1
|
Su Z, Chen T, Liu X, Kang X. Size-tunable transmembrane nanopores assembled from decomposable molecular templates. Biosens Bioelectron 2025; 267:116780. [PMID: 39277918 DOI: 10.1016/j.bios.2024.116780] [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: 05/18/2024] [Revised: 09/03/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Transmembrane nanopores, as key elements in molecular transport and single-molecule sensors, are assembled naturally from multiple monomers in the presence of lipid bilayers. The nanopore size, especially the precise diameter of the inner space, determines its sensing targets and further biological application. In this paper, we introduce a template molecule-aided assembly strategy for constructing size-tunable transmembrane nanopores. Inspired by the barrel-like structure, similar to many transmembrane proteins, cyclodextrin molecules of different sizes are utilized as templates and modulators to assemble the α-helical barreled peptide of polysaccharide transporters (Wza). The functional nanopores assembled by this strategy possess high biological and chemical activity and can be inserted into lipid bilayers, forming stable single channels for single-molecule sensing. After enzyme digestion, the cyclodextrins on protein nanopores can be degraded, and the remaining nontemplate transmembrane protein nanopores can also preserve the integrity of their structure and function. The template molecule-aided assembly strategy employed a simple and convenient method for fully artificially synthesizing transmembrane protein nanopores; the pore size is completely dependent on the size of the template molecule and controllable, ranging from 1.1 to 1.8 nm. Furthermore, by chemically synthesized peptides and modifications, the pore function is easily modulated and does not involve the cumbersome genetic mutations of other biological techniques.
Collapse
Affiliation(s)
- Zhuoqun Su
- College of Chemistry & Materials Science, Key Laboratory of Synthetic and Natural Functional Molecular Chemistry, Northwest University, Xi'an, 710127, China; School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, 710021, China
| | - Tingting Chen
- College of Chemistry & Materials Science, Key Laboratory of Synthetic and Natural Functional Molecular Chemistry, Northwest University, Xi'an, 710127, China
| | - Xingtong Liu
- College of Chemistry & Materials Science, Key Laboratory of Synthetic and Natural Functional Molecular Chemistry, Northwest University, Xi'an, 710127, China
| | - Xiaofeng Kang
- College of Chemistry & Materials Science, Key Laboratory of Synthetic and Natural Functional Molecular Chemistry, Northwest University, Xi'an, 710127, China.
| |
Collapse
|
2
|
El-Ashmawy NE, Al-Ashmawy GM, Hamada OB, Khedr NF. The role of ABCG2 in health and disease: Linking cancer therapy resistance and other disorders. Life Sci 2025; 360:123245. [PMID: 39561874 DOI: 10.1016/j.lfs.2024.123245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/13/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024]
Abstract
All biological systems have adenosine triphosphate (ATP) binding cassette (ABC) transporters, one of the significant protein superfamilies involved in transport across membranes. ABC transporters have been implicated in the etiology of diseases like metabolic disorders, cancer, and Alzheimer's disease. ATP-binding cassette superfamily G member 2 (ABCG2), one of the ABC transporters, is necessary for the ATP-dependent efflux of several endogenous and exogenous substances. Consequently, it maintained cellular homeostasis and shielded tissue from xenobiotic substances. ABCG2 was initially identified in an Adriamycin-selected breast cancer cell line (MCF-7/AdrVp) and was linked to the emergence of multidrug resistance (MDR) in cancerous cells. Under many pathophysiological conditions, including inflammation, disease pathology, tissue injury, infection, and in response to xenobiotics and endogenous substances, the expression of ABCG2 undergoes alterations that result in modifications in its function and activity. Genetic variants in the ABCG2 transporter can potentially impact its expression and function, contributing to the development of many disorders. This review aimed to illustrate the impact of ABCG2 expression and its variants on oral drug bioavailability, MDR in specific cancer cells, explore the relationship between ABCG2 expression and other disorders such as gout, Alzheimer's disease, epilepsy, and erythropoietic protoporphyria, and demonstrate the influence of various synthetic and natural compounds in regulating ABCG2 expression.
Collapse
Affiliation(s)
- Nahla E El-Ashmawy
- Tanta University, Faculty of Pharmacy, Department of Biochemistry, Tanta Postal Code: 31527, Egypt; The British University in Egypt, Faculty of Pharmacy, Department of Pharmacology & Biochemistry, El Sherouk City, Cairo Postal Code: 11837, Egypt.
| | - Ghada M Al-Ashmawy
- Tanta University, Faculty of Pharmacy, Department of Biochemistry, Tanta Postal Code: 31527, Egypt; Alsalam University in Egypt, Faculty of Pharmacy, Department of Biochemistry, Kafr El Zayat, Egypt.
| | - Omnia B Hamada
- Tanta University, Faculty of Pharmacy, Department of Biochemistry, Tanta Postal Code: 31527, Egypt.
| | - Naglaa F Khedr
- Tanta University, Faculty of Pharmacy, Department of Biochemistry, Tanta Postal Code: 31527, Egypt.
| |
Collapse
|
3
|
Lakli M, Onnée M, Carrez T, Becq F, Falguières T, Fanen P. ABC transporters involved in respiratory and cholestatic diseases: From rare to very rare monogenic diseases. Biochem Pharmacol 2024; 229:116468. [PMID: 39111603 DOI: 10.1016/j.bcp.2024.116468] [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: 02/26/2024] [Revised: 07/16/2024] [Accepted: 08/03/2024] [Indexed: 08/24/2024]
Abstract
ATP-binding cassette (ABC) transporters constitute a 49-member superfamily in humans. These proteins, most of them being transmembrane, allow the active transport of an important variety of substrates across biological membranes, using ATP hydrolysis as an energy source. For an important proportion of these ABC transporters, genetic variations of the loci encoding them have been correlated with rare genetic diseases, including cystic fibrosis and interstitial lung disease (variations in CFTR/ABCC7 and ABCA3) as well as cholestatic liver diseases (variations in ABCB4 and ABCB11). In this review, we first describe these ABC transporters and how their molecular dysfunction may lead to human diseases. Then, we propose a classification of the genetic variants according to their molecular defect (expression, traffic, function and/or stability), which may be considered as a general guideline for all ABC transporters' variants. Finally, we discuss recent progress in the field of targeted pharmacotherapy, which aim to correct specific molecular defects using small molecules. In conclusion, we are opening the path to treatment repurposing for diseases involving similar deficiencies in other ABC transporters.
Collapse
Affiliation(s)
- Mounia Lakli
- Inserm, Université Paris-Saclay, Physiopathogenèse et traitement des maladies du foie, UMR_S 1193, Hepatinov, 91400 Orsay, France
| | - Marion Onnée
- Univ Paris Est Creteil, INSERM, IMRB, F-94010, Créteil, France
| | - Thomas Carrez
- Université de Poitiers, Laboratoire Physiopathologie et Régulation des Transports Ioniques, Pôle Biologie Santé, 86000 Poitiers, France; ManRos Therapeutics, Hôtel de Recherche, Centre de Perharidy, 29680, Roscoff, France
| | - Frédéric Becq
- Université de Poitiers, Laboratoire Physiopathologie et Régulation des Transports Ioniques, Pôle Biologie Santé, 86000 Poitiers, France
| | - Thomas Falguières
- Inserm, Université Paris-Saclay, Physiopathogenèse et traitement des maladies du foie, UMR_S 1193, Hepatinov, 91400 Orsay, France
| | - Pascale Fanen
- Univ Paris Est Creteil, INSERM, IMRB, F-94010, Créteil, France; AP-HP, Département de Génétique Médicale, Hôpital Henri Mondor, F-94010, Créteil, France.
| |
Collapse
|
4
|
De Vecchis D, Schäfer LV. Coupling the role of lipids to the conformational dynamics of the ABC transporter P-glycoprotein. Biophys J 2024; 123:2522-2536. [PMID: 38909280 PMCID: PMC11365111 DOI: 10.1016/j.bpj.2024.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/31/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024] Open
Abstract
The ATP-binding cassette transporter P-glycoprotein (P-gp) is a multidrug efflux pump that is overexpressed in a variety of cancers and associated with the drug-resistance phenomenon. P-gp structures were previously determined in detergent and in nanodiscs, in which different transmembrane helix conformations were found, "straight" and "kinked," respectively, indicating a possible role of the lipid environment on the P-gp structural ensemble. Here, we investigate the dynamic conformational ensembles and protein-lipid interactions of two human P-gp inward-open conformers, straight and kinked, employing all-atom molecular dynamics (MD) simulations in asymmetric multicomponent lipid bilayers that mimic the highly specialized hepatocyte membrane in which P-gp is expressed. The two conformers are found to differ in terms of the accessibility of the substrate cavity. The MD simulations show how cholesterol and different lipid species wedge, snorkel, and partially enter into the cavity of the straight P-gp conformer solved in detergent. However, access to the cavity of the kinked P-gp conformer solved in nanodiscs is restricted. Furthermore, the volume and dynamic fluctuations of the substrate cavity largely differ between the two P-gp conformers and are modulated by the presence (or absence) of cholesterol in the membrane and/or of ATP. From the mechanistic perspective, the findings indicate that the straight conformer likely precedes the kinked conformer in the functional working cycle of P-gp, with the latter conformation representing a post substrate-bound state. The inaccessibility of the main transmembrane cavity in the kinked conformer might be crucial in preventing substrate disengagement and transport withdrawal. Remarkably, in our unbiased MD simulations, one transmembrane helix (TM10) of the straight conformer underwent a spontaneous transition to a kinked conformation, underlining the relevance of both conformations in a native phospholipid environment and revealing structural descriptors defining the transition between the two P-gp conformers.
Collapse
Affiliation(s)
- Dario De Vecchis
- Center for Theoretical Chemistry, Ruhr University Bochum, Bochum, Germany.
| | - Lars V Schäfer
- Center for Theoretical Chemistry, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
5
|
Korn LL, Kutyavin VI, Bachtel ND, Medzhitov R. Adverse Food Reactions: Physiological and Ecological Perspectives. Annu Rev Nutr 2024; 44:155-178. [PMID: 38724028 DOI: 10.1146/annurev-nutr-061021-022909] [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] [Indexed: 05/15/2024]
Abstract
While food is essential for survival, it can also cause a variety of harmful effects, ranging from intolerance to specific nutrients to celiac disease and food allergies. In addition to nutrients, foods contain myriads of substances that can have either beneficial or detrimental effects on the animals consuming them. Consequently, all animals evolved defense mechanisms that protect them from harmful food components. These "antitoxin" defenses have some parallels with antimicrobial defenses and operate at a cost to the animal's fitness. These costs outweigh benefits when defense responses are exaggerated or mistargeted, resulting in adverse reactions to foods. Additionally, pathological effects of foods can stem from insufficient defenses, due to unabated toxicity of harmful food components. We discuss the structure of antitoxin defenses and how their failures can lead to a variety of adverse food reactions.
Collapse
Affiliation(s)
- Lisa L Korn
- Department of Medicine, Section of Rheumatology, Allergy, and Clinical Immunology, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - Vassily I Kutyavin
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - Nathaniel D Bachtel
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - Ruslan Medzhitov
- Tananbaum Center for Theoretical and Analytical Human Biology, Yale University School of Medicine, New Haven, Connecticut, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| |
Collapse
|
6
|
Lakli M, Dumont J, Vauthier V, Charton J, Crespi V, Banet M, Riahi Y, Ben Saad A, Mareux E, Lapalus M, Gonzales E, Jacquemin E, Di Meo F, Deprez B, Leroux F, Falguières T. Identification of new correctors for traffic-defective ABCB4 variants by a high-content screening approach. Commun Biol 2024; 7:898. [PMID: 39048674 PMCID: PMC11269752 DOI: 10.1038/s42003-024-06590-y] [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: 02/06/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024] Open
Abstract
ABCB4 is located at the canalicular membrane of hepatocytes and is responsible for the secretion of phosphatidylcholine into bile. Genetic variations of this transporter are correlated with rare cholestatic liver diseases, the most severe being progressive familial intrahepatic cholestasis type 3 (PFIC3). PFIC3 patients most often require liver transplantation. In this context of unmet medical need, we developed a high-content screening approach to identify small molecules able to correct ABCB4 molecular defects. Intracellularly-retained variants of ABCB4 were expressed in cell models and their maturation, cellular localization and function were analyzed after treatment with the molecules identified by high-content screening. In total, six hits were identified by high-content screening. Three of them were able to correct the maturation and canalicular localization of two distinct intracellularly-retained ABCB4 variants; one molecule was able to significantly restore the function of two ABCB4 variants. In addition, in silico molecular docking calculations suggest that the identified hits may interact with wild type ABCB4 residues involved in ATP binding/hydrolysis. Our results pave the way for their optimization in order to provide new drug candidates as potential alternative to liver transplantation for patients with severe forms of ABCB4-related diseases, including PFIC3.
Collapse
Affiliation(s)
- Mounia Lakli
- Inserm, Université Paris-Saclay, Physiopathogénèse et traitement des maladies du foie, UMR_S 1193, Hepatinov, F-91400, Orsay, France
| | - Julie Dumont
- Université de Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, F-59000, Lille, France
| | - Virginie Vauthier
- Inserm, Sorbonne Université, Centre de Recherche Saint-Antoine (CRSA), UMR_S 938, Institute of Cardiometabolism and Nutrition (ICAN), F-75012, Paris, France
| | - Julie Charton
- Université de Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Veronica Crespi
- Inserm, Université de Limoges, Pharmacology & Transplantation, UMR 1248, Centre de Biologie et Recherche en Santé, F-87000, Limoges, France
| | - Manon Banet
- Inserm, Université Paris-Saclay, Physiopathogénèse et traitement des maladies du foie, UMR_S 1193, Hepatinov, F-91400, Orsay, France
| | - Yosra Riahi
- Inserm, Université Paris-Saclay, Physiopathogénèse et traitement des maladies du foie, UMR_S 1193, Hepatinov, F-91400, Orsay, France
| | - Amel Ben Saad
- Inserm, Université Paris-Saclay, Physiopathogénèse et traitement des maladies du foie, UMR_S 1193, Hepatinov, F-91400, Orsay, France
| | - Elodie Mareux
- Inserm, Université Paris-Saclay, Physiopathogénèse et traitement des maladies du foie, UMR_S 1193, Hepatinov, F-91400, Orsay, France
| | - Martine Lapalus
- Inserm, Université Paris-Saclay, Physiopathogénèse et traitement des maladies du foie, UMR_S 1193, Hepatinov, F-91400, Orsay, France
| | - Emmanuel Gonzales
- Inserm, Université Paris-Saclay, Physiopathogénèse et traitement des maladies du foie, UMR_S 1193, Hepatinov, F-91400, Orsay, France
- Assistance Publique - Hôpitaux de Paris, Paediatric Hepatology & Paediatric Liver Transplant Department, Reference Center for Rare Paediatric Liver Diseases, FILFOIE, ERN RARE LIVER, Faculté de Médecine Paris-Saclay, CHU Bicêtre, F-94270, Le Kremlin-Bicêtre, France
| | - Emmanuel Jacquemin
- Inserm, Université Paris-Saclay, Physiopathogénèse et traitement des maladies du foie, UMR_S 1193, Hepatinov, F-91400, Orsay, France
- Assistance Publique - Hôpitaux de Paris, Paediatric Hepatology & Paediatric Liver Transplant Department, Reference Center for Rare Paediatric Liver Diseases, FILFOIE, ERN RARE LIVER, Faculté de Médecine Paris-Saclay, CHU Bicêtre, F-94270, Le Kremlin-Bicêtre, France
| | - Florent Di Meo
- Inserm, Université de Limoges, Pharmacology & Transplantation, UMR 1248, Centre de Biologie et Recherche en Santé, F-87000, Limoges, France
| | - Benoit Deprez
- Université de Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, F-59000, Lille, France
| | - Florence Leroux
- Université de Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for Living Systems, F-59000, Lille, France
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, F-59000, Lille, France
| | - Thomas Falguières
- Inserm, Université Paris-Saclay, Physiopathogénèse et traitement des maladies du foie, UMR_S 1193, Hepatinov, F-91400, Orsay, France.
| |
Collapse
|
7
|
Sostare E, Bowen TJ, Lawson TN, Freier A, Li X, Lloyd GR, Najdekr L, Jankevics A, Smith T, Varshavi D, Ludwig C, Colbourne JK, Weber RJM, Crizer DM, Auerbach SS, Bucher JR, Viant MR. Metabolomics Simultaneously Derives Benchmark Dose Estimates and Discovers Metabolic Biotransformations in a Rat Bioassay. Chem Res Toxicol 2024; 37:923-934. [PMID: 38842447 PMCID: PMC11187623 DOI: 10.1021/acs.chemrestox.4c00002] [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: 01/03/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/07/2024]
Abstract
Benchmark dose (BMD) modeling estimates the dose of a chemical that causes a perturbation from baseline. Transcriptional BMDs have been shown to be relatively consistent with apical end point BMDs, opening the door to using molecular BMDs to derive human health-based guidance values for chemical exposure. Metabolomics measures the responses of small-molecule endogenous metabolites to chemical exposure, complementing transcriptomics by characterizing downstream molecular phenotypes that are more closely associated with apical end points. The aim of this study was to apply BMD modeling to in vivo metabolomics data, to compare metabolic BMDs to both transcriptional and apical end point BMDs. This builds upon our previous application of transcriptomics and BMD modeling to a 5-day rat study of triphenyl phosphate (TPhP), applying metabolomics to the same archived tissues. Specifically, liver from rats exposed to five doses of TPhP was investigated using liquid chromatography-mass spectrometry and 1H nuclear magnetic resonance spectroscopy-based metabolomics. Following the application of BMDExpress2 software, 2903 endogenous metabolic features yielded viable dose-response models, confirming a perturbation to the liver metabolome. Metabolic BMD estimates were similarly sensitive to transcriptional BMDs, and more sensitive than both clinical chemistry and apical end point BMDs. Pathway analysis of the multiomics data sets revealed a major effect of TPhP exposure on cholesterol (and downstream) pathways, consistent with clinical chemistry measurements. Additionally, the transcriptomics data indicated that TPhP activated xenobiotic metabolism pathways, which was confirmed by using the underexploited capability of metabolomics to detect xenobiotic-related compounds. Eleven biotransformation products of TPhP were discovered, and their levels were highly correlated with multiple xenobiotic metabolism genes. This work provides a case study showing how metabolomics and transcriptomics can estimate mechanistically anchored points-of-departure. Furthermore, the study demonstrates how metabolomics can also discover biotransformation products, which could be of value within a regulatory setting, for example, as an enhancement of OECD Test Guideline 417 (toxicokinetics).
Collapse
Affiliation(s)
- Elena Sostare
- Michabo
Health Science Ltd., Union House, 111 New Union Street, Coventry CV1 2NT, U.K.
| | - Tara J. Bowen
- School
of Biosciences, University of Birmingham, Birmingham B15 2TT, U.K.
| | - Thomas N. Lawson
- Michabo
Health Science Ltd., Union House, 111 New Union Street, Coventry CV1 2NT, U.K.
| | - Anne Freier
- School
of Biosciences, University of Birmingham, Birmingham B15 2TT, U.K.
| | - Xiaojing Li
- School
of Biosciences, University of Birmingham, Birmingham B15 2TT, U.K.
| | - Gavin R. Lloyd
- Phenome
Centre Birmingham, University of Birmingham, Birmingham B15 2TT, U.K.
| | - Lukáš Najdekr
- Phenome
Centre Birmingham, University of Birmingham, Birmingham B15 2TT, U.K.
| | - Andris Jankevics
- Phenome
Centre Birmingham, University of Birmingham, Birmingham B15 2TT, U.K.
| | - Thomas Smith
- Phenome
Centre Birmingham, University of Birmingham, Birmingham B15 2TT, U.K.
| | - Dorsa Varshavi
- Phenome
Centre Birmingham, University of Birmingham, Birmingham B15 2TT, U.K.
| | - Christian Ludwig
- Phenome
Centre Birmingham, University of Birmingham, Birmingham B15 2TT, U.K.
| | - John K. Colbourne
- Michabo
Health Science Ltd., Union House, 111 New Union Street, Coventry CV1 2NT, U.K.
- School
of Biosciences, University of Birmingham, Birmingham B15 2TT, U.K.
| | - Ralf J. M. Weber
- Michabo
Health Science Ltd., Union House, 111 New Union Street, Coventry CV1 2NT, U.K.
- School
of Biosciences, University of Birmingham, Birmingham B15 2TT, U.K.
- Phenome
Centre Birmingham, University of Birmingham, Birmingham B15 2TT, U.K.
| | - David M. Crizer
- Division
of Translational Toxicology, National Institute
of Environmental Health Sciences, Research Triangle Park NC 27709, North Carolina, United
States
| | - Scott S. Auerbach
- Division
of Translational Toxicology, National Institute
of Environmental Health Sciences, Research Triangle Park NC 27709, North Carolina, United
States
| | - John R. Bucher
- Division
of Translational Toxicology, National Institute
of Environmental Health Sciences, Research Triangle Park NC 27709, North Carolina, United
States
| | - Mark R. Viant
- Michabo
Health Science Ltd., Union House, 111 New Union Street, Coventry CV1 2NT, U.K.
- School
of Biosciences, University of Birmingham, Birmingham B15 2TT, U.K.
- Phenome
Centre Birmingham, University of Birmingham, Birmingham B15 2TT, U.K.
| |
Collapse
|
8
|
Thévenod F, Lee WK. Cadmium transport by mammalian ATP-binding cassette transporters. Biometals 2024; 37:697-719. [PMID: 38319451 PMCID: PMC11101381 DOI: 10.1007/s10534-024-00582-5] [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: 09/15/2023] [Accepted: 01/04/2024] [Indexed: 02/07/2024]
Abstract
Cellular responses to toxic metals depend on metal accessibility to intracellular targets, reaching interaction sites, and the intracellular metal concentration, which is mainly determined by uptake pathways, binding/sequestration and efflux pathways. ATP-binding cassette (ABC) transporters are ubiquitous in the human body-usually in epithelia-and are responsible for the transfer of indispensable physiological substrates (e.g. lipids and heme), protection against potentially toxic substances, maintenance of fluid composition, and excretion of metabolic waste products. Derailed regulation and gene variants of ABC transporters culminate in a wide array of pathophysiological disease states, such as oncogenic multidrug resistance or cystic fibrosis. Cadmium (Cd) has no known physiological role in mammalians and poses a health risk due to its release into the environment as a result of industrial activities, and eventually passes into the food chain. Epithelial cells, especially within the liver, lungs, gastrointestinal tract and kidneys, are particularly susceptible to the multifaceted effects of Cd because of the plethora of uptake pathways available. Pertinent to their broad substrate spectra, ABC transporters represent a major cellular efflux pathway for Cd and Cd complexes. In this review, we summarize current knowledge concerning transport of Cd and its complexes (mainly Cd bound to glutathione) by the ABC transporters ABCB1 (P-glycoprotein, MDR1), ABCB6, ABCC1 (multidrug resistance related protein 1, MRP1), ABCC7 (cystic fibrosis transmembrane regulator, CFTR), and ABCG2 (breast cancer related protein, BCRP). Potential detoxification strategies underlying ABC transporter-mediated efflux of Cd and Cd complexes are discussed.
Collapse
Affiliation(s)
- Frank Thévenod
- Institute for Physiology, Pathophysiology and Toxicology & ZBAF, Witten/Herdecke University, 58453, Witten, Germany
- Physiology and Pathophysiology of Cells and Membranes, Medical School OWL, Bielefeld University, Morgenbreede 1, 33615, Bielefeld, Germany
| | - Wing-Kee Lee
- Physiology and Pathophysiology of Cells and Membranes, Medical School OWL, Bielefeld University, Morgenbreede 1, 33615, Bielefeld, Germany.
| |
Collapse
|
9
|
Maru K, Singh A, Jangir R, Jangir KK. Amyloid detection in neurodegenerative diseases using MOFs. J Mater Chem B 2024; 12:4553-4573. [PMID: 38646795 DOI: 10.1039/d4tb00373j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Neurodegenerative diseases (amyloid diseases such as Alzheimer's and Parkinson's), stemming from protein misfolding and aggregation, encompass a spectrum of disorders with severe systemic implications. Timely detection is pivotal in managing these diseases owing to their significant impact on organ function and high mortality rates. The diverse array of amyloid disorders, spanning localized and systemic manifestations, underscores the complexity of these conditions and highlights the need for advanced detection methods. Traditional approaches have focused on identifying biomarkers using imaging techniques (PET and MRI) or invasive procedures. However, recent efforts have focused on the use of metal-organic frameworks (MOFs), a versatile class of materials known for their unique properties, in revolutionizing amyloid disease detection. The high porosity, customizable structures, and biocompatibility of MOFs enable their integration with biomolecules, laying the groundwork for highly sensitive and specific biosensors. These sensors have been employed using electrochemical and photophysical techniques that target amyloid species under neurodegenerative conditions. The adaptability of MOFs allows for the precise detection and quantification of amyloid proteins, offering potential advancements in early diagnosis and disease management. This review article delves into how MOFs contribute to detecting amyloid diseases by categorizing their uses based on different sensing methods, such as electrochemical (EC), electrochemiluminescence (ECL), fluorescence, Förster resonance energy transfer (FRET), up-conversion luminescence resonance energy transfer (ULRET), and photoelectrochemical (PEC) sensing. The drawbacks of MOF biosensors and the challenges encountered in the field are also briefly explored from our perspective.
Collapse
Affiliation(s)
- Ketan Maru
- Sardar Vallabhbhai National Institute of Technology, Ichchanath, Surat-395 007, Gujarat, India.
| | - Amarendra Singh
- Sardar Vallabhbhai National Institute of Technology, Ichchanath, Surat-395 007, Gujarat, India.
| | - Ritambhara Jangir
- Sardar Vallabhbhai National Institute of Technology, Ichchanath, Surat-395 007, Gujarat, India.
| | | |
Collapse
|
10
|
Sarma H, Gogoi B, Guan CY, Yu CP. Nitro-PAHs: Occurrences, ecological consequences, and remediation strategies for environmental restoration. CHEMOSPHERE 2024; 356:141795. [PMID: 38548078 DOI: 10.1016/j.chemosphere.2024.141795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 12/24/2023] [Accepted: 03/23/2024] [Indexed: 04/12/2024]
Abstract
Nitrated polycyclic aromatic hydrocarbons (nitro-PAHs) are persistent pollutants that have been introduced into the environment as a result of human activities. They are produced when PAHs undergo oxidation and are highly resistant to degradation, resulting in prolonged exposure and significant health risks for wildlife and humans. Nitro-PAHs' potential to induce cancer and mutations has raised concerns about their harmful effects. Furthermore, their ability to accumulate in the food chain seriously threatens the ecosystem and human health. Moreover, nitro-PAHs can disrupt the normal functioning of the endocrine system, leading to reproductive and developmental problems in humans and other organisms. Reducing nitro-PAHs in the environment through source management, physical removal, and chemical treatment is essential to mitigate the associated environmental and human health risks. Recent studies have focused on improving nitro-PAHs' phytoremediation by incorporating microorganisms and biostimulants. Microbes can break down nitro-PAHs into less harmful substances, while biostimulants can enhance plant growth and metabolic activity. By combining these elements, the effectiveness of phytoremediation for nitro-PAHs can be increased. This study aimed to investigate the impact of introducing microbial and biostimulant agents on the phytoremediation process for nitro-PAHs and identify potential solutions for addressing the environmental risks associated with these pollutants.
Collapse
Affiliation(s)
- Hemen Sarma
- Bioremediation Technology Research Group, Department of Botany, Bodoland University, Rangalikhata, Deborgaon, Kokrajhar (BTR), Assam, 783370, India.
| | - Bhoirob Gogoi
- Bioremediation Technology Research Group, Department of Botany, Bodoland University, Rangalikhata, Deborgaon, Kokrajhar (BTR), Assam, 783370, India
| | - Chung-Yu Guan
- Department of Environmental Engineering, National Ilan University, Yilan, 260, Taiwan
| | - Chang-Ping Yu
- Graduate Institute of Environmental Engineering, National Taiwan University. B.S., Civil Engineering, National Taiwan University, Taiwan
| |
Collapse
|
11
|
Gandhi N, Wills L, Akers K, Su Y, Niccum P, Murali TM, Rajagopalan P. Comparative transcriptomic and phenotypic analysis of induced pluripotent stem cell hepatocyte-like cells and primary human hepatocytes. Cell Tissue Res 2024; 396:119-139. [PMID: 38369646 DOI: 10.1007/s00441-024-03868-9] [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: 02/13/2023] [Accepted: 01/22/2024] [Indexed: 02/20/2024]
Abstract
Primary human hepatocytes (PHHs) are used extensively for in vitro liver cultures to study hepatic functions. However, limited availability and invasive retrieval prevent their widespread use. Induced pluripotent stem cells exhibit significant potential since they can be obtained non-invasively and differentiated into hepatic lineages, such as hepatocyte-like cells (iHLCs). However, there are concerns about their fetal phenotypic characteristics and their hepatic functions compared to PHHs in culture. Therefore, we performed an RNA-sequencing (RNA-seq) analysis to understand pathways that are either up- or downregulated in each cell type. Analysis of the RNA-seq data showed an upregulation in the bile secretion pathway where genes such as AQP9 and UGT1A1 were higher expressed in PHHs compared to iHLCs by 455- and 15-fold, respectively. Upon immunostaining, bile canaliculi were shown to be present in PHHs. The TCA cycle in PHHs was upregulated compared to iHLCs. Cellular analysis showed a 2-2.5-fold increase in normalized urea production in PHHs compared to iHLCs. In addition, drug metabolism pathways, including cytochrome P450 (CYP450) and UDP-glucuronosyltransferase enzymes, were upregulated in PHHs compared to iHLCs. Of note, CYP2E1 gene expression was significantly higher (21,810-fold) in PHHs. Acetaminophen and ethanol were administered to PHH and iHLC cultures to investigate differences in biotransformation. CYP450 activity of baseline and toxicant-treated samples was significantly higher in PHHs compared to iHLCs. Our analysis revealed that iHLCs have substantial differences from PHHs in critical hepatic functions. These results have highlighted the differences in gene expression and hepatic functions between PHHs and iHLCs to motivate future investigation.
Collapse
Affiliation(s)
- Neeti Gandhi
- Department of Chemical Engineering, Virginia Tech, 333 Kelly Hall, Blacksburg, VA, 24061, USA
| | - Lauren Wills
- School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, USA
| | - Kyle Akers
- Genetics, Bioinformatics, and Computational Biology Ph.D. Program, Virginia Tech, Blacksburg, VA, USA
| | - Yiqi Su
- Department of Computer Science, Virginia Tech, Blacksburg, VA, USA
| | - Parker Niccum
- Genetics, Bioinformatics, and Computational Biology Ph.D. Program, Virginia Tech, Blacksburg, VA, USA
| | - T M Murali
- Department of Computer Science, Virginia Tech, Blacksburg, VA, USA
| | - Padmavathy Rajagopalan
- Department of Chemical Engineering, Virginia Tech, 333 Kelly Hall, Blacksburg, VA, 24061, USA.
- School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, USA.
| |
Collapse
|
12
|
Fan W, Shao K, Luo M. Structural View of Cryo-Electron Microscopy-Determined ATP-Binding Cassette Transporters in Human Multidrug Resistance. Biomolecules 2024; 14:231. [PMID: 38397468 PMCID: PMC10886794 DOI: 10.3390/biom14020231] [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: 12/19/2023] [Revised: 02/01/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
ATP-binding cassette (ABC) transporters, acting as cellular "pumps," facilitate solute translocation through membranes via ATP hydrolysis. Their overexpression is closely tied to multidrug resistance (MDR), a major obstacle in chemotherapy and neurological disorder treatment, hampering drug accumulation and delivery. Extensive research has delved into the intricate interplay between ABC transporter structure, function, and potential inhibition for MDR reversal. Cryo-electron microscopy has been instrumental in unveiling structural details of various MDR-causing ABC transporters, encompassing ABCB1, ABCC1, and ABCG2, as well as the recently revealed ABCC3 and ABCC4 structures. The newly obtained structural insight has deepened our understanding of substrate and drug binding, translocation mechanisms, and inhibitor interactions. Given the growing body of structural information available for human MDR transporters and their associated mechanisms, we believe it is timely to compile a comprehensive review of these transporters and compare their functional mechanisms in the context of multidrug resistance. Therefore, this review primarily focuses on the structural aspects of clinically significant human ABC transporters linked to MDR, with the aim of providing valuable insights to enhance the effectiveness of MDR reversal strategies in clinical therapies.
Collapse
Affiliation(s)
| | | | - Min Luo
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore 117543, Singapore; (W.F.); (K.S.)
| |
Collapse
|
13
|
Gao Y, Deng H, Zhao Y, Li M, Wang L, Zhang Y. Gene Expression of Abcc2 and Its Regulation by Chicken Xenobiotic Receptor. TOXICS 2024; 12:55. [PMID: 38251011 PMCID: PMC10818656 DOI: 10.3390/toxics12010055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/26/2023] [Accepted: 01/09/2024] [Indexed: 01/23/2024]
Abstract
Membrane transporter multidrug resistance-associated protein 2 (MRP2/Abcc2) exhibits high pharmaco-toxicological relevance because it exports multiple cytotoxic compounds from cells. However, no detailed information about the gene expression and regulation of MRP2 in chickens is yet available. Here, we sought to investigate the expression distribution of Abcc2 in different tissues of chicken and then determine whether Abcc2 expression is induced by chicken xenobiotic receptor (CXR). The bioinformatics analyses showed that MRP2 transporters have three transmembrane structural domains (MSDs) and two highly conserved nucleotide structural domains (NBDs), and a close evolutionary relationship with turkeys. Tissue distribution analysis indicated that Abcc2 was highly expressed in the liver, kidney, duodenum, and jejunum. When exposed to metyrapone (an agonist of CXR) and ketoconazole (an antagonist of CXR), Abcc2 expression was upregulated and downregulated correspondingly. We further confirmed that Abcc2 gene regulation is dependent on CXR, by overexpressing and interfering with CXR, respectively. We also demonstrated the induction of Abcc2 expression and the activity of ivermectin, with CXR being a likely mediator. Animal experiments demonstrated that metyrapone and ivermectin induced Abcc2 in the liver, kidney, and duodenum of chickens. Together, our study identified the gene expression of Abcc2 and its regulation by CXR in chickens, which may provide novel targets for the reasonable usage of veterinary drugs.
Collapse
Affiliation(s)
- Yanhong Gao
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.G.); (H.D.); (Y.Z.); (M.L.)
| | - Huacheng Deng
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.G.); (H.D.); (Y.Z.); (M.L.)
| | - Yuying Zhao
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.G.); (H.D.); (Y.Z.); (M.L.)
| | - Mei Li
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.G.); (H.D.); (Y.Z.); (M.L.)
| | - Liping Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China;
| | - Yujuan Zhang
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.G.); (H.D.); (Y.Z.); (M.L.)
| |
Collapse
|
14
|
Cavicchioli Azevedo V, Johnston CU, Kennedy CJ. Ivermectin Toxicokinetics in Rainbow Trout (Oncorhynchus mykiss) following P-glycoprotein Induction. ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2024; 86:58-72. [PMID: 38103085 DOI: 10.1007/s00244-023-01045-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2023]
Abstract
Alterations in ivermectin (IVM, 22,23-dihydro avermectin B1a+22,23-dihydro avermectin B1b) toxicokinetics following P-glycoprotein (P-gp) induction by clotrimazole (CTZ) were examined in rainbow trout (Oncorhynchus mykiss) to assess the potential importance of P-gp activity levels in xenobiotic distribution and kinetics in fish. Control and fish pretreated with CTZ (30 µmol/kg) were administered 175 µg/kg 3H-IVM into the caudal vasculature. At various time points (0.25, 0.5, 1, 3, 24, 48, 96, and 168 h) following injection, tissues (blood, liver, kidney, gill, intestines, brain [5 regions], eye, gonad and fat) were removed analyzed for IVM-derived radioactivity. IVM concentration declined in blood, liver, kidney and gill, and concentrations in other tissues remained constant over the sampling period. The highest measured concentrations were found in kidney, followed by liver, with the lowest values found in brain, eye and gonad. The highest % of the administered dose was found in the liver and kidney in the immediate hours post-administration, and in the intestines and fat at 24 h post-administration. P-gp induction by CTZ did not alter IVM distribution or any calculated toxicokinetic parameter (AUC, mean residence time, T1/2, clearance rate, volume of distribution), suggesting that P-gp induction may be limited or that P-gp plays a lesser role in xenobiotic kinetics in fish compared to mammals.
Collapse
Affiliation(s)
| | - Christina U Johnston
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC, Canada
| | - Christopher J Kennedy
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC, Canada.
| |
Collapse
|
15
|
Johnston CU, Kennedy CJ. Effects of the chemosensitizer verapamil on P-glycoprotein substrate efflux in rainbow trout hepatocytes. Comp Biochem Physiol C Toxicol Pharmacol 2024; 275:109763. [PMID: 37820937 DOI: 10.1016/j.cbpc.2023.109763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/29/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
The ATP-dependent membrane transporter P-glycoprotein (P-gp) is associated with resistance to a wide variety of chemical substrates, as well as the multi-drug resistance (MDR) phenotype in mammals. Less is known regarding P-gp's function and relevance in teleosts; this study expanded the range of known substrates and the inhibitory effects of a model chemosensitizer verapamil. The P-gp-mediated uptake and efflux dynamics of 5 known mammalian substrates (berberine, cortisol, doxorubicin, rhodamine 123 [R123], and vinorelbine) were examined in isolated rainbow trout (Oncorhynchus mykiss) hepatocytes with and without co-exposure to varying doses of verapamil. Initial substrate uptake rates (pmol/106 cells/min) varied widely and were in order: berberine (482 ± 94) > R123 (364 ± 67) > doxorubicin (158 ± 41) > cortisol (20.3 ± 5.9) > vinorelbine (15.3 ± 3.5). Initial efflux rates (pmol/106 cells/min) were highest in berberine (464 ± 110) > doxorubicin (341 ± 57) > R123 (106 ± 33) > cortisol (26.6 ± 6.1) > vinorelbine (9.0 ± 2.4). Transport of vinorelbine and R123 is verapamil sensitive, but verapamil had no effect on transport of berberine, cortisol, or doxorubicin. Cortisol and doxorubicin showed evidence of high P-gp affinity, thus displacing verapamil from their shared P-gp binding site. Cortisol, doxorubicin, R123, and vinorelbine transport by rainbow trout P-gp was confirmed, while berberine could not be confirmed or excluded as a substrate. Binding sites and affinities were similar between mammalian and trout P-gp for doxorubicin, R123, and vinorelbine, while fish P-gp had a higher affinity for cortisol than mammalian P-gp. This study demonstrated that the range of substrates, as well as binding sites and affinities, of fish P-gp are well-aligned with those in mammals.
Collapse
Affiliation(s)
- Christina U Johnston
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Christopher J Kennedy
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada.
| |
Collapse
|
16
|
Kumar H, Gupta NV, Jain R, Madhunapantula SV, Babu CS, Kesharwani SS, Dey S, Jain V. A review of biological targets and therapeutic approaches in the management of triple-negative breast cancer. J Adv Res 2023; 54:271-292. [PMID: 36791960 DOI: 10.1016/j.jare.2023.02.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/23/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a heterogeneous, aggressive phenotype of breast cancer with associated chemoresistance. The development of chemo- or radioresistance could be attributed to diverse tumor microenvironments, overexpression of membrane proteins (transporters), epigenetic changes, and alteration of the cell signaling pathways/genes associated with the development of cancer stem cells (CSCs). AIM OF REVIEW Due to the diverse and heterogeneous nature of TNBC, therapeutic response to the existing modalities offers limited scope and thus results in reccurance after therapy. To establish landmark therapeutic efficacy, a number of novel therapeutic modalities have been proposed. In addition, reversal of the resistance that developed during treatment may be altered by employing appropriate therapeutic modalities. This review aims to discuss the plethora of investigations carried out, which will help readers understand and make an appropriate choice of therapy directed toward complete elimination of TNBC. KEY SCIENTIFIC CONCEPTS OF REVIEW This manuscript addresses the major contributory factors from the tumor microenvironment that are responsible for the development of chemoresistance and poor prognosis. The associated cellular events and molecular mechanism-based therapeutic interventions have been explained in detail. Inhibition of ABC transporters, cell signaling pathways associated with CSCs, and epigenetic modification offers promising results in this regard. TNBC progression, invasion, metastasis and recurrence can also be inhibited by blocking multiple cell signaling pathways, targeting specific receptors/epigenetic targets, disrupting bioenergetics and generating reactive oxygen species (ROS).
Collapse
Affiliation(s)
- Hitesh Kumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - N Vishal Gupta
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - Rupshee Jain
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - SubbaRao V Madhunapantula
- Department of Biochemistry, Centre of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - C Saravana Babu
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | | | - Surajit Dey
- Roseman University of Health Sciences, College of Pharmacy, Henderson, NV, USA
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India.
| |
Collapse
|
17
|
Wu S, Yan M, Liang M, Yang W, Chen J, Zhou J. Supramolecular host-guest nanosystems for overcoming cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:805-827. [PMID: 38263983 PMCID: PMC10804391 DOI: 10.20517/cdr.2023.77] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/31/2023] [Accepted: 11/15/2023] [Indexed: 01/25/2024]
Abstract
Cancer drug resistance has become one of the main challenges for the failure of chemotherapy, greatly limiting the selection and use of anticancer drugs and dashing the hopes of cancer patients. The emergence of supramolecular host-guest nanosystems has brought the field of supramolecular chemistry into the nanoworld, providing a potential solution to this challenge. Compared with conventional chemotherapeutic platforms, supramolecular host-guest nanosystems can reverse cancer drug resistance by increasing drug uptake, reducing drug efflux, activating drugs, and inhibiting DNA repair. Herein, we summarize the research progress of supramolecular host-guest nanosystems for overcoming cancer drug resistance and discuss the future research direction in this field. It is hoped that this review will provide more positive references for overcoming cancer drug resistance and promoting the development of supramolecular host-guest nanosystems.
Collapse
Affiliation(s)
- Sha Wu
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, Liaoning, China
| | - Miaomiao Yan
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, Liaoning, China
| | - Minghao Liang
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, Liaoning, China
| | - Wenzhi Yang
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, Liaoning, China
| | - Jingyu Chen
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, Liaoning, China
| | - Jiong Zhou
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, Liaoning, China
- Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou 510632, Guangdong, China
| |
Collapse
|
18
|
Solan ME, Schackmuth B, Bruce ED, Pradhan S, Sayes CM, Lavado R. Effects of short-chain per- and polyfluoroalkyl substances (PFAS) on toxicologically relevant gene expression profiles in a liver-on-a-chip model. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 337:122610. [PMID: 37742859 DOI: 10.1016/j.envpol.2023.122610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/23/2023] [Accepted: 09/22/2023] [Indexed: 09/26/2023]
Abstract
Short-chain per- and polyfluoroalkyl substances (PFAS) are highly stable and widely used environmental contaminants that pose potential health risks to humans. Aggregating reliable mechanistic information for safety assessments necessitates physiologically relevant high-throughput screening approaches. Here, we demonstrated the utility of a liver-on-a-chip model to investigate the effects of five short-chain PFAS at low (1 nM) and high (1 μM) concentrations on toxicologically-relevant gene expression profiles using the QuantiGene® Plex Assay. We found that the short-chain PFAS tested in this study modulated the expression of ABCG2, a gene encoding for the breast cancer resistance protein (BCRP), with marked and significant upregulation (up to 4-fold) observed for all but one of the short-chain PFAS tested. PFBS and HFPO-DA repressed SLCO1B3 expression, a gene that encodes for an essential liver-specific organic anion transporter. High concentrations of PFBS, PFHxA, and PFHxS upregulated the expression of genes encCYP1A1,CYP2B6 and CYP2C19 with the same treatments resulting in the repression of the expression of the gene encoding CYP1A2. This dysregulation could have consequences for the clearance of endogenous compounds and xenobiotics. However, we acknowledge that increased expression of genes encoding for transporters and biotransformation enzymes may or may not indicate changes to their protein expression or activity. Overall, our study provides important insights into the effects of short-chain PFAS on liver function and their potential implications for human health. The use of the liver-on-a-chip model in combination with the QuantiGene® Plex Assay may be a valuable tool for future high-throughput screening and gene expression profiling in toxicology studies.
Collapse
Affiliation(s)
- Megan E Solan
- Department of Environmental Science, Baylor University, Waco, TX, 76798, USA
| | - Bennett Schackmuth
- Department of Environmental Science, Baylor University, Waco, TX, 76798, USA
| | - Erica D Bruce
- Department of Environmental Science, Baylor University, Waco, TX, 76798, USA
| | - Sahar Pradhan
- Department of Environmental Science, Baylor University, Waco, TX, 76798, USA
| | - Christie M Sayes
- Department of Environmental Science, Baylor University, Waco, TX, 76798, USA
| | - Ramon Lavado
- Department of Environmental Science, Baylor University, Waco, TX, 76798, USA.
| |
Collapse
|
19
|
Alhattab DM, Isaioglou I, Alshehri S, Khan ZN, Susapto HH, Li Y, Marghani Y, Alghuneim AA, Díaz-Rúa R, Abdelrahman S, Al-Bihani S, Ahmed F, Felimban RI, Alkhatabi H, Alserihi R, Abedalthagafi M, AlFadel A, Awidi A, Chaudhary AG, Merzaban J, Hauser CAE. Fabrication of a three-dimensional bone marrow niche-like acute myeloid Leukemia disease model by an automated and controlled process using a robotic multicellular bioprinting system. Biomater Res 2023; 27:111. [PMID: 37932837 PMCID: PMC10626721 DOI: 10.1186/s40824-023-00457-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/29/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a hematological malignancy that remains a therapeutic challenge due to the high incidence of disease relapse. To better understand resistance mechanisms and identify novel therapies, robust preclinical models mimicking the bone marrow (BM) microenvironment are needed. This study aimed to achieve an automated fabrication process of a three-dimensional (3D) AML disease model that recapitulates the 3D spatial structure of the BM microenvironment and applies to drug screening and investigational studies. METHODS To build this model, we investigated a unique class of tetramer peptides with an innate ability to self-assemble into stable hydrogel. An automated robotic bioprinting process was established to fabricate a 3D BM (niche-like) multicellular AML disease model comprised of leukemia cells and the BM's stromal and endothelial cellular fractions. In addition, monoculture and dual-culture models were also fabricated. Leukemia cell compatibility, functionalities (in vitro and in vivo), and drug assessment studies using our model were performed. In addition, RNAseq and gene expression analysis using TaqMan arrays were also performed on 3D cultured stromal cells and primary leukemia cells. RESULTS The selected peptide hydrogel formed a highly porous network of nanofibers with mechanical properties similar to the BM extracellular matrix. The robotic bioprinter and the novel quadruple coaxial nozzle enabled the automated fabrication of a 3D BM niche-like AML disease model with controlled deposition of multiple cell types into the model. This model supported the viability and growth of primary leukemic, endothelial, and stromal cells and recapitulated cell-cell and cell-ECM interactions. In addition, AML cells in our model possessed quiescent characteristics with improved chemoresistance attributes, resembling more the native conditions as indicated by our in vivo results. Moreover, the whole transcriptome data demonstrated the effect of 3D culture on enhancing BM niche cell characteristics. We identified molecular pathways upregulated in AML cells in our 3D model that might contribute to AML drug resistance and disease relapse. CONCLUSIONS Our results demonstrate the importance of developing 3D biomimicry models that closely recapitulate the in vivo conditions to gain deeper insights into drug resistance mechanisms and novel therapy development. These models can also improve personalized medicine by testing patient-specific treatments.
Collapse
Affiliation(s)
- Dana M Alhattab
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- KAUST Smart Health Initiative (KSHI), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Ioannis Isaioglou
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Salwa Alshehri
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Zainab N Khan
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Red Sea Research Center (RSRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Hepi H Susapto
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Yanyan Li
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Yara Marghani
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Arwa A Alghuneim
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Rubén Díaz-Rúa
- Core Laboratories, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Sherin Abdelrahman
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Shuroug Al-Bihani
- Core Laboratories, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Farid Ahmed
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Raed I Felimban
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Heba Alkhatabi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Hematology Research Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Raed Alserihi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Malak Abedalthagafi
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, USA
| | - AlShaibani AlFadel
- Division of Hematology, Stem Cell Transplantation & Cellular Therapy, Oncology Center, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Abdalla Awidi
- Cell Therapy Center, The University of Jordan, Amman, Jordan
- Medical School, The University of Jordan, Amman, Jordan
- Jordan University Hospital, Amman, Jordan
| | - Adeel Gulzar Chaudhary
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Jasmeen Merzaban
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Charlotte A E Hauser
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
- KAUST Smart Health Initiative (KSHI), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
- Red Sea Research Center (RSRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
| |
Collapse
|
20
|
Tóth Á, Crespi V, Janaszkiewicz A, Di Meo F. Computational and structural insights into the pre- and post-hydrolysis states of bovine multidrug resistance-associated protein 1. Basic Clin Pharmacol Toxicol 2023; 133:508-525. [PMID: 37038087 DOI: 10.1111/bcpt.13871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/12/2023]
Abstract
ATP-binding cassette C-family drug membrane transporters play an important role in local pharmacokinetics, that is, drug concentration in cellular compartments. From the structural point of view, only the bovine ortholog of the multidrug resistance-associated protein 1 (bMRP1) has been resolved. We here used μs-scaled molecular dynamics simulations to investigate the structure and dynamics of the bovine multidrug resistance-associated protein 1 in pre- and post-hydrolysis functional states. The present work aims to examine the slight but likely relevant structural differences between pre- and post-hydrolysis states of outward-facing conformations as well as the interactions between the multidrug resistance-associated protein 1 and the surrounding lipid bilayer. Global conformational dynamics show unfavourable extracellular opening associated with nucleotide-binding domain dimerization indicating that the post-hydrolysis state adopts a close-cleft conformation rather than an outward-open conformation. Our present simulations also highlight persistent interactions with annular cholesterol molecules and the expected active role of lipid bilayer in the allosteric communication between distant domains of the transporter.
Collapse
Affiliation(s)
- Ágota Tóth
- Inserm UMR 1248 Pharmacology & Transplantation, Univ. Limoges, Limoges, France
| | - Veronica Crespi
- Inserm UMR 1248 Pharmacology & Transplantation, Univ. Limoges, Limoges, France
| | | | - Florent Di Meo
- Inserm UMR 1248 Pharmacology & Transplantation, Univ. Limoges, Limoges, France
| |
Collapse
|
21
|
Fu T, Zeng S, Zheng Q, Zhu F. The Important Role of Transporter Structures in Drug Disposition, Efficacy, and Toxicity. Drug Metab Dispos 2023; 51:1316-1323. [PMID: 37295948 DOI: 10.1124/dmd.123.001275] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/27/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
The ATP-binding cassette (ABC) and solute carrier (SLC) transporters are critical determinants of drug disposition, clinical efficacy, and toxicity as they specifically mediate the influx and efflux of various substrates and drugs. ABC transporters can modulate the pharmacokinetics of many drugs via mediating the translocation of drugs across biologic membranes. SLC transporters are important drug targets involved in the uptake of a broad range of compounds across the membrane. However, high-resolution experimental structures have been reported for a very limited number of transporters, which limits the study of their physiologic functions. In this review, we collected structural information on ABC and SLC transporters and described the application of computational methods in structure prediction. Taking P-glycoprotein (ABCB1) and serotonin transporter (SLC6A4) as examples, we assessed the pivotal role of structure in transport mechanisms, details of ligand-receptor interactions, drug selectivity, the molecular mechanisms of drug-drug interactions, and differences caused by genetic polymorphisms. The data collected contributes toward safer and more effective pharmacological treatments. SIGNIFICANCE STATEMENT: The experimental structure of ATP-binding cassette and solute carrier transporters was collected, and the application of computational methods in structure prediction was described. P-glycoprotein and serotonin transporter were used as examples to reveal the pivotal role of structure in transport mechanisms, drug selectivity, the molecular mechanisms of drug-drug interactions, and differences caused by genetic polymorphisms.
Collapse
Affiliation(s)
- Tingting Fu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China (F.Z.); School of Pharmaceutical Sciences, Jilin University, Changchun, China (T.F., Q.Z.); College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (S.Z., F.Z.); and Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China (F.Z.)
| | - Su Zeng
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China (F.Z.); School of Pharmaceutical Sciences, Jilin University, Changchun, China (T.F., Q.Z.); College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (S.Z., F.Z.); and Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China (F.Z.)
| | - Qingchuan Zheng
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China (F.Z.); School of Pharmaceutical Sciences, Jilin University, Changchun, China (T.F., Q.Z.); College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (S.Z., F.Z.); and Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China (F.Z.)
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China (F.Z.); School of Pharmaceutical Sciences, Jilin University, Changchun, China (T.F., Q.Z.); College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China (S.Z., F.Z.); and Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China (F.Z.)
| |
Collapse
|
22
|
Romeo M, Dallio M, Federico A. Is it time to reconsider the MRP6 role in bile secretion and LPAC genetic signature? J Hepatol 2023; 79:e152-e154. [PMID: 37392836 DOI: 10.1016/j.jhep.2023.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 06/15/2023] [Indexed: 07/03/2023]
Affiliation(s)
- Mario Romeo
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Marcello Dallio
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy.
| | - Alessandro Federico
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
23
|
Badiee SA, Isu UH, Khodadadi E, Moradi M. The Alternating Access Mechanism in Mammalian Multidrug Resistance Transporters and Their Bacterial Homologs. MEMBRANES 2023; 13:568. [PMID: 37367772 PMCID: PMC10305233 DOI: 10.3390/membranes13060568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/23/2023] [Accepted: 05/27/2023] [Indexed: 06/28/2023]
Abstract
Multidrug resistance (MDR) proteins belonging to the ATP-Binding Cassette (ABC) transporter group play a crucial role in the export of cytotoxic drugs across cell membranes. These proteins are particularly fascinating due to their ability to confer drug resistance, which subsequently leads to the failure of therapeutic interventions and hinders successful treatments. One key mechanism by which multidrug resistance (MDR) proteins carry out their transport function is through alternating access. This mechanism involves intricate conformational changes that enable the binding and transport of substrates across cellular membranes. In this extensive review, we provide an overview of ABC transporters, including their classifications and structural similarities. We focus specifically on well-known mammalian multidrug resistance proteins such as MRP1 and Pgp (MDR1), as well as bacterial counterparts such as Sav1866 and lipid flippase MsbA. By exploring the structural and functional features of these MDR proteins, we shed light on the roles of their nucleotide-binding domains (NBDs) and transmembrane domains (TMDs) in the transport process. Notably, while the structures of NBDs in prokaryotic ABC proteins, such as Sav1866, MsbA, and mammalian Pgp, are identical, MRP1 exhibits distinct characteristics in its NBDs. Our review also emphasizes the importance of two ATP molecules for the formation of an interface between the two binding sites of NBD domains across all these transporters. ATP hydrolysis occurs following substrate transport and is vital for recycling the transporters in subsequent cycles of substrate transportation. Specifically, among the studied transporters, only NBD2 in MRP1 possesses the ability to hydrolyze ATP, while both NBDs of Pgp, Sav1866, and MsbA are capable of carrying out this reaction. Furthermore, we highlight recent advancements in the study of MDR proteins and the alternating access mechanism. We discuss the experimental and computational approaches utilized to investigate the structure and dynamics of MDR proteins, providing valuable insights into their conformational changes and substrate transport. This review not only contributes to an enhanced understanding of multidrug resistance proteins but also holds immense potential for guiding future research and facilitating the development of effective strategies to overcome multidrug resistance, thus improving therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | - Mahmoud Moradi
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA; (S.A.B.); (U.H.I.); (E.K.)
| |
Collapse
|
24
|
Yin Y, Xin Y, Zhang F, An D, Fan H, Qin M, Xia J, Xi T, Xiong J. Overcoming ABCB1-mediated multidrug resistance by transcription factor BHLHE40. Neoplasia 2023; 39:100891. [PMID: 36931039 PMCID: PMC10025992 DOI: 10.1016/j.neo.2023.100891] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 02/21/2023] [Accepted: 02/21/2023] [Indexed: 03/17/2023]
Abstract
Multidrug resistance (MDR) hinders treatment efficacy in cancer therapy. One typical mechanism contributing to MDR is the overexpression of permeability-glycoprotein (P-gp) encoded by ATP-binding cassette subfamily B member 1 (ABCB1). Basic helix-loop-helix family member e40 (BHLHE40) is a well-known transcription factor that has pleiotropic effects including the regulation of cancer-related processes. However, whether BHLHE40 regulates MDR is still unknown. Chromatin immunoprecipitation-seq study revealed BHLHE40 occupancy in the promoter of ABCB1 gene. Adriamycin (ADM)-resistant human chronic myeloid leukemia cells (K562/A) and human breast cancer cells (MCF-7/A) were established. BHLHE40 expression was downregulated in the ADM-resistant cell lines. Overexpression of BHLHE40 resensitized resistant cells to ADM, promoted cell apoptosis in vitro and suppressed tumor growth in vivo, whereas BHLHE40 knockdown induced resistance to ADM in parental cells. Moreover, we found that BHLHE40 regulated drug resistance by directly binding to the ABCB1 promoter (-1605 to -1597) and inactivating its transcription. In consistence, the expression of BHLHE40 was negatively correlated with ABCB1 in various cancer cells, while positively with cancer cell chemosensitivity and better prognosis of patients with breast cancer. The study reveals the role of BHLHE40 as a transcriptional suppressor on the expression of ABCB1, major ABC transporter in chemoresistance. The findings extend the function of BHLHE40 in tumor progression and provides a novel mechanism for the reversal of multidrug resistance.
Collapse
Affiliation(s)
- Yongmei Yin
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu 210009, China; Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Yu Xin
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu 210009, China
| | - Feng Zhang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu 210009, China
| | - Donghao An
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu 210009, China
| | - Hui Fan
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu 210009, China
| | - Mengyao Qin
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu 210009, China
| | - Jinxin Xia
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu 210009, China
| | - Tao Xi
- Research Center of Biotechnology, School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu 210009, China
| | - Jing Xiong
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
25
|
Danner L, Malard F, Valdes R, Olivier-Van Stichelen S. Non-Nutritive Sweeteners Acesulfame Potassium and Sucralose Are Competitive Inhibitors of the Human P-glycoprotein/Multidrug Resistance Protein 1 (PGP/MDR1). Nutrients 2023; 15:1118. [PMID: 36904118 PMCID: PMC10005754 DOI: 10.3390/nu15051118] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Non-nutritive sweeteners (NNS) are popular sugar replacements used in foods, beverages, and medications. Although NNS are considered safe by regulatory organizations, their effects on physiological processes such as detoxification are incompletely understood. Previous studies revealed that the NNS sucralose (Sucr) altered P-glycoprotein (PGP) expression in rat colon. We also demonstrated that early-life exposure to NNS Sucr and acesulfame potassium (AceK) compromises mouse liver detoxification. Building upon these initial discoveries, we investigated the impact of AceK and Sucr on the PGP transporter in human cells to assess whether NNS influence its key role in cellular detoxification and drug metabolism. We showed that AceK and Sucr acted as PGP inhibitors, competing for the natural substrate-binding pocket of PGP. Most importantly, this was observed after exposure to concentrations of NNS within expected levels from common foods and beverage consumption. This may suggest risks for NNS consumers, either when taking medications that require PGP as the primary detoxification transporter or during exposure to toxic compounds.
Collapse
Affiliation(s)
- Laura Danner
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Florian Malard
- INSERM U1212, CNRS UMR5320, ARNA Laboratory, University of Bordeaux, 33000 Bordeaux, France
| | - Raquel Valdes
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Stephanie Olivier-Van Stichelen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Obstetrics & Gynecology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
26
|
Tóth Á, Janaszkiewicz A, Crespi V, Di Meo F. On the interplay between lipids and asymmetric dynamics of an NBS degenerate ABC transporter. Commun Biol 2023; 6:149. [PMID: 36737455 PMCID: PMC9898250 DOI: 10.1038/s42003-023-04537-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
Multidrug resistance-associated proteins are ABC C-family exporters. They are crucial in pharmacology as they transport various substrates across membranes. However, the role of the degenerate nucleotide-binding site (NBS) remains unclear likewise the interplay with the surrounding lipid environment. Here, we propose a dynamic and structural overview of MRP1 from ca. 110 μs molecular dynamics simulations. ATP binding to NBS1 is likely maintained along several transport cycles. Asymmetric NBD behaviour is ensured by lower signal transduction from NBD1 to the rest of the protein owing to the absence of ball-and-socket conformation between NBD1 and coupling helices. Even though surrounding lipids play an active role in the allosteric communication between the substrate-binding pocket and NBDs, our results suggest that lipid composition has a limited impact, mostly by affecting transport kinetics. We believe that our work can be extended to other degenerate NBS ABC proteins and provide hints for deciphering mechanistic differences among ABC transporters.
Collapse
Affiliation(s)
- Ágota Tóth
- grid.9966.00000 0001 2165 4861Inserm U1248 Pharmacology & Transplantation, ΩHealth Institute—Univ. Limoges, 2 rue du Prof. Descottes, 87000 F Limoges, France
| | - Angelika Janaszkiewicz
- grid.9966.00000 0001 2165 4861Inserm U1248 Pharmacology & Transplantation, ΩHealth Institute—Univ. Limoges, 2 rue du Prof. Descottes, 87000 F Limoges, France
| | - Veronica Crespi
- grid.9966.00000 0001 2165 4861Inserm U1248 Pharmacology & Transplantation, ΩHealth Institute—Univ. Limoges, 2 rue du Prof. Descottes, 87000 F Limoges, France
| | - Florent Di Meo
- Inserm U1248 Pharmacology & Transplantation, ΩHealth Institute-Univ. Limoges, 2 rue du Prof. Descottes, 87000 F, Limoges, France.
| |
Collapse
|
27
|
Moore JM, Bell EL, Hughes RO, Garfield AS. ABC transporters: human disease and pharmacotherapeutic potential. Trends Mol Med 2023; 29:152-172. [PMID: 36503994 DOI: 10.1016/j.molmed.2022.11.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/24/2022] [Accepted: 11/01/2022] [Indexed: 12/12/2022]
Abstract
Adenosine triphosphate (ATP)-binding cassette (ABC) transporters are a 48-member superfamily of membrane proteins that actively transport a variety of biological substrates across lipid membranes. Their functional diversity defines an expansive involvement in myriad aspects of human biology. At least 21 ABC transporters underlie rare monogenic disorders, with even more implicated in the predisposition to and symptomology of common and complex diseases. Such broad (patho)physiological relevance places this class of proteins at the intersection of disease causation and therapeutic potential, underlining them as promising targets for drug discovery, as exemplified by the transformative CFTR (ABCC7) modulator therapies for cystic fibrosis. This review will explore the growing relevance of ABC transporters to human disease and their potential as small-molecule drug targets.
Collapse
|
28
|
Ivacaftor-Mediated Potentiation of ABCB4 Missense Mutations Affecting Critical Motifs of the NBDs: Repositioning Perspectives for Hepatobiliary Diseases. Int J Mol Sci 2023; 24:ijms24021236. [PMID: 36674751 PMCID: PMC9867378 DOI: 10.3390/ijms24021236] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/15/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
ABCB4 (ATP-binding cassette subfamily B member 4) is a hepatocanalicular floppase involved in biliary phosphatidylcholine (PC) secretion. Variations in the ABCB4 gene give rise to several biliary diseases, including progressive familial intrahepatic cholestasis type 3 (PFIC3), an autosomal recessive disease that can be lethal in the absence of liver transplantation. In this study, we investigated the effect and potential rescue of ten ABCB4 missense variations in NBD1:NBD2 homologous positions (Y403H/Y1043H, K435M/K1075M, E558K/E1200A, D564G/D1206G and H589Y/H1231Y) all localized at the conserved and functionally critical motifs of ABC transporters, six of which are mutated in patients. By combining structure analysis and in vitro studies, we found that all ten mutants were normally processed and localized at the canalicular membrane of HepG2 cells, but showed dramatically impaired PC transport activity that was significantly rescued by treatment with the clinically approved CFTR potentiator ivacaftor. Our results provide evidence that functional ABCB4 mutations are rescued by ivacaftor, paving the way for the repositioning of this potentiator for the treatment of selected patients with PFIC3 caused by mutations in the ATP-binding sites of ABCB4.
Collapse
|
29
|
Identification of potential M2 macrophage-associated diagnostic biomarkers in coronary artery disease. Biosci Rep 2022; 42:231928. [PMID: 36222281 DOI: 10.1042/bsr20221394] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/29/2022] [Accepted: 10/06/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND M2 macrophages have been reported to be important in the progression of coronary artery disease (CAD). Thus, the present study aims at exploring the diagnostic value of M2 macrophage-associated genes in CAD. METHODS Transcriptome profile of CAD and control samples were downloaded from Gene Expression Omnibus database. The proportion of immune cells was analyzed using cell type identification by estimating relative subsets of RNA transcripts. Weighted Gene Co-expression Network Analysis (WGCNA) was carried out to screen the relevant module associated with M2 macrophages. Differential CAD and control samples of expressed genes (DEGs) were identified by the limma R package. Functional enrichment analysis by means of the clusterProfiler R package. Least absolute shrinkage and selection operator (LASSO) and random forest (RF) algorithms were carried out to select signature genes. Receiver operating curves (ROC) were plotted to evaluate the diagnostic value of selected signature genes. The expressions of potential diagnostic markers were validated by RT-qPCR. The ceRNA network of diagnostic biomarkers was constructed via miRwalk and Starbase database. CMap database was used to screen candidate drugs in the treatment of CAD by targeting diagnostic biomarkers. RESULTS A total of 166 M2 macrophage-associated genes were identified by WGCNA. By intersecting those genes with 879 DEGs, 53 M2 macrophage-associated DEGs were obtained in the present study. By LASSO, RF, and ROC analyses, C1orf105, CCL22, CRYGB, FRK, GAP43, REG1P, CALB1, and PTPN21 were identified as potential diagnostic biomarkers. RT-qPCR showed the consistent expression patterns of diagnostic biomarkers between GEO dataset and clinical samples. Perhexiline, alimemazine and mecamylamine were found to be potential drugs in the treatment of CAD. CONCLUSION We identified eight M2 macrophage-associated diagnostic biomarkers and candidate drugs for the CAD treatment.
Collapse
|
30
|
Majsterek M, Wierzchowska-Opoka M, Makosz I, Kreczyńska L, Kimber-Trojnar Ż, Leszczyńska-Gorzelak B. Bile Acids in Intrahepatic Cholestasis of Pregnancy. Diagnostics (Basel) 2022; 12:2746. [PMID: 36359589 PMCID: PMC9688989 DOI: 10.3390/diagnostics12112746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/05/2022] [Accepted: 11/06/2022] [Indexed: 11/12/2023] Open
Abstract
Intrahepatic cholestasis of pregnancy (ICP) is the most common, reversible, and closely related to pregnancy condition characterized by elevated levels of bile acids (BAs) in blood serum and an increased risk of adverse perinatal outcomes. Due to the complex interactions between the mother and the fetus in metabolism and transplacental BAs transport, ICP is classified as a fetal-maternal disease. The disease is usually mild in pregnant women, but it can be fatal to the fetus, leading to numerous complications, including intrauterine death. The pathophysiology of the disease is based on inflammatory mechanisms caused by elevated BA levels. Although ICP cannot be completely prevented, its early diagnosis and prompt management significantly reduce the risk of fetal complications, the most serious of which is unexpected intrauterine death. It is worth emphasizing that all diagnostics and management of ICP during pregnancy are based on BA levels. Therefore, it is important to standardize the criteria for diagnosis, as well as recommendations for management depending on the level of BAs, which undoubtedly determines the impact on the fetus. The purpose of this review is to present the potential and importance of BAs in the detection and rules of medical procedure in ICP.
Collapse
Affiliation(s)
| | | | | | | | - Żaneta Kimber-Trojnar
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland
| | | |
Collapse
|
31
|
Ghanem CI, Manautou JE. Role and Regulation of Hepatobiliary ATP-Binding Cassette Transporters during Chemical-Induced Liver Injury. Drug Metab Dispos 2022; 50:1376-1388. [PMID: 35914951 PMCID: PMC9513844 DOI: 10.1124/dmd.121.000450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/20/2022] [Indexed: 11/22/2022] Open
Abstract
Severity of drug-induced liver injury (DILI) ranges from mild, asymptomatic, and transient elevations in liver function tests to irreversible liver damage, often needing transplantation. Traditionally, DILI is classified mechanistically as high-frequency intrinsic DILI, commonly dose dependent or DILI that rarely occurs and is idiosyncratic in nature. This latter form is not dose dependent and has a pattern of histopathological manifestation that is not always uniform. Currently, a third type of DILI called indirect hepatotoxicity has been described that is associated with the pharmacological action of the drug. Historically, DILI was primarily linked to drug metabolism events; however, the impact of transporter-mediated rates of drug uptake and excretion has gained greater prominence in DILI research. This review provides a comprehensive view of the major findings from studies examining the contribution of hepatic ATP-binding cassette transporters as key contributors to DILI and how changes in their expression and function influence the development, severity, and overall toxicity outcome. SIGNIFICANCE STATEMENT: Drug-induced liver injury (DILI) continues to be a focal point in drug development research. ATP-binding cassette (ABC) transporters have emerged as important determinants of drug detoxification, disposition, and safety. This review article provides a comprehensive analysis of the literature addressing: (a) the role of hepatic ABC transporters in DILI, (b) the influence of genetic mutations in ABC transporters on DILI, and (c) new areas of research emphasis, such as the influence of the gut microbiota and epigenetic regulation, on ABC transporters.
Collapse
Affiliation(s)
- Carolina I Ghanem
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET) (C.I.G.) and Cátedra de Fisiopatología (C.I.G.), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina; and Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (J.E.M.)
| | - Jose E Manautou
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET) (C.I.G.) and Cátedra de Fisiopatología (C.I.G.), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina; and Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (J.E.M.)
| |
Collapse
|
32
|
Revisiting Quantification of Phenylalanine/Tyrosine Flux in the Ochronotic Pathway during Long-Term Nitisinone Treatment of Alkaptonuria. Metabolites 2022; 12:metabo12100920. [PMID: 36295821 PMCID: PMC9610527 DOI: 10.3390/metabo12100920] [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: 08/17/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Changes in the phenylalanine (PHE)/tyrosine (TYR) pathway metabolites before and during homogentisic acid (HGA)-lowering by nitisinone in the Suitability of Nitisinone in Alkaptonuria (AKU) 2 (SONIA 2) study enabled the magnitude of the flux in the pathway to be examined. SONIA 2 was a 48-month randomised, open-label, evaluator-blinded, parallel-group study performed in the UK, France and Slovakia recruiting patients with confirmed AKU to receive either 10 mg nitisinone or no treatment. Site visits were performed at 3 months and yearly thereafter. Results from history, photographs of eyes/ears, whole body scintigraphy, echocardiography and abdomen/pelvis ultrasonography were combined to produce the Alkaptonuria Severity Score Index (cAKUSSI). PHE, TYR, hydroxyphenylpyruvate (HPPA), hydroxyphenyllactate (HPLA) and HGA metabolites were analysed by liquid chromatography/tandem mass spectrometry in 24 h urine and serum samples collected before and during nitisinone. Serum metabolites were corrected for total body water (TBW), and the sum of 24 h urine plus total body water metabolites of PHE, TYR, HPPA, HPLA and HGA were determined. The sum of urine metabolites (PHE, TYR, HPPA, HPLA and HGA) were similar pre- and peri-nitisinone. The sum of TBW metabolites and sum TBW + URINE metabolites were significantly higher peri-nitisinone (p < 0.001 for both) compared with pre-nitisinone baseline. Significantly higher concentrations of metabolites from the tyrosine metabolic pathway were observed during treatment with nitisinone. Arguments for unmasking of the ochronotic pathway and biliary elimination of HGA are put forward.
Collapse
|
33
|
Mohi-Ud-Din R, Mir RH, Mir PA, Banday N, Shah AJ, Sawhney G, Bhat MM, Batiha GE, Pottoo FH, Pottoo FH. Dysfunction of ABC Transporters at the Surface of BBB: Potential Implications in Intractable Epilepsy and Applications of Nanotechnology Enabled Drug Delivery. Curr Drug Metab 2022; 23:735-756. [PMID: 35980054 DOI: 10.2174/1389200223666220817115003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/10/2022] [Accepted: 05/31/2022] [Indexed: 01/05/2023]
Abstract
Epilepsy is a chronic neurological disorder affecting 70 million people globally. One of the fascinating attributes of brain microvasculature is the (BBB), which controls a chain of distinct features that securely regulate the molecules, ions, and cells movement between the blood and the parenchyma. The barrier's integrity is of paramount importance and essential for maintaining brain homeostasis, as it offers both physical and chemical barriers to counter pathogens and xenobiotics. Dysfunction of various transporters in the (BBB), mainly ATP binding cassette (ABC), is considered to play a vital role in hampering the availability of antiepileptic drugs into the brain. ABC (ATP-binding cassette) transporters constitute a most diverse protein superfamily, which plays an essential part in various biological processes, including cell homeostasis, cell signaling, uptake of nutrients, and drug metabolism. Moreover, it plays a crucial role in neuroprotection by out-flowing various internal and external toxic substances from the interior of a cell, thus decreasing their buildup inside the cell. In humans, forty-eight ABC transporters have been acknowledged and categorized into subfamilies A to G based on their phylogenetic analysis. ABC subfamilies B, C, and G, impart a vital role at the BBB in guarding the brain against the entrance of various xenobiotic and their buildup. The illnesses of the central nervous system have received a lot of attention lately Owing to the existence of the BBB, the penetration effectiveness of most CNS medicines into the brain parenchyma is very limited (BBB). In the development of neurological therapies, BBB crossing for medication delivery to the CNS continues to be a major barrier. Nanomaterials with BBB cross ability have indeed been extensively developed for the treatment of CNS diseases due to their advantageous properties. This review will focus on multiple possible factors like inflammation, oxidative stress, uncontrolled recurrent seizures, and genetic polymorphisms that result in the deregulation of ABC transporters in epilepsy and nanotechnology-enabled delivery across BBB in epilepsy.
Collapse
Affiliation(s)
- Roohi Mohi-Ud-Din
- Department of General Medicine, Sher-I-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, Jammu & Kashmir, 190011, India.,Department of Pharmaceutical Sciences, School of Applied Sciences & Technology, University of Kashmir, Hazratbal, Srinagar-190006, Jammu & Kashmir, India
| | - Reyaz Hassan Mir
- Pharmaceutical Chemistry Division, Chandigarh College of Pharmacy, Landran, Punjab-140301, India.,Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir, India
| | - Prince Ahad Mir
- Department of Pharmaceutical Sciences, Khalsa College of Pharmacy, G.T. Road, Amritsar-143002, Punjab, India
| | - Nazia Banday
- Department of Pharmaceutical Sciences, School of Applied Sciences & Technology, University of Kashmir, Hazratbal, Srinagar-190006, Jammu & Kashmir, India
| | - Abdul Jalil Shah
- Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Kashmir, Hazratbal, Srinagar-190006, Kashmir, India
| | - Gifty Sawhney
- Inflammation Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-Tawi, Jammu 180001, India
| | - Mudasir Maqbool Bhat
- Department of Pharmaceutical Sciences, Pharmacy Practice Division, University of Kashmir, Hazratbal, Srinagar-190006, Jammu & Kashmir, India
| | - Gaber E Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, 31441, Dammam, Saudi Arabia
| |
Collapse
|
34
|
ABC Transporters in Human Diseases: Future Directions and Therapeutic Perspectives. Int J Mol Sci 2022; 23:ijms23084250. [PMID: 35457067 PMCID: PMC9028344 DOI: 10.3390/ijms23084250] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 01/27/2023] Open
|
35
|
Li H, Kou B, Yuan Y, Chai Y, Yuan R. Porous Fe 3O 4@COF-Immobilized gold nanoparticles with excellent catalytic performance for sensitive electrochemical detection of ATP. Biosens Bioelectron 2022; 197:113758. [PMID: 34798499 DOI: 10.1016/j.bios.2021.113758] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 12/20/2022]
Abstract
In this work, a "signal-off" electrochemical biosensor was established for sensitive detection of adenosine triphosphate (ATP) based on Fe3O4@covalent organic framework-immobilized gold nanoparticles (Fe3O4@COF-Au NPs) porous composite material as a nanocarrier. The proposed Fe3O4@COF-Au NPs could effectively confine Au NPs in the uniform channels of the Fe3O4@COF, which successfully avoided Au NPs aggregation to a certain extent and provided a comparatively independent and stable micro-environment via its hydrophobic porous nanochannels, thereby owning excellent electro-catalytic performance for the reduction of 4-nitrophenol. Moreover, the Fe3O4@COF-Au NPs nanomaterials were served as functional platform for immobilizing DNA substrate (S0), which was used to bind with the conversion product (S1) of the target ATP for subsequent branched hybridization chain reaction (b-HCR) to form dendritic DNA strands to hinder electron transfer between Fe3O4@COF-Au NPs and 4-nitrophenol, finally achieving sensitive detection of ATP with a wide linear range of 5 pM-50 μM and a low detection limit of 1.6 pM. Such strategy provides a multifunctional immobilized platform for the sensitive detection of ATP and a versatile strategy for monitoring other biomolecules.
Collapse
Affiliation(s)
- Hao Li
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Beibei Kou
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Yali Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Yaqin Chai
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China.
| | - Ruo Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China.
| |
Collapse
|
36
|
Gertzen CGW, Gohlke H, Häussinger D, Herebian D, Keitel V, Kubitz R, Mayatepek E, Schmitt L. The many facets of bile acids in the physiology and pathophysiology of the human liver. Biol Chem 2021; 402:1047-1062. [PMID: 34049433 DOI: 10.1515/hsz-2021-0156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/14/2021] [Indexed: 12/12/2022]
Abstract
Bile acids perform vital functions in the human liver and are the essential component of bile. It is therefore not surprising that the biology of bile acids is extremely complex, regulated on different levels, and involves soluble and membrane receptors as well as transporters. Hereditary disorders of these proteins manifest in different pathophysiological processes that result in liver diseases of varying severity. In this review, we summarize our current knowledge of the physiology and pathophysiology of bile acids with an emphasis on recently established analytical approaches as well as the molecular mechanisms that underlie signaling and transport of bile acids. In this review, we will focus on ABC transporters of the canalicular membrane and their associated diseases. As the G protein-coupled receptor, TGR5, receives increasing attention, we have included aspects of this receptor and its interaction with bile acids.
Collapse
Affiliation(s)
- Christoph G W Gertzen
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Center for Structural Studies (CSS), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Holger Gohlke
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- John von Neumann Institute for Computing (NIC), Jülich Supercomputing Centre (JSC), Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Institute of Bio- and Geosciences (IBG-4: Bioinformatics), Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Dieter Häussinger
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Diran Herebian
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Verena Keitel
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Ralf Kubitz
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Lutz Schmitt
- Institute of Biochemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
37
|
RAB10 Interacts with ABCB4 and Regulates Its Intracellular Traffic. Int J Mol Sci 2021; 22:ijms22137087. [PMID: 34209301 PMCID: PMC8268348 DOI: 10.3390/ijms22137087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/11/2022] Open
Abstract
ABCB4 (ATP-binding cassette subfamily B member 4) is an ABC transporter expressed at the canalicular membrane of hepatocytes where it ensures phosphatidylcholine secretion into bile. Genetic variations of ABCB4 are associated with several rare cholestatic diseases. The available treatments are not efficient for a significant proportion of patients with ABCB4-related diseases and liver transplantation is often required. The development of novel therapies requires a deep understanding of the molecular mechanisms regulating ABCB4 expression, intracellular traffic, and function. Using an immunoprecipitation approach combined with mass spectrometry analyses, we have identified the small GTPase RAB10 as a novel molecular partner of ABCB4. Our results indicate that the overexpression of wild type RAB10 or its dominant-active mutant significantly increases the amount of ABCB4 at the plasma membrane expression and its phosphatidylcholine floppase function. Contrariwise, RAB10 silencing induces the intracellular retention of ABCB4 and then indirectly diminishes its secretory function. Taken together, our findings suggest that RAB10 regulates the plasma membrane targeting of ABCB4 and consequently its capacity to mediate phosphatidylcholine secretion.
Collapse
|
38
|
Ben Saad A, Vauthier V, Tóth Á, Janaszkiewicz A, Durand-Schneider AM, Bruneau A, Delaunay JL, Lapalus M, Mareux E, Garcin I, Gonzales E, Housset C, Aït-Slimane T, Jacquemin E, Di Meo F, Falguières T. Effect of CFTR correctors on the traffic and the function of intracellularly retained ABCB4 variants. Liver Int 2021; 41:1344-1357. [PMID: 33650203 DOI: 10.1111/liv.14839] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/25/2021] [Accepted: 02/23/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIM ABCB4 is expressed at the canalicular membrane of hepatocytes. This ATP-binding cassette (ABC) transporter is responsible for the secretion of phosphatidylcholine into bile canaliculi. Missense genetic variations of ABCB4 are correlated with several rare cholestatic liver diseases, the most severe being progressive familial intrahepatic cholestasis type 3 (PFIC3). In a repurposing strategy to correct intracellularly retained ABCB4 variants, we tested 16 compounds previously validated as cystic fibrosis transmembrane conductance regulator (CFTR) correctors. METHODS The maturation, intracellular localization and activity of intracellularly retained ABCB4 variants were analyzed in cell models after treatment with CFTR correctors. In addition, in silico molecular docking calculations were performed to test the potential interaction of CFTR correctors with ABCB4. RESULTS We observed that the correctors C10, C13, and C17, as well as the combinations of C3 + C18 and C4 + C18, allowed the rescue of maturation and canalicular localization of four distinct traffic-defective ABCB4 variants. However, such treatments did not permit a rescue of the phosphatidylcholine secretion activity of these defective variants and were also inhibitory of the activity of wild type ABCB4. In silico molecular docking analyses suggest that these CFTR correctors might directly interact with transmembrane domains and/or ATP-binding sites of the transporter. CONCLUSION Our results illustrate the uncoupling between the traffic and the activity of ABCB4 because the same molecules can rescue the traffic of defective variants while they inhibit the secretion activity of the transporter. We expect that this study will help to design new pharmacological tools with potential clinical interest.
Collapse
Affiliation(s)
- Amel Ben Saad
- Inserm, Université Paris-Saclay, Physiopathogénèse et traitement des maladies du foie, UMR_S 1193, Orsay, France.,Inserm, Sorbonne Université, Centre de Recherche Saint-Antoine (CRSA), UMR_S 938, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Virginie Vauthier
- Inserm, Sorbonne Université, Centre de Recherche Saint-Antoine (CRSA), UMR_S 938, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France.,Université de Paris, Institut Cochin, Inserm U1016, CNRS UMR 8104, Paris, France
| | - Ágota Tóth
- Inserm, Université de Limoges, UMR 1248 IPPRITT, Limoges, France
| | | | - Anne-Marie Durand-Schneider
- Inserm, Sorbonne Université, Centre de Recherche Saint-Antoine (CRSA), UMR_S 938, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Alix Bruneau
- Inserm, Sorbonne Université, Centre de Recherche Saint-Antoine (CRSA), UMR_S 938, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France.,Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jean-Louis Delaunay
- Inserm, Sorbonne Université, Centre de Recherche Saint-Antoine (CRSA), UMR_S 938, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Martine Lapalus
- Inserm, Université Paris-Saclay, Physiopathogénèse et traitement des maladies du foie, UMR_S 1193, Orsay, France
| | - Elodie Mareux
- Inserm, Université Paris-Saclay, Physiopathogénèse et traitement des maladies du foie, UMR_S 1193, Orsay, France
| | - Isabelle Garcin
- Inserm, Université Paris-Saclay, Physiopathogénèse et traitement des maladies du foie, UMR_S 1193, Orsay, France
| | - Emmanuel Gonzales
- Inserm, Université Paris-Saclay, Physiopathogénèse et traitement des maladies du foie, UMR_S 1193, Orsay, France.,Assistance Publique - Hôpitaux de Paris, CHU Bicêtre, Paediatric Hepatology & Paediatric Liver Transplant Department, Reference Center for Rare Paediatric Liver Diseases, FILFOIE, ERN Rare-Liver, Faculté de Médecine Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Chantal Housset
- Inserm, Sorbonne Université, Centre de Recherche Saint-Antoine (CRSA), UMR_S 938, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France.,Assistance Publique - Hôpitaux de Paris, Hôpital Saint-Antoine, Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis, FILFOIE, ERN Rare-Liver, Paris, France
| | - Tounsia Aït-Slimane
- Inserm, Sorbonne Université, Centre de Recherche Saint-Antoine (CRSA), UMR_S 938, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Emmanuel Jacquemin
- Inserm, Université Paris-Saclay, Physiopathogénèse et traitement des maladies du foie, UMR_S 1193, Orsay, France.,Assistance Publique - Hôpitaux de Paris, CHU Bicêtre, Paediatric Hepatology & Paediatric Liver Transplant Department, Reference Center for Rare Paediatric Liver Diseases, FILFOIE, ERN Rare-Liver, Faculté de Médecine Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Florent Di Meo
- Inserm, Université de Limoges, UMR 1248 IPPRITT, Limoges, France
| | - Thomas Falguières
- Inserm, Université Paris-Saclay, Physiopathogénèse et traitement des maladies du foie, UMR_S 1193, Orsay, France
| |
Collapse
|
39
|
Prescher M, Bonus M, Stindt J, Keitel-Anselmino V, Smits SHJ, Gohlke H, Schmitt L. Evidence for a credit-card-swipe mechanism in the human PC floppase ABCB4. Structure 2021; 29:1144-1155.e5. [PMID: 34107287 DOI: 10.1016/j.str.2021.05.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/27/2021] [Accepted: 05/17/2021] [Indexed: 10/21/2022]
Abstract
ABCB4 is described as an ATP-binding cassette (ABC) transporter that primarily transports lipids of the phosphatidylcholine (PC) family but is also capable of translocating a subset of typical multidrug-resistance-associated drugs. The high degree of amino acid identity of 76% for ABCB4 and ABCB1, which is a prototype multidrug-resistance-mediating protein, results in ABCB4's second subset of substrates, which overlap with ABCB1's substrates. This often leads to incomplete annotations of ABCB4, in which it was described as exclusively PC-lipid specific. When the hydrophilic amino acids from ABCB4 are changed to the analogous but hydrophobic ones from ABCB1, the stimulation of ATPase activity by 1,2-dioleoyl-sn-glycero-3-phosphocholine, as a prime example of PC lipids, is strongly diminished, whereas the modulation capability of ABCB1 substrates remains unchanged. This indicates two distinct and autonomous substrate binding sites in ABCB4.
Collapse
Affiliation(s)
- Martin Prescher
- Institute of Biochemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Michele Bonus
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jan Stindt
- Clinic for Gastroenterology, Hepatology and Infectious Diseases University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Verena Keitel-Anselmino
- Clinic for Gastroenterology, Hepatology and Infectious Diseases University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sander H J Smits
- Institute of Biochemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Center for Structural Studies, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Holger Gohlke
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; John von Neumann Institute for Computing (NIC), Jülich Supercomputing Centre (JSC), Institute of Biological Information Processing (IBI-7: Structural Biochemistry) and Institute of Bio- and Geosciences (IBG-4: Bioinformatics), Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Lutz Schmitt
- Institute of Biochemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
40
|
Kroll T, Smits SHJ, Schmitt L. Monomeric bile acids modulate the ATPase activity of detergent-solubilized ABCB4/MDR3. J Lipid Res 2021; 62:100087. [PMID: 34022183 PMCID: PMC8233136 DOI: 10.1016/j.jlr.2021.100087] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/28/2021] [Accepted: 05/10/2021] [Indexed: 12/19/2022] Open
Abstract
ABCB4, also called multidrug-resistant protein 3 (MDR3), is an ATP binding cassette transporter located in the canalicular membrane of hepatocytes that specifically translocates phosphatidylcholine (PC) lipids from the cytoplasmic to the extracellular leaflet. Due to the harsh detergent effect of bile acids, PC lipids provided by ABCB4 are extracted into the bile. While it is well known that bile acids are the major extractor of PC lipids from the membrane into bile, it is unknown whether only PC lipid extraction is improved or whether bile acids also have a direct effect on ABCB4. Using in vitro experiments, we investigated the modulation of ATP hydrolysis of ABC by different bile acids commonly present in humans. We demonstrated that all tested bile acids stimulated ATPase activity except for taurolithocholic acid, which inhibited ATPase activity due to its hydrophobic nature. Additionally, we observed a nearly linear correlation between the critical micelle concentration and maximal stimulation by each bile acid, and that this modulation was maintained in the presence of PC lipids. This study revealed a large effect of 24-nor-ursodeoxycholic acid, suggesting a distinct mode of regulation of ATPase activity compared with other bile acids. In addition, it sheds light on the molecular cross talk of canalicular ABC transporters of the human liver.
Collapse
Affiliation(s)
- Tim Kroll
- Institute of Biochemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sander H J Smits
- Institute of Biochemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Lutz Schmitt
- Institute of Biochemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
41
|
Alav I, Kobylka J, Kuth MS, Pos KM, Picard M, Blair JMA, Bavro VN. Structure, Assembly, and Function of Tripartite Efflux and Type 1 Secretion Systems in Gram-Negative Bacteria. Chem Rev 2021; 121:5479-5596. [PMID: 33909410 PMCID: PMC8277102 DOI: 10.1021/acs.chemrev.1c00055] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Indexed: 12/11/2022]
Abstract
Tripartite efflux pumps and the related type 1 secretion systems (T1SSs) in Gram-negative organisms are diverse in function, energization, and structural organization. They form continuous conduits spanning both the inner and the outer membrane and are composed of three principal components-the energized inner membrane transporters (belonging to ABC, RND, and MFS families), the outer membrane factor channel-like proteins, and linking the two, the periplasmic adaptor proteins (PAPs), also known as the membrane fusion proteins (MFPs). In this review we summarize the recent advances in understanding of structural biology, function, and regulation of these systems, highlighting the previously undescribed role of PAPs in providing a common architectural scaffold across diverse families of transporters. Despite being built from a limited number of basic structural domains, these complexes present a staggering variety of architectures. While key insights have been derived from the RND transporter systems, a closer inspection of the operation and structural organization of different tripartite systems reveals unexpected analogies between them, including those formed around MFS- and ATP-driven transporters, suggesting that they operate around basic common principles. Based on that we are proposing a new integrated model of PAP-mediated communication within the conformational cycling of tripartite systems, which could be expanded to other types of assemblies.
Collapse
Affiliation(s)
- Ilyas Alav
- Institute
of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Jessica Kobylka
- Institute
of Biochemistry, Biocenter, Goethe Universität
Frankfurt, Max-von-Laue-Straße 9, D-60438 Frankfurt, Germany
| | - Miriam S. Kuth
- Institute
of Biochemistry, Biocenter, Goethe Universität
Frankfurt, Max-von-Laue-Straße 9, D-60438 Frankfurt, Germany
| | - Klaas M. Pos
- Institute
of Biochemistry, Biocenter, Goethe Universität
Frankfurt, Max-von-Laue-Straße 9, D-60438 Frankfurt, Germany
| | - Martin Picard
- Laboratoire
de Biologie Physico-Chimique des Protéines Membranaires, CNRS
UMR 7099, Université de Paris, 75005 Paris, France
- Fondation
Edmond de Rothschild pour le développement de la recherche
Scientifique, Institut de Biologie Physico-Chimique, 75005 Paris, France
| | - Jessica M. A. Blair
- Institute
of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Vassiliy N. Bavro
- School
of Life Sciences, University of Essex, Colchester, CO4 3SQ United Kingdom
| |
Collapse
|
42
|
Molecular Regulation of Canalicular ABC Transporters. Int J Mol Sci 2021; 22:ijms22042113. [PMID: 33672718 PMCID: PMC7924332 DOI: 10.3390/ijms22042113] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 12/17/2022] Open
Abstract
The ATP-binding cassette (ABC) transporters expressed at the canalicular membrane of hepatocytes mediate the secretion of several compounds into the bile canaliculi and therefore play a key role in bile secretion. Among these transporters, ABCB11 secretes bile acids, ABCB4 translocates phosphatidylcholine and ABCG5/G8 is responsible for cholesterol secretion, while ABCB1 and ABCC2 transport a variety of drugs and other compounds. The dysfunction of these transporters leads to severe, rare, evolutionary biliary diseases. The development of new therapies for patients with these diseases requires a deep understanding of the biology of these transporters. In this review, we report the current knowledge regarding the regulation of canalicular ABC transporters' folding, trafficking, membrane stability and function, and we highlight the role of molecular partners in these regulating mechanisms.
Collapse
|