1
|
Tamas C, Tamas F, Kovecsi A, Cehan A, Balasa A. Metabolic Contrasts: Fatty Acid Oxidation and Ketone Bodies in Healthy Brains vs. Glioblastoma Multiforme. Int J Mol Sci 2024; 25:5482. [PMID: 38791520 PMCID: PMC11122426 DOI: 10.3390/ijms25105482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/09/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
The metabolism of glucose and lipids plays a crucial role in the normal homeostasis of the body. Although glucose is the main energy substrate, in its absence, lipid metabolism becomes the primary source of energy. The main means of fatty acid oxidation (FAO) takes place in the mitochondrial matrix through β-oxidation. Glioblastoma (GBM) is the most common form of primary malignant brain tumor (45.6%), with an incidence of 3.1 per 100,000. The metabolic changes found in GBM cells and in the surrounding microenvironment are associated with proliferation, migration, and resistance to treatment. Tumor cells show a remodeling of metabolism with the use of glycolysis at the expense of oxidative phosphorylation (OXPHOS), known as the Warburg effect. Specialized fatty acids (FAs) transporters such as FAT, FABP, or FATP from the tumor microenvironment are overexpressed in GBM and contribute to the absorption and storage of an increased amount of lipids that will provide sufficient energy used for tumor growth and invasion. This review provides an overview of the key enzymes, transporters, and main regulatory pathways of FAs and ketone bodies (KBs) in normal versus GBM cells, highlighting the need to develop new therapeutic strategies to improve treatment efficacy in patients with GBM.
Collapse
Affiliation(s)
- Corina Tamas
- Doctoral School of Medicine and Pharmacy, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania;
- Department of Neurosurgery, Emergency Clinical County Hospital, 540136 Targu Mures, Romania;
- Department of Neurosurgery, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania
| | - Flaviu Tamas
- Doctoral School of Medicine and Pharmacy, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania;
- Department of Neurosurgery, Emergency Clinical County Hospital, 540136 Targu Mures, Romania;
- Department of Neurosurgery, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania
| | - Attila Kovecsi
- Department of Morphopathology, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania;
- Department of Morphopathology, Emergency Clinical County Hospital, 540136 Targu Mures, Romania
| | - Alina Cehan
- Department of Plastic, Esthetics and Reconstructive Surgery, Emergency Clinical County Hospital, 540136 Targu Mures, Romania;
| | - Adrian Balasa
- Department of Neurosurgery, Emergency Clinical County Hospital, 540136 Targu Mures, Romania;
- Department of Neurosurgery, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania
| |
Collapse
|
2
|
Ni C, Hong M. Oligomerization of drug transporters: Forms, functions, and mechanisms. Acta Pharm Sin B 2024; 14:1924-1938. [PMID: 38799641 PMCID: PMC11119549 DOI: 10.1016/j.apsb.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/07/2023] [Accepted: 01/05/2024] [Indexed: 05/29/2024] Open
Abstract
Drug transporters are essential players in the transmembrane transport of a wide variety of clinical drugs. The broad substrate spectra and versatile distribution pattern of these membrane proteins infer their pharmacological and clinical significance. With our accumulating knowledge on the three-dimensional structure of drug transporters, their oligomerization status has become a topic of intense study due to the possible functional roles carried out by such kind of post-translational modification (PTM). In-depth studies of oligomeric complexes formed among drug transporters as well as their interactions with other regulatory proteins can help us better understand the regulatory mechanisms of these membrane proteins, provide clues for the development of novel drugs, and improve the therapeutic efficacy. In this review, we describe different oligomerization forms as well as their structural basis of major drug transporters in the ATP-binding cassette and solute carrier superfamilies, summarize our current knowledge on the influence of oligomerization for protein expression level and transport function of these membrane proteins, and discuss the regulatory mechanisms of oligomerization. Finally, we highlight the challenges associated with the current oligomerization studies and propose some thoughts on the pharmaceutical application of this important drug transporter PTM.
Collapse
Affiliation(s)
- Chunxu Ni
- College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Mei Hong
- College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
3
|
Anagnostakis F, Kokkorakis M, Markouli M, Piperi C. Impact of Solute Carrier Transporters in Glioma Pathology: A Comprehensive Review. Int J Mol Sci 2023; 24:ijms24119393. [PMID: 37298344 DOI: 10.3390/ijms24119393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Solute carriers (SLCs) are essential for brain physiology and homeostasis due to their role in transporting necessary substances across cell membranes. There is an increasing need to further unravel their pathophysiological implications since they have been proposed to play a pivotal role in brain tumor development, progression, and the formation of the tumor microenvironment (TME) through the upregulation and downregulation of various amino acid transporters. Due to their implication in malignancy and tumor progression, SLCs are currently positioned at the center of novel pharmacological targeting strategies and drug development. In this review, we discuss the key structural and functional characteristics of the main SLC family members involved in glioma pathogenesis, along with their potential targeting options to provide new opportunities for CNS drug design and more effective glioma management.
Collapse
Affiliation(s)
- Filippos Anagnostakis
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Michail Kokkorakis
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Mariam Markouli
- Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
4
|
Papierniak-Wyglądała A, Lamch W, Jurewicz E, Nałęcz KA. The activity and surface presence of organic cation/carnitine transporter OCTN2 (SLC22A5) in breast cancer cells depends on AKT kinase. Arch Biochem Biophys 2023; 742:109616. [PMID: 37187422 DOI: 10.1016/j.abb.2023.109616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/06/2023] [Accepted: 04/26/2023] [Indexed: 05/17/2023]
Abstract
l-carnitine is indispensable for transfer of fatty acids to mitochondria for the process of β-oxidation, a process, whose significance in cancer has drawn attention in recent years. In humans majority of carnitine is delivered by diet and enters the cell due to activity of solute carriers (SLCs), mainly by ubiquitously expressed organic cation/carnitine transporter (OCTN2/SLC22A5). In control and cancer human breast epithelial cell lines the major fraction of OCTN2 is present as a not matured non-glycosylated form. Studies on overexpressed OCTN2 demonstrated an exclusive interaction with SEC24C, as the cargo-recognizing subunit of coatomer II in transporter exit from endoplasmic reticulum. Co-transfection with SEC24C dominant negative mutant completely abolished presence of the mature form of OCTN2, pointing to a possibility of trafficking regulation. SEC24C was previously shown to be phosphorylated by serine/threonine kinase AKT, known to be activated in cancer. Further studies on breast cell lines showed that inhibition of AKT with MK-2206 in control and cancer lines decreased level of OCTN2 mature form. Proximity ligation assay showed that phosphorylation of OCTN2 on threonine was significantly abolished by AKT inhibition with MK-2206. Carnitine transport was positively correlated with the level of OCTN2 phosphorylated by AKT on threonine moiety. The observed regulation of OCTN2 by AKT places this kinase in the center of metabolic control. This points to both proteins, AKT and OCTN2, as druggable targets, in particular in a combination therapy of breast cancer.
Collapse
Affiliation(s)
- Anna Papierniak-Wyglądała
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland.
| | - Weronika Lamch
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland.
| | - Ewelina Jurewicz
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland.
| | - Katarzyna A Nałęcz
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland.
| |
Collapse
|
5
|
Zhang L, Gui T, Console L, Scalise M, Indiveri C, Hausler S, Kullak-Ublick GA, Gai Z, Visentin M. Cholesterol stimulates the cellular uptake of L-carnitine by the carnitine/organic cation transporter novel 2 (OCTN2). J Biol Chem 2020; 296:100204. [PMID: 33334877 PMCID: PMC7948396 DOI: 10.1074/jbc.ra120.015175] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 12/14/2020] [Accepted: 12/17/2020] [Indexed: 12/19/2022] Open
Abstract
The carnitine/organic cation transporter novel 2 (OCTN2) is responsible for the cellular uptake of carnitine in most tissues. Being a transmembrane protein OCTN2 must interact with the surrounding lipid microenvironment to function. Among the main lipid species that constitute eukaryotic cells, cholesterol has highly dynamic levels under a number of physiopathological conditions. This work describes how plasma membrane cholesterol modulates OCTN2 transport of L-carnitine in human embryonic kidney 293 cells overexpressing OCTN2 (OCTN2-HEK293) and in proteoliposomes harboring human OCTN2. We manipulated the cholesterol content of intact cells, assessed by thin layer chromatography, through short exposures to empty and/or cholesterol-saturated methyl-β-cyclodextrin (mβcd), whereas free cholesterol was used to enrich reconstituted proteoliposomes. We measured OCTN2 transport using [3H]L-carnitine, and expression levels and localization by surface biotinylation and Western blotting. A 20-min preincubation with mβcd reduced the cellular cholesterol content and inhibited L-carnitine influx by 50% in comparison with controls. Analogously, the insertion of cholesterol in OCTN2-proteoliposomes stimulated L-carnitine uptake in a dose-dependent manner. Carnitine uptake in cells incubated with empty mβcd and cholesterol-saturated mβcd to preserve the cholesterol content was comparable with controls, suggesting that the mβcd effect on OCTN2 was cholesterol dependent. Cholesterol stimulated L-carnitine influx in cells by markedly increasing the affinity for L-carnitine and in proteoliposomes by significantly enhancing the affinity for Na+ and, in turn, the L-carnitine maximal transport capacity. Because of the antilipogenic and antioxidant features of L-carnitine, the stimulatory effect of cholesterol on L-carnitine uptake might represent a novel protective effect against lipid-induced toxicity and oxidative stress.
Collapse
Affiliation(s)
- Lu Zhang
- College of Traditional Chinese Medicine, Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China; Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ting Gui
- College of Traditional Chinese Medicine, Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lara Console
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Mariafrancesca Scalise
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Stephanie Hausler
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Gerd A Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Mechanistic Safety, CMO & Patient Safety, Global Drug Development, Novartis Pharma, Basel, Switzerland
| | - Zhibo Gai
- College of Traditional Chinese Medicine, Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China; Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Michele Visentin
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
6
|
Nałęcz KA. Amino Acid Transporter SLC6A14 (ATB 0,+) - A Target in Combined Anti-cancer Therapy. Front Cell Dev Biol 2020; 8:594464. [PMID: 33195271 PMCID: PMC7609839 DOI: 10.3389/fcell.2020.594464] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer cells are characterized by quick growth and proliferation, demanding constant supply of various nutrients. Several plasma membrane transporters delivering such compounds are upregulated in cancer. Solute carrier family 6 member 14 (SLC6A14), known as amino acid transporter B0,+ (ATB0,+) transports all amino acids with exception of the acidic ones: aspartate and glutamate. Its malfunctioning is correlated with several pathological states and it is upregulated in solid tumors. The high expression of SLC6A14 is prognostic and unfavorable in pancreatic cancer, while in breast cancer it is expressed in estrogen receptor positive cells. As many plasma membrane transporters it resides in endoplasmic reticulum (ER) membrane after translation before further trafficking through Golgi to the cell surface. Transporter exit from ER is strictly controlled. The proper folding of SLC6A14 was shown to be controlled from the cytoplasmic side by heat shock proteins, further exit from ER and formation of coatomer II (COPII) coated vesicles depends on specific interaction with COPII cargo-recognizing subunit SEC24C, phosphorylated by kinase AKT. Inhibition of heat shock proteins, known to be upregulated in cancer, directs SLC6A14 to degradation. Targeting proteins regulating SLC6A14 trafficking is proposed as an additional pharmacological treatment of cancer.
Collapse
Affiliation(s)
- Katarzyna A Nałęcz
- Laboratory of Transport Through Biomembranes, Nencki Institute of Experimental Biology, Warsaw, Poland
| |
Collapse
|
7
|
Juraszek B, Czarnecka-Herok J, Nałęcz KA. Glioma cells survival depends both on fatty acid oxidation and on functional carnitine transport by SLC22A5. J Neurochem 2020; 156:642-657. [PMID: 32654140 DOI: 10.1111/jnc.15124] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023]
Abstract
Gliomas are the most common primary malignant brain tumor in adults, but current treatment for glioblastoma multiforme (GBM) is insufficient. Even though glucose is the primary energetic substrate of glioma cells, they are capable of using fatty acids to generate energy. Fatty acid oxidation (FAO) in mitochondria requires L-carnitine for the formation of acylcarnitines by carnitine palmitoylotransferase 1 (CPT1) and further transport of acyl carnitine esters to mitochondrial matrix. Carnitine can be delivered to the cell by an organic cation/carnitine transporter-SLC22A5/OCTN2. In this study, we show that SLC22A5 is up-regulated in glioma cells and that they vary in the amount of SLC22A5 in the plasma membrane. Research on glioma cells (lines U87MG, LN229, T98G) with various expression levels of SLC22A5 demonstrated a correlation between the FAO rate, the level of the transporter, and the carnitine transport. Inhibition of carnitine transport by chemotherapeutics, such as vinorelbine and vincristine, led to inhibition of FAO, which was further intensified by etomoxir-a CPT1 inhibitor. This led to reduced viability and increased apoptosis in glioma cells. Modulation of SLC22A5 level by either silencing or up-regulation of SLC22A5 also affected glioma cell survival in a FAO-dependent way. These observations suggest that the survival of glioma cells is heavily reliant on both FAO and SLC22A5 activity, as well as that CPT1 and SLC22A5 might be possible drug targets.
Collapse
Affiliation(s)
- Barbara Juraszek
- Laboratory of Transport through Biomembranes, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Joanna Czarnecka-Herok
- Laboratory of Molecular Bases of Ageing, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna A Nałęcz
- Laboratory of Transport through Biomembranes, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
8
|
Zhang Y, Ruggiero M, Hagenbuch B. OATP1B3 Expression and Function is Modulated by Coexpression with OCT1, OATP1B1, and NTCP. Drug Metab Dispos 2020; 48:622-630. [PMID: 32482756 DOI: 10.1124/dmd.119.089847] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 05/13/2020] [Indexed: 12/29/2022] Open
Abstract
Organic anion transporting polypeptide (OATP) 1B3 is a drug transporter expressed at the basolateral membrane of human hepatocytes. Along with other transporters, including OATP1B1, Na+/taurocholate cotransporting polypeptide (NTCP), and organic cation transporter (OCT) 1, it is responsible for the uptake of endo- and xenobiotics into hepatocytes. Our previous studies demonstrated that OATP1B3 can form hetero-oligomers with OATP1B1 in human embryonic kidney 293T (HEK293) cells and with NTCP in both HEK293 cells and frozen human liver sections. To further characterize the hetero-oligomerization of OATP1B3, we investigated OCT1 as a potential interacting partner and determined the functional consequences of OATP1B3 hetero-oligomerization. We demonstrated interactions between OATP1B3 and OCT1 by coimmunoprecipitation with an anti-OATP1B3 antibody from human hepatocytes. In addition, we visualized the interaction using the proximity ligation assay in both HEK293 cells and in frozen human liver sections. We investigated the functional consequences of OATP1B3 hetero-oligomerization by measuring the OATP1B3 plasma membrane expression and the uptake of the OATP1B3 selective substrate cholecystokinin-8 (CCK-8) in the absence and presence of OATP1B1, NTCP, and OCT1. A significant decrease of OATP1B3 plasma membrane expression was observed after coexpression with OCT1, whereas coexpression with OATP1B1 or NTCP resulted in an increase of plasma membrane expression. With respect to transport, coexpression of OCT1 increased the apparent turnover rate of OATP1B3, whereas coexpression of OATP1B1 or NTCP decreased it. These findings demonstrated that coexpression of OATP1B3 with OATP1B1, NTCP, and OCT1 in HEK293 cells results in a transporter-dependent modification of OATP1B3-mediated CCK-8 transport and suggest that functional results obtained in single transporter overexpressing cell lines over- or underestimate OATP1B3 function in human hepatocytes. SIGNIFICANCE STATEMENT: Coexpression of organic anion transporting polypeptide (OATP) 1B3 with organic cation transporter (OCT) 1, Na+/taurocholate cotransporting polypeptide, or OATP1B1 in human embryonic kidney 293T cells affects its expression level and function. When OCT1 is knocked down in human hepatocytes, function of OATP1B3 goes up. These results suggest that protein-protein interactions can affect the expression and function of the involved proteins, and thus single transporter expression systems might lead to over- or underestimation of drug-drug interactions.
Collapse
Affiliation(s)
- Yuchen Zhang
- Department of Pharmacology, Toxicology, and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas
| | - Melissa Ruggiero
- Department of Pharmacology, Toxicology, and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas
| | - Bruno Hagenbuch
- Department of Pharmacology, Toxicology, and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
9
|
Juraszek B, Nałęcz KA. SLC22A5 (OCTN2) Carnitine Transporter-Indispensable for Cell Metabolism, a Jekyll and Hyde of Human Cancer. Molecules 2019; 25:molecules25010014. [PMID: 31861504 PMCID: PMC6982704 DOI: 10.3390/molecules25010014] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/12/2019] [Accepted: 12/14/2019] [Indexed: 12/26/2022] Open
Abstract
Oxidation of fatty acids uses l-carnitine to transport acyl moieties to mitochondria in a so-called carnitine shuttle. The process of β-oxidation also takes place in cancer cells. The majority of carnitine comes from the diet and is transported to the cell by ubiquitously expressed organic cation transporter novel family member 2 (OCTN2)/solute carrier family 22 member 5 (SLC22A5). The expression of SLC22A5 is regulated by transcription factors peroxisome proliferator-activated receptors (PPARs) and estrogen receptor. Transporter delivery to the cell surface, as well as transport activity are controlled by OCTN2 interaction with other proteins, such as PDZ-domain containing proteins, protein phosphatase PP2A, caveolin-1, protein kinase C. SLC22A5 expression is altered in many types of cancer, giving an advantage to some of them by supplying carnitine for β-oxidation, thus providing an alternative to glucose source of energy for growth and proliferation. On the other hand, SLC22A5 can also transport several chemotherapeutics used in clinics, leading to cancer cell death.
Collapse
|
10
|
Zhang Y, Hagenbuch B. Protein-protein interactions of drug uptake transporters that are important for liver and kidney. Biochem Pharmacol 2019; 168:384-391. [PMID: 31381872 DOI: 10.1016/j.bcp.2019.07.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/31/2019] [Indexed: 12/28/2022]
Abstract
Drug uptake transporters are membrane proteins responsible for the trans-membrane transport of endo- and xenobiotics, including numerous drugs. They are important for the uptake of drugs into target tissues or into organs for metabolism and excretion. Many drug uptake transporters have a broad spectrum of structural-independent substrates, which make them vulnerable to drug-drug interactions. Recent studies have shown more and more complex pharmacokinetics involving transporters, and regulatory agencies now require studies to be performed to measure the involvement of transporters in drug development. A better understanding of the factors affecting the expression of transporters is needed. Despite many efforts devoted to the functional characterization of different drug uptake transporters, transporter in vitro to in vivo extrapolations are far from predicting the behavior under physiological conditions. There is an increasing number of uptake transporters demonstrated to form protein-protein interactions or to oligomerize. This raises the possibility that these interactions between or among transporters could help explaining the gap between in vitro and in vivo measurement of drug transporters. In this review, we summarized protein-protein interactions of drug uptake transporters that are important for pharmacokinetics, especially those in the liver and the kidneys.
Collapse
Affiliation(s)
- Yuchen Zhang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, United States
| | - Bruno Hagenbuch
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, United States.
| |
Collapse
|
11
|
Pochini L, Galluccio M, Scalise M, Console L, Indiveri C. OCTN: A Small Transporter Subfamily with Great Relevance to Human Pathophysiology, Drug Discovery, and Diagnostics. SLAS DISCOVERY 2018; 24:89-110. [PMID: 30523710 DOI: 10.1177/2472555218812821] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OCTN is a small subfamily of membrane transport proteins that belongs to the larger SLC22 family. Two of the three members of the subfamily, namely, OCTN2 and OCTN1, are present in humans. OCTN2 plays a crucial role in the absorption of carnitine from diet and in its distribution to tissues, as demonstrated by the occurrence of severe pathologies caused by malfunctioning or altered expression of this transporter. These findings suggest avoiding a strict vegetarian diet during pregnancy and in childhood. Other roles of OCTN2 are related to the traffic of carnitine derivatives in many tissues. The role of OCTN1 is still unclear, despite the identification of some substrates such as ergothioneine, acetylcholine, and choline. Plausibly, the transporter acts on the control of inflammation and oxidative stress, even though knockout mice do not display phenotypes. A clear role of both transporters has been revealed in drug interaction and delivery. The polyspecificity of the OCTNs is at the base of the interactions with drugs. Interestingly, OCTN2 has been recently exploited in the prodrug approach and in diagnostics. A promising application derives from the localization of OCTN2 in exosomes that represent a noninvasive diagnostic tool.
Collapse
Affiliation(s)
- Lorena Pochini
- 1 Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Michele Galluccio
- 1 Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Mariafrancesca Scalise
- 1 Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Lara Console
- 1 Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy
| | - Cesare Indiveri
- 1 Department DiBEST (Biologia, Ecologia, Scienze della Terra), Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Arcavacata di Rende, Italy.,2 CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, Bari, Italy
| |
Collapse
|
12
|
Lamhonwah AM, Barić I, Lamhonwah J, Grubić M, Tein I. Attention deficit/hyperactivity disorder as an associated feature in OCTN2 deficiency with novel deletion (p.T440-Y449). Clin Case Rep 2018; 6:585-591. [PMID: 29636919 PMCID: PMC5889263 DOI: 10.1002/ccr3.1316] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 10/09/2017] [Accepted: 10/31/2017] [Indexed: 01/15/2023] Open
Abstract
This boy presented with ADHD at 3 years and at 8 years was hyperactive with no documented hypoglycemia and had myopathy, cardiomyopathy, and very low serum carnitine. L‐carnitine improved his exercise intolerance, cardiomyopathy, and behavior. Analysis of SLC22A5 revealed a premature stop codon (p.R282*) and a novel in‐frame deletion (p.T440‐Y449).
Collapse
Affiliation(s)
- Anne-Marie Lamhonwah
- Division of Neurology Department of Pediatrics, and Genetics and Genome Biology Program The Research Institute The Hospital for Sick Children University of Toronto Toronto Ontario M5G 1X8 Canada
| | - Ivo Barić
- Department of Pediatrics University Hospital Center Zagreb Zagreb 10000 Croatia.,School of Medicine University of Zagreb Zagreb 10000 Croatia
| | - Jessica Lamhonwah
- Division of Neurology Department of Pediatrics, and Genetics and Genome Biology Program The Research Institute The Hospital for Sick Children University of Toronto Toronto Ontario M5G 1X8 Canada
| | - Marina Grubić
- Department of Pediatrics University Hospital Center Zagreb Zagreb 10000 Croatia
| | - Ingrid Tein
- Division of Neurology Department of Pediatrics, and Genetics and Genome Biology Program The Research Institute The Hospital for Sick Children University of Toronto Toronto Ontario M5G 1X8 Canada.,Department of Laboratory Medicine and Pathobiology University of Toronto Toronto Ontario M5G 1X8 Canada
| |
Collapse
|
13
|
L-Carnitine and Acetyl-L-carnitine Roles and Neuroprotection in Developing Brain. Neurochem Res 2017; 42:1661-1675. [PMID: 28508995 DOI: 10.1007/s11064-017-2288-7] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 12/30/2022]
Abstract
L-Carnitine functions to transport long chain fatty acyl-CoAs into the mitochondria for degradation by β-oxidation. Treatment with L-carnitine can ameliorate metabolic imbalances in many inborn errors of metabolism. In recent years there has been considerable interest in the therapeutic potential of L-carnitine and its acetylated derivative acetyl-L-carnitine (ALCAR) for neuroprotection in a number of disorders including hypoxia-ischemia, traumatic brain injury, Alzheimer's disease and in conditions leading to central or peripheral nervous system injury. There is compelling evidence from preclinical studies that L-carnitine and ALCAR can improve energy status, decrease oxidative stress and prevent subsequent cell death in models of adult, neonatal and pediatric brain injury. ALCAR can provide an acetyl moiety that can be oxidized for energy, used as a precursor for acetylcholine, or incorporated into glutamate, glutamine and GABA, or into lipids for myelination and cell growth. Administration of ALCAR after brain injury in rat pups improved long-term functional outcomes, including memory. Additional studies are needed to better explore the potential of L-carnitine and ALCAR for protection of developing brain as there is an urgent need for therapies that can improve outcome after neonatal and pediatric brain injury.
Collapse
|
14
|
Protein Kinases C-Mediated Regulations of Drug Transporter Activity, Localization and Expression. Int J Mol Sci 2017; 18:ijms18040764. [PMID: 28375174 PMCID: PMC5412348 DOI: 10.3390/ijms18040764] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 03/25/2017] [Accepted: 03/27/2017] [Indexed: 01/05/2023] Open
Abstract
Drug transporters are now recognized as major actors in pharmacokinetics, involved notably in drug–drug interactions and drug adverse effects. Factors that govern their activity, localization and expression are therefore important to consider. In the present review, the implications of protein kinases C (PKCs) in transporter regulations are summarized and discussed. Both solute carrier (SLC) and ATP-binding cassette (ABC) drug transporters can be regulated by PKCs-related signaling pathways. PKCs thus target activity, membrane localization and/or expression level of major influx and efflux drug transporters, in various normal and pathological types of cells and tissues, often in a PKC isoform-specific manner. PKCs are notably implicated in membrane insertion of bile acid transporters in liver and, in this way, are thought to contribute to cholestatic or choleretic effects of endogenous compounds or drugs. The exact clinical relevance of PKCs-related regulation of drug transporters in terms of drug resistance, pharmacokinetics, drug–drug interactions and drug toxicity remains however to be precisely determined. This issue is likely important to consider in the context of the development of new drugs targeting PKCs-mediated signaling pathways, for treating notably cancers, diabetes or psychiatric disorders.
Collapse
|
15
|
Tight junction protein ZO-1 controls organic cation/carnitine transporter OCTN2 (SLC22A5) in a protein kinase C-dependent way. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:797-805. [PMID: 28257821 DOI: 10.1016/j.bbamcr.2017.02.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 02/07/2017] [Accepted: 02/27/2017] [Indexed: 12/20/2022]
Abstract
OCTN2 (SLC22A5) is an organic cation/carnitine transporter belonging to the solute carrier transporters (SLC) family. OCTN2 is ubiquitously expressed and its presence was shown in various brain cells, including the endothelial cells forming blood-brain barrier, where it was mainly detected at abluminal membrane and in proximity of tight junctions (TJ). Since OCTN2 contains a PDZ-binding domain, the present study was focused on a possible role of transporter interaction with a TJ-associated protein ZO-1, containing PDZ domains and detected in rat Octn2 proteome. We showed previously that activation of protein kinase C (PKC) in rat astrocytes regulates Octn2 surface presence and activity. Regulation of a wild type Octn2 and its deletion mutant without a PDZ binding motif were studied in heterologous expression system in HEK293 cells. Plasma membrane presence of overexpressed Octn2 did not depend on either PKC activation or presence of PDZ-binding motif, anyhow, as assayed in proximity ligation assay, the truncation of PDZ binding motif resulted in a strongly diminished Octn2/ZO-1 interaction and in a decreased transporter activity. The same effects on Octn2 activity were detected upon PKC activation, what correlated with ZO-1 phosphorylation. It is postulated that ZO-1, when not phosphorylated by PKC, keeps Octn2 in an active state, while elimination of this binding in ΔPDZ mutant or after ZO-1 phosphorylation leads to diminution of Octn2 activity.
Collapse
|
16
|
Juraszek B, Nałęcz KA. Protein phosphatase PP2A - a novel interacting partner of carnitine transporter OCTN2 (SLC22A5) in rat astrocytes. J Neurochem 2016; 139:537-551. [DOI: 10.1111/jnc.13777] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 07/15/2016] [Accepted: 08/08/2016] [Indexed: 11/29/2022]
Affiliation(s)
- Barbara Juraszek
- Laboratory of Transport through Biomembranes; Department of Molecular and Cellular Neurobiology; Nencki Institute of Experimental Biology of Polish Academy of Sciences; Warsaw Poland
| | - Katarzyna A. Nałęcz
- Laboratory of Transport through Biomembranes; Department of Molecular and Cellular Neurobiology; Nencki Institute of Experimental Biology of Polish Academy of Sciences; Warsaw Poland
| |
Collapse
|
17
|
Szabó K, Nagy Z, Juhász V, Zolnerciks JK, Csorba A, Tímár Z, Molnár É, Pádár P, Johnson W, Beéry E, Krajcsi P. Species specificity profiling of rat and human organic cation/carnitine transporter Slc22a5/SLC22A5 (Octn2/OCTN2). Drug Metab Pharmacokinet 2016; 32:165-171. [PMID: 28365301 DOI: 10.1016/j.dmpk.2016.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 07/25/2016] [Accepted: 08/18/2016] [Indexed: 01/09/2023]
Abstract
The purpose of this study was to characterize the uptake of carnitine, the physiological substrate, and the uptake of 3-(2,2,2-trimethylhydrazinium)propionate, a consensus substrate by rat Octn2 and human OCTN2 transporters as well as to characterize drug-mediated inhibition of l-carnitine uptake by the rat and human orthologs overexpressed in CHO-K1 cells. l-carnitine and 3-(2,2,2-trimethylhydrazinium)propionate were found to be a lower affinity substrate for rat Octn2 (KM = 32.66 ± 5.11 μM and 23.62 ± 4.99 μM respectively) than for human OCTN2 (KM = 3.08 ± 0.74 μM and 7.98 ± 0.63 μM). The intrinsic clearance (CLint) value for carnitine was higher for the human than for the rat transporter (22.82 ± 5.57 ml/min*mg vs 4.008 ± 0.675 ml/min*mg). For 3-(2,2,2-trimethylhydrazinium)propionate, in contrast, the CLint value for rat Octn2 was higher than for human OCTN2 (323.9 ± 72.8 ml/min*mg vs 65.11 ± 5.33 ml/min*mg). Furthermore, many pharmacologically important drugs were shown to affect l-carnitine transport by Octn2/OCTN2. The correlation between the IC50 datasets for the rat and human transporter resulted in an r value of 0.47 (p > 0.05). However, the greatest difference was less than seven-fold and 13 of 15 compounds yielded a difference less than 3-fold. Thus, the transporters from these two species showed an overlapping but somewhat different substrate and inhibitor specificity.
Collapse
Affiliation(s)
- Kitti Szabó
- SOLVO Biotechnology, 2 Gyár utca, Budaörs 2040, Hungary; SOLVO Biotechnology, 52 Közép fasor, Szeged 6726, Hungary.
| | - Zoltán Nagy
- SOLVO Biotechnology, 2 Gyár utca, Budaörs 2040, Hungary.
| | | | | | - Attila Csorba
- SOLVO Biotechnology, 52 Közép fasor, Szeged 6726, Hungary.
| | - Zoltán Tímár
- SOLVO Biotechnology, 52 Közép fasor, Szeged 6726, Hungary.
| | - Éva Molnár
- SOLVO Biotechnology, 52 Közép fasor, Szeged 6726, Hungary.
| | - Petra Pádár
- SOLVO Biotechnology, 52 Közép fasor, Szeged 6726, Hungary.
| | | | - Erzsébet Beéry
- SOLVO Biotechnology, 2 Gyár utca, Budaörs 2040, Hungary.
| | - Péter Krajcsi
- SOLVO Biotechnology, 2 Gyár utca, Budaörs 2040, Hungary.
| |
Collapse
|
18
|
Nałęcz KA. Solute Carriers in the Blood–Brain Barier: Safety in Abundance. Neurochem Res 2016; 42:795-809. [DOI: 10.1007/s11064-016-2030-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/29/2016] [Accepted: 08/02/2016] [Indexed: 12/22/2022]
|
19
|
Michalec K, Mysiorek C, Kuntz M, Bérézowski V, Szczepankiewicz AA, Wilczyński GM, Cecchelli R, Nałęcz KA. Protein kinase C restricts transport of carnitine by amino acid transporter ATB(0,+) apically localized in the blood-brain barrier. Arch Biochem Biophys 2014; 554:28-35. [PMID: 24823859 DOI: 10.1016/j.abb.2014.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 04/29/2014] [Accepted: 05/03/2014] [Indexed: 10/25/2022]
Abstract
Carnitine (3-hydroxy-4-trimethylammoniobutyrate) is necessary for transfer of fatty acids through the inner mitochondrial membrane. Carnitine, not synthesized in the brain, is delivered there through the strongly polarized blood-brain barrier (BBB). Expression and presence of two carnitine transporters - organic cation/carnitine transporter (OCTN2) and amino acid transporter B(0,+) (ATB(0,+)) have been demonstrated previously in an in vitro model of the BBB. Due to potential protein kinase C (PKC) phosphorylation sites within ATB(0,+) sequence, the present study verified effects of this kinase on transporter function and localization in the BBB. ATB(0,+) can be regulated by estrogen receptor α and up-regulated in vitro, therefore its presence in vivo was verified with the transmission electron microscopy. The analyses of brain slices demonstrated ATB(0,+) luminal localization in brain capillaries, confirmed by biotinylation experiments in an in vitro model of the BBB. Brain capillary endothelial cells were shown to control carnitine gradient. ATB(0,+) was phosphorylated by PKC, what correlated with inhibition of carnitine transport. PKC activation did not change the amount of ATB(0,+) present in the apical membrane of brain endothelial cells, but resulted in transporter exclusion from raft microdomains. ATB(0,+) inactivation by a lateral movement in plasma membrane after transporter phosphorylation has been postulated.
Collapse
Affiliation(s)
- Katarzyna Michalec
- Laboratory of Mechanisms of Transport Through Biomembranes, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Caroline Mysiorek
- Univ Lille Nord de France, Lille, France; UArtois, LBHE, Lens, France; IMPRT-IFR114, Lille, France
| | - Mélanie Kuntz
- Univ Lille Nord de France, Lille, France; UArtois, LBHE, Lens, France; IMPRT-IFR114, Lille, France
| | - Vincent Bérézowski
- Univ Lille Nord de France, Lille, France; UArtois, LBHE, Lens, France; IMPRT-IFR114, Lille, France
| | - Andrzej A Szczepankiewicz
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Grzegorz M Wilczyński
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Roméo Cecchelli
- Univ Lille Nord de France, Lille, France; UArtois, LBHE, Lens, France; IMPRT-IFR114, Lille, France
| | - Katarzyna A Nałęcz
- Laboratory of Mechanisms of Transport Through Biomembranes, Nencki Institute of Experimental Biology, Warsaw, Poland.
| |
Collapse
|