1
|
Del Chierico F, Masi L, Petito V, Baldelli V, Puca P, Benvenuto R, Fidaleo M, Palucci I, Lopetuso LR, Caristo ME, Carrozza C, Giustiniani MC, Nakamichi N, Kato Y, Putignani L, Gasbarrini A, Pani G, Scaldaferri F. Solute Transporter OCTN1/Slc22a4 Affects Disease Severity and Response to Infliximab in Experimental Colitis: Role of Gut Microbiota and Immune Modulation. Inflamm Bowel Dis 2024; 30:2259-2270. [PMID: 38944815 PMCID: PMC11630256 DOI: 10.1093/ibd/izae135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Indexed: 07/01/2024]
Abstract
BACKGROUND Inflammatory bowel diseases are chronic disabling conditions with a complex and multifactorial etiology, still incompletely understood. OCTN1, an organic cation transporter, could have a role in modulating the inflammatory response, and some genetic polymorphisms of this molecule have been associated with increased risk of inflammatory bowel diseases. Until now, limited information exists on its potential in predicting/modulating patient's response to therapies. The aim of this study was to evaluate the role of OCTN1 in modifying gut microbiota and mucosal immunity in response to infliximab therapy in murine colitis. METHODS A dextran sodium sulphate model of colitis was used to assess the clinical efficacy of infliximab administered intravenously in ocnt1 gene knockout mice and their C57BL/6 controls. Stool, colon, and mesenteric lymph node samples were collected to evaluate differences in gut microbiota composition, histology, and T cell populations, respectively. RESULTS Octn1 -/- influences the microbiota profile and is associated with a worse dysbiosis in mice with colitis. Infliximab treatment attenuates colitis-associated dysbiosis, with an increase of bacterial richness and evenness in both strains. In comparison with wild type, octn1-/- mice have milder disease and a higher baseline percentage of Treg, Tmemory, Th2 and Th17 cells. CONCLUSIONS Our data support the murine model to study OCTN1 genetic contribution to inflammatory bowel diseases. This could be the first step towards the recognition of this membrane transporter as a biomarker in inflammatory conditions and a predictor of response to therapies.
Collapse
Affiliation(s)
- Federica Del Chierico
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Letizia Masi
- Department of Medical and Surgical Science, Digestive Disease Center (CeMAD) Translational Research Laboratories, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy
| | - Valentina Petito
- Department of Medical and Surgical Science, Digestive Disease Center (CeMAD) Translational Research Laboratories, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy
| | - Valerio Baldelli
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Pierluigi Puca
- Department of Medical and Surgical Sciences, UOS Inflammatory Bowel Diseases, Center for Diseases of Digestive System (CeMAD), Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, Rome, Italy
| | - Roberta Benvenuto
- Department of Pathology, Fondazione Policlinico Universitario A.Gemelli IRCCS, Rome, Italy
| | - Marco Fidaleo
- Department of Biology and Biotechnologies Charles Darwin, Università La Sapienza, Rome, Italy
| | - Ivana Palucci
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Fondazione Policlinico Universitario A.Gemelli IRCCS, Rome, Italy
- Institute of Microbiology, Catholic University of the Sacred Heart, Rome, Italy
| | - Loris Riccardo Lopetuso
- Department of Medical and Surgical Sciences, UOS Inflammatory Bowel Diseases, Center for Diseases of Digestive System (CeMAD), Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Medicine and Ageing Sciences, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | | | - Cinzia Carrozza
- Department of Clinical Biochemistry, Laboratory and Infectious Science, Fondazione Policlinico Universitario A.Gemelli IRCCS, Rome, Italy
| | | | - Noritaka Nakamichi
- Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, 370-0033, Takasaki, Gunma, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Lorenza Putignani
- Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Science, Digestive Disease Center (CeMAD) Translational Research Laboratories, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, Rome, Italy
- Department of Medical and Surgical Science, Digestive Disease Center (CeMAD), Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy
| | - Giovambattista Pani
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, Rome, Italy
| | - Franco Scaldaferri
- Department of Medical and Surgical Sciences, UOS Inflammatory Bowel Diseases, Center for Diseases of Digestive System (CeMAD), Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
2
|
Pochini L. Involvement of mammalian SoLute Carriers (SLC) in the traffic of polyamines. Front Mol Biosci 2024; 11:1452184. [PMID: 39130372 PMCID: PMC11310933 DOI: 10.3389/fmolb.2024.1452184] [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: 06/20/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024] Open
Abstract
Polyamines interact with different molecular targets to regulate a vast range of cellular processes. A network of enzymes and transport systems is crucial for the maintenance of polyamine homeostasis. Indeed, polyamines after synthesis must be distributed to the various tissues and some intracellular organelles. Differently from the well characterized enzymes devoted to polyamine synthesis, the transport systems are not unequivocally identified or characterized. Besides some ATPases which have been identified as polyamine transporters, much less is known about solute carriers (SLC) involved in the transport of these compounds. Only two SLCs have been unequivocally identified as polyamine transporters: SLC18B1 (VPAT) and SLC22A4 (OCTN1). Transport studies have been performed with cells transfected with the cDNAs encoding the two and other SLCs or, in the case of OCTN1, also by in vitro assay using proteoliposomes harboring the recombinant human protein. According to the role proposed for OCTN1, polyamines have been associated with prolonged and quality of life. This review provides an update on the most recent findings concerning the polyamine transporters or the prediction of the putative ones.
Collapse
Affiliation(s)
- Lorena Pochini
- Laboratory of Biochemistry, Molecular Biotechnology and Molecular Biology, Department DiBEST (Biologia, Ecologia, Scienze Della Terra), University of Calabria, Rende, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Bari, Italy
| |
Collapse
|
3
|
Ben Mariem O, Palazzolo L, Torre B, Wei Y, Bianchi D, Guerrini U, Laurenzi T, Saporiti S, De Fabiani E, Pochini L, Indiveri C, Eberini I. Atomistic description of the OCTN1 recognition mechanism via in silico methods. PLoS One 2024; 19:e0304512. [PMID: 38829838 PMCID: PMC11146731 DOI: 10.1371/journal.pone.0304512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
The Organic Cation Transporter Novel 1 (OCTN1), also known as SLC22A4, is widely expressed in various human tissues, and involved in numerous physiological and pathological processes remains. It facilitates the transport of organic cations, zwitterions, with selectivity for positively charged solutes. Ergothioneine, an antioxidant compound, and acetylcholine (Ach) are among its substrates. Given the lack of experimentally solved structures of this protein, this study aimed at generating a reliable 3D model of OCTN1 to shed light on its substrate-binding preferences and the role of sodium in substrate recognition and transport. A chimeric model was built by grafting the large extracellular loop 1 (EL1) from an AlphaFold-generated model onto a homology model. Molecular dynamics simulations revealed domain-specific mobility, with EL1 exhibiting the highest impact on overall stability. Molecular docking simulations identified cytarabine and verapamil as highest affinity ligands, consistent with their known inhibitory effects on OCTN1. Furthermore, MM/GBSA analysis allowed the categorization of substrates into weak, good, and strong binders, with molecular weight strongly correlating with binding affinity to the recognition site. Key recognition residues, including Tyr211, Glu381, and Arg469, were identified through interaction analysis. Ach demonstrated a low interaction energy, supporting the hypothesis of its one-directional transport towards to outside of the membrane. Regarding the role of sodium, our model suggested the involvement of Glu381 in sodium binding. Molecular dynamics simulations of systems at increasing levels of Na+ concentrations revealed increased sodium occupancy around Glu381, supporting experimental data associating Na+ concentration to molecule transport. In conclusion, this study provides valuable insights into the 3D structure of OCTN1, its substrate-binding preferences, and the role of sodium in the recognition. These findings contribute to the understanding of OCTN1 involvement in various physiological and pathological processes and may have implications for drug development and disease management.
Collapse
Affiliation(s)
- Omar Ben Mariem
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Luca Palazzolo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Beatrice Torre
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Yao Wei
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Davide Bianchi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Uliano Guerrini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Tommaso Laurenzi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Simona Saporiti
- Analytical Excellence and Program Management, Merck Serono S.p.A., Rome, Italy
| | - Emma De Fabiani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Lorena Pochini
- Dipartimento di Biologia, Ecologia e Scienze della Terra, Università della Calabria, Arcavacata CS, Italy
| | - Cesare Indiveri
- Dipartimento di Biologia, Ecologia e Scienze della Terra, Università della Calabria, Arcavacata CS, Italy
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Bari, Italy
| | - Ivano Eberini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
- DSRC, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
4
|
Pochini L, Galluccio M, Console L, Scalise M, Eberini I, Indiveri C. Inflammation and Organic Cation Transporters Novel (OCTNs). Biomolecules 2024; 14:392. [PMID: 38672410 PMCID: PMC11048549 DOI: 10.3390/biom14040392] [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: 02/15/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Inflammation is a physiological condition characterized by a complex interplay between different cells handled by metabolites and specific inflammatory-related molecules. In some pathological situations, inflammation persists underlying and worsening the pathological state. Over the years, two membrane transporters namely OCTN1 (SLC22A4) and OCTN2 (SLC22A5) have been shown to play specific roles in inflammation. These transporters form the OCTN subfamily within the larger SLC22 family. The link between these proteins and inflammation has been proposed based on their link to some chronic inflammatory diseases such as asthma, Crohn's disease (CD), and rheumatoid arthritis (RA). Moreover, the two transporters show the ability to mediate the transport of several compounds including carnitine, carnitine derivatives, acetylcholine, ergothioneine, and gut microbiota by-products, which have been specifically associated with inflammation for their anti- or proinflammatory action. Therefore, the absorption and distribution of these molecules rely on the presence of OCTN1 and OCTN2, whose expression is modulated by inflammatory cytokines and transcription factors typically activated by inflammation. In the present review, we wish to provide a state of the art on OCTN1 and OCTN2 transport function and regulation in relationships with inflammation and inflammatory diseases focusing on the metabolic signature collected in different body districts and gene polymorphisms related to inflammatory diseases.
Collapse
Affiliation(s)
- Lorena Pochini
- Laboratory of Biochemistry, Molecular Biotechnology and Molecular Biology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (M.G.); (L.C.); (M.S.)
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via Amendola 122/O, 70126 Bari, Italy
| | - Michele Galluccio
- Laboratory of Biochemistry, Molecular Biotechnology and Molecular Biology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (M.G.); (L.C.); (M.S.)
| | - Lara Console
- Laboratory of Biochemistry, Molecular Biotechnology and Molecular Biology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (M.G.); (L.C.); (M.S.)
| | - Mariafrancesca Scalise
- Laboratory of Biochemistry, Molecular Biotechnology and Molecular Biology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (M.G.); (L.C.); (M.S.)
| | - Ivano Eberini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Cesare Indiveri
- Laboratory of Biochemistry, Molecular Biotechnology and Molecular Biology, Department DiBEST (Biologia, Ecologia, Scienze della Terra), University of Calabria, Via Bucci 4C, 6C, 87036 Arcavacata di Rende, Italy; (M.G.); (L.C.); (M.S.)
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via Amendola 122/O, 70126 Bari, Italy
| |
Collapse
|
5
|
Pochini L, Galluccio M, Scalise M, Console L, Pappacoda G, Indiveri C. OCTN1: A Widely Studied but Still Enigmatic Organic Cation Transporter Linked to Human Pathology and Drug Interactions. Int J Mol Sci 2022; 23:ijms23020914. [PMID: 35055100 PMCID: PMC8776198 DOI: 10.3390/ijms23020914] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/05/2022] [Accepted: 01/12/2022] [Indexed: 01/27/2023] Open
Abstract
The Novel Organic Cation Transporter, OCTN1, is the first member of the OCTN subfamily; it belongs to the wider Solute Carrier family SLC22, which counts many members including cation and anion organic transporters. The tertiary structure has not been resolved for any cation organic transporter. The functional role of OCNT1 is still not well assessed despite the many functional studies so far conducted. The lack of a definitive identification of OCTN1 function can be attributed to the different experimental systems and methodologies adopted for studying each of the proposed ligands. Apart from the contradictory data, the international scientific community agrees on a role of OCTN1 in protecting cells and tissues from oxidative and/or inflammatory damage. Moreover, the involvement of this transporter in drug interactions and delivery has been well clarified, even though the exact profile of the transported/interacting molecules is still somehow confusing. Therefore, OCTN1 continues to be a hot topic in terms of its functional role and structure. This review focuses on the most recent advances on OCTN1 in terms of functional aspects, physiological roles, substrate specificity, drug interactions, tissue expression, and relationships with pathology.
Collapse
Affiliation(s)
- Lorena Pochini
- Unit of Biochemistry, Molecular Biotechnology and Molecular Biology, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, Via P. Bucci 4c, Arcavacata di Rende, 87036 Cosenza, Italy; (L.P.); (M.G.); (M.S.); (L.C.); (G.P.)
| | - Michele Galluccio
- Unit of Biochemistry, Molecular Biotechnology and Molecular Biology, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, Via P. Bucci 4c, Arcavacata di Rende, 87036 Cosenza, Italy; (L.P.); (M.G.); (M.S.); (L.C.); (G.P.)
| | - Mariafrancesca Scalise
- Unit of Biochemistry, Molecular Biotechnology and Molecular Biology, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, Via P. Bucci 4c, Arcavacata di Rende, 87036 Cosenza, Italy; (L.P.); (M.G.); (M.S.); (L.C.); (G.P.)
| | - Lara Console
- Unit of Biochemistry, Molecular Biotechnology and Molecular Biology, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, Via P. Bucci 4c, Arcavacata di Rende, 87036 Cosenza, Italy; (L.P.); (M.G.); (M.S.); (L.C.); (G.P.)
| | - Gilda Pappacoda
- Unit of Biochemistry, Molecular Biotechnology and Molecular Biology, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, Via P. Bucci 4c, Arcavacata di Rende, 87036 Cosenza, Italy; (L.P.); (M.G.); (M.S.); (L.C.); (G.P.)
| | - Cesare Indiveri
- Unit of Biochemistry, Molecular Biotechnology and Molecular Biology, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, Via P. Bucci 4c, Arcavacata di Rende, 87036 Cosenza, Italy; (L.P.); (M.G.); (M.S.); (L.C.); (G.P.)
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), National Research Council—CNR, Via Amendola 122/O, 70126 Bari, Italy
- Correspondence:
| |
Collapse
|
6
|
Gründemann D, Hartmann L, Flögel S. The Ergothioneine Transporter (ETT): Substrates and Locations, an Inventory. FEBS Lett 2021; 596:1252-1269. [PMID: 34958679 DOI: 10.1002/1873-3468.14269] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 11/07/2022]
Abstract
In all vertebrates including mammals, the ergothioneine transporter ETT (obsolete name OCTN1; human gene symbol SLC22A4) is a powerful and highly specific transporter for the uptake of ergothioneine (ET). ETT is not expressed ubiquitously and only cells with high ETT cell-surface levels can accumulate ET to high concentration. Without ETT, there is no uptake because the plasma membrane is essentially impermeable to this hydrophilic zwitterion. Here, we review the substrate specificity and localization of ETT, which is prominently expressed in neutrophils, monocytes/macrophages, and developing erythrocytes. Most sites of strong expression are conserved across species, but there are also major differences. In particular, we critically analyze the evidence for the expression of ETT in the brain as well as recent data suggesting that the transporter SLC22A15 may transport also ET. We conclude that, to date, ETT remains the only well-defined biomarker for intracellular ET activity. In humans, the ability to take up, distribute, and retain ET depends principally on this transporter.
Collapse
Affiliation(s)
- Dirk Gründemann
- Department of Pharmacology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Gleueler Straße 24, 50931, Cologne, Germany
| | - Lea Hartmann
- Department of Pharmacology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Gleueler Straße 24, 50931, Cologne, Germany
| | - Svenja Flögel
- Department of Pharmacology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Gleueler Straße 24, 50931, Cologne, Germany
| |
Collapse
|
7
|
Liu Y, Li C, Su R, Yin Z, Huang G, Yang J, Li Z, Zhang K, Fei J. Targeting SOS1 overcomes imatinib resistance with BCR-ABL independence through uptake transporter SLC22A4 in CML. Mol Ther Oncolytics 2021; 23:560-570. [PMID: 34938856 PMCID: PMC8654699 DOI: 10.1016/j.omto.2021.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 11/16/2021] [Indexed: 12/28/2022] Open
Abstract
Resistance to the BCR-ABL inhibitor imatinib mesylate poses a major problem for the treatment of chronic myeloid leukemia. Imatinib resistance often results from a secondary mutation in BCR-ABL that interferes with drug binding. However, sometimes there is no mutation in BCR-ABL, and the basis of such BCR-ABL-independent imatinib mesylate resistance remains to be elucidated. SOS1, a guanine nucleotide exchange factor for Ras protein, affects drug sensitivity and resistance to imatinib. The depletion of SOS1 markedly inhibits cell growth either in vitro or in vivo and significantly increases the sensitivity of chronic myeloid leukemia cells to imatinib. Furthermore, LC-MS/MS and RNA-seq assays reveal that SOS1 negatively regulates the expression of SLC22A4, a member of the carnitine/organic cation transporter family, which mediates the active uptake of imatinib into chronic myeloid leukemia cells. HPLC assay confirms that intracellular accumulation of imatinib is accompanied by upregulation of SLC22A4 through SOS1 inhibition in both sensitive and resistant chronic myeloid leukemia cells. BAY-293, an inhibitor of SOS1/Ras, was found to depress proliferation and colony formation in chronic myeloid leukemia cells with resistance and BCR-ABL independence. Altogether these findings indicate that targeting SOS1 inhibition promotes imatinib sensitivity and overcomes resistance with BCR-ABL independence by SLC22A4-mediated uptake transport.
Collapse
Affiliation(s)
- Yanjun Liu
- Department of Biochemistry and Molecular Biology, Medical College of Jinan University, Guangzhou 510632, China.,Engineering Technology Research Center of Guangdong Province for Small Nucleic Acids Drug Development, Guangzhou 510632, China.,Antisense Biopharmaceutical Technology Co., Ltd., Guangzhou 510632, China
| | - Chuting Li
- Department of Biochemistry and Molecular Biology, Medical College of Jinan University, Guangzhou 510632, China.,Engineering Technology Research Center of Guangdong Province for Small Nucleic Acids Drug Development, Guangzhou 510632, China.,Antisense Biopharmaceutical Technology Co., Ltd., Guangzhou 510632, China
| | - Rui Su
- Department of Biochemistry and Molecular Biology, Medical College of Jinan University, Guangzhou 510632, China.,Engineering Technology Research Center of Guangdong Province for Small Nucleic Acids Drug Development, Guangzhou 510632, China.,Antisense Biopharmaceutical Technology Co., Ltd., Guangzhou 510632, China
| | - Zhao Yin
- Department of Biochemistry and Molecular Biology, Medical College of Jinan University, Guangzhou 510632, China.,Engineering Technology Research Center of Guangdong Province for Small Nucleic Acids Drug Development, Guangzhou 510632, China.,Antisense Biopharmaceutical Technology Co., Ltd., Guangzhou 510632, China
| | - Guiping Huang
- Department of Biochemistry and Molecular Biology, Medical College of Jinan University, Guangzhou 510632, China.,Engineering Technology Research Center of Guangdong Province for Small Nucleic Acids Drug Development, Guangzhou 510632, China.,Antisense Biopharmaceutical Technology Co., Ltd., Guangzhou 510632, China
| | - Juhua Yang
- Department of Biochemistry and Molecular Biology, Medical College of Jinan University, Guangzhou 510632, China.,Engineering Technology Research Center of Guangdong Province for Small Nucleic Acids Drug Development, Guangzhou 510632, China.,Antisense Biopharmaceutical Technology Co., Ltd., Guangzhou 510632, China
| | - Zhendong Li
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510632, Guangdong, China
| | - Keda Zhang
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, Guangdong, China
| | - Jia Fei
- Department of Biochemistry and Molecular Biology, Medical College of Jinan University, Guangzhou 510632, China.,Engineering Technology Research Center of Guangdong Province for Small Nucleic Acids Drug Development, Guangzhou 510632, China.,Antisense Biopharmaceutical Technology Co., Ltd., Guangzhou 510632, China
| |
Collapse
|
8
|
Yee SW, Buitrago D, Stecula A, Ngo HX, Chien HC, Zou L, Koleske ML, Giacomini KM. Deorphaning a solute carrier 22 family member, SLC22A15, through functional genomic studies. FASEB J 2020; 34:15734-15752. [PMID: 33124720 DOI: 10.1096/fj.202001497r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/10/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022]
Abstract
The human solute carrier 22A (SLC22A) family consists of 23 members, representing one of the largest families in the human SLC superfamily. Despite their pharmacological and physiological importance in the absorption and disposition of a range of solutes, eight SLC22A family members remain classified as orphans. In this study, we used a multifaceted approach to identify ligands of orphan SLC22A15. Ligands of SLC22A15 were proposed based on phylogenetic analysis and comparative modeling. The putative ligands were then confirmed by metabolomic screening and uptake assays in SLC22A15 transfected HEK293 cells. Metabolomic studies and transporter assays revealed that SLC22A15 prefers zwitterionic compounds over cations and anions. We identified eight zwitterions, including ergothioneine, carnitine, carnosine, gabapentin, as well as four cations, including MPP+ , thiamine, and cimetidine, as substrates of SLC22A15. Carnosine was a specific substrate of SLC22A15 among the transporters in the SLC22A family. SLC22A15 transport of several substrates was sodium-dependent and exhibited a higher Km for ergothioneine, carnitine, and carnosine compared to previously identified transporters for these ligands. This is the first study to characterize the function of SLC22A15. Our studies demonstrate that SLC22A15 may play an important role in determining the systemic and tissue levels of ergothioneine, carnosine, and other zwitterions.
Collapse
Affiliation(s)
- Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Dina Buitrago
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Adrian Stecula
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Huy X Ngo
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Huan-Chieh Chien
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Ling Zou
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Megan L Koleske
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA.,Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
9
|
Nishiyama M, Nakamichi N, Yoshimura T, Masuo Y, Komori T, Ishimoto T, Matsuo JI, Kato Y. Homostachydrine is a Xenobiotic Substrate of OCTN1/SLC22A4 and Potentially Sensitizes Pentylenetetrazole-Induced Seizures in Mice. Neurochem Res 2020; 45:2664-2678. [PMID: 32844295 DOI: 10.1007/s11064-020-03118-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/10/2020] [Accepted: 08/15/2020] [Indexed: 12/23/2022]
Abstract
Understanding of the underlying mechanism of epilepsy is desired since some patients fail to control their seizures. The carnitine/organic cation transporter OCTN1/SLC22A4 is expressed in brain neurons and transports food-derived antioxidant ergothioneine (ERGO), L-carnitine, and spermine, all of which may be associated with epilepsy. This study aimed to clarify the possible association of this transporter with epileptic seizures. In both pentylenetetrazole (PTZ)-induced acute seizure and kindling models, ocnt1 gene knockout mice (octn1-/-) showed lower seizure scores compared with wild-type mice. Up-regulation of the epilepsy-related genes, c-fos and Arc, and the neurotrophic factor BDNF following PTZ administration was observed in the hippocampus of wild-type, but not octn1-/- mice. To find the OCTN1 substrate associated with the seizure, untargeted metabolomics analysis using liquid chromatography-quadrupole time-of-flight mass spectrometry was conducted on extracts from the hippocampus, frontal cortex, and plasma of both strains, leading to the identification of a plant alkaloid homostachydrine as a compound present in a lower concentration in octn1-/- mice. OCTN1-mediated uptake of deuterium-labeled homostachydrine was confirmed in OCTN1-transfected HEK293 cells, suggesting that this compound is a substrate of OCTN1. Homostachydrine administration increased PTZ-induced acute seizure scores and the expression of Arc in the hippocampus and that of Arc, Egr1, and BDNF in the frontal cortex. Conversely, administration of the OCTN1 substrate/inhibitor ERGO inhibited PTZ-induced kindling and reduced the plasma homostachydrine concentration. Thus, these results suggest that OCTN1 is at least partially associated with PTZ-induced seizures, which is potentially deteriorated by treatment with homostachydrine, a newly identified food-derived OCTN1 substrate.
Collapse
Affiliation(s)
- Misa Nishiyama
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Noritaka Nakamichi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan. .,Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki, Gunma, 370-0033, Japan.
| | - Tomoyuki Yoshimura
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Yusuke Masuo
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Tomoe Komori
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Takahiro Ishimoto
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Jun-Ichi Matsuo
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| |
Collapse
|
10
|
Structural Characterization of an S-enantioselective Imine Reductase from Mycobacterium Smegmatis. Biomolecules 2020; 10:biom10081130. [PMID: 32751900 PMCID: PMC7465668 DOI: 10.3390/biom10081130] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/21/2020] [Accepted: 07/27/2020] [Indexed: 11/17/2022] Open
Abstract
NADPH-dependent imine reductases (IREDs) are enzymes capable of enantioselectively reducing imines to chiral secondary amines, which represent important building blocks in the chemical and pharmaceutical industry. Since their discovery in 2011, many previously unknown IREDs have been identified, biochemically and structurally characterized and categorized into families. However, the catalytic mechanism and guiding principles for substrate specificity and stereoselectivity remain disputed. Herein, we describe the crystal structure of S-IRED-Ms from Mycobacterium smegmatis together with its cofactor NADPH. S-IRED-Ms belongs to the S-enantioselective superfamily 3 (SFam3) and is the first IRED from SFam3 to be structurally described. The data presented provide further evidence for the overall high degree of structural conservation between different IREDs of various superfamilies. We discuss the role of Asp170 in catalysis and the importance of hydrophobic amino acids in the active site for stereospecificity. Moreover, a separate entrance to the active site, potentially functioning according to a gatekeeping mechanism regulating access and, therefore, substrate specificity is described.
Collapse
|
11
|
Pochini L, Pappacoda G, Galluccio M, Pastore F, Scalise M, Indiveri C. Effect of Cholesterol on the Organic Cation Transporter OCTN1 (SLC22A4). Int J Mol Sci 2020; 21:ijms21031091. [PMID: 32041338 PMCID: PMC7037232 DOI: 10.3390/ijms21031091] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/02/2020] [Accepted: 02/03/2020] [Indexed: 02/07/2023] Open
Abstract
The effect of cholesterol was investigated on the OCTN1 transport activity measured as [14C]-tetraethylamonium or [3H]-acetylcholine uptake in proteoliposomes reconstituted with native transporter extracted from HeLa cells or the human recombinant OCTN1 over-expressed in E. coli. Removal of cholesterol from the native transporter by MβCD before reconstitution led to impairment of transport activity. A similar activity impairment was observed after treatment of proteoliposomes harboring the recombinant (cholesterol-free) protein by MβCD, suggesting that the lipid mixture used for reconstitution contained some cholesterol. An enzymatic assay revealed the presence of 10 µg cholesterol/mg total lipids corresponding to 1% cholesterol in the phospholipid mixture used for the proteoliposome preparation. On the other way around, the activity of the recombinant OCTN1 was stimulated by adding the cholesterol analogue, CHS to the proteoliposome preparation. Optimal transport activity was detected in the presence of 83 µg CHS/ mg total lipids for both [14C]-tetraethylamonium or [3H]-acetylcholine uptake. Kinetic analysis of transport demonstrated that the stimulation of transport activity by CHS consisted in an increase of the Vmax of transport with no changes of the Km. Altogether, the data suggests a direct interaction of cholesterol with the protein. A further support to this interpretation was given by a docking analysis indicating the interaction of cholesterol with some protein sites corresponding to CARC-CRAC motifs. The observed direct interaction of cholesterol with OCTN1 points to a possible direct influence of cholesterol on tumor cells or on acetylcholine transport in neuronal and non-neuronal cells via OCTN1.
Collapse
|
12
|
Yee SW, Stecula A, Chien HC, Zou L, Feofanova EV, van Borselen M, Cheung KWK, Yousri NA, Suhre K, Kinchen JM, Boerwinkle E, Irannejad R, Yu B, Giacomini KM. Unraveling the functional role of the orphan solute carrier, SLC22A24 in the transport of steroid conjugates through metabolomic and genome-wide association studies. PLoS Genet 2019; 15:e1008208. [PMID: 31553721 PMCID: PMC6760779 DOI: 10.1371/journal.pgen.1008208] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/17/2019] [Indexed: 12/16/2022] Open
Abstract
Variation in steroid hormone levels has wide implications for health and disease. The genes encoding the proteins involved in steroid disposition represent key determinants of interindividual variation in steroid levels and ultimately, their effects. Beginning with metabolomic data from genome-wide association studies (GWAS), we observed that genetic variants in the orphan transporter, SLC22A24 were significantly associated with levels of androsterone glucuronide and etiocholanolone glucuronide (sentinel SNPs p-value <1x10-30). In cells over-expressing human or various mammalian orthologs of SLC22A24, we showed that steroid conjugates and bile acids were substrates of the transporter. Phylogenetic, genomic, and transcriptomic analyses suggested that SLC22A24 has a specialized role in the kidney and appears to function in the reabsorption of organic anions, and in particular, anionic steroids. Phenome-wide analysis showed that functional variants of SLC22A24 are associated with human disease such as cardiovascular diseases and acne, which have been linked to dysregulated steroid metabolism. Collectively, these functional genomic studies reveal a previously uncharacterized protein involved in steroid homeostasis, opening up new possibilities for SLC22A24 as a pharmacological target for regulating steroid levels.
Collapse
Affiliation(s)
- Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, California, United States of America
| | - Adrian Stecula
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, California, United States of America
| | - Huan-Chieh Chien
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, California, United States of America
| | - Ling Zou
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, California, United States of America
| | - Elena V. Feofanova
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Marjolein van Borselen
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, California, United States of America
| | - Kit Wun Kathy Cheung
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, California, United States of America
| | - Noha A. Yousri
- Genetic Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
- Computer and Systems Engineering, Alexandria University, Alexandria, Egypt
| | - Karsten Suhre
- Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha, Qatar
| | | | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Roshanak Irannejad
- The Cardiovascular Research Institute, University of California, San Francisco, California, United States of America
| | - Bing Yu
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Kathleen M. Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, California, United States of America
| |
Collapse
|