1
|
Zhou Z, Zhu T, Zheng W, Zou Z, Shan Q, Chen Q, Wang G, Wang Y. LAT1 transporter as a target for breast cancer diagnosis and therapy. Eur J Med Chem 2025; 283:117064. [PMID: 39631100 DOI: 10.1016/j.ejmech.2024.117064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/09/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024]
Abstract
Breast cancer is the main cause of female malignant tumor death in China. Numerous cellular molecules are associated with the onset and progression of breast cancer. However, these molecules have proven ineffective for the diagnosis and treatment of the disease, indicating a need for the identification of new biomarkers. LAT1 (SLC7A5) plays a crucial role in mediating the uptake of amino acids into breast cancer cells, influencing proliferation, invasion, migration, drug resistance, and prognosis through the mTOR signaling pathway. Notably, LAT1 exhibits differential expression across various types of breast cancer, positioning it as a promising candidate for diagnostic and therapeutic applications. Recent advancements in LAT1-targeting strategies for breast cancer have been made, particularly with the rapid developments in small molecular inhibitors and nanotechnology. In this article, we review the structure and function of LAT1, its relationship with breast cancer, and LAT1-mediated diagnostic and treatment strategies. This article specifically focuses on the LAT1-targeting strategy in breast tumors, aiming to evaluate its potential role as a novel biomarker for diagnosis and treatment.
Collapse
Affiliation(s)
- Zheyang Zhou
- Institute of Traditional Chinese and Zhuang-Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China; Guang Xi Zhuang Yao Medicine Center of Engineering and Technology, Wuhe Rode, Nanning, 530200, China
| | - Tao Zhu
- Institute of Traditional Chinese and Zhuang-Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China; Guang Xi Zhuang Yao Medicine Center of Engineering and Technology, Wuhe Rode, Nanning, 530200, China
| | - Wenlong Zheng
- Guang Xi Zhuang Yao Medicine Center of Engineering and Technology, Wuhe Rode, Nanning, 530200, China; Pharmaceutical College, Guangxi University of Chinese Medicine, China
| | - Zhixiang Zou
- Institute of Traditional Chinese and Zhuang-Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China; Guang Xi Zhuang Yao Medicine Center of Engineering and Technology, Wuhe Rode, Nanning, 530200, China
| | - Qingfei Shan
- Institute of Traditional Chinese and Zhuang-Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China; Guang Xi Zhuang Yao Medicine Center of Engineering and Technology, Wuhe Rode, Nanning, 530200, China
| | - Qing Chen
- Institute of Traditional Chinese and Zhuang-Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China; Guang Xi Zhuang Yao Medicine Center of Engineering and Technology, Wuhe Rode, Nanning, 530200, China
| | - Gang Wang
- Institute of Traditional Chinese and Zhuang-Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China; Guang Xi Zhuang Yao Medicine Center of Engineering and Technology, Wuhe Rode, Nanning, 530200, China
| | - Yang Wang
- Institute of Traditional Chinese and Zhuang-Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China; Guang Xi Zhuang Yao Medicine Center of Engineering and Technology, Wuhe Rode, Nanning, 530200, China.
| |
Collapse
|
2
|
Li J, Yang D, Lyu W, Yuan Y, Han X, Yue W, Jiang J, Xiao Y, Fang Z, Lu X, Wang W, Huang W. A Bioinspired Photosensitizer Performs Tumor Thermoresistance Reversion to Optimize the Atraumatic Mild-Hyperthermia Photothermal Therapy for Breast Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405890. [PMID: 39045923 DOI: 10.1002/adma.202405890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/08/2024] [Indexed: 07/25/2024]
Abstract
Mild-hyperthermia photothermal therapy (mPTT) has therapeutic potential with minimized damage to normal tissues. However, the poorly vascularized tumor area severely hampers the penetration of photothermal agents (PTAs), resulting in their heterogeneous distribution and the subsequent heterogeneous local temperature during mPTT. The presence of regions below the therapeutic 42 °C threshold can lead to incomplete tumor ablation and potential recurrence. Additionally, tumor anti-apoptosis and cytoprotection pathways, particularly activated thermoresistance, can nullify mild hyperthermia-induced tumor damage. Therefore, a bioinspired photosensitizer decorated with leucine to form biomimetic nanoclusters (CP-PLeu nanoparticles (NPs)) aimed at achieving rapid and homogeneous accumulation in tumors, is introduced. Moreover, CP-PLeu exhibits photodynamic effects that reverse tumor thermoresistance and physiological repair mechanisms, thereby inhibiting tumor resistance to hyperthermia. With the addition of NIR-II laser irradiation, CP-PLeu optimizes the therapeutic efficacy of mPTT and contributes to a minimally invasive therapeutic process for breast cancer. This therapeutic strategy, utilizing a biomimetic photosensitizer for homogeneous distribution of therapeutic temperature and photoactivated reversal of tumor thermoresistance, successfully achieves efficient breast tumor inhibition through an atraumatic mPTT process.
Collapse
Affiliation(s)
- Jie Li
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM) and School of Flexible Electronics (Future Technologies), Nanjing Tech University (Nanjing Tech), Nanjing, 211800, China
| | - Die Yang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM) and School of Flexible Electronics (Future Technologies), Nanjing Tech University (Nanjing Tech), Nanjing, 211800, China
| | - Wentao Lyu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Yan Yuan
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM) and School of Flexible Electronics (Future Technologies), Nanjing Tech University (Nanjing Tech), Nanjing, 211800, China
| | - Xiao Han
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM) and School of Flexible Electronics (Future Technologies), Nanjing Tech University (Nanjing Tech), Nanjing, 211800, China
| | - Weiqing Yue
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM) and School of Flexible Electronics (Future Technologies), Nanjing Tech University (Nanjing Tech), Nanjing, 211800, China
| | - Jian Jiang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM) and School of Flexible Electronics (Future Technologies), Nanjing Tech University (Nanjing Tech), Nanjing, 211800, China
| | - Yingping Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Zhijie Fang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM) and School of Flexible Electronics (Future Technologies), Nanjing Tech University (Nanjing Tech), Nanjing, 211800, China
| | - Xiaomei Lu
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM) and School of Flexible Electronics (Future Technologies), Nanjing Tech University (Nanjing Tech), Nanjing, 211800, China
- Zhengzhou Institute of Biomedical Engineering and Technology, Zhengzhou, 450001, P. R. China
| | - Wen Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM) and School of Flexible Electronics (Future Technologies), Nanjing Tech University (Nanjing Tech), Nanjing, 211800, China
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, China
| |
Collapse
|
3
|
Gyimesi G, Hediger MA. Transporter-Mediated Drug Delivery. Molecules 2023; 28:molecules28031151. [PMID: 36770817 PMCID: PMC9919865 DOI: 10.3390/molecules28031151] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 01/27/2023] Open
Abstract
Transmembrane transport of small organic and inorganic molecules is one of the cornerstones of cellular metabolism. Among transmembrane transporters, solute carrier (SLC) proteins form the largest, albeit very diverse, superfamily with over 400 members. It was recognized early on that xenobiotics can directly interact with SLCs and that this interaction can fundamentally determine their efficacy, including bioavailability and intertissue distribution. Apart from the well-established prodrug strategy, the chemical ligation of transporter substrates to nanoparticles of various chemical compositions has recently been used as a means to enhance their targeting and absorption. In this review, we summarize efforts in drug design exploiting interactions with specific SLC transporters to optimize their therapeutic effects. Furthermore, we describe current and future challenges as well as new directions for the advanced development of therapeutics that target SLC transporters.
Collapse
|
4
|
Huttunen J, Kronenberger T, Montaser AB, Králová A, Terasaki T, Poso A, Huttunen KM. Sodium-Dependent Neutral Amino Acid Transporter 2 Can Serve as a Tertiary Carrier for l-Type Amino Acid Transporter 1-Utilizing Prodrugs. Mol Pharm 2023; 20:1331-1346. [PMID: 36688491 PMCID: PMC9906736 DOI: 10.1021/acs.molpharmaceut.2c00948] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Membrane transporters are the key determinants of the homeostasis of endogenous compounds in the cells and their exposure to drugs. However, the substrate specificities of distinct transporters can overlap. In the present study, the interactions of l-type amino acid transporter 1 (LAT1)-utilizing prodrugs with sodium-coupled neutral amino acid transporter 2 (SNAT2) were explored. The results showed that the cellular uptake of LAT1-utilizing prodrugs into a human breast cancer cell line, MCF-7 cells, was mediated via SNATs as the uptake was increased at higher pH (8.5), decreased in the absence of sodium, and inhibited in the presence of unselective SNAT-inhibitor, (α-(methylamino)isobutyric acid, MeAIB). Moreover, docking the compounds to a SNAT2 homology model (inward-open conformation) and further molecular dynamics simulations and the subsequent trajectory and principal component analyses confirmed the chemical features supporting the interactions of the studied compounds with SNAT2, which was found to be the main SNAT expressed in MCF-7 cells.
Collapse
Affiliation(s)
- Johanna Huttunen
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.
Box 1627, FI-70211 Kuopio, Finland
| | - Thales Kronenberger
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.
Box 1627, FI-70211 Kuopio, Finland,Department
of Internal Medicine VIII, University Hospital
Tübingen, Otfried-Müller-Strasse
14, DE 72076 Tübingen, Germany,Department
of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical
Sciences, Eberhard-Karls-Universität,
Tübingen, Auf
der Morgenstelle 8, 72076 Tübingen, Germany,Cluster
of Excellence iFIT (EXC 2180) “Image-Guided and Functionally
Instructed Tumor Therapies”, University
of Tübingen, 72076 Tübingen, Germany,Tübingen
Center for Academic Drug Discovery & Development (TüCAD2), 72076 Tübingen, Germany
| | - Ahmed B. Montaser
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.
Box 1627, FI-70211 Kuopio, Finland
| | - Adéla Králová
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.
Box 1627, FI-70211 Kuopio, Finland
| | - Tetsuya Terasaki
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.
Box 1627, FI-70211 Kuopio, Finland
| | - Antti Poso
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.
Box 1627, FI-70211 Kuopio, Finland,Department
of Internal Medicine VIII, University Hospital
Tübingen, Otfried-Müller-Strasse
14, DE 72076 Tübingen, Germany,Department
of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical
Sciences, Eberhard-Karls-Universität,
Tübingen, Auf
der Morgenstelle 8, 72076 Tübingen, Germany,Cluster
of Excellence iFIT (EXC 2180) “Image-Guided and Functionally
Instructed Tumor Therapies”, University
of Tübingen, 72076 Tübingen, Germany,Tübingen
Center for Academic Drug Discovery & Development (TüCAD2), 72076 Tübingen, Germany
| | - Kristiina M. Huttunen
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.
Box 1627, FI-70211 Kuopio, Finland,
| |
Collapse
|
5
|
Hutchinson K, Silva DB, Bohlke J, Clausen C, Thomas AA, Bonomi M, Schlessinger A. Describing inhibitor specificity for the amino acid transporter LAT1 from metainference simulations. Biophys J 2022; 121:4476-4491. [PMID: 36369754 PMCID: PMC9748366 DOI: 10.1016/j.bpj.2022.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 09/09/2022] [Accepted: 10/31/2022] [Indexed: 11/13/2022] Open
Abstract
The human L-type amino acid transporter 1 (LAT1; SLC7A5) is a membrane transporter of amino acids, thyroid hormones, and drugs such as the Parkinson's disease drug levodopa (L-Dopa). LAT1 is found in the blood-brain barrier, testis, bone marrow, and placenta, and its dysregulation has been associated with various neurological diseases, such as autism and epilepsy, as well as cancer. In this study, we combine metainference molecular dynamics simulations, molecular docking, and experimental testing, to characterize LAT1-inhibitor interactions. We first conducted a series of molecular docking experiments to identify the most relevant interactions between LAT1's substrate-binding site and ligands, including both inhibitors and substrates. We then performed metainference molecular dynamics simulations using cryoelectron microscopy structures in different conformations of LAT1 with the electron density map as a spatial restraint, to explore the inherent heterogeneity in the structures. We analyzed the LAT1 substrate-binding site to map important LAT1-ligand interactions as well as newly described druggable pockets. Finally, this analysis guided the discovery of previously unknown LAT1 ligands using virtual screening and cellular uptake experiments. Our results improve our understanding of LAT1-inhibitor recognition, providing a framework for rational design of future lead compounds targeting this key drug target.
Collapse
Affiliation(s)
- Keino Hutchinson
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Dina Buitrago Silva
- Department of Bioengineering and Therapeutic Sciences University of California, San Francisco, San Francisco, California
| | - Joshua Bohlke
- Department of Chemistry, University of Nebraska at Kearney, Kearney, Nebraska
| | - Chase Clausen
- Department of Chemistry, University of Nebraska at Kearney, Kearney, Nebraska
| | - Allen A Thomas
- Department of Chemistry, University of Nebraska at Kearney, Kearney, Nebraska
| | - Massimiliano Bonomi
- Department of Structural Biology and Chemistry, Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Paris, France.
| | - Avner Schlessinger
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
6
|
Hugele A, Löffler S, Molina BH, Guillon M, Montaser AB, Auriola S, Huttunen KM. Aminopeptidase B can bioconvert L-type amino acid transporter 1 (LAT1)-utilizing amide prodrugs in the brain. Front Pharmacol 2022; 13:1034964. [PMID: 36339537 PMCID: PMC9631218 DOI: 10.3389/fphar.2022.1034964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/05/2022] [Indexed: 11/29/2022] Open
Abstract
A prodrug approach is a powerful method to temporarily change the physicochemical and thus, pharmacokinetic properties of drugs. However, in site-selective targeted prodrug delivery, tissue or cell-specific bioconverting enzyme is needed to be utilized to release the active parent drug at a particular location. Unfortunately, ubiquitously expressed enzymes, such as phosphatases and carboxylesterases are well used in phosphate and ester prodrug applications, but less is known about enzymes selectively expressed, e.g., in the brain and enzymes that can hydrolyze more stable prodrug bonds, such as amides and carbamates. In the present study, L-type amino acid transporter 1 (LAT1)-utilizing amide prodrugs bioconverting enzyme was identified by gradually exploring the environment and possible determinants, such as pH and metal ions, that affect amide prodrug hydrolysis. Based on inducement by cobalt ions and slightly elevated pH (8.5) as well as localization in plasma, liver, and particularly in the brain, aminopeptidase B was proposed to be responsible for the bioconversion of the majority of the studied amino acid amide prodrugs. However, this enzyme hydrolyzed only those prodrugs that contained an aromatic promoiety (L-Phe), while leaving the aliphatic promoeities (L-Lys) and the smallest prodrug (with L-Phe promoiety) intact. Moreover, the parent drugs’ structure (flexibility and the number of aromatic rings) largely affected the bioconversion rate. It was also noticed in this study, that there were species differences in the bioconversion rate by aminopeptidase B (rodents > human), although the in vitro–in vivo correlation of the studied prodrugs was relatively accurate.
Collapse
|
7
|
Spicer JA, Huttunen KM, Jose J, Dimitrov I, Akhlaghi H, Sutton VR, Voskoboinik I, Trapani J. Small Molecule Inhibitors of Lymphocyte Perforin as Focused Immunosuppressants for Infection and Autoimmunity. J Med Chem 2022; 65:14305-14325. [PMID: 36263926 DOI: 10.1021/acs.jmedchem.2c01338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
New drugs that precisely target the immune mechanisms critical for cytotoxic T lymphocyte (CTL) and natural killer (NK) cell driven pathologies are desperately needed. In this perspective, we explore the cytolytic protein perforin as a target for therapeutic intervention. Perforin plays an indispensable role in CTL/NK killing and controls a range of immune pathologies, while being encoded by a single copy gene with no redundancy of function. An immunosuppressant targeting this protein would provide the first-ever therapy focused specifically on one of the principal cell death pathways contributing to allotransplant rejection and underpinning multiple autoimmune and postinfectious diseases. No drugs that selectively block perforin-dependent cell death are currently in clinical use, so this perspective will review published novel small molecule inhibitors, concluding with in vivo proof-of-concept experiments performed in mouse models of perforin-mediated immune pathologies that provide a potential pathway toward a clinically useful therapeutic agent.
Collapse
Affiliation(s)
- Julie A Spicer
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, A New Zealand Centre for Research Excellence, Auckland 1142, New Zealand
| | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Jiney Jose
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, A New Zealand Centre for Research Excellence, Auckland 1142, New Zealand
| | - Ivo Dimitrov
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, A New Zealand Centre for Research Excellence, Auckland 1142, New Zealand
| | - Hedieh Akhlaghi
- Cancer Immunology Program, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Vivien R Sutton
- Cancer Immunology Program, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Ilia Voskoboinik
- Cancer Immunology Program, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Joseph Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
8
|
Tampio J, Markowicz-Piasecka M, Montaser A, Rysä J, Kauppinen A, Huttunen KM. L-type Amino Acid Transporter 1 Utilizing Ferulic Acid Derivatives Show Increased Drug Delivery in the Mouse Pancreas Along with Decreased Lipid Peroxidation and Prostaglandin Production. Mol Pharm 2022; 19:3806-3819. [PMID: 36027044 PMCID: PMC9644403 DOI: 10.1021/acs.molpharmaceut.2c00328] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
![]()
Oxidative stress and pathological changes of Alzheimer’s
disease (AD) overlap with metabolic diseases, such as diabetes mellitus
(DM). Therefore, tackling oxidative stress with antioxidants is a
compelling drug target against multiple chronic diseases simultaneously.
Ferulic acid (FA), a natural antioxidant, has previously been studied
as a therapeutic agent against both AD and DM. However, FA suffers
from poor bioavailability and delivery. As a solution, we have previously
reported about L-type amino acid transporter 1 (LAT1)-utilizing derivatives
with increased brain delivery and efficacy. In the present study,
we evaluated the pharmacokinetics and antioxidative efficacy of the
two derivatives in peripheral mouse tissues. Furthermore, we quantified
the LAT1 expression in studied tissues with a targeted proteomics
method to verify the transporter expression in mouse tissues. Additionally,
the safety of the derivatives was assessed by exploring their effects
on hemostasis in human plasma, erythrocytes, and endothelial cells.
We found that both derivatives accumulated substantially in the pancreas,
with over a 100-times higher area under curve compared to the FA.
Supporting the pharmacokinetics, the LAT1 was highly expressed in
the mouse pancreas. Treating mice with the LAT1-utilizing derivative
of FA lowered malondialdehyde and prostaglandin E2 production
in the pancreas, highlighting its antioxidative efficacy. Additionally,
the LAT1-utilizing derivatives were found to be hemocompatible in
human plasma and endothelial cells. Since antioxidative derivative
1 was substantially delivered into the pancreas along the previously
studied brain, the derivative can be considered as a safe dual-targeting
drug candidate in both the pancreas and the brain.
Collapse
Affiliation(s)
- Janne Tampio
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211Kuopio, Finland
| | - Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151Lodz, Poland
| | - Ahmed Montaser
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211Kuopio, Finland
| | - Jaana Rysä
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211Kuopio, Finland
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211Kuopio, Finland
| | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211Kuopio, Finland
| |
Collapse
|
9
|
Targeting Transporters for Drug Delivery to the Brain: Can We Do Better? Pharm Res 2022; 39:1415-1455. [PMID: 35359241 PMCID: PMC9246765 DOI: 10.1007/s11095-022-03241-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/21/2022] [Indexed: 12/11/2022]
Abstract
Limited drug delivery to the brain is one of the major reasons for high failure rates of central nervous system (CNS) drug candidates. The blood–brain barrier (BBB) with its tight junctions, membrane transporters, receptors and metabolizing enzymes is a main player in drug delivery to the brain, restricting the entrance of the drugs and other xenobiotics. Current knowledge about the uptake transporters expressed at the BBB and brain parenchymal cells has been used for delivery of CNS drugs to the brain via targeting transporters. Although many transporter-utilizing (pro)drugs and nanocarriers have been developed to improve the uptake of drugs to the brain, their success rate of translation from preclinical development to humans is negligible. In the present review, we provide a systematic summary of the current progress in development of transporter-utilizing (pro)drugs and nanocarriers for delivery of drugs to the brain. In addition, we applied CNS pharmacokinetic concepts for evaluation of the limitations and gaps in investigation of the developed transporter-utilizing (pro)drugs and nanocarriers. Finally, we give recommendations for a rational development of transporter-utilizing drug delivery systems targeting the brain based on CNS pharmacokinetic principles.
Collapse
|
10
|
Huttunen KM, Terasaki T, Urtti A, Montaser AB, Uchida Y. Pharmacoproteomics of Brain Barrier Transporters and Substrate Design for the Brain Targeted Drug Delivery. Pharm Res 2022; 39:1363-1392. [PMID: 35257288 PMCID: PMC9246989 DOI: 10.1007/s11095-022-03193-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/08/2022] [Indexed: 12/12/2022]
Abstract
One of the major reasons why central nervous system (CNS)-drug development has been challenging in the past, is the barriers that prevent substances entering from the blood circulation into the brain. These barriers include the blood-brain barrier (BBB), blood-spinal cord barrier (BSCB), blood-cerebrospinal fluid barrier (BCSFB), and blood-arachnoid barrier (BAB), and they differ from each other in their transporter protein expression and function as well as among the species. The quantitative expression profiles of the transporters in the CNS-barriers have been recently revealed, and in this review, it is described how they affect the pharmacokinetics of compounds and how these expression differences can be taken into account in the prediction of brain drug disposition in humans, an approach called pharmacoproteomics. In recent years, also structural biology and computational resources have progressed remarkably, enabling a detailed understanding of the dynamic processes of transporters. Molecular dynamics simulations (MDS) are currently used commonly to reveal the conformational changes of the transporters and to find the interactions between the substrates and the protein during the binding, translocation in the transporter cavity, and release of the substrate on the other side of the membrane. The computational advancements have also aided in the rational design of transporter-utilizing compounds, including prodrugs that can be actively transported without losing potency towards the pharmacological target. In this review, the state-of-art of these approaches will be also discussed to give insights into the transporter-mediated drug delivery to the CNS.
Collapse
Affiliation(s)
- Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| | - Tetsuya Terasaki
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| | - Arto Urtti
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Ahmed B Montaser
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Yasuo Uchida
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| |
Collapse
|
11
|
Bhilare NV, Marulkar VS, Kumar D, Chatap VK, Patil KS, Shirote PJ. An insight into prodrug strategy for the treatment of Alzheimer’s disease. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02859-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
12
|
Huttunen J, Agami M, Tampio J, Montaser AB, Huttunen KM. Comparison of Experimental Strategies to Study l-Type Amino Acid Transporter 1 (LAT1) Utilization by Ligands. Molecules 2021; 27:molecules27010037. [PMID: 35011270 PMCID: PMC8746705 DOI: 10.3390/molecules27010037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 01/08/2023] Open
Abstract
l-Type amino acid transporter 1 (LAT1), expressed abundantly in the brain and placenta and overexpressed in several cancer cell types, has gained a lot of interest in drug research and development, as it can be utilized for brain-targeted drug delivery, as well as inhibiting the essential amino acid supply to cancer cells. The structure of LAT1 is today very well-known and the interactions of ligands at the binding site of LAT1 can be modeled and explained. However, less is known of LAT1′s life cycle within the cells. Moreover, the functionality of LAT1 can be measured by several different methods, which may vary between the laboratories and make the comparison of the results challenging. In the present study, the usefulness of indirect cis-inhibition methods and direct cellular uptake methods and their variations to interpret the interactions of LAT1-ligands were evaluated. Moreover, this study also highlights the importance of understanding the intracellular kinetics of LAT1-ligands, and how they can affect the regular function of LAT1 in critical tissues, such as the brain. Hence, it is discussed herein how the selected methodology influences the outcome and created knowledge of LAT1-utilizing compounds.
Collapse
|
13
|
Chemical Approaches for Studying the Biology and Pharmacology of Membrane Transporters: The Histidine/Large Amino Acid Transporter SLC7A5 as a Benchmark. Molecules 2021; 26:molecules26216562. [PMID: 34770970 PMCID: PMC8588388 DOI: 10.3390/molecules26216562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
The localization of membrane transporters at the forefront of natural barriers makes these proteins very interesting due to their involvement in the absorption and distribution of nutrients and xenobiotics, including drugs. Over the years, structure/function relationship studies have been performed employing several strategies, including chemical modification of exposed amino acid residues. These approaches are very meaningful when applied to membrane transporters, given that these proteins are characterized by both hydrophobic and hydrophilic domains with a different degree of accessibility to employed chemicals. Besides basic features, the chemical targeting approaches can disclose information useful for pharmacological applications as well. An eminent example of this picture is the histidine/large amino acid transporter SLC7A5, known as LAT1 (Large Amino Acid Transporter 1). This protein is crucial in cell life because it is responsible for mediating the absorption and distribution of essential amino acids in peculiar body districts, such as the blood brain barrier and placenta. Furthermore, LAT1 can recognize a large variety of molecules of pharmacological interest and is also considered a hot target for drugs due to its over-expression in virtually all human cancers. Therefore, it is not surprising that the chemical targeting approach, coupled with bioinformatics, site-directed mutagenesis and transport assays, proved fundamental in describing features of LAT1 such as the substrate binding site, regulatory domains and interactions with drugs that will be discussed in this review. The results on LAT1 can be considered to have general applicability to other transporters linked with human diseases.
Collapse
|
14
|
Tampio J, Markowicz-Piasecka M, Huttunen KM. Hemocompatible L-Type amino acid transporter 1 (LAT1)-Utilizing prodrugs of perforin inhibitors can accumulate into the pancreas and alleviate inflammation-induced apoptosis. Chem Biol Interact 2021; 345:109560. [PMID: 34153225 DOI: 10.1016/j.cbi.2021.109560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/20/2021] [Accepted: 06/15/2021] [Indexed: 11/26/2022]
Abstract
Cytolytic pore-forming protein, perforin, has been associated with autoimmune destruction of pancreatic β-cells in type 1 diabetes mellitus (T1DM) once released from CD8+ T cells. Curiously, perforinopathy has also been implicated in numerous brain diseases. Therefore, inhibitors of perforin have been in demand with targeted delivery in mind. l-Type amino acid transporter 1 (LAT1) is known to be expressed in both the above-mentioned target tissues, in the pancreas as well as in the brain. Thus, in the present study, the distribution of two LAT1-utilizing prodrugs of investigational perforin inhibitors into the pancreas was explored after intraperitoneal (i.p., 30 μmol/kg) bolus injection to mice. The effects of prodrug 1 were also studied in lipopolysaccharide (LPS)-induced in vitro (50 μg/mL) and in vivo (250 μg/kg x 3 days) apoptosis and pancreatitis models by determining the cellular apoptotic levels with human umbilical vein endothelial cells (HUVEC) and pancreatic caspase-3/-7 activity in mice. Furthermore, the biocompatibility of prodrug 1 was explored in human plasma and towards red blood cells. According to the results, both prodrugs were accumulated more effectively into the pancreas than their parent drugs (in addition to the brain that has been previously reported). Prodrug 1 (30 μmol/kg) also decreased the pancreatic caspase-3/-7 activity (52%) and with 2.5 μM concentration, the number of early and late apoptotic cells (32-53%). Since prodrug 1 was also found to be hemocompatible and not affecting human plasma hemostasis or inducing hemolysis of erythrocytes at the concentration <50 μM, it can be considered biocompatible in systemic circulation and ready to be studied in the future as a dual-acting drug candidate (in the pancreas and brain) in diseases like T1DM with neurodegenerative comorbidities.
Collapse
Affiliation(s)
- Janne Tampio
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, Ul. Muszyńskiego 1, 90-151, Lodz, Poland
| | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
15
|
Venteicher B, Merklin K, Ngo HX, Chien HC, Hutchinson K, Campbell J, Way H, Griffith J, Alvarado C, Chandra S, Hill E, Schlessinger A, Thomas AA. The Effects of Prodrug Size and a Carbonyl Linker on l-Type Amino Acid Transporter 1-Targeted Cellular and Brain Uptake. ChemMedChem 2020; 16:869-880. [PMID: 33230949 DOI: 10.1002/cmdc.202000824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/23/2020] [Indexed: 11/08/2022]
Abstract
The l-type amino acid transporter 1 (LAT1, SLC7A5) imports dietary amino acids and amino acid drugs (e. g., l-DOPA) into the brain, and plays a role in cancer metabolism. Though there have been numerous reports of LAT1-targeted amino acid-drug conjugates (prodrugs), identifying the structural determinants to enhance substrate activity has been challenging. In this work, we investigated the position and orientation of a carbonyl group in linking hydrophobic moieties including the anti-inflammatory drug ketoprofen to l-tyrosine and l-phenylalanine. We found that esters of meta-carboxyl l-phenylalanine had better LAT1 transport rates than the corresponding acylated l-tyrosine analogues. However, as the size of the hydrophobic moiety increased, we observed a decrease in LAT1 transport rate with a concomitant increase in potency of inhibition. Our results have important implications for designing amino acid prodrugs that target LAT1 at the blood-brain barrier or on cancer cells.
Collapse
Affiliation(s)
- Brooklynn Venteicher
- Department of Chemistry, University of Nebraska at Kearney, 2401 11th Ave, Bruner Hall of Science, Kearney, NE 68849, USA
| | - Kasey Merklin
- Department of Chemistry, University of Nebraska at Kearney, 2401 11th Ave, Bruner Hall of Science, Kearney, NE 68849, USA
| | - Huy X Ngo
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 1550 4th St, Rm RH581, San Francisco, CA 94143, USA
| | - Huan-Chieh Chien
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 1550 4th St, Rm RH581, San Francisco, CA 94143, USA
| | - Keino Hutchinson
- Department of Pharmacological Sciences, Icahn School of Medicine at Mt. Sinai, 1468 Madison Ave, Annenberg Building Floor 19, New York, NY 10029, USA
| | - Jerome Campbell
- Department of Chemistry, University of Nebraska at Kearney, 2401 11th Ave, Bruner Hall of Science, Kearney, NE 68849, USA
| | - Hannah Way
- Department of Chemistry, University of Nebraska at Kearney, 2401 11th Ave, Bruner Hall of Science, Kearney, NE 68849, USA
| | - Joseph Griffith
- Department of Chemistry, University of Nebraska at Kearney, 2401 11th Ave, Bruner Hall of Science, Kearney, NE 68849, USA
| | - Cesar Alvarado
- Department of Chemistry, University of Nebraska at Kearney, 2401 11th Ave, Bruner Hall of Science, Kearney, NE 68849, USA
| | - Surabhi Chandra
- Department of Biology, University of Nebraska at Kearney, 2401 11th Ave, Bruner Hall of Science, Kearney, NE 68849, USA
| | - Evan Hill
- Department of Psychology, University of Nebraska, at Kearney 2507 11th Ave, Copeland Hall, Kearney, NE, 68849, (USA)
| | - Avner Schlessinger
- Department of Pharmacological Sciences, Icahn School of Medicine at Mt. Sinai, 1468 Madison Ave, Annenberg Building Floor 19, New York, NY 10029, USA
| | - Allen A Thomas
- Department of Chemistry, University of Nebraska at Kearney, 2401 11th Ave, Bruner Hall of Science, Kearney, NE 68849, USA
| |
Collapse
|
16
|
Tampio J, Huttunen J, Montaser A, Huttunen KM. Targeting of Perforin Inhibitor into the Brain Parenchyma Via a Prodrug Approach Can Decrease Oxidative Stress and Neuroinflammation and Improve Cell Survival. Mol Neurobiol 2020; 57:4563-4577. [PMID: 32754897 PMCID: PMC7515946 DOI: 10.1007/s12035-020-02045-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/28/2020] [Indexed: 12/31/2022]
Abstract
The cytolytic protein perforin has a crucial role in infections and tumor surveillance. Recently, it has also been associated with many brain diseases, such as neurodegenerative diseases and stroke. Therefore, inhibitors of perforin have attracted interest as novel drug candidates. We have previously reported that converting a perforin inhibitor into an L-type amino acid transporter 1 (LAT1)-utilizing prodrug can improve the compound's brain drug delivery not only across the blood-brain barrier (BBB) but also into the brain parenchymal cells: neurons, astrocytes, and microglia. The present study evaluated whether the increased uptake into mouse primary cortical astrocytes and subsequently improvements in the cellular bioavailability of this brain-targeted perforin inhibitor prodrug could enhance its pharmacological effects, such as inhibition of production of caspase-3/-7, lipid peroxidation products and prostaglandin E2 (PGE2) in the lipopolysaccharide (LPS)-induced neuroinflammation mouse model. It was demonstrated that increased brain and cellular drug delivery could improve the ability of perforin inhibitors to elicit their pharmacological effects in the brain at nano- to picomolar levels. Furthermore, the prodrug displayed multifunctional properties since it also inhibited the activity of several key enzymes related to Alzheimer's disease (AD), such as the β-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1), acetylcholinesterase (AChE), and most probably also cyclooxygenases (COX) at micromolar concentrations. Therefore, this prodrug is a potential drug candidate for preventing Aβ-accumulation and ACh-depletion in addition to combatting neuroinflammation, oxidative stress, and neural apoptosis within the brain. Graphical abstract.
Collapse
Affiliation(s)
- Janne Tampio
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Johanna Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Ahmed Montaser
- 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
|
Colas C. Toward a Systematic Structural and Functional Annotation of Solute Carriers Transporters-Example of the SLC6 and SLC7 Families. Front Pharmacol 2020; 11:1229. [PMID: 32973497 PMCID: PMC7466448 DOI: 10.3389/fphar.2020.01229] [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: 06/04/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
SLC transporters are emerging key drug targets. One important step for drug development is the profound understanding of the structural determinants defining the substrate selectivity of each transporter. Recently, the improvement of computational power and experimental methods such as X-ray and cryo-EM crystallography permitted to conduct structure-based studies on specific transporters having important pharmacological impact. However, a lot remains to be discovered regarding their dynamics, transport modulation and ligand recognition. A detailed functional characterization of transporters would provide opportunities to develop new compounds targeting these key drug targets. Here, we are giving an overview of two major human LeuT-fold families, SLC6 and SLC7, with an emphasis on the most relevant members of each family for drug development. We gather the most recent understanding on the structural determinants of selectivity within and across the two families. We then use this information to discuss the benefits of a more generalized structural and functional annotation of the LeuT fold and the implications of such mapping for drug discovery.
Collapse
Affiliation(s)
- Claire Colas
- University of Vienna, Department of Pharmaceutical Chemistry, Vienna, Austria
| |
Collapse
|
18
|
Puris E, Gynther M, Auriola S, Huttunen KM. L-Type amino acid transporter 1 as a target for drug delivery. Pharm Res 2020; 37:88. [PMID: 32377929 PMCID: PMC7203094 DOI: 10.1007/s11095-020-02826-8] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/15/2020] [Indexed: 12/13/2022]
Abstract
Our growing understanding of membrane transporters and their substrate specificity has opened a new avenue in the field of targeted drug delivery. The L-type amino acid transporter 1 (LAT1) has been one of the most extensively investigated transporters for delivering drugs across biological barriers. The transporter is predominantly expressed in cerebral cortex, blood-brain barrier, blood-retina barrier, testis, placenta, bone marrow and several types of cancer. Its physiological function is to mediate Na+ and pH independent exchange of essential amino acids: leucine, phenylalanine, etc. Several drugs and prodrugs designed as LAT1 substrates have been developed to improve targeted delivery into the brain and cancer cells. Thus, the anti-parkinsonian drug, L-Dopa, the anti-cancer drug, melphalan and the anti-epileptic drug gabapentin, all used in clinical practice, utilize LAT1 to reach their target site. These examples provide supporting evidence for the utility of the LAT1-mediated targeted delivery of the (pro)drug. This review comprehensively summarizes recent advances in LAT1-mediated targeted drug delivery. In addition, the use of LAT1 is critically evaluated and limitations of the approach are discussed.
Collapse
Affiliation(s)
- Elena Puris
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, 69120, Heidelberg, Germany.
| | - Mikko Gynther
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Kristiina M Huttunen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| |
Collapse
|
19
|
Li S, Su W, Wu H, Yuan T, Yuan C, Liu J, Deng G, Gao X, Chen Z, Bao Y, Yuan F, Zhou S, Tan H, Li Y, Li X, Fan L, Zhu J, Chen AT, Liu F, Zhou Y, Li M, Zhai X, Zhou J. Targeted tumour theranostics in mice via carbon quantum dots structurally mimicking large amino acids. Nat Biomed Eng 2020; 4:704-716. [PMID: 32231314 PMCID: PMC7197249 DOI: 10.1038/s41551-020-0540-y] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 02/20/2020] [Indexed: 12/28/2022]
Abstract
Strategies for selectively imaging and delivering drugs to tumours typically leverage differentially upregulated surface molecules on cancer cells. Here, we show that intravenously injected carbon quantum dots, functionalized with multiple paired α-carboxyl and amino groups that bind to the large neutral amino acid transporter 1 (which is expressed in most tumours), selectively accumulate in human tumour xenografts in mice and in an orthotopic mouse model of human glioma. The functionalized quantum dots, which structurally mimic large amino acids and can be loaded with aromatic drugs through π–π stacking interactions, enabled—in the absence of detectable toxicity—near-infrared fluorescence and photoacoustic imaging of the tumours and a reduction in tumour burden after the targeted delivery of chemotherapeutics to the tumours. The versatility of functionalization and high tumour selectivity of the quantum dots make them broadly suitable for tumour-specific imaging and drug delivery. Intravenously injected functionalized carbon quantum dots that bind to the large neutral amino acid transporter 1 and that structurally mimic large amino acids selectively accumulate in human tumours in mice, facilitating targeted theranostics.
Collapse
Affiliation(s)
- Shuhua Li
- College of Chemistry, Key Laboratories of Theoretical and Computational Photochemistry, and Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, China
| | - Wen Su
- College of Chemistry, Key Laboratories of Theoretical and Computational Photochemistry, and Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, China
| | - Hao Wu
- College of Chemistry, Key Laboratories of Theoretical and Computational Photochemistry, and Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, China
| | - Ting Yuan
- College of Chemistry, Key Laboratories of Theoretical and Computational Photochemistry, and Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, China
| | - Chang Yuan
- College of Chemistry, Key Laboratories of Theoretical and Computational Photochemistry, and Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, China
| | - Jun Liu
- Department of Neurosurgery, Yale University, New Haven, CT, USA
| | - Gang Deng
- Department of Neurosurgery, Yale University, New Haven, CT, USA
| | - Xingchun Gao
- Department of Neurosurgery, Yale University, New Haven, CT, USA
| | - Zeming Chen
- Department of Neurosurgery, Yale University, New Haven, CT, USA
| | - Youmei Bao
- Department of Neurosurgery, Yale University, New Haven, CT, USA
| | - Fanglong Yuan
- College of Chemistry, Key Laboratories of Theoretical and Computational Photochemistry, and Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, China
| | - Shixin Zhou
- Department of Cell Biology and Stem Cell Research Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Hongwei Tan
- College of Chemistry, Key Laboratories of Theoretical and Computational Photochemistry, and Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, China
| | - Yunchao Li
- College of Chemistry, Key Laboratories of Theoretical and Computational Photochemistry, and Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, China
| | - Xiaohong Li
- College of Chemistry, Key Laboratories of Theoretical and Computational Photochemistry, and Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, China
| | - Louzhen Fan
- College of Chemistry, Key Laboratories of Theoretical and Computational Photochemistry, and Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, China.
| | - Jia Zhu
- College of Chemistry, Key Laboratories of Theoretical and Computational Photochemistry, and Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, China.
| | - Ann T Chen
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Fuyao Liu
- Department of Neurosurgery, Yale University, New Haven, CT, USA
| | - Yu Zhou
- Department of Neurosurgery, Yale University, New Haven, CT, USA
| | - Miao Li
- Department of Neurosurgery, Yale University, New Haven, CT, USA
| | - Xingchen Zhai
- Department of Neurosurgery, Yale University, New Haven, CT, USA
| | - Jiangbing Zhou
- Department of Neurosurgery, Yale University, New Haven, CT, USA. .,Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
20
|
Pocasap P, Weerapreeyakul N, Timonen J, Järvinen J, Leppänen J, Kärkkäinen J, Rautio J. Tyrosine-Chlorambucil Conjugates Facilitate Cellular Uptake through L-Type Amino Acid Transporter 1 (LAT1) in Human Breast Cancer Cell Line MCF-7. Int J Mol Sci 2020; 21:ijms21062132. [PMID: 32244913 PMCID: PMC7139360 DOI: 10.3390/ijms21062132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/19/2020] [Accepted: 03/19/2020] [Indexed: 12/27/2022] Open
Abstract
l-type amino acid transporter 1 (LAT1) is an amino acid transporter that is overexpressed in several types of cancer and, thus, it can be a potential target for chemotherapy. The objectives of this study were to (a) synthesize LAT1-targeted chlorambucil derivatives and (b) evaluate their LAT1-mediated cellular uptake as well as antiproliferative activity in vitro in the human breast cancer MCF-7 cell line. Chlorambucil was conjugated to l-tyrosine—an endogenous LAT1 substrate—via either ester or amide linkage (compounds 1 and 2, respectively). While chlorambucil itself did not bind to LAT1, its derivatives 1 and 2 bound to LAT1 with a similar affinity as with l-tyrosine and their respective cellular uptake was significantly higher than that of chlorambucil in MCF-7. The results of our cellular uptake study are indicative of antiproliferative activity, as a higher intracellular uptake of chlorambucil derivatives resulted in greater cytotoxicity than chlorambucil by itself. LAT1 thus contributes to intracellular uptake of chlorambucil derivatives and, therefore, increases antiproliferative activity. The understanding gained from our research can be used in the development of LAT1-targeted anticancer drugs and prodrugs for site-selective and enhanced chemotherapeutic activity.
Collapse
Affiliation(s)
- Piman Pocasap
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand;
- Human High Performance and Health Promotion Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Natthida Weerapreeyakul
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand;
- Human High Performance and Health Promotion Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
- Correspondence: (N.W.); (J.R.)
| | - Juri Timonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (J.T.); (J.J.); (J.L.); (J.K.)
| | - Juulia Järvinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (J.T.); (J.J.); (J.L.); (J.K.)
| | - Jukka Leppänen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (J.T.); (J.J.); (J.L.); (J.K.)
| | - Jussi Kärkkäinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (J.T.); (J.J.); (J.L.); (J.K.)
| | - Jarkko Rautio
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (J.T.); (J.J.); (J.L.); (J.K.)
- Correspondence: (N.W.); (J.R.)
| |
Collapse
|
21
|
Astrocyte-Targeted Transporter-Utilizing Derivatives of Ferulic Acid Can Have Multifunctional Effects Ameliorating Inflammation and Oxidative Stress in the Brain. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3528148. [PMID: 31814871 PMCID: PMC6877910 DOI: 10.1155/2019/3528148] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 09/19/2019] [Accepted: 10/03/2019] [Indexed: 12/27/2022]
Abstract
Ferulic acid (FA) is a natural phenolic antioxidant, which can exert also several other beneficial effects to combat neuroinflammation and neurodegenerative diseases, such as Alzheimer's disease. One of these properties is the inhibition of several enzymes and factors, such as β-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1), cyclooxygenases (COXs), lipoxygenases (LOXs), mammalian (or mechanistic) target for rapamycin (mTOR), and transcription factor NF-κB. We have previously synthesized three L-type amino acid transporter 1- (LAT1-) utilizing FA-derivatives with the aim to develop brain-targeted prodrugs of FA. In the present study, the cellular uptake and bioavailability of these FA-derivatives were evaluated in mouse primary astrocytic cell cultures together with their inhibitory effects towards BACE1, COX/LOX, mTOR, NF-κB, acetylcholinesterase (AChE), and oxidative stress. According to the results, all three FA-derivatives were taken up 200–600 times more effectively at 10 μM concentration into the astrocytes than FA, with one derivative having a high intracellular bioavailability (Kp,uu), particularly at low concentrations. Moreover, all of the derivatives were able to inhibit BACE1, COX/LOX, AChE, and oxidative stress measured as decreased cellular lipid peroxidation. Furthermore, one of the derivatives modified the total mTOR amount. Therefore, these derivatives have the potential to act as multifunctional compounds preventing β-amyloid accumulation as well as combating inflammation and reducing oxidative stress in the brain. Thus, this study shows that converting a parent drug into a transporter-utilizing derivative not only may increase its brain and cellular uptake, and bioavailability but can also broaden the spectrum of pharmacological effects elicited by the derivative.
Collapse
|
22
|
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: 1.8] [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]
|
23
|
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: 8.3] [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
|
24
|
Markowicz-Piasecka M, Huttunen J, Sikora J, Huttunen KM. Sulfenamide derivatives can improve transporter-mediated cellular uptake of metformin and induce cytotoxicity in human breast adenocarcinoma cell lines. Bioorg Chem 2019; 87:321-334. [PMID: 30913467 DOI: 10.1016/j.bioorg.2019.03.036] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/05/2019] [Accepted: 03/14/2019] [Indexed: 12/14/2022]
Abstract
Metformin, the most frequently administered oral anti-diabetic drug, is a substrate for organic cation transporters (OCTs). This determines not only its pharmacokinetic properties but also its biochemical effects in humans, including its recently-discovered antiproliferative properties. The aim of the study was to verify the hypothesis whether chemical modification of its biguanide backbone may increase the cellular uptake and antiproliferative efficacy of metformin. The study examines five sulfenamide derivatives of metformin with differing lengths of alkyl chains. It determines their cellular uptake and the role of OCTs in their transport in human breast adenocarcinoma cells (epithelial-like MCF-7, and MDA-MB-231). It also evaluates whether increased cellular uptake of metformin derivatives is associated with their cytotoxic properties. Sulfenamide derivatives were characterized by a greater ability to bind to OCTs than metformin. Compound 2 with n-octyl alkyl chain was found to possess the greatest affinity towards OCTs, as measured by determination of [14C]choline uptake inhibition (IC50 = 236.1 ± 1.28 μmol/L, and 217.4 ± 1.33 μmol/L, for MCF-7 and MDA-MB-231 respectively). Sulfenamides were also found to exhibit better cellular uptake in comparison with the parent drug, metformin. For instance, the uptake of cyclohexyl derivative 1 was 1.28 ± 0.19 nmol/min/mg of proteins and thus was 12-fold higher than the metformin in MCF-7 cells. Furthermore, higher uptake was associated with the greatest antiproliferative properties expressed as the lowest IC50 value i.e. inhibiting the growth of 50% of the cells (IC50 = 0.72 ± 1.31 μmol/L). Collectively, chemical modification of metformin into sulfenamides with different alkyl substituents obtains better substrates for OCTs, and subsequently higher cellular uptake in MCF-7 and MDA-MB-231 cells. Additionally, the length of alkyl chain introduced to the sulfenamides was found to influence selectivity and transport efficiency via OCT1 compared to other possible transporters, as well as potential intracellular activity and cytotoxicity.
Collapse
Affiliation(s)
- Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151 Lodz, Poland.
| | - Johanna Huttunen
- School Of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211 Kuopio, Finland
| | - Joanna Sikora
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151 Lodz, Poland
| | - Kristiina M Huttunen
- School Of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211 Kuopio, Finland.
| |
Collapse
|
25
|
Huttunen K. Identification of human, rat and mouse hydrolyzing enzymes bioconverting amino acid ester prodrug of ketoprofen. Bioorg Chem 2018; 81:494-503. [DOI: 10.1016/j.bioorg.2018.09.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 09/10/2018] [Indexed: 12/25/2022]
|
26
|
Secondary carbamate linker can facilitate the sustained release of dopamine from brain-targeted prodrug. Bioorg Med Chem Lett 2018; 28:2856-2860. [DOI: 10.1016/j.bmcl.2018.07.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/16/2018] [Accepted: 07/18/2018] [Indexed: 11/21/2022]
|
27
|
Huttunen J, Gynther M, Huttunen KM. Targeted efflux transporter inhibitors - A solution to improve poor cellular accumulation of anti-cancer agents. Int J Pharm 2018; 550:278-289. [PMID: 30149128 DOI: 10.1016/j.ijpharm.2018.08.047] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/20/2018] [Accepted: 08/24/2018] [Indexed: 12/27/2022]
Abstract
Efflux transporters function as vacuum cleaners of xenobiotics and therefore they hinder drugs to reach their targets at effective enough concentrations. Efflux pump inhibitors can be used to improve the cell accumulation of drugs, however all the current inhibitors lack selectivity towards cancer cells. l-Type amino acid transporter 1 (LAT1), which is expressed in many types of cancer cells can be utilized to target inhibitors of efflux transporters to these cells by converting the inhibitors into LAT1-utilizing prodrugs. In this study, we prepared 5 LAT1-utilizing prodrugs of an efflux pump inhibitor, probenecid (PRB). All novel compounds were transported into human breast cancer cells (MCF-7) mainly via LAT1. The compounds also interacted with either multiresistant proteins (MRPs), P-glycoprotein (P-gp) or breast cancer resistant protein (BCRP) and increased significantly (3-4-fold) the cellular accumulation of anti-cancer agent vinblastine (VBL). Consequently, this improved the anti-proliferative efficacy of VBL by decreasing the cell growth after 72 h from 100% (VBL treatment alone) to 48-75% (combination treatment). However, the same phenomenon was not seen with other chemotherapeutic, methotrexate (MTX). Therefore, the chemotherapeutics need to be selected carefully based on their uptake mechanism to the combinations with LAT1-utilizing prodrugs of efflux pump inhibitors to defeat effectively the multidrug resistance (MDR) of chemotherapy.
Collapse
Affiliation(s)
- Johanna Huttunen
- 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
| | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
28
|
Chien HC, Colas C, Finke K, Springer S, Stoner L, Zur AA, Venteicher B, Campbell J, Hall C, Flint A, Augustyn E, Hernandez C, Heeren N, Hansen L, Anthony A, Bauer J, Fotiadis D, Schlessinger A, Giacomini KM, Thomas AA. Reevaluating the Substrate Specificity of the L-Type Amino Acid Transporter (LAT1). J Med Chem 2018; 61:7358-7373. [PMID: 30048132 DOI: 10.1021/acs.jmedchem.8b01007] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The L-type amino acid transporter 1 (LAT1, SLC7A5) transports essential amino acids across the blood-brain barrier (BBB) and into cancer cells. To utilize LAT1 for drug delivery, potent amino acid promoieties are desired, as prodrugs must compete with millimolar concentrations of endogenous amino acids. To better understand ligand-transporter interactions that could improve potency, we developed structural LAT1 models to guide the design of substituted analogues of phenylalanine and histidine. Furthermore, we evaluated the structure-activity relationship (SAR) for both enantiomers of naturally occurring LAT1 substrates. Analogues were tested in cis-inhibition and trans-stimulation cell assays to determine potency and uptake rate. Surprisingly, LAT1 can transport amino acid-like substrates with wide-ranging polarities including those containing ionizable substituents. Additionally, the rate of LAT1 transport was generally nonstereoselective even though enantiomers likely exhibit different binding modes. Our findings have broad implications to the development of new treatments for brain disorders and cancer.
Collapse
Affiliation(s)
- Huan-Chieh Chien
- Department of Bioengineering and Therapeutic Sciences , University of California, San Francisco , San Francisco , California 94158 , United States
| | - Claire Colas
- Department of Pharmacological Sciences , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Karissa Finke
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Seth Springer
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Laura Stoner
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Arik A Zur
- Department of Bioengineering and Therapeutic Sciences , University of California, San Francisco , San Francisco , California 94158 , United States
| | - Brooklynn Venteicher
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Jerome Campbell
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Colton Hall
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Andrew Flint
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Evan Augustyn
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Christopher Hernandez
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Nathan Heeren
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Logan Hansen
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Abby Anthony
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Justine Bauer
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| | - Dimitrios Fotiadis
- Institute of Biochemistry and Molecular Medicine, and Swiss National Centre of Competence in Research (NCCR) TransCure , University of Bern , 3012 Bern , Switzerland
| | - Avner Schlessinger
- Department of Pharmacological Sciences , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences , University of California, San Francisco , San Francisco , California 94158 , United States
| | - Allen A Thomas
- Department of Chemistry , University of Nebraska at Kearney , Kearney , Nebraska 68849 , United States
| |
Collapse
|
29
|
Scalise M, Galluccio M, Console L, Pochini L, Indiveri C. The Human SLC7A5 (LAT1): The Intriguing Histidine/Large Neutral Amino Acid Transporter and Its Relevance to Human Health. Front Chem 2018; 6:243. [PMID: 29988369 PMCID: PMC6023973 DOI: 10.3389/fchem.2018.00243] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/06/2018] [Indexed: 12/22/2022] Open
Abstract
SLC7A5, known as LAT1, belongs to the APC superfamily and forms a heterodimeric amino acid transporter interacting with the glycoprotein CD98 (SLC3A2) through a conserved disulfide. The complex is responsible for uptake of essential amino acids in crucial body districts such as placenta and blood brain barrier. LAT1/CD98 heterodimer has been studied over the years to unravel the transport mechanism and the role of each subunit. Studies conducted in intact cells demonstrated that LAT1/CD98 mediates a Na+ and pH independent antiport of amino acids. Some novel insights into the function of LAT1 derived from studies conducted in proteoliposomes reconstituted with the recombinant human LAT1. Using this experimental tool, it has been demonstrated that the preferred substrate is histidine and that CD98 is not required for transport being, plausibly, involved in routing LAT1 to the plasma membrane. Since a 3D structure of LAT1 is not available, homology models have been built on the basis of the AdiC transporter from E.coli. Crucial residues for substrate recognition and gating have been identified using a combined approach of bioinformatics and site-directed mutagenesis coupled to functional assays. Over the years, the interest around LAT1 increased because this transporter is involved in important human diseases such as neurological disorders and cancer. Therefore, LAT1 became an important pharmacological target together with other nutrient membrane transporters. Moving from knowledge on structure/function relationships, two cysteine residues, lying on the substrate binding site, have been exploited for designing thiol reacting covalent inhibitors. Some lead compounds have been characterized whose efficacy has been tested in a cancer cell line.
Collapse
Affiliation(s)
- Mariafrancesca Scalise
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Rende, Italy
| | - Michele Galluccio
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Rende, Italy
| | - Lara Console
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Rende, Italy
| | - Lorena Pochini
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Rende, Italy
| | - Cesare Indiveri
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Rende, Italy.,CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, Bari, Italy
| |
Collapse
|
30
|
Kärkkäinen J, Gynther M, Kokkola T, Petsalo A, Auriola S, Lahtela-Kakkonen M, Laine K, Rautio J, Huttunen KM. Structural properties for selective and efficient l-type amino acid transporter 1 (LAT1) mediated cellular uptake. Int J Pharm 2018; 544:91-99. [DOI: 10.1016/j.ijpharm.2018.04.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/13/2018] [Accepted: 04/13/2018] [Indexed: 12/24/2022]
|
31
|
Puris E, Gynther M, Huttunen J, Petsalo A, Huttunen KM. L-type amino acid transporter 1 utilizing prodrugs: How to achieve effective brain delivery and low systemic exposure of drugs. J Control Release 2017; 261:93-104. [DOI: 10.1016/j.jconrel.2017.06.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 06/18/2017] [Accepted: 06/24/2017] [Indexed: 10/19/2022]
|
32
|
Zhao LY, Zhang WM. Recent progress in drug delivery of pluronic P123: pharmaceutical perspectives. J Drug Target 2017; 25:471-484. [PMID: 28135859 DOI: 10.1080/1061186x.2017.1289538] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
This review focuses on recent investigations that used Pluronic P123 (P123) as pharmaceutical ingredients in vesicle, micelle, mixed micelle, in situ gel, tablet and emulsion. The main results from these studies show that P123 can significantly increase the stability of incorporated hydrophobic drugs with enhanced in vitro cytotoxicity and cellular uptake of anticancer drugs. Moreover, modified forms of P123 with RGD, folate or other targeted marker have shown its therapeutic potentials in various types of tumors and cancers. Furthermore, modified forms of P123 alone and/or mixed with other copolymers have less toxic effects and more tumor-specific delivery of anticancer drugs. They are promising materials as a nanoplatform for the drug delivery. Finally, the future perspectives of the field are briefly discussed.
Collapse
Affiliation(s)
- Li-Yan Zhao
- a Department of Pharmacy , Hebei North University , Zhangjiakou , PR China
| | - Wan-Ming Zhang
- a Department of Pharmacy , Hebei North University , Zhangjiakou , PR China
| |
Collapse
|
33
|
Kellici TF, Chatziathanasiadou MV, Lee MS, Sayyad N, Geromichalou EG, Vrettos EI, Tsiailanis AD, Chi SW, Geromichalos GD, Mavromoustakos T, Tzakos AG. Rational design and structure–activity relationship studies of quercetin–amino acid hybrids targeting the anti-apoptotic protein Bcl-xL. Org Biomol Chem 2017; 15:7956-7976. [DOI: 10.1039/c7ob02045g] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Anti-apoptotic proteins, like the Bcl-2 family proteins, present an important therapeutic cancer drug target.
Collapse
Affiliation(s)
- Tahsin F. Kellici
- Department of Chemistry
- University of Ioannina
- Ioannina 45110
- Greece
- Department of Chemistry
| | | | - Min-Sung Lee
- Disease Target Structure Research Center
- KRIBB
- Daejeon 34141
- Republic of Korea
| | - Nisar Sayyad
- Department of Chemistry
- University of Ioannina
- Ioannina 45110
- Greece
| | - Elena G. Geromichalou
- Laboratory of Pharmacology
- Medical School National and Kapodistrian University of Athens
- Athens 11527
- Greece
| | | | | | - Seung-Wook Chi
- Disease Target Structure Research Center
- KRIBB
- Daejeon 34141
- Republic of Korea
| | - George D. Geromichalos
- Laboratory of Inorganic Chemistry
- Faculty of Chemistry
- Aristotle University of Thessaloniki
- GR-54124 Thessaloniki
- Greece
| | | | | |
Collapse
|
34
|
Gynther M, Pickering DS, Spicer JA, Denny WA, Huttunen KM. Systemic and Brain Pharmacokinetics of Perforin Inhibitor Prodrugs. Mol Pharm 2016; 13:2484-91. [DOI: 10.1021/acs.molpharmaceut.6b00217] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Mikko Gynther
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.
Box 1627, FI-70211 Kuopio, Finland
| | - Darryl S. Pickering
- Department of Drug Design and Pharmacology, Faculty of Health & Medical Sciences, University of Copenhagen, Jagtvej 160, 1165 Copenhagen, Denmark
| | - Julie A. Spicer
- Auckland
Cancer Society Research Centre, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - William A. Denny
- Auckland
Cancer Society Research Centre, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Kristiina M. Huttunen
- School
of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O.
Box 1627, FI-70211 Kuopio, Finland
| |
Collapse
|
35
|
Estudante M, Soveral G, Morais JG, Benet LZ. Insights into solute carriers: physiological functions and implications in disease and pharmacokinetics. MEDCHEMCOMM 2016. [DOI: 10.1039/c6md00188b] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SLCs transport many endogenous and exogenous compounds including drugs; SLCs dysfunction has implications in pharmacokinetics, drug toxicity or lack of efficacy.
Collapse
Affiliation(s)
- Margarida Estudante
- Department of Pharmacological Sciences
- Faculty of Pharmacy
- Universidade de Lisboa
- Portugal
- Research Institute for Medicines (iMed.ULisboa)
| | - Graça Soveral
- Research Institute for Medicines (iMed.ULisboa)
- Faculty of Pharmacy
- Universidade de Lisboa
- Portugal
| | - José G. Morais
- Department of Pharmacological Sciences
- Faculty of Pharmacy
- Universidade de Lisboa
- Portugal
- Research Institute for Medicines (iMed.ULisboa)
| | - Leslie Z. Benet
- Department of Bioengineering and Therapeutic Sciences
- University of California
- San Francisco
- USA
| |
Collapse
|