, Okayasu I. Positive correlation of expression of L-type amino-acid transporter 1 with colorectal tumor progression and prognosis: Higher expression in sporadic colorectal tumors compared with ulcerative colitis-associated neoplasia. Pathol Res Pract 2020; 216:152972. [PMID: 32359697 DOI: 10.1016/j.prp.2020.152972] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 04/04/2020] [Accepted: 04/11/2020] [Indexed: 01/06/2023]
Abstract
The role of L-type amino-acid transporter 1 (LAT1), an oncofetal protein, in tumor progression is not well known, although it is important for the survival and proliferation of cancer cells. LAT1 expression was immunohistochemically analyzed and compared in sporadic (conventional) colorectal tumors and ulcerative colitis (UC)-associated neoplasia development and progression. LAT1 expression showed a significant stepwise increase in the order: conventional low-grade tubular adenoma, high-grade tubular adenoma, and invasive adenocarcinoma. Similarly, the same increasing trend in LAT1 expression was found in UC-associated low-grade dysplasia, high-grade dysplasia, and adenocarcinoma, whereas expression was significantly lower compared with that in an adenoma-adenocarcinoma series. LAT1 expression was predominant in the upper half of mucosal lesions in low-grade adenoma. This localized difference in LAT1 expression between the upper and lower halves of mucosal lesions disappeared in conventional high-grade adenoma and adenocarcinoma. LAT1 expression in the colorectal mucosa was significantly increased in the order: nontumor mucosa, quiescent phase of UC, and active phase of UC. Considering the histological pattern of Ki-67 labeling, LAT1 expression appeared partly related to cell proliferation, but this was not significant. In relation to the prognosis of patients with sporadic phase IV colorectal adenocarcinoma, this was significantly poorer in the group with high LAT1 expression compared with that with low LAT1 expression. This suggests LAT1 expression may be used as a companion biomarker for anti-cancer therapy targeting the LAT1 molecule in colorectal cancers.
Collapse
Affiliation(s)
| | | | - Tetuo Mikami
- Department of Pathology, Toho University School of Medicine, Tokyo, Japan
| | - Tutomu Yoshida
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | | | - Isao Okayasu
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan; Division of Nutrition, Faculty of Health Care, Kiryu University, Midori, Japan.
| |
Collapse
|
13
|
Lieu EL, Nguyen T, Rhyne S, Kim J. Amino acids in cancer. Exp Mol Med 2020; 52:15-30. [PMID: 31980738 PMCID: PMC7000687 DOI: 10.1038/s12276-020-0375-3] [Citation(s) in RCA: 400] [Impact Index Per Article: 100.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/24/2019] [Accepted: 12/02/2019] [Indexed: 01/22/2023] Open
Abstract
Over 90 years ago, Otto Warburg's seminal discovery of aerobic glycolysis established metabolic reprogramming as one of the first distinguishing characteristics of cancer1. The field of cancer metabolism subsequently revealed additional metabolic alterations in cancer by focusing on central carbon metabolism, including the citric acid cycle and pentose phosphate pathway. Recent reports have, however, uncovered substantial non-carbon metabolism contributions to cancer cell viability and growth. Amino acids, nutrients vital to the survival of all cell types, experience reprogrammed metabolism in cancer. This review outlines the diverse roles of amino acids within the tumor and in the tumor microenvironment. Beyond their role in biosynthesis, they serve as energy sources and help maintain redox balance. In addition, amino acid derivatives contribute to epigenetic regulation and immune responses linked to tumorigenesis and metastasis. Furthermore, in discussing the transporters and transaminases that mediate amino acid uptake and synthesis, we identify potential metabolic liabilities as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Elizabeth L. Lieu
- 0000 0001 2175 0319grid.185648.6Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL USA
| | - Tu Nguyen
- 0000 0001 2175 0319grid.185648.6Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL USA
| | - Shawn Rhyne
- 0000 0001 2175 0319grid.185648.6Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL USA
| | - Jiyeon Kim
- 0000 0001 2175 0319grid.185648.6Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL USA
| |
Collapse
|
14
|
Rahaman KA, Hasan M, Seo JE, Muresan AR, Song HJ, Min H, Son J, Lee J, Lee J, Kim B, Kwon OS. Severity of the autoimmune encephalomyelitis symptoms in mouse model by inhibition of LAT-1 transporters. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2019. [DOI: 10.1007/s40005-019-00468-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
15
|
Huttunen J, Gynther M, Vellonen KS, Huttunen KM. L-Type amino acid transporter 1 (LAT1)-utilizing prodrugs are carrier-selective despite having low affinity for organic anion transporting polypeptides (OATPs). Int J Pharm 2019; 571:118714. [DOI: 10.1016/j.ijpharm.2019.118714] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/22/2019] [Accepted: 09/16/2019] [Indexed: 12/16/2022]
|
16
|
Huttunen J, Peltokangas S, Gynther M, Natunen T, Hiltunen M, Auriola S, Ruponen M, Vellonen KS, Huttunen KM. L-Type Amino Acid Transporter 1 (LAT1/Lat1)-Utilizing Prodrugs Can Improve the Delivery of Drugs into Neurons, Astrocytes and Microglia. Sci Rep 2019; 9:12860. [PMID: 31492955 PMCID: PMC6731241 DOI: 10.1038/s41598-019-49009-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 08/18/2019] [Indexed: 12/27/2022] Open
Abstract
l-Type Amino Acid Transporter 1 (LAT1/Lat1) is responsible for carrying large, neutral l-amino acids as well as several drugs and prodrugs across the blood-brain barrier (BBB). However, the BBB is not the only barrier that hinders drugs acting effectively within the brain; the brain parenchymal cell membranes represent a secondary barrier for the drugs with intracellular target sites. In this study, expression and function of Lat1 was quantified in mouse primary neuron, astrocyte and immortalized microglia (BV2) cultures. Moreover, ability of Lat1 to carry prodrugs inside these brain cells was evaluated. The results showed that Lat1 was localized at the similar level in all studied cells (3.07 ± 0.92–3.77 ± 0.91 fmol/µg protein). The transporter was also functional in all three cell types, astrocytes having the highest transport capacity and affinity for the LAT1/Lat1-substrate, [14C]-l-leucine, followed by neurons and microglia. The designed prodrugs (1-6) were able to utilize Lat1 for their cellular uptake and it was mainly much higher than the one of their parent drugs. Interestingly, improved cellular uptake was also achieved in cells representing Alzheimer’s Disease phenotype. Therefore, improved delivery and intra-brain targeting of drugs can be attained by utilizing LAT1/Lat1 and prodrug approach.
Collapse
Affiliation(s)
- Johanna Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Soile Peltokangas
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Mikko Gynther
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Teemu Natunen
- Institute of Biomedicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Mikko Hiltunen
- Institute of Biomedicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Seppo Auriola
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Marika Ruponen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Kati-Sisko Vellonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
17
|
Abstract
PURPOSE This pilot study aimed to evaluate the amino acid tracer F-FACBC with simultaneous PET/MRI in diagnostic assessment and neurosurgery of gliomas. MATERIALS AND METHODS Eleven patients with suspected primary or recurrent low- or high-grade glioma received an F-FACBC PET/MRI examination before surgery. PET and MRI were used for diagnostic assessment, and for guiding tumor resection and histopathological tissue sampling. PET uptake, tumor-to-background ratios (TBRs), time-activity curves, as well as PET and MRI tumor volumes were evaluated. The sensitivities of lesion detection and to detect glioma tissue were calculated for PET, MRI, and combined PET/MRI with histopathology (biopsies for final diagnosis and additional image-localized biopsies) as reference. RESULTS Overall sensitivity for lesion detection was 54.5% (95% confidence interval [CI], 23.4-83.3) for PET, 45.5% (95% CI, 16.7-76.6) for contrast-enhanced MRI (MRICE), and 100% (95% CI, 71.5-100.0) for combined PET/MRI, with a significant difference between MRICE and combined PET/MRI (P = 0.031). TBRs increased with tumor grade (P = 0.004) and were stable from 10 minutes post injection. PET tumor volumes enclosed most of the MRICE volumes (>98%) and were generally larger (1.5-2.8 times) than the MRICE volumes. Based on image-localized biopsies, combined PET/MRI demonstrated higher concurrence with malignant findings at histopathology (89.5%) than MRICE (26.3%). CONCLUSIONS Low- versus high-grade glioma differentiation may be possible with F-FACBC using TBR. F-FACBC PET/MRI outperformed MRICE in lesion detection and in detection of glioma tissue. More research is required to evaluate F-FACBC properties, especially in grade II and III tumors, and for different subtypes of gliomas.
Collapse
|
18
|
Taslimifar M, Buoso S, Verrey F, Kurtcuoglu V. Propagation of Plasma L-Phenylalanine Concentration Fluctuations to the Neurovascular Unit in Phenylketonuria: An in silico Study. Front Physiol 2019; 10:360. [PMID: 31105574 PMCID: PMC6454150 DOI: 10.3389/fphys.2019.00360] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 03/14/2019] [Indexed: 01/21/2023] Open
Abstract
Phenylketonuria (PKU) is an inherited metabolic disease characterized by abnormally high concentrations of the essential amino acid L-phenylalanine (Phe) in blood plasma caused by reduced activity of phenylalanine hydroxylase (PAH). While numerous studies have shown association between high plasma Phe concentration and intellectual impairment, it is not clear whether increased Phe fluctuations also observed in PKU affect the brain as well. To investigate this, time-resolved in vivo data on Phe and competing large neutral amino acid (LNAA) concentrations in neurons are needed, but cannot be acquired readily with current methods. We have used in silico modeling as an alternative approach to characterize the interactive dynamics of Phe and competing LNAAs (CL) in the neurovascular unit (NVU). Our results suggest that plasma Phe fluctuations can propagate into the NVU cells and change there the concentration of LNAAs, with the highest magnitude of this effect observed at low frequency and high amplitude-to-mean ratio of the plasma Phe concentration fluctuations. Our model further elucidates the effect of therapeutic LNAA supplementation in PKU, showing how abnormal concentrations of Phe and CL in the NVU move thereby toward normal physiologic levels.
Collapse
Affiliation(s)
- Mehdi Taslimifar
- The Interface Group, Institute of Physiology, University of Zurich, Zurich, Switzerland.,Epithelial Transport Group, Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Stefano Buoso
- The Interface Group, Institute of Physiology, University of Zurich, Zurich, Switzerland.,Institute for Diagnostic and Interventional Radiology, Zurich University Hospital, Zurich, Switzerland
| | - Francois Verrey
- Epithelial Transport Group, Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,National Center of Competence in Research, Kidney Control of Homeostasis, Zurich, Switzerland
| | - Vartan Kurtcuoglu
- The Interface Group, Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,National Center of Competence in Research, Kidney Control of Homeostasis, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
19
|
In vivo assessment of increased oxidation of branched-chain amino acids in glioblastoma. Sci Rep 2019; 9:340. [PMID: 30674979 PMCID: PMC6344513 DOI: 10.1038/s41598-018-37390-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 12/04/2018] [Indexed: 01/01/2023] Open
Abstract
Altered branched-chain amino acids (BCAAs) metabolism is a distinctive feature of various cancers and plays an important role in sustaining tumor proliferation and aggressiveness. Despite the therapeutic and diagnostic potentials, the role of BCAA metabolism in cancer and the activities of associated enzymes remain unclear. Due to its pivotal role in BCAA metabolism and rapid cellular transport, hyperpolarized 13C-labeled α-ketoisocaproate (KIC), the α-keto acid corresponding to leucine, can assess both BCAA aminotransferase (BCAT) and branched-chain α-keto acid dehydrogenase complex (BCKDC) activities via production of [1-13C]leucine or 13CO2 (and thus H13CO3−), respectively. Here, we investigated BCAA metabolism of F98 rat glioma model in vivo using hyperpolarized 13C-KIC. In tumor regions, we observed a decrease in 13C-leucine production from injected hyperpolarized 13C-KIC via BCAT compared to the contralateral normal-appearing brain, and an increase in H13CO3−, a catabolic product of KIC through the mitochondrial BCKDC. A parallel ex vivo13C NMR isotopomer analysis following steady-state infusion of [U-13C]leucine to glioma-bearing rats verified the increased oxidation of leucine in glioma tissue. Both the in vivo hyperpolarized KIC imaging and the leucine infusion study indicate that KIC catabolism is upregulated through BCAT/BCKDC and further oxidized via the citric acid cycle in F98 glioma.
Collapse
|
20
|
Gaonkar RH, Baishya R, Paul B, Dewanjee S, Ganguly S, Debnath MC, Ganguly S. Development of a peptide-based bifunctional chelator conjugated to a cytotoxic drug for the treatment of melanotic melanoma. MEDCHEMCOMM 2018; 9:812-826. [PMID: 30108970 PMCID: PMC6071751 DOI: 10.1039/c7md00638a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/28/2018] [Indexed: 01/02/2023]
Abstract
The cytotoxic drug gemcitabine (GEM) has been conjugated to receptor-binding peptides to target melanoma tumors. A hexapeptide having a Lys-Gly-His-Lys sequence (pep-1), an octapeptide with an Arg-Gly-Asp-Lys-Gly-His-Lys sequence (pep-2), a GEM-conjugated Lys-Gly-His-Lys peptide (GEM-pep-3) and a GEM-conjugated Asp-Gly-Arg peptide (GEM-pep-4) were synthesized and characterized. In vitro uptake of fluorescently labeled GEM-pep-3 and GEM-pep-4 on B16F10 cells was investigated. Fluorescence microscopy studies demonstrated significant uptake of GEM-pep-3 in the B16F10 mouse melanoma cell line. The peptides and GEM-coupled peptides were radiolabeled with [99mTc(CO)3(H2O)3]+ and examined for in vitro cell binding in the B16F10 melanoma cell line and in vivo biodistribution and scintigraphic studies in a B16F10 melanoma tumor-bearing mice model. In vitro cellular uptake studies and biological evaluation confirmed significant deposition of GEM-pep-3 at the melanoma tumor site. The MTT assay depicted higher cytotoxic behaviour of GEM-pep-3 than free GEM. A considerable amount of cell apoptosis was also observed in B16F10 cells. Finally, the in vivo therapeutic efficacy study revealed a significant decrease in tumor growth in the GEM-pep-3-treated animal model. These studies reveal enough potentiality of GEM-pep-3 to treat melanoma and underline the need for further evaluation.
Collapse
Affiliation(s)
- Raghuvir H Gaonkar
- Infectious Diseases and Immunology Division , CSIR-Indian Institute of Chemical Biology , Kolkata , India .
| | - Rinku Baishya
- Natural Product Chemistry Group , Chemical Science and Technology Division , North East Institute of Science and Technology , Assam , India
| | - Brahamacharry Paul
- Infectious Diseases and Immunology Division , CSIR-Indian Institute of Chemical Biology , Kolkata , India .
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory , Department of Pharmaceutical Technology , Jadavpur University , Kolkata , India
| | - Shantanu Ganguly
- Regional Radiation Medicine Center , Thakurpukur Cancer Center and Welfare Home Campus , Kolkata , India
| | - Mita C Debnath
- Infectious Diseases and Immunology Division , CSIR-Indian Institute of Chemical Biology , Kolkata , India .
| | - Soumya Ganguly
- Infectious Diseases and Immunology Division , CSIR-Indian Institute of Chemical Biology , Kolkata , India .
| |
Collapse
|
21
|
l-Dopa and Fluoxetine Upregulate Astroglial 5-HT2B Receptors and Ameliorate Depression in Parkinson’s Disease Mice. ACTA ACUST UNITED AC 2018. [DOI: 10.3390/neuroglia1010006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Here, we report the association between depressive behavior (anhedonia) and astroglial expression of 5-hydroxytryptamine receptor 2B (5-HT2B) in an animal model of Parkinson’s disease, induced by bilateral injection of 6-hydroxydopamine (6-OHDA) into the striatum. Expression of the 5-HT2B receptor at the mRNA and protein level was decreased in the brain tissue of 6-OHDA-treated animals with anhedonia. Expression of the 5-HT2B receptor was corrected by four weeks treatment with either l-3,4-dihydroxyphenylalanine (l-dopa) or fluoxetine. Simultaneously, treatment with l-dopa abolished 6-OHDA effects on both depressive behavior and motor activity. In contrast, fluoxetine corrected 6-OHDA-induced depression but did not affect 6-OHDA-induced motor deficiency. In addition, 6-OHDA downregulated gene expression of the 5-HT2B receptor in astrocytes in purified cell culture and this downregulation was corrected by both l-dopa and fluoxetine. Our findings suggest that 6-OHDA-induced depressive behavior may be related to the downregulation of gene expression of the 5-HT2B receptor but 6-OHDA-induced motor deficiency reflects, arguably, dopamine depletion. Previously, we demonstrated that fluoxetine regulates gene expression in astrocytes by 5-HT2B receptor-mediated transactivation of epidermal growth factor receptor (EGFR). However, the underlying mechanism of l-dopa action remains unclear. The present work indicates that the decrease of gene expression of the astroglial 5-HT2B receptor may contribute to development of depressive behavior in Parkinson’s disease.
Collapse
|
22
|
Taslimifar M, Buoso S, Verrey F, Kurtcuoglu V. Functional Polarity of Microvascular Brain Endothelial Cells Supported by Neurovascular Unit Computational Model of Large Neutral Amino Acid Homeostasis. Front Physiol 2018; 9:171. [PMID: 29593549 PMCID: PMC5859092 DOI: 10.3389/fphys.2018.00171] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 02/20/2018] [Indexed: 11/13/2022] Open
Abstract
The homeostatic regulation of large neutral amino acid (LNAA) concentration in the brain interstitial fluid (ISF) is essential for proper brain function. LNAA passage into the brain is primarily mediated by the complex and dynamic interactions between various solute carrier (SLC) transporters expressed in the neurovascular unit (NVU), among which SLC7A5/LAT1 is considered to be the major contributor in microvascular brain endothelial cells (MBEC). The LAT1-mediated trans-endothelial transport of LNAAs, however, could not be characterized precisely by available in vitro and in vivo standard methods so far. To circumvent these limitations, we have incorporated published in vivo data of rat brain into a robust computational model of NVU-LNAA homeostasis, allowing us to evaluate hypotheses concerning LAT1-mediated trans-endothelial transport of LNAAs across the blood brain barrier (BBB). We show that accounting for functional polarity of MBECs with either asymmetric LAT1 distribution between membranes and/or intrinsic LAT1 asymmetry with low intraendothelial binding affinity is required to reproduce the experimentally measured brain ISF response to intraperitoneal (IP) L-tyrosine and L-phenylalanine injection. On the basis of these findings, we have also investigated the effect of IP administrated L-tyrosine and L-phenylalanine on the dynamics of LNAAs in MBECs, astrocytes and neurons. Finally, the computational model was shown to explain the trans-stimulation of LNAA uptake across the BBB observed upon ISF perfusion with a competitive LAT1 inhibitor.
Collapse
Affiliation(s)
- Mehdi Taslimifar
- The Interface Group, Institute of Physiology, University of Zurich, Zurich, Switzerland.,Epithelial Transport Group, Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Stefano Buoso
- The Interface Group, Institute of Physiology, University of Zurich, Zurich, Switzerland.,Institute for Diagnostic and Interventional Radiology, Zurich University Hospital, Zurich, Switzerland
| | - Francois Verrey
- Epithelial Transport Group, Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,National Center of Competence in Research, Kidney.CH, Zurich, Switzerland
| | - Vartan Kurtcuoglu
- The Interface Group, Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,National Center of Competence in Research, Kidney.CH, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
24
|
Verkhratsky A, Nedergaard M. Physiology of Astroglia. Physiol Rev 2018; 98:239-389. [PMID: 29351512 PMCID: PMC6050349 DOI: 10.1152/physrev.00042.2016] [Citation(s) in RCA: 961] [Impact Index Per Article: 160.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/22/2017] [Accepted: 04/27/2017] [Indexed: 02/07/2023] Open
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
25
|
Phenylglycine analogs are inhibitors of the neutral amino acid transporters ASCT1 and ASCT2 and enhance NMDA receptor-mediated LTP in rat visual cortex slices. Neuropharmacology 2017; 126:70-83. [PMID: 28807674 DOI: 10.1016/j.neuropharm.2017.08.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 07/31/2017] [Accepted: 08/09/2017] [Indexed: 01/18/2023]
Abstract
The N-methyl-d-aspartate receptor (NMDA) co-agonist d-serine is a substrate for the neutral amino acid transporters ASCT1 (SLC1A4) and ASCT2 (SLC1A5). We identified l-phenylglycine (PG) and its analogs as inhibitors of ASCT1 and ASCT2. PG analogs were shown to be non-substrate inhibitors of ASCT1 and ASCT2 with a range of activities relative to other amino acid transport systems, including sodium-dependent glutamate transporters, the sodium-independent d-serine transporter asc-1 and system L. L-4-chloroPG was the most potent and selective ASCT1/2 inhibitor identified. The PG analogs facilitated theta-burst induced long-term potentiation in rat visual cortex slices in a manner that was dependent on extracellular d-serine. For structurally-related PG analogs, there was an excellent correlation between ASCT1/2 transport inhibition and enhancement of LTP which was not the case for inhibition of asc-1 or system L. The ability of PG analogs to enhance LTP is likely due to inhibition of d-serine transport by ASCT1/2, leading to elevated extracellular levels of d-serine and increased NMDA receptor activity. These results suggest that ASCT1/2 may play an important role in regulating extracellular d-serine and NMDA receptor-mediated physiological effects and that ASCT1/2 inhibitors have the potential for therapeutic benefit.
Collapse
|
26
|
Felber M, Bauwens M, Imstepf S, Fox T, Mottaghy FM, Alberto R. Biological Evaluation of
l
‐Tyrosine Labelled with
fac
‐[
99m
Tc(CO)
3
]
+
at a
para
‐OH‐Coupled 2,3‐Diaminopropionic Acid Based Chelator. Eur J Inorg Chem 2017. [DOI: 10.1002/ejic.201601316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Michael Felber
- Department of Chemistry University of Zürich Winterthurerstr. 190 8057 Zürich Switzerland
| | - Matthias Bauwens
- Department of Nuclear Medicine Maastricht University Medical Center P. Debeyelaan 25 6229 Maastricht The Netherlands
| | - Sebastian Imstepf
- Department of Chemistry University of Zürich Winterthurerstr. 190 8057 Zürich Switzerland
| | - Thomas Fox
- Department of Chemistry University of Zürich Winterthurerstr. 190 8057 Zürich Switzerland
| | - Felix M. Mottaghy
- Department of Nuclear Medicine Maastricht University Medical Center P. Debeyelaan 25 6229 Maastricht The Netherlands
| | - Roger Alberto
- Department of Chemistry University of Zürich Winterthurerstr. 190 8057 Zürich Switzerland
| |
Collapse
|
27
|
Taslimifar M, Oparija L, Verrey F, Kurtcuoglu V, Olgac U, Makrides V. Quantifying the relative contributions of different solute carriers to aggregate substrate transport. Sci Rep 2017; 7:40628. [PMID: 28091567 PMCID: PMC5238446 DOI: 10.1038/srep40628] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 12/07/2016] [Indexed: 02/07/2023] Open
Abstract
Determining the contributions of different transporter species to overall cellular transport is fundamental for understanding the physiological regulation of solutes. We calculated the relative activities of Solute Carrier (SLC) transporters using the Michaelis-Menten equation and global fitting to estimate the normalized maximum transport rate for each transporter (Vmax). Data input were the normalized measured uptake of the essential neutral amino acid (AA) L-leucine (Leu) from concentration-dependence assays performed using Xenopus laevis oocytes. Our methodology was verified by calculating Leu and L-phenylalanine (Phe) data in the presence of competitive substrates and/or inhibitors. Among 9 potentially expressed endogenous X. laevis oocyte Leu transporter species, activities of only the uniporters SLC43A2/LAT4 (and/or SLC43A1/LAT3) and the sodium symporter SLC6A19/B0AT1 were required to account for total uptake. Furthermore, Leu and Phe uptake by heterologously expressed human SLC6A14/ATB0,+ and SLC43A2/LAT4 was accurately calculated. This versatile systems biology approach is useful for analyses where the kinetics of each active protein species can be represented by the Hill equation. Furthermore, its applicable even in the absence of protein expression data. It could potentially be applied, for example, to quantify drug transporter activities in target cells to improve specificity.
Collapse
Affiliation(s)
- Mehdi Taslimifar
- The Interface Group, Institute of Physiology, University of Zurich, Switzerland.,Epithelial Transport Group, Institute of Physiology, University of Zurich, Switzerland
| | - Lalita Oparija
- Epithelial Transport Group, Institute of Physiology, University of Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Francois Verrey
- Epithelial Transport Group, Institute of Physiology, University of Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland.,National Center of Competence in Research, Kidney CH, Switzerland
| | - Vartan Kurtcuoglu
- The Interface Group, Institute of Physiology, University of Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland.,National Center of Competence in Research, Kidney CH, Switzerland
| | - Ufuk Olgac
- The Interface Group, Institute of Physiology, University of Zurich, Switzerland.,National Center of Competence in Research, Kidney CH, Switzerland
| | - Victoria Makrides
- Epithelial Transport Group, Institute of Physiology, University of Zurich, Switzerland
| |
Collapse
|
28
|
Abstract
Glycine, besides exerting essential metabolic functions, is an important inhibitory neurotransmitter in caudal areas of the central nervous system and also a positive neuromodulator at excitatory glutamate-mediated synapses. Glial cells provide metabolic support to neurons and modulate synaptic activity. Six transporters belonging to three solute carrier families (SLC6, SLC38, and SLC7) are capable of transporting glycine across the glial plasma membrane. The unique glial glycine-selective transporter GlyT1 (SLC6) is the main regulator of synaptic glycine concentrations, assisted by the neuronal GlyT2. The five additional glycine transporters ATB0,+, SNAT1, SNAT2, SNAT5, and LAT2 display broad amino acid specificity and have differential contributions to glial glycine transport. Glial glycine transporters are divergent in sequence but share a similar architecture displaying the 5 + 5 inverted fold originally characterized in the leucine transporter LeuT. The availability of protein crystals solved at high resolution for prokaryotic and, more recently, eukaryotic homologues of this superfamily has advanced significantly our understanding of the mechanism of glycine transport.
Collapse
|
29
|
18F-FBPA as a tumor-specific probe of L-type amino acid transporter 1 (LAT1): a comparison study with 18F-FDG and 11C-Methionine PET. Eur J Nucl Med Mol Imaging 2016; 44:321-331. [PMID: 27550420 DOI: 10.1007/s00259-016-3487-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 08/05/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE The purpose of this study was to evaluate the usefulness of L-4-borono-2-18F-fluoro-phenylalanine (18F-FBPA) as a tumor-specific probe, in comparison to 18F-FDG and 11C-methionine (Met), focusing on its transport selectivity by L-type amino acid transporter 1 (LAT1), which is highly upregulated in cancers. METHODS Cellular analyses of FBPA were performed to evaluate the transportablity and Km value. PET studies were performed in rat xenograft models of C6 glioma (n = 12) and in rat models of turpentine oil-induced subcutaneous inflammation (n = 9). The kinetic parameters and uptake values on static PET images were compared using the one-tissue compartment model (K1, k2) and maximum standardized uptake value (SUVmax). RESULTS The cellular analyses showed that FBPA had a lower affinity to a normal cell-type transporter LAT2 and induced less efflux through LAT2 among FBPA, Met, and BPA, while the efflux through LAT1 induced by FBPA was similar among the three compounds. The Km value of 18F-FBPA for LAT1 (196.8 ± 11.4 μM) was dramatically lower than that for LAT2 (2813.8 ± 574.5 μM), suggesting the higher selectivity of 18F-FBPA for LAT1. K1 and k2 values were significantly smaller in 18F-FBPA PET (K1 = 0.04 ± 0.01 ml/ccm/min and k2 = 0.07 ± 0.01 /min) as compared to 11C-Met PET (0.22 ± 0.09 and 0.52 ± 0.10, respectively) in inflammatory lesions. Static PET analysis based on the SUVmax showed significantly higher accumulation of 18F-FDG in the tumor and inflammatory lesions (7.2 ± 2.1 and 4.6 ± 0.63, respectively) as compared to both 18F-FBPA (3.2 ± 0.40 and 1.9 ± 0.19) and 11C-Met (3.4 ± 0.43 and 1.6 ± 0.11). No significant difference was observed between 18F-FBPA and 11C-Met in the static PET images. CONCLUSION This study shows the utility of 18F-FBPA as a tumor-specific probe of LAT1 with low accumulation in the inflammatory lesions.
Collapse
|
30
|
Mechanisms involved in the transport of mercuric ions in target tissues. Arch Toxicol 2016; 91:63-81. [PMID: 27422290 DOI: 10.1007/s00204-016-1803-y] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 07/07/2016] [Indexed: 01/16/2023]
Abstract
Mercury exists in the environment in various forms, all of which pose a risk to human health. Despite guidelines regulating the industrial release of mercury into the environment, humans continue to be exposed regularly to various forms of this metal via inhalation or ingestion. Following exposure, mercuric ions are taken up by and accumulate in numerous organs, including brain, intestine, kidney, liver, and placenta. In order to understand the toxicological effects of exposure to mercury, a thorough understanding of the mechanisms that facilitate entry of mercuric ions into target cells must first be obtained. A number of mechanisms for the transport of mercuric ions into target cells and organs have been proposed in recent years. However, the ability of these mechanisms to transport mercuric ions and the regulatory features of these carriers have not been characterized completely. The purpose of this review is to summarize the current findings related to the mechanisms that may be involved in the transport of inorganic and organic forms of mercury in target tissues and organs. This review will describe mechanisms known to be involved in the transport of mercury and will also propose additional mechanisms that may potentially be involved in the transport of mercuric ions into target cells.
Collapse
|
31
|
Chiotellis A, Müller Herde A, Rössler SL, Brekalo A, Gedeonova E, Mu L, Keller C, Schibli R, Krämer SD, Ametamey SM. Synthesis, Radiolabeling, and Biological Evaluation of 5-Hydroxy-2-[18F]fluoroalkyl-tryptophan Analogues as Potential PET Radiotracers for Tumor Imaging. J Med Chem 2016; 59:5324-40. [DOI: 10.1021/acs.jmedchem.6b00057] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Aristeidis Chiotellis
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| | - Adrienne Müller Herde
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| | - Simon L. Rössler
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| | - Ante Brekalo
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| | - Erika Gedeonova
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| | - Linjing Mu
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Department of Nuclear
Medicine, University Hospital Zurich, Zurich 8091, Switzerland
| | - Claudia Keller
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| | - Roger Schibli
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| | - Stefanie D. Krämer
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| | - Simon M. Ametamey
- Center
for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 1-5/10, Zurich 8093, Switzerland
| |
Collapse
|
32
|
Barollo S, Bertazza L, Watutantrige-Fernando S, Censi S, Cavedon E, Galuppini F, Pennelli G, Fassina A, Citton M, Rubin B, Pezzani R, Benna C, Opocher G, Iacobone M, Mian C. Overexpression of L-Type Amino Acid Transporter 1 (LAT1) and 2 (LAT2): Novel Markers of Neuroendocrine Tumors. PLoS One 2016; 11:e0156044. [PMID: 27224648 PMCID: PMC4880303 DOI: 10.1371/journal.pone.0156044] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 05/09/2016] [Indexed: 12/22/2022] Open
Abstract
Background 6-18F-fluoro-L-3,4-dihydroxyphenylalanine (18F-FDOPA) PET is a useful tool in the clinical management of pheochromocytoma (PHEO) and medullary thyroid carcinoma (MTC). 18F-FDOPA is a large neutral amino acid biochemically resembling endogenous L-DOPA and taken up by the L-type amino acid transporters (LAT1 and LAT2). This study was conducted to examine the expression of the LAT system in PHEO and MTC. Methods Real-time PCR and Western blot analyses were used to assess LAT1 and LAT2 gene and protein expression in 32 PHEO, 38 MTC, 16 normal adrenal medulla and 15 normal thyroid tissue samples. Immunohistochemistry method was applied to identify the proteins’ subcellular localization. Results LAT1 and LAT2 were overexpressed in both PHEO and MTC by comparison with normal tissues. LAT1 presented a stronger induction than LAT2, and their greater expression was more evident in PHEO (15.1- and 4.1-fold increases, respectively) than in MTC (9.9- and 4.1-fold increases, respectively). Furthermore we found a good correlation between LAT1/2 and GLUT1 expression levels. A positive correlation was also found between urinary noradrenaline and adrenaline levels and LAT1 gene expression in PHEO. The increased expression of LAT1 is also confirmed at the protein level, in both PHEO and MTC, with a strong cytoplasmic localization. Conclusions The present study is the first to provide experimental evidence of the overexpression in some NET cancers (such as PHEO or MTC) of L-type amino acid transporters, and the LAT1 isoform in particular, giving the molecular basis to explain the increase of the DOPA uptake seen in such tumor cells.
Collapse
Affiliation(s)
- Susi Barollo
- Endocrinology Unit, Department of Medicine, University-Hospital of Padua, Padua, Italy
| | - Loris Bertazza
- Endocrinology Unit, Department of Medicine, University-Hospital of Padua, Padua, Italy
| | | | - Simona Censi
- Endocrinology Unit, Department of Medicine, University-Hospital of Padua, Padua, Italy
| | - Elisabetta Cavedon
- Endocrinology Unit, Department of Medicine, University-Hospital of Padua, Padua, Italy
| | - Francesca Galuppini
- II Pathology Unit, Department of Medicine, University-Hospital of Padua, Padua, Italy
| | - Gianmaria Pennelli
- II Pathology Unit, Department of Medicine, University-Hospital of Padua, Padua, Italy
| | - Ambrogio Fassina
- II Pathology Unit, Department of Medicine, University-Hospital of Padua, Padua, Italy
| | - Marilisa Citton
- Surgical Pathology Unit, Department of Surgery, Oncology and Gastroenterology Sciences, University-Hospital of Padua, Padua, Italy
| | - Beatrice Rubin
- Endocrinology Unit, Department of Medicine, University-Hospital of Padua, Padua, Italy
| | - Raffaele Pezzani
- Endocrinology Unit, Department of Medicine, University-Hospital of Padua, Padua, Italy
| | - Clara Benna
- Surgical Pathology Unit, Department of Surgery, Oncology and Gastroenterology Sciences, University-Hospital of Padua, Padua, Italy
| | - Giuseppe Opocher
- Familial Cancer Clinic and Oncoendocrinology, Veneto Institute of Oncology, Padova, Italy
| | - Maurizio Iacobone
- Surgical Pathology Unit, Department of Surgery, Oncology and Gastroenterology Sciences, University-Hospital of Padua, Padua, Italy
| | - Caterina Mian
- Endocrinology Unit, Department of Medicine, University-Hospital of Padua, Padua, Italy
- * E-mail:
| |
Collapse
|
33
|
Uetsuka S, Ogata G, Nagamori S, Isozumi N, Nin F, Yoshida T, Komune S, Kitahara T, Kikkawa Y, Inohara H, Kanai Y, Hibino H. Molecular architecture of the stria vascularis membrane transport system, which is essential for physiological functions of the mammalian cochlea. Eur J Neurosci 2015; 42:1984-2002. [PMID: 26060893 DOI: 10.1111/ejn.12973] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 06/02/2015] [Accepted: 06/02/2015] [Indexed: 11/29/2022]
Abstract
Stria vascularis of the mammalian cochlea transports K(+) to establish the electrochemical property in the endolymph crucial for hearing. This epithelial tissue also transports various small molecules. To clarify the profile of proteins participating in the transport system in the stria vascularis, membrane components purified from the stria of adult rats were analysed by liquid chromatography tandem mass spectrometry. Of the 3236 proteins detected in the analysis, 1807 were membrane proteins. Ingenuity Knowledge Base and literature data identified 513 proteins as being expressed on the 'plasma membrane', these included 25 ion channels and 79 transporters. Sixteen of the former and 62 of the latter had not yet been identified in the stria. Unexpectedly, many Cl(-) and Ca(2+) transport systems were found, suggesting that the dynamics of these ions play multiple roles. Several transporters for organic substances were also detected. Network analysis demonstrated that a few kinases, including protein kinase A, and Ca(2+) were key regulators for the strial transports. In the library of channels and transporters, 19 new candidates for uncloned deafness-related genes were identified. These resources provide a platform for understanding the molecular mechanisms underlying the epithelial transport essential for cochlear function and the pathophysiological processes involved in hearing disorders.
Collapse
Affiliation(s)
- Satoru Uetsuka
- Department of Molecular Physiology, Niigata University School of Medicine, 1-757 Asahimachi-dori, Niigata, 951-8510, Japan.,Center for Transdisciplinary Research, Niigata University, Niigata, Japan.,Department of Otorhinolaryngology - Head and Neck Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Genki Ogata
- Department of Molecular Physiology, Niigata University School of Medicine, 1-757 Asahimachi-dori, Niigata, 951-8510, Japan.,Center for Transdisciplinary Research, Niigata University, Niigata, Japan
| | - Shushi Nagamori
- Division of Bio-system Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Noriyoshi Isozumi
- Division of Bio-system Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Fumiaki Nin
- Department of Molecular Physiology, Niigata University School of Medicine, 1-757 Asahimachi-dori, Niigata, 951-8510, Japan.,Center for Transdisciplinary Research, Niigata University, Niigata, Japan
| | - Takamasa Yoshida
- Department of Molecular Physiology, Niigata University School of Medicine, 1-757 Asahimachi-dori, Niigata, 951-8510, Japan.,Center for Transdisciplinary Research, Niigata University, Niigata, Japan.,Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shizuo Komune
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tadashi Kitahara
- Department of Otorhinolaryngology - Head and Neck Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Otorhinolaryngology - Head and Neck Surgery, Nara Medical University, Nara, Japan
| | - Yoshiaki Kikkawa
- Mammalian Genetics Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hidenori Inohara
- Department of Otorhinolaryngology - Head and Neck Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshikatsu Kanai
- Division of Bio-system Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiroshi Hibino
- Department of Molecular Physiology, Niigata University School of Medicine, 1-757 Asahimachi-dori, Niigata, 951-8510, Japan.,Center for Transdisciplinary Research, Niigata University, Niigata, Japan
| |
Collapse
|
34
|
Asanuma M, Miyazaki I, Murakami S, Diaz-Corrales FJ, Ogawa N. Striatal astrocytes act as a reservoir for L-DOPA. PLoS One 2014; 9:e106362. [PMID: 25188235 PMCID: PMC4154692 DOI: 10.1371/journal.pone.0106362] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 07/29/2014] [Indexed: 11/18/2022] Open
Abstract
L-DOPA is therapeutically efficacious in patients with Parkinson’s disease (PD), although dopamine (DA) neurons are severely degenerated. Since cortical astrocytes express neutral amino acid transporter (LAT) and DA transporter (DAT), the uptake and metabolism of L-DOPA and DA in striatal astrocytes may influence their availability in the dopaminergic system of PD. To assess possible L-DOPA- and DA-uptake and metabolic properties of striatal astrocytes, we examined the expression of L-DOPA, DA and DAT in striatal astrocytes of hemi-parkinsonian model rats after repeated L-DOPA administration, and measured the contents of L-DOPA, DA and their metabolite in primary cultured striatal astrocytes after L-DOPA/DA treatment. Repeated injections of L-DOPA induced apparent L-DOPA- and DA-immunoreactivities and marked expression of DAT in reactive astrocytes on the lesioned side of the striatum in hemi-parkinsonian rats. Exposure to DA for 4h significantly increased the levels of DA and its metabolite DOPAC in cultured striatal astrocytes. L-DOPA was also markedly increased in cultured striatal astrocytes after 4-h L-DOPA exposure, but DA was not detected 4 or 8h after L-DOPA treatment, despite the expression of aromatic amino acid decarboxylase in astrocytes. Furthermore, the intracellular level of L-DOPA in cultured striatal astrocytes decreased rapidly after removal of extracellular L-DOPA. The results suggest that DA uptaken into striatal astrocytes is rapidly metabolized and that striatal astrocytes act as a reservoir of L-DOPA that govern the uptake or release of L-DOPA depending on extracellular L-DOPA concentration, but are less capable of converting L-DOPA to DA.
Collapse
Affiliation(s)
- Masato Asanuma
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- * E-mail:
| | - Ikuko Miyazaki
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinki Murakami
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Francisco J. Diaz-Corrales
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Norio Ogawa
- Department of Brain Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
35
|
Synthesis and in vitro evaluation of thiododecaborated α, α- cycloalkylamino acids for the treatment of malignant brain tumors by boron neutron capture therapy. Amino Acids 2014; 46:2715-20. [PMID: 25173737 DOI: 10.1007/s00726-014-1829-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 08/21/2014] [Indexed: 10/24/2022]
Abstract
Boron-neutron capture therapy (BNCT) is an attractive technique for cancer treatment. As such, α, α-cycloalkyl amino acids containing thiododecaborate ([B12H11](2-)-S-) units were designed and synthesized as novel boron delivery agents for BNCT. In the present study, new thiododecaborate α, α-cycloalkyl amino acids were synthesized, and biological evaluation of the boron compounds as boron carrier for BNCT was carried out.
Collapse
|
36
|
Nedergaard MK, Kristoffersen K, Michaelsen SR, Madsen J, Poulsen HS, Stockhausen MT, Lassen U, Kjaer A. The use of longitudinal 18F-FET MicroPET imaging to evaluate response to irinotecan in orthotopic human glioblastoma multiforme xenografts. PLoS One 2014; 9:e100009. [PMID: 24918622 PMCID: PMC4053391 DOI: 10.1371/journal.pone.0100009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 05/21/2014] [Indexed: 11/26/2022] Open
Abstract
Objectives Brain tumor imaging is challenging. Although 18F-FET PET is widely used in the clinic, the value of 18F-FET MicroPET to evaluate brain tumors in xenograft has not been assessed to date. The aim of this study therefore was to evaluate the performance of in vivo18F-FET MicroPET in detecting a treatment response in xenografts. In addition, the correlations between the 18F-FET tumor accumulation and the gene expression of Ki67 and the amino acid transporters LAT1 and LAT2 were investigated. Furthermore, Ki67, LAT1 and LAT2 gene expression in xenograft and archival patient tumors was compared. Methods Human GBM cells were injected orthotopically in nude mice and 18F-FET uptake was followed by weekly MicroPET/CT. When tumor take was observed, mice were treated with CPT-11 or saline weekly. After two weeks of treatment the brain tumors were isolated and quantitative polymerase chain reaction were performed on the xenograft tumors and in parallel on archival patient tumor specimens. Results The relative tumor-to-brain (T/B) ratio of SUVmax was significantly lower after one week (123±6%, n = 7 vs. 147±6%, n = 7; p = 0.018) and after two weeks (142±8%, n = 5 vs. 204±27%, n = 4; p = 0.047) in the CPT-11 group compared with the control group. Strong negative correlations between SUVmax T/B ratio and LAT1 (r = −0.62, p = 0.04) and LAT2 (r = −0.67, p = 0.02) were observed. In addition, a strong positive correlation between LAT1 and Ki67 was detected in xenografts. Furthermore, a 1.6 fold higher expression of LAT1 and a 23 fold higher expression of LAT2 were observed in patient specimens compared to xenografts. Conclusions 18F-FET MicroPET can be used to detect a treatment response to CPT-11 in GBM xenografts. The strong negative correlation between SUVmax T/B ratio and LAT1/LAT2 indicates an export transport function. We suggest that 18F-FET PET may be used for detection of early treatment response in patients.
Collapse
Affiliation(s)
- Mette K. Nedergaard
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| | - Karina Kristoffersen
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Copenhagen, Denmark
| | - Signe R. Michaelsen
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Copenhagen, Denmark
| | - Jacob Madsen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Hans S. Poulsen
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Copenhagen, Denmark
| | | | - Ulrik Lassen
- Phase 1 Unit, Department of Oncology, The Finsen Center, Rigshospitalet, Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
37
|
Yun DW, Lee SA, Park MG, Kim JS, Yu SK, Park MR, Kim SG, Oh JS, Kim CS, Kim HJ, Kim JS, Chun HS, Kanai Y, Endou H, Wempe MF, Kim DK. JPH203, an L-type amino acid transporter 1-selective compound, induces apoptosis of YD-38 human oral cancer cells. J Pharmacol Sci 2014; 124:208-17. [PMID: 24492461 DOI: 10.1254/jphs.13154fp] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Compared to most normal cells that express L-type amino acid transporter 2, L-type amino acid transporter 1 is highly expressed in cancer cells and presumed to support their elevated growth and proliferation. This study examined JPH203, a potent and selective L-type amino acid transporter 1 inhibitor, and its ability to suppress YD-38 human oral cancer cell growth. The YD-38 cells express L-type amino acid transporter 1 with its associating protein 4F2 heavy chain, but not L-type amino acid transporter 2. JPH203 and BCH, a non-selective L-type amino acid transporter inhibitor, completely inhibited l-leucine uptake in YD-38 cells. As expected, the intrinsic affinity of JPH203 to inhibit l-leucine uptake was far more efficient than BCH. Likewise, JPH203 and BCH inhibited YD-38 cell growth, with JPH203 being superior to BCH. JPH203 up-regulated the population of apoptotic YD-38 cells through the activation of apoptotic factors, including caspases and PARP. These results suggest that the inhibition of L-type amino acid transporter 1 activity via JPH203, which may act as a potential novel anti-oral-cancer agent, leads to apoptosis by inducing the intracellular depletion of the neutral amino acids essential for cancer cell growth in YD-38 human oral cancer cells.
Collapse
Affiliation(s)
- Dae-Woong Yun
- Oral Biology Research Institute, Chosun University School of Dentistry, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Andersson M, Karlsson O, Bergström U, Brittebo EB, Brandt I. Maternal transfer of the cyanobacterial neurotoxin β-N-methylamino-L-alanine (BMAA) via milk to suckling offspring. PLoS One 2013; 8:e78133. [PMID: 24194910 PMCID: PMC3806833 DOI: 10.1371/journal.pone.0078133] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Accepted: 09/17/2013] [Indexed: 12/14/2022] Open
Abstract
The cyanobacterial neurotoxin β-N-methylamino-L-alanine (BMAA) has been implicated in the etiology of neurodegenerative disease and proposed to be biomagnified in terrestrial and aquatic food chains. We have previously shown that the neonatal period in rats, which in humans corresponds to the last trimester of pregnancy and the first few years of age, is a particularly sensitive period for exposure to BMAA. The present study aimed to examine the secretion of 14C-labeled L- and D-BMAA into milk in lactating mice and the subsequent transfer of BMAA into the developing brain. The results suggest that secretion into milk is an important elimination pathway of BMAA in lactating mothers and an efficient exposure route predominantly for L-BMAA but also for D-BMAA in suckling mice. Following secretion of [14C]L-BMAA into milk, the levels of [14C]L-BMAA in the brains of the suckling neonatal mice significantly exceeded the levels in the maternal brains. In vitro studies using the mouse mammary epithelial HC11 cell line confirmed a more efficient influx and efflux of L-BMAA than of D-BMAA in cells, suggesting enantiomer-selective transport. Competition experiments with other amino acids and a low sodium dependency of the influx suggests that the amino acid transporters LAT1 and LAT2 are involved in the transport of L-BMAA into milk. Given the persistent neurodevelopmental toxicity following injection of L-BMAA to neonatal rodent pups, the current results highlight the need to determine whether BMAA is enriched mother's and cow's milk.
Collapse
Affiliation(s)
- Marie Andersson
- Department of Environmental Toxicology, Uppsala University, Uppsala, Sweden
| | - Oskar Karlsson
- Department of Environmental Toxicology, Uppsala University, Uppsala, Sweden
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Ulrika Bergström
- Department of Environmental Toxicology, Uppsala University, Uppsala, Sweden
| | - Eva B. Brittebo
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Ingvar Brandt
- Department of Environmental Toxicology, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
39
|
Synthesis and biological evaluation of O-[3-18F-fluoropropyl]-α-methyl tyrosine in mesothelioma-bearing rodents. BIOMED RESEARCH INTERNATIONAL 2013; 2013:460619. [PMID: 23936803 PMCID: PMC3722965 DOI: 10.1155/2013/460619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 06/06/2013] [Indexed: 12/27/2022]
Abstract
Radiolabeled tyrosine analogs enter cancer cells via upregulated amino acid transporter system and have been shown to be superior to 18F-fluoro-2-deoxy-D-glucose (18F-FDG) in differential diagnosis in cancers. In this study, we synthesized O-[3-19F-fluoropropyl]-α-methyl tyrosine (19F-FPAMT) and used manual and automated methods to synthesize O-[3-18F-fluoropropyl]-α-methyl tyrosine (18F-FPAMT) in three steps: nucleophilic substitution, deprotection of butoxycarbonyl, and deesterification. Manual and automated synthesis methods produced 18F-FPAMT with a radiochemical purity >96%. The decay-corrected yield of 18F-FPAMT by manual synthesis was 34% at end-of-synthesis (88 min). The decay-corrected yield of 18F-FPAMT by automated synthesis was 15% at end-of-synthesis (110 min). 18F-FDG and 18F-FPAMT were used for in vitro and in vivo studies to evaluate the feasibility of 18F-FPAMT for imaging rat mesothelioma (IL-45). In vitro studies comparing 18F-FPAMT with 18F-FDG revealed that 18F-FDG had higher uptake than that of 18F-FPAMT, and the uptake ratio of 18F-FPAMT reached the plateau after being incubated for 60 min. Biodistribution studies revealed that the accumulation of 18F-FPAMT in the heart, lungs, thyroid, spleen, and brain was significantly lower than that of 18F-FDG. There was poor bone uptake in 18F-FPAMT for up to 3 hrs suggesting its in vivo stability. The imaging studies showed good visualization of tumors with 18F-FPAMT. Together, these results suggest that 18F-FPAMT can be successfully synthesized and has great potential in mesothelioma imaging.
Collapse
|
40
|
Shearer KD, Fragoso YD, Clagett-Dame M, McCaffery PJ. Astrocytes as a regulated source of retinoic acid for the brain. Glia 2012; 60:1964-76. [PMID: 22930583 DOI: 10.1002/glia.22412] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 08/02/2012] [Indexed: 11/09/2022]
Abstract
Retinaldehyde dehydrogenases (RALDH) catalyze the synthesis of the regulatory factor retinoic acid (RA). Cultured astrocytes express several of the RALDH enzyme family, and it has been assumed that this can be extrapolated to astrocytes in vivo. However, this study finds that few astrocytes in the rodent brain express detectable RALDH enzymes, and only when these cells are grown in culture are these enzymes upregulated. Factors controlling the expression of the RALDHs in cultured astrocytes were explored to determine possible reasons for differences between in vitro versus in vivo expression. Retinoids were found to feedback to suppress several of the RALDHs, and physiological levels of retinoids may be one route by which astrocytic RALDHs are maintained at low levels. In the case of RALDH2, in vivo reduction of vitamin A levels in rats resulted in an increase in astrocyte RALDH2 expression in the hippocampus. Other factors though are likely to control RALDH expression. A shift in astrocytic RALDH subcellular localization is a potential mechanism for regulating RA signaling. Under conditions of vitamin A deficiency, RALDH2 protein moved from the cytoplasm to the nucleus where it may synthesize RA at the site of the nuclear RA receptors. Similarly, in conditions of oxidative stress RALDH1 and RALDH2 moved from the cytoplasm to a predominantly nuclear position. Thus, the RALDHs have been revealed to be dynamic in their expression in astrocytes where they may maintain retinoid homeostasis in the brain.
Collapse
Affiliation(s)
- Kirsty D Shearer
- Translational Neuroscience, Institute of Medical Sciences, School of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | | | | | | |
Collapse
|
41
|
L-DOPA Uptake in Astrocytic Endfeet Enwrapping Blood Vessels in Rat Brain. PARKINSONS DISEASE 2012; 2012:321406. [PMID: 22888467 PMCID: PMC3409556 DOI: 10.1155/2012/321406] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 06/13/2012] [Indexed: 11/23/2022]
Abstract
Astrocyte endfeet surround brain blood vessels and can play a role in the delivery of therapeutic drugs for Parkinson's disease. However, there is no previous evidence of the presence of LAT transporter for L-DOPA in brain astrocytes except in culture. Using systemic L-DOPA administration and a combination of patch clamp, histochemistry and confocal microscopy we found that L-DOPA is accumulated mainly in astrocyte cell bodies, astrocytic endfeet surrounding blood vessels, and pericytes. In brain slices: (1) astrocytes were exposed to ASP+, a fluorescent monoamine analog of MPP+; (2) ASP+ taken up by astrocytes was colocalized with L-DOPA fluorescence in (3) glial somata and in the endfeet attached to blood vessels; (4) these astrocytes have an electrogenic transporter current elicited by ASP+, but intriguingly not by L-DOPA, suggesting a different pathway for monoamines and L-DOPA via astrocytic membrane. (5) The pattern of monoamine oxidase (MAO type B) allocation in pericytes and astrocytic endfeet was similar to that of L-DOPA accumulation. We conclude that astrocytes control L-DOPA uptake and metabolism and, therefore, may play a key role in regulating brain dopamine level during dopamine-associated diseases. These data also suggest that different transporter mechanisms may exist for monoamines and L-DOPA.
Collapse
|
42
|
Abstract
For tumors of the central nervous system (CNS), the ability to accurately delineate the extent of tumor has implications for diagnosis, prognosis, and treatment. PET, mainly with (18)F-fluorodeoxyglucose (FDG), has become commonplace in the work-up of many extracranial tumors. However, the relative high background of FDG-PET activity of normal brain tissue has limited the applicability of this modality in CNS tumors to date. More recently, novel PET tracers for imaging of CNS tumors have been developed. This article outlines recent advances in PET as a complementary imaging modality with implications for diagnosis, prognosis, surgical and radiation treatment planning, and post-therapy surveillance in malignancies of the CNS. Pharmacokinetic properties of the radiotracers and the influence of blood-brain-barrier integrity are also incorporated into the discussion.
Collapse
Affiliation(s)
- Donald M Cannon
- Department of Human Oncology and Radiation Oncology, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, K4/B100, Madison, WI 53792, USA
| | | | | |
Collapse
|
43
|
Lim BC, Cho KY, Lim JS, Lee RS, Kim HS, Kim MK, Kim JH, Woo YJ, Kim JK, Kim DK, Kim HI, Lee KW, Lee MC. Increased expression of L-amino acid transporters in balloon cells of tuberous sclerosis. Childs Nerv Syst 2011; 27:63-70. [PMID: 20680301 DOI: 10.1007/s00381-010-1239-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 07/15/2010] [Indexed: 10/19/2022]
Abstract
OBJECTS Tuberous sclerosis complex (TSC) is a dysgenetic syndrome involved in multiple organs, and the pathognomonic cortical tuber act as an epileptic substrate. The amino acid transport system L (LAT) is a major nutrient transport system, and LAT1 is highly expressed in malignant tumors to support tumor cell growth. To study the life-long epilepsy from the cortical tuber, the expression of LAT1 in balloon cells and dysplastic neurons of the cortical tuber is investigated. MATERIALS AND METHODS LAT1 expression was investigated by LAT1 mRNA using reverse transcription-polymerase chain reaction and immunohistochemical staining with anti-human LAT1 antibody in nine patients with TSC and three control brains. CONCLUSION LAT1 mRNA was detectable only in fresh-frozen tissues of TSC, and it was upregulated in the cortical tuber lesion. While the LAT1 immunopositivity of control brains was limited in the capillary endothelial cells in the gray matter, increased LAT1 immunopositivity was noted in balloon cells of the cortical tubers in addition to the capillary endothelial cells as shown in control brains. Linear and strong immunopositivity along the cell membrane and cytoplasm of the balloon cells, and weakly granular immunopositivity in their cytoplasm were noted. Increased expression of LAT1 in the balloon cells is important for the active transport of large neutral amino acids into the balloon cells, and that the biologic process may play an important role in the active protein synthesis with metabolic maintenance of balloon cells in cortical tubers of patients with TSC.
Collapse
Affiliation(s)
- Byung-Chan Lim
- Department of Neurosurgery, Gwangju Christian Hospital, Gwangju, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Samluk Ł, Czeredys M, Nałęcz KA. Regulation of amino acid/carnitine transporter B 0,+ (ATB 0,+) in astrocytes by protein kinase C: independent effects on raft and non-raft transporter subpopulations. J Neurochem 2010; 115:1386-97. [PMID: 20977479 DOI: 10.1111/j.1471-4159.2010.07040.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Neutral and basic amino acid transporter B(0,+) belongs to a Na,Cl-dependent superfamily of proteins transporting neurotransmitters, amino acids and osmolytes, known to be regulated by protein kinase C (PKC). The present study demonstrates an increased phosphorylation of B(0,+) on serine moiety after treatment of rat astrocytes with phorbol 12-myristate 13-acetate, a process correlated with an augmented activity of l-leucine transport and an enhanced presence of the transporter at the cell surface. After solubilization with Triton X-100 and sucrose gradient centrifugation, B(0,+) was detected in non-raft as well as in detergent-resistant raft fractions under control conditions, while phorbol 12-myristate 13-acetate treatment resulted in a complete disappearance of the transporter from the raft fraction. B(0,+) was observed to interact with caveolin-1 and flotillin-1 (reggie-2) proteins, the markers of detergent-resistant microdomains of plasma membrane. As verified in immunocytochemistry and immunoprecipitation experiments, modification of PKC activity did not affect these interactions. It is proposed that PKC reveals different effects on raft and non-raft subpopulations of B(0,+). Phorbol ester treatment results in trafficking of the transporter from the intracellular pool to non-raft microdomains and increased activity, while B(0,+) present in raft microdomains undergoes either internalization or is transferred laterally to non-raft domains.
Collapse
Affiliation(s)
- Łukasz Samluk
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | | | | |
Collapse
|
45
|
Xia Luo, Coon JS, Su E, Kerry Pearson E, Ping Yin, Ishikawa H, Bulun SE. LAT1 Regulates Growth of Uterine Leiomyoma Smooth Muscle Cells. Reprod Sci 2010; 17:791-7. [DOI: 10.1177/1933719110372419] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Xia Luo
- Division of Reproductive Biology Research, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA, Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong, People's Republic of China
| | - John S. Coon
- Division of Reproductive Biology Research, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Emily Su
- Division of Reproductive Biology Research, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Elizabeth Kerry Pearson
- Division of Reproductive Biology Research, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ping Yin
- Division of Reproductive Biology Research, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hiroshi Ishikawa
- Division of Reproductive Biology Research, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Serdar E. Bulun
- Division of Reproductive Biology Research, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA,
| |
Collapse
|
46
|
Hirooka T, Fujiwara Y, Minami Y, Ishii A, Ishigooka M, Shinkai Y, Yamamoto C, Satoh M, Yasutake A, Eto K, Kaji T. Cell-density-dependent methylmercury susceptibility of cultured human brain microvascular pericytes. Toxicol In Vitro 2010; 24:835-41. [DOI: 10.1016/j.tiv.2009.12.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Revised: 10/08/2009] [Accepted: 12/09/2009] [Indexed: 10/20/2022]
|
47
|
Abstract
Despite the recognized limitations of (18)Fluorodeoxyglucose positron emission tomography (FDG-PET) in brain tumor imaging due to the high background of normal gray matter, this imaging modality provides critical information for the management of patients with cerebral neoplasms with regard to the following aspects: (1) providing a global picture of the tumor and thus guiding the appropriate site for stereotactic biopsy, and thereby enhancing its accuracy and reducing the number of biopsy samples; and (2) prediction of biologic behavior and aggressiveness of the tumor, thereby aiding in prognosis. Another area, which has been investigated extensively, includes differentiating recurrent tumor from treatment-related changes (eg, radiation necrosis and postsurgical changes). Furthermore, FDG-PET has demonstrated its usefulness in differentiating lymphoma from toxoplasmosis in patients with acquired immune deficiency syndrome with great accuracy, and is used as the investigation of choice in this setting. Image coregistration with magnetic resonance imaging and delayed FDG-PET imaging are 2 maneuvers that substantially improve the accuracy of interpretation, and hence should be routinely employed in clinical settings. In recent years an increasing number of brain tumor PET studies has used other tracers (like labeled methionine, tyrosine, thymidine, choline, fluoromisonidazole, EF5, and so forth), of which positron-labeled amino acid analogues, nucleotide analogues, and the hypoxia imaging tracers are of special interest. The major advantage of these radiotracers over FDG is the markedly lower background activity in normal brain tissue, which allows detection of small lesions and low-grade tumors. The promise of the amino acid PET tracers has been emphasized due to their higher sensitivity in imaging recurrent tumors (particularly the low-grade ones) and better accuracy for differentiating between recurrent tumors and treatment-related changes compared with FDG. The newer PET tracers have also shown great potential to image important aspects of tumor biology and thereby demonstrate ability to forecast prognosis. The value of hypoxia imaging tracers (such as fluoromisonidazole or more recently EF5) is substantial in radiotherapy planning and predicting treatment response. In addition, they may play an important role in the future in directing and monitoring targeted hypoxic therapy for tumors with hypoxia. Development of optimal image segmentation strategy with novel PET tracers and multimodality imaging is an approach that deserves mention in the era of intensity modulated radiotherapy, and which is likely to have important clinical and research applications in radiotherapy planning in patients with brain tumor.
Collapse
Affiliation(s)
- Sandip Basu
- Radiation Medicine Centre (BARC), Tata Memorial Hospital Annexe, Parel, Bombay 400012, India
| | | |
Collapse
|
48
|
Hazari PP, Shukla G, Goel V, Chuttani K, Kumar N, Sharma R, Mishra AK. Synthesis of Specific SPECT-Radiopharmaceutical for Tumor Imaging Based on Methionine: 99mTc-DTPA-bis(methionine). Bioconjug Chem 2010; 21:229-39. [DOI: 10.1021/bc900197n] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Puja Panwar Hazari
- Division Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig SK Mazumdar Road, Delhi-110054, India
| | - Gauri Shukla
- Division Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig SK Mazumdar Road, Delhi-110054, India
| | - Vijay Goel
- Division Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig SK Mazumdar Road, Delhi-110054, India
| | - Krishna Chuttani
- Division Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig SK Mazumdar Road, Delhi-110054, India
| | - Nitin Kumar
- Division Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig SK Mazumdar Road, Delhi-110054, India
| | - Rajnish Sharma
- Division Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig SK Mazumdar Road, Delhi-110054, India
| | - Anil Kumar Mishra
- Division Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig SK Mazumdar Road, Delhi-110054, India
| |
Collapse
|
49
|
Luo X, Yin P, Reierstad S, Ishikawa H, Lin Z, Pavone ME, Zhao H, Marsh EE, Bulun SE. Progesterone and mifepristone regulate L-type amino acid transporter 2 and 4F2 heavy chain expression in uterine leiomyoma cells. J Clin Endocrinol Metab 2009; 94:4533-9. [PMID: 19808856 PMCID: PMC2775649 DOI: 10.1210/jc.2009-1286] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CONTEXT Progesterone and its receptor (PR) play key roles in uterine leiomyoma growth. Previously, using chromatin immunoprecipitation-based cloning, we uncovered L-type amino acid transporter 2 (LAT2) as a novel PR target gene. LAT2 forms heterodimeric complexes with 4F2 heavy chain (4F2hc), a single transmembrane domain protein essential for LAT2 to exert its function in the plasma membrane. Until now, little is known about the roles of LAT2/4F2hc in the regulation of the growth of human uterine leiomyoma. OBJECTIVE The aim of the study is to investigate the regulation of LAT2 and 4F2hc by progesterone and the antiprogestin mifepristone and their functions in primary human uterine leiomyoma smooth muscle (LSM) cells and tissues from 39 premenopausal women. RESULTS In primary LSM cells, progesterone significantly induced LAT2 mRNA levels, and this was blocked by cotreatment with mifepristone. Progesterone did not alter 4F2hc mRNA levels, whereas mifepristone significantly induced 4F2hc mRNA expression. Small interfering RNA knockdown of LAT2 or 4F2hc markedly increased LSM cell proliferation. LAT2, PR-B, and PR-A levels were significantly higher in freshly isolated LSM cells vs. adjacent myometrial cells. In vivo, mRNA levels of LAT2 and PR but not 4F2hc were significantly higher in leiomyoma tissues compared with matched myometrial tissues. CONCLUSION We present evidence that progesterone and its antagonist mifepristone regulate the amino acid transporter system LAT2/4F2hc in leiomyoma tissues and cells. Our findings suggest that products of the LAT2/4F2hc genes may play important roles in leiomyoma cell proliferation. We speculate that critical ratios of LAT2 to 4F2hc regulate leiomyoma growth.
Collapse
Affiliation(s)
- Xia Luo
- Division of Reproductive Biology Research, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Heggland I, Kaur P, Syversen T. Uptake and efflux of methylmercury in vitro: Comparison of transport mechanisms in C6, B35 and RBE4 cells. Toxicol In Vitro 2009; 23:1020-7. [DOI: 10.1016/j.tiv.2009.06.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2009] [Revised: 06/10/2009] [Accepted: 06/15/2009] [Indexed: 10/20/2022]
|