1
|
Sorkina T, Bagalkot T, Cheng MH, Guthrie DA, Newman AH, Watkins SC, Sorkin A. Monoamine transporter ubiquitination and inward-open conformation synergistically maximize transporter endocytosis. SCIENCE ADVANCES 2024; 10:eadq9793. [PMID: 39576869 PMCID: PMC11584022 DOI: 10.1126/sciadv.adq9793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024]
Abstract
Monoamine transporters function in neuronal membranes to control extracellular concentrations of their substrates. Cell-surface expression of transporters is regulated by substrates and intracellular signaling, but the underlying mechanisms remain unclear. Here, we found that substrates of the dopamine transporter (DAT), amphetamine and dopamine, synergize with protein kinase C (PKC)-dependent DAT ubiquitination to markedly elevate clathrin-mediated endocytosis of DAT, which is accompanied by DAT movement out of plasma membrane protrusions with a negative curvature. Disruption of the outward-open (OO) DAT conformation or its stabilization in the inward-open (IO) conformation recapitulates substrate effects on DAT endocytosis. Amphetamine strongly increases PKC-dependent endocytosis of norepinephrine transporter (NET) but not of serotonin transporter (SERT), correlating with a substantially weaker ubiquitination of SERT compared to NET. We propose a "shape-transition" model whereby shifting from convex-shaped OO conformers to IO conformers minimizes retention of transporters in negatively curved membranes, which facilitates their PKC-dependent ubiquitination and recruitment to positively invaginated clathrin-coated membranes, driving robust transporter endocytosis.
Collapse
Affiliation(s)
- Tatiana Sorkina
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tarique Bagalkot
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mary Hongying Cheng
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Daryl A Guthrie
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute of Drug Abuse-Intramural Research Program, Baltimore, MD, USA
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute of Drug Abuse-Intramural Research Program, Baltimore, MD, USA
| | - Simon C Watkins
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alexander Sorkin
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Ma CIJ, Steinfeld N, Wang WA, Maxfield FR. A high-content microscopy drug screening platform for regulators of the extracellular digestion of lipoprotein aggregates by macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615160. [PMID: 39605493 PMCID: PMC11601252 DOI: 10.1101/2024.09.26.615160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The recruitment of macrophages to the intima of arteries is a critical event in atherosclerotic progression. These macrophages accumulate excessive lipid droplets and become "foam cells", a hallmark of atherosclerosis. Most studies focus on lipid accumulation through macrophage interaction with modified monomeric low-density lipoprotein (LDL). However, in the intima, macrophages predominantly encounter aggregated LDL (agLDL), an interaction that has been studied far less. Macrophages digest agLDL and generate free cholesterol in an extracellular, acidic, hydrolytic compartment. They form a tight seal around agLDL through actin polymerization and deliver lysosomal contents into this space in a process termed digestive exophagy. There is some evidence that inhibiting digestive exophagy to slow cholesterol accumulation in macrophages protects them from becoming foam cells and slows the progression of atherosclerotic lesions. Thus, understanding the mechanisms of digestive exophagy is critical. Here, we describe a high-content microscopy screen on a library of repurposed drugs for compounds that inhibit lysosome exocytosis during digestive exophagy. We identified many hit compounds and further characterized the effects that five of these compounds have on various aspects of digestive exophagy. In addition, three of the five compounds do not inhibit oxidized LDL-induced foam cell formation, indicating the two pathways to foam cell formation can be targeted independently. We demonstrate that this high-content screening platform has great potential as a drug discovery tool with the ability to effectively and efficiently screen for modulators of digestive exophagy.
Collapse
Affiliation(s)
- Cheng-I J Ma
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Noah Steinfeld
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Weixiang A Wang
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | | |
Collapse
|
3
|
Kurtyka M, Wessely F, Bau S, Ifie E, He L, de Wit NM, Pedersen ABV, Keller M, Webber C, de Vries HE, Ansorge O, Betsholtz C, De Bock M, Chaves C, Brodin B, Nielsen MS, Neuhaus W, Bell RD, Letoha T, Meyer AH, Leparc G, Lenter M, Lesuisse D, Cader ZM, Buckley ST, Loryan I, Pietrzik CU. The solute carrier SLC7A1 may act as a protein transporter at the blood-brain barrier. Eur J Cell Biol 2024; 103:151406. [PMID: 38547677 DOI: 10.1016/j.ejcb.2024.151406] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/02/2024] [Accepted: 03/20/2024] [Indexed: 06/29/2024] Open
Abstract
Despite extensive research, targeted delivery of substances to the brain still poses a great challenge due to the selectivity of the blood-brain barrier (BBB). Most molecules require either carrier- or receptor-mediated transport systems to reach the central nervous system (CNS). These transport systems form attractive routes for the delivery of therapeutics into the CNS, yet the number of known brain endothelium-enriched receptors allowing the transport of large molecules into the brain is scarce. Therefore, to identify novel BBB targets, we combined transcriptomic analysis of human and murine brain endothelium and performed a complex screening of BBB-enriched genes according to established selection criteria. As a result, we propose the high-affinity cationic amino acid transporter 1 (SLC7A1) as a novel candidate for transport of large molecules across the BBB. Using RNA sequencing and in situ hybridization assays, we demonstrated elevated SLC7A1 gene expression in both human and mouse brain endothelium. Moreover, we confirmed SLC7A1 protein expression in brain vasculature of both young and aged mice. To assess the potential of SLC7A1 as a transporter for larger proteins, we performed internalization and transcytosis studies using a radiolabelled or fluorophore-labelled anti-SLC7A1 antibody. Our results showed that SLC7A1 internalised a SLC7A1-specific antibody in human colorectal carcinoma (HCT116) cells. Moreover, transcytosis studies in both immortalised human brain endothelial (hCMEC/D3) cells and primary mouse brain endothelial cells clearly demonstrated that SLC7A1 effectively transported the SLC7A1-specific antibody from luminal to abluminal side. Therefore, here in this study, we present for the first time the SLC7A1 as a novel candidate for transport of larger molecules across the BBB.
Collapse
Affiliation(s)
- Magdalena Kurtyka
- Institute for Pathobiochemistry, University Medical Center Mainz, Mainz, Germany
| | - Frank Wessely
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Sarah Bau
- Pathology & Imaging, Novo Nordisk A/S, Måløv, Denmark
| | - Eseoghene Ifie
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Nienke M de Wit
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam, the Netherlands; Amsterdam Neuroscience, Amsterdam, the Netherlands
| | | | - Maximilian Keller
- Institute for Pathobiochemistry, University Medical Center Mainz, Mainz, Germany
| | - Caleb Webber
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Helga E de Vries
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam, the Netherlands; Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Olaf Ansorge
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden; Department of Medicine (Huddinge), Karolinska Institutet, Huddinge, Sweden
| | - Marijke De Bock
- Neuroscience Discovery, Janssen Research & Development, Janssen Pharmaceutica, Beerse, Belgium
| | - Catarina Chaves
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, Chilly Mazarin, France
| | - Birger Brodin
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | - Morten S Nielsen
- Department of Biomedicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Winfried Neuhaus
- Austrian Institute of Technology GmbH, Vienna, Austria; Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, Krems, Austria
| | | | | | - Axel H Meyer
- AbbVie Deutschland GmbH & Co. KG, Quantitative, Translational & ADME Sciences, Ludwigshafen, Germany
| | - Germán Leparc
- Boehringer Ingelheim Pharma GmbH & Co. KG, Translational Medicine & Clinical Pharmacology, Biberach, Germany
| | - Martin Lenter
- Boehringer Ingelheim Pharma GmbH & Co. KG, Drug Discovery Sciences, Biberach, Germany
| | - Dominique Lesuisse
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, Chilly Mazarin, France
| | - Zameel M Cader
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | | | - Irena Loryan
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Claus U Pietrzik
- Institute for Pathobiochemistry, University Medical Center Mainz, Mainz, Germany.
| |
Collapse
|
4
|
Ho HMK, Day RM, Craig DQM. An Investigation into the Effects of Processing Factors on the Properties and Scaling-Up Potential of Propranolol-Loaded Chitosan Nanogels. Pharmaceutics 2024; 16:662. [PMID: 38794324 PMCID: PMC11125439 DOI: 10.3390/pharmaceutics16050662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/05/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Chitosan-triphosphate (TPP) nanogels are widely studied drug delivery carrier systems, typically prepared via a simple mixing process. However, the effects of the processing factors on nanogel production have not been extensively explored, despite the importance of understanding and standardising such factors to allow upscaling and commercial usage. This study aims to systematically evaluate the effects of various fabrication and processing factors on the properties of nanogels using a Design of Experiment approach. Hydrodynamic size, polydispersity index (PDI), zeta potential, and encapsulation efficiency were determined as the dependent factors. The temperature, stirring rate, chitosan grade, crosslinker choice, and the interaction term between temperature and chitosan grade were found to have a significant effect on the particle size, whereas the effect of temperature and the addition rate of crosslinker on the PDI was also noteworthy. Moreover, the addition rate of the crosslinker and the volume of the reaction vessel were found to impact the encapsulation efficiency. The zeta potential of the nanogels was found to be governed by the chitosan grade. The optimal fabrication conditions for the development of medium molecular weight chitosan and TPP nanogels included the following: the addition rate for TPP solution was set at 2 mL/min, while the solution was then stirred at a temperature of 50 °C and a stirring speed of 600 rpm. The volume of the glass vial used was 28 mL, while the stirrer size was 20 mm. The second aim of the study was to evaluate the potential for scaling up the nanogels. Size and PDI were found to increase from 128 nm to 151 nm and from 0.232 to 0.267, respectively, when the volume of the reaction mixture was increased from 4 to 20 mL and other processing factors were kept unchanged. These results indicate that caution is required when scaling up as the nanogel properties may be significantly altered with an increasing production scale.
Collapse
Affiliation(s)
- Hei Ming Kenneth Ho
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
- Centre for Precision Healthcare, UCL Division of Medicine, University College London, 5 University Street, London WC1E 6JF, UK
| | - Richard M. Day
- Centre for Precision Healthcare, UCL Division of Medicine, University College London, 5 University Street, London WC1E 6JF, UK
| | - Duncan Q. M. Craig
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
- Faculty of Science, University of Bath, Claverton Down, Bath BA2 7AY, UK
| |
Collapse
|
5
|
Koe JC, Parker SJ. The posttranslational regulation of amino acid transporters is critical for their function in the tumor microenvironment. Curr Opin Biotechnol 2024; 85:103022. [PMID: 38056204 DOI: 10.1016/j.copbio.2023.103022] [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: 09/09/2023] [Revised: 10/28/2023] [Accepted: 11/06/2023] [Indexed: 12/08/2023]
Abstract
Amino acid transporters (AATs) facilitate nutrient uptake and nutrient exchange between cancer and stromal cells. The posttranslational modification (PTM) of transporters is an important mechanism that tumor-associated cells use to dynamically regulate their function and stability in response to microenvironmental cues. In this review, we summarize recent findings that demonstrate the significance of N-glycosylation, phosphorylation, and ubiquitylation for the function of AATs. We also highlight powerful approaches that hijack the PTM machinery that could be used as therapeutics or tools to modulate transporter activity.
Collapse
Affiliation(s)
- Jessica C Koe
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC, Canada; Centre for Molecular Medicine and Therapeutics, Vancouver, BC, Canada
| | - Seth J Parker
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC, Canada; Centre for Molecular Medicine and Therapeutics, Vancouver, BC, Canada; British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada.
| |
Collapse
|
6
|
Sun LM, Yu B, Luo YH, Zheng P, Huang Z, Yu J, Mao X, Yan H, Luo J, He J. Effect of small peptide chelated iron on growth performance, immunity and intestinal health in weaned pigs. Porcine Health Manag 2023; 9:32. [PMID: 37420289 DOI: 10.1186/s40813-023-00327-9] [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: 02/21/2023] [Accepted: 06/21/2023] [Indexed: 07/09/2023] Open
Abstract
BACKGROUND Small peptide chelated iron (SPCI), a novel iron supplementation in pig diets, owns growth-enhancing characteristics. Although a number of researches have been performed, there is no clear-cut evidence to show the exact relationship between the dose and effects of small peptide chelated minerals. Therefore, we investigated the effect of dietary supplementation of SPCI at different doses in the growth performance, immunity, and intestinal health in weaned pigs. METHODS Thirty weaned pigs were randomly assigned into five groups and feed with basal diet or the basal diet containing 50, 75, 100, or 125 mg/kg Fe as SPCI diets. The experiment lasted for 21 d and on day 22, blood samples were collected 1 h later. The tissue and intestinal mucosa samples were collected following. RESULTS Our results showed that the feed to gain ratio (F:G) decreased with different levels of SPCI addition (P < 0.05). The average daily gain (ADG) (P < 0.05) and digestibility of crude protein (P < 0.01) decreased with 125 mg/kg SPCI addition. With dietary different levels of SPCI addition, the serum concentrations of ferritin (quadratic, P < 0.001), transferrin (quadratic, P < 0.001), iron content in liver (quadratic, P < 0.05), gallbladder (quadratic, P < 0.01) and fecal (quadratic, P < 0.01) increased quadraticly. While the iron content in tibia (P < 0.01) increased by 100 mg/kg SPCI supplementation. Dietary 75 mg/kg SPCI addition increased the serum insulin-like growth factor I (IGF-I) (P < 0.01) and SPCI (75 ~ 100 mg/kg) addition also increased the serum content of IgA (P < 0.01). The serum concentrations of IgG (quadratic, P < 0.05) and IgM (quadratic, P < 0.01) increased quadraticly by different levels of SPCI supplementation. Moreover, different levels of SPCI supplementation decreased the serum concentration of D-lactic acid (P < 0.01). The serum glutathione peroxidase (GSH-Px) (P < 0.01) elevated but the malondialdehyde (MDA) (P < 0.05) decreased by 100 mg/kg SPCI addition. Interestingly, SPCI supplementation at 75 ~ 100 mg/kg improved the intestinal morphology and barrier function, as suggested by enhanced villus height (P < 0.01) and villus height/crypt depth (V/C) (P < 0.01) in duodenum, as well as jejunum epithelium tight-junction protein ZO-1 (P < 0.01). Moreover, SPCI supplementation at 75 ~ 100 mg/kg increased the activity of duodenal lactase (P < 0.01), jejunal sucrase (P < 0.01) and ileal maltase (P < 0.01). Importantly, the expression levels of divalent metal transporter-1(DMT1) decreased with different levels of SPCI addition (P < 0.01). In addition, dietary SPCI supplementation at 75 mg/kg elevated the expression levels of critical functional genes such as peptide transporter-1(PePT1) (P = 0.06) and zinc transporter 1 (ZnT1) (P < 0.01) in ileum. The expression levels of sodium/glucose co-transporter-1 (SGLT1) in ileum (quadratic, P < 0.05) increased quadraticly by different levels of SPCI addition and amino acid transporter-1 (CAT1) in jejunum(P < 0.05) also increased by 100 mg/kg SPCI addition. CONCLUSIONS Dietary SPCI supplementation at 75 ~ 100 mg/kg improved growth performance by elevated immunity and intestinal health.
Collapse
Affiliation(s)
- Limei M Sun
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan Province, P. R. China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, 611130, Sichuan Province, P. R. China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan Province, P. R. China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, 611130, Sichuan Province, P. R. China
| | - Yuheng H Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan Province, P. R. China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, 611130, Sichuan Province, P. R. China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan Province, P. R. China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, 611130, Sichuan Province, P. R. China
| | - Zhiqing Huang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan Province, P. R. China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, 611130, Sichuan Province, P. R. China
| | - Jie Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan Province, P. R. China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, 611130, Sichuan Province, P. R. China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan Province, P. R. China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, 611130, Sichuan Province, P. R. China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan Province, P. R. China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, 611130, Sichuan Province, P. R. China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan Province, P. R. China
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, 611130, Sichuan Province, P. R. China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, Sichuan Province, P. R. China.
- Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, 611130, Sichuan Province, P. R. China.
| |
Collapse
|
7
|
Afshinpour M, Mahdiuni H. Arginine transportation mechanism through cationic amino acid transporter 1: insights from molecular dynamics studies. J Biomol Struct Dyn 2023; 41:13580-13594. [PMID: 36762692 DOI: 10.1080/07391102.2023.2175374] [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: 07/21/2022] [Accepted: 01/28/2023] [Indexed: 02/11/2023]
Abstract
Metabolic and signaling mechanisms in mammalian cells are facilitated by the transportation of L-arginine (Arg) across the plasma membrane through cationic amino acid transporter (CAT) proteins. Due to a lack of argininosuccinate synthase (ASS) activity in various tumor cells such as acute myeloid leukemia, acute lymphocytic leukemia, and chronic lymphocytic leukemia, these tumor entities are arginine-auxotrophic and therefore depend on the uptake of the amino acid arginine. Cationic amino acid transporter-1 (CAT-1) is the leading arginine importer expressed in the aforementioned tumor entities. Hence, in the present study, to investigate the transportation mechanism of arginine in CAT-1, we performed molecular dynamics (MD) simulation methods on the modeled human CAT-1. The MM-PBSA approach was conducted to determine the critical residues interacting with arginine within the corresponding binding site of CAT-1. In addition, we found out that the water molecules have the leading role in forming the transportation channel within CAT-1. The conductive structure of CAT-1 was formed only when the water molecules were continuously distributed across the channel. Steered molecular dynamics (SMD) simulation approach showed various energy barriers against arginine transportation through CAT-1, especially while crossing the bottlenecks of the related channel. These findings at the molecular level might shed light on identifying the crucial amino acids in the binding of arginine to eukaryotic CATs and also provide fundamental insights into the arginine transportation mechanisms through CAT-1. Understanding the transportation mechanism of arginine is essential to developing CAT-1 blockers, which can be potential medications for some types of cancers.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Maral Afshinpour
- Bioinformatics Lab., Department of Biology, School of Sciences, Razi University, Kermanshah, Iran
| | - Hamid Mahdiuni
- Bioinformatics Lab., Department of Biology, School of Sciences, Razi University, Kermanshah, Iran
| |
Collapse
|
8
|
Kearney PJ, Bolden NC, Kahuno E, Conklin TL, Martin GE, Lubec G, Melikian HE. Presynaptic Gq-coupled receptors drive biphasic dopamine transporter trafficking that modulates dopamine clearance and motor function. J Biol Chem 2023; 299:102900. [PMID: 36640864 PMCID: PMC9943899 DOI: 10.1016/j.jbc.2023.102900] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/23/2022] [Accepted: 01/03/2023] [Indexed: 01/13/2023] Open
Abstract
Extracellular dopamine (DA) levels are constrained by the presynaptic DA transporter (DAT), a major psychostimulant target. Despite its necessity for DA neurotransmission, DAT regulation in situ is poorly understood, and it is unknown whether regulated DAT trafficking impacts dopaminergic signaling and/or behaviors. Leveraging chemogenetics and conditional gene silencing, we found that activating presynaptic Gq-coupled receptors, either hM3Dq or mGlu5, drove rapid biphasic DAT membrane trafficking in ex vivo striatal slices, with region-specific differences between ventral and dorsal striata. DAT insertion required D2 DA autoreceptors and intact retromer, whereas DAT retrieval required PKC activation and Rit2. Ex vivo voltammetric studies revealed that DAT trafficking impacts DA clearance. Furthermore, dopaminergic mGlu5 silencing elevated DAT surface expression and abolished motor learning, which was rescued by inhibiting DAT with a subthreshold CE-158 dose. We discovered that presynaptic DAT trafficking is complex, multimodal, and region specific, and for the first time, we identified cell autonomous mechanisms that govern presynaptic DAT tone. Importantly, the findings are consistent with a role for regulated DAT trafficking in DA clearance and motor function.
Collapse
Affiliation(s)
- Patrick J. Kearney
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA,Morningside Graduate School of Biomedical Sciences, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Nicholas C. Bolden
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA,Morningside Graduate School of Biomedical Sciences, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Elizabeth Kahuno
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Tucker L. Conklin
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Gilles E. Martin
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Private Medical University, Salzburg, Austria
| | - Haley E. Melikian
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA,For correspondence: Haley E. Melikian
| |
Collapse
|
9
|
Nepal B, Das S, Reith ME, Kortagere S. Overview of the structure and function of the dopamine transporter and its protein interactions. Front Physiol 2023; 14:1150355. [PMID: 36935752 PMCID: PMC10020207 DOI: 10.3389/fphys.2023.1150355] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The dopamine transporter (DAT) plays an integral role in dopamine neurotransmission through the clearance of dopamine from the extracellular space. Dysregulation of DAT is central to the pathophysiology of numerous neuropsychiatric disorders and as such is an attractive therapeutic target. DAT belongs to the solute carrier family 6 (SLC6) class of Na+/Cl- dependent transporters that move various cargo into neurons against their concentration gradient. This review focuses on DAT (SCL6A3 protein) while extending the narrative to the closely related transporters for serotonin and norepinephrine where needed for comparison or functional relevance. Cloning and site-directed mutagenesis experiments provided early structural knowledge of DAT but our contemporary understanding was achieved through a combination of crystallization of the related bacterial transporter LeuT, homology modeling, and subsequently the crystallization of drosophila DAT. These seminal findings enabled a better understanding of the conformational states involved in the transport of substrate, subsequently aiding state-specific drug design. Post-translational modifications to DAT such as phosphorylation, palmitoylation, ubiquitination also influence the plasma membrane localization and kinetics. Substrates and drugs can interact with multiple sites within DAT including the primary S1 and S2 sites involved in dopamine binding and novel allosteric sites. Major research has centered around the question what determines the substrate and inhibitor selectivity of DAT in comparison to serotonin and norepinephrine transporters. DAT has been implicated in many neurological disorders and may play a role in the pathology of HIV and Parkinson's disease via direct physical interaction with HIV-1 Tat and α-synuclein proteins respectively.
Collapse
Affiliation(s)
- Binod Nepal
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Sanjay Das
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Maarten E. Reith
- Department of Psychiatry, New York University School of Medicine, New York City, NY, United States
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- *Correspondence: Sandhya Kortagere,
| |
Collapse
|
10
|
Kumar A, Kanak KR, Arunachalam A, Dass RS, Lakshmi PTV. Comparative transcriptome profiling and weighted gene co-expression network analysis to identify core genes in maize ( Zea mays L.) silks infected by multiple fungi. FRONTIERS IN PLANT SCIENCE 2022; 13:985396. [PMID: 36388593 PMCID: PMC9647128 DOI: 10.3389/fpls.2022.985396] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/23/2022] [Indexed: 06/16/2023]
Abstract
Maize (Zea mays L.) is the third most popular Poaceae crop after wheat and rice and used in feed and pharmaceutical sectors. The maize silk contains bioactive components explored by traditional Chinese herbal medicine for various pharmacological activities. However, Fusarium graminearum, Fusarium verticillioides, Trichoderma atroviride, and Ustilago maydis can infect the maize, produce mycotoxins, hamper the quantity and quality of silk production, and further harm the primary consumer's health. However, the defense mechanism is not fully understood in multiple fungal infections in the silk of Z. mays. In this study, we applied bioinformatics approaches to use the publicly available transcriptome data of Z. mays silk affected by multiple fungal flora to identify core genes involved in combatting disease response. Differentially expressed genes (DEGs) were identified among intra- and inter-transcriptome data sets of control versus infected Z. mays silks. Upon further comparison between up- and downregulated genes within the control of datasets, 4,519 upregulated and 5,125 downregulated genes were found. The DEGs have been compared with genes in the modules of weighted gene co-expression network analysis to relevant specific traits towards identifying core genes. The expression pattern of transcription factors, carbohydrate-active enzymes (CAZyme), and resistance genes was analyzed. The present investigation is supportive of our findings that the gene ontology, immunity stimulus, and resistance genes are upregulated, but physical and metabolic processes such as cell wall organizations and pectin synthesis were downregulated respectively. Our results are indicative that terpene synthase TPS6 and TPS11 are involved in the defense mechanism against fungal infections in maize silk.
Collapse
Affiliation(s)
- Amrendra Kumar
- Phytomatics Lab, Department of Bioinformatics, School of Life Sciences, Pondicherry University, Pondicherry, India
| | - Kanak Raj Kanak
- Fungal Genetics and Mycotoxicology Laboratory, Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Annamalai Arunachalam
- Postgraduate and Research Department of Botany, Arignar Anna Government Arts College, Villupuram, Tamil Nadu, India
| | - Regina Sharmila Dass
- Fungal Genetics and Mycotoxicology Laboratory, Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry, India
| | - P. T. V. Lakshmi
- Phytomatics Lab, Department of Bioinformatics, School of Life Sciences, Pondicherry University, Pondicherry, India
| |
Collapse
|
11
|
Wang C, Zhao F, Liu J, Liu H. The ubiquitin ligase Nedd4-2 mediates the regulation of PepT2 by mTORC1 in bovine mammary epithelial cells. ANIMAL NUTRITION 2022; 10:12-18. [PMID: 35601254 PMCID: PMC9111928 DOI: 10.1016/j.aninu.2021.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/21/2021] [Accepted: 11/23/2021] [Indexed: 11/16/2022]
Abstract
Peptide transporter 2 (PepT2) transports short peptides from the blood into bovine mammary epithelial cells (BMEC) to stimulate milk protein synthesis. Despite the fact that the effect of PepT2 is acknowledged in BMEC, little is known about its regulation. This study was completed to investigate the role of mammalian target of the rapamycin (mTOR) signaling in regulating the expression and function of PepT2 in BMEC. The regulation of PepT2 by mTOR in BMEC was studied in vitro using peptide transport assay, gene silencing, Western blot. The membrane expression of PepT2 and the uptake of β-Ala-Lys-N-7-amino-4-methylcoumarin-3-acetic acid (β-Ala-Lys-AMCA), a model dipeptide, in BMEC were reduced by rapamycin (a mTOR inhibitor) and silencing of either mTOR complex 1 (mTORC1) or mTOR complex 2 (mTORC2), stimulated by DEP domain-containing mTOR-interacting protein (DEPTOR, endogenous inhibitor of mTORC1 and mTORC2) silencing. The trafficking of PepT2 to the membrane and the uptake of β-Ala-Lys-AMCA was promoted by neuronal precursor cell-expressed developmentally down-regulated 4 isoform 2 (Nedd4-2) silencing. The effects of knockdown of mTORC1, but not mTORC2, on cell membrane expression and transport activity of PepT2 was abolished by Nedd4-2 silencing. With immunofluorescence staining, PepT2 was identified to be interacting with Nedd4-2. The Nedd4-2 expression and the interaction between PepT2 and Nedd4-2 was increased through mTORC1 knockdown, indicating an increased ubiquitination of PepT2. The results revealed that mTORC1 can regulate the expression and function of PepT2 through Nedd4-2 in BMEC.
Collapse
|
12
|
Pneumolysin boosts the neuroinflammatory response to Streptococcus pneumoniae through enhanced endocytosis. Nat Commun 2022; 13:5032. [PMID: 36028511 PMCID: PMC9418233 DOI: 10.1038/s41467-022-32624-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/09/2022] [Indexed: 11/08/2022] Open
Abstract
In pneumococcal meningitis, bacterial growth in the cerebrospinal fluid results in lysis, the release of toxic factors, and subsequent neuroinflammation. Exposure of primary murine glia to Streptococcus pneumoniae lysates leads to strong proinflammatory cytokine and chemokine production, blocked by inhibition of the intracellular innate receptor Nod1. Lysates enhance dynamin-dependent endocytosis, and dynamin inhibition reduces neuroinflammation, blocking ligand internalization. Here we identify the cholesterol-dependent cytolysin pneumolysin as a pro-endocytotic factor in lysates, its elimination reduces their proinflammatory effect. Only pore-competent pneumolysin enhances endocytosis in a dynamin-, phosphatidylinositol-3-kinase- and potassium-dependent manner. Endocytic enhancement is limited to toxin-exposed parts of the membrane, the effect is rapid and pneumolysin permanently alters membrane dynamics. In a murine model of pneumococcal meningitis, mice treated with chlorpromazine, a neuroleptic with a complementary endocytosis inhibitory effect show reduced neuroinflammation. Thus, the dynamin-dependent endocytosis emerges as a factor in pneumococcal neuroinflammation, and its enhancement by a cytolysin represents a proinflammatory control mechanism.
Collapse
|
13
|
The NEDD4 ubiquitin E3 ligase: a snapshot view of its functional activity and regulation. Biochem Soc Trans 2022; 50:473-485. [PMID: 35129615 DOI: 10.1042/bst20210731] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 11/17/2022]
Abstract
Due to its fundamental role in all eukaryotic cells, a deeper understanding of the molecular mechanisms underlying ubiquitination is of central importance. Being responsible for chain specificity and substrate recognition, E3 ligases are the selective elements of the ubiquitination process. In this review, we discuss different cellular pathways regulated by one of the first identified E3 ligase, NEDD4, focusing on its pathophysiological role, its known targets and modulators. In addition, we highlight small molecule inhibitors that act on NEDD4 and discuss new strategies to effectively target this E3 enzyme.
Collapse
|
14
|
Alves SAS, Florentino LS, Teixeira DE, Silva-Aguiar RP, Peruchetti DB, Oliveira AC, Scharfstein J, Marzolo MP, Pinheiro AAS, Caruso-Neves C. Surface megalin expression is a target to the inhibitory effect of bradykinin on the renal albumin endocytosis. Peptides 2021; 146:170646. [PMID: 34500007 DOI: 10.1016/j.peptides.2021.170646] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/31/2022]
Abstract
Megalin-mediated albumin endocytosis plays a critical role in albumin reabsorption in proximal tubule (PT) epithelial cells (PTECs). Some studies have pointed out the modulatory effect of bradykinin (BK) on urinary protein excretion, but its role in PT protein endocytosis has not yet been determined. Here, we studied the possible correlation between BK and albumin endocytosis in PT. Using LLC-PK1 cells, a model of PTECs, we showed that BK specifically inhibited megalin-mediated albumin endocytosis. This inhibitory effect of BK was mediated by B2 receptor (B2R) because it was abolished by HOE140, an antagonist of B2R, but it was not affected by Lys-des-Arg9-BK, an antagonist of B1. BK induced the stall of megalin in EEA1+ endosomes, but not in LAMP1+ lysosomes, leading to a decrease in surface megalin expression. In addition, we showed that BK, through B2R, activated calphostin C-sensitive protein kinase C, which mediated its effect on the surface megalin expression and albumin endocytosis. These results reveal an important modulatory mechanism of PT albumin endocytosis by BK, which opens new possibilities to understanding the effect of BK on urinary albumin excretion.
Collapse
Affiliation(s)
- Sarah A S Alves
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucas S Florentino
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Douglas E Teixeira
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo P Silva-Aguiar
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diogo B Peruchetti
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Carolina Oliveira
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Julio Scharfstein
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - María-Paz Marzolo
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ana Acacia S Pinheiro
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
| | - Celso Caruso-Neves
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.
| |
Collapse
|
15
|
Jenkin KA, Han Y, Lin S, He P, Yun CC. Nedd4-2-dependent Ubiquitination Potentiates the Inhibition of Human NHE3 by Cholera Toxin and Enteropathogenic Escherichia coli. Cell Mol Gastroenterol Hepatol 2021; 13:695-716. [PMID: 34823064 PMCID: PMC8789535 DOI: 10.1016/j.jcmgh.2021.11.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Diarrhea is one of the most common illnesses and is often caused by bacterial infection. Recently, we have shown that human Na+/H+ exchanger NHE3 (hNHE3), but not non-human NHE3s, interacts with the E3 ubiquitin ligase Nedd4-2. We hypothesize that this property of hNHE3 contributes to the increased severity of diarrhea in humans. METHODS We used humanized mice expressing hNHE3 in the intestine (hNHE3int) to compare the contribution of hNHE3 and mouse NHE3 to diarrhea induced by cholera toxin (CTX) and enteropathogenic Escherichia coli (EPEC). We measured Na+/H+ exchange activity and fluid absorption. The role of Nedd4-2 on hNHE3 activity and ubiquitination was determined by knockdown in Caco-2bbe cells. The effects of protein kinase A (PKA), the primary mediator of CTX-induced diarrhea, on Nedd4-2 and hNHE3 phosphorylation and their interaction were determined. RESULTS The effects of CTX and EPEC were greater in hNHE3int mice than in control wild-type (WT) mice, resulting in greater inhibition of NHE3 activity and increased fluid accumulation in the intestine, the hallmark of diarrhea. Activation of PKA increased ubiquitination of hNHE3 and enhanced interaction of Nedd4-2 with hNHE3 via phosphorylation of Nedd4-2 at S342. S342A mutation mitigated the Nedd4-2-hNHE3 interaction and blocked PKA-induced inhibition of hNHE3. Unlike non-human NHE3s, inhibition of hNHE3 by PKA is independent of NHE3 phosphorylation, suggesting a distinct mechanism of hNHE3 regulation. CONCLUSIONS The effects of CTX and EPEC on hNHE3 are amplified, and the unique properties of hNHE3 may contribute to diarrheal symptoms occurring in humans.
Collapse
Affiliation(s)
- Kayte A. Jenkin
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,School of Science, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Yiran Han
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,Atlanta VA Medical Center, Decatur, Georgia
| | - Songbai Lin
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,Atlanta VA Medical Center, Decatur, Georgia
| | - Peijian He
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - C. Chris Yun
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia,Atlanta VA Medical Center, Decatur, Georgia,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia,Correspondence Address correspondence to: Chris Yun, PhD, Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia 30324. fax: (404) 727-5767.
| |
Collapse
|
16
|
Abbiati RA, Wientjes MG, Au JLS. Is It Time to Use Modeling of Cellular Transporter Homeostasis to Inform Drug-Drug Interaction Studies: Theoretical Considerations. AAPS J 2021; 23:102. [PMID: 34435271 PMCID: PMC11048728 DOI: 10.1208/s12248-021-00635-4] [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: 06/08/2021] [Accepted: 08/06/2021] [Indexed: 11/30/2022] Open
Abstract
Mathematical modeling has been an important tool in pharmaceutical research for 50 + years and there is increased emphasis over the last decade on using modeling to improve the efficiency and effectiveness of drug development. In an earlier commentary, we applied a multiscale model linking 6 scales (whole body, tumor, vasculature, cell, spatial location, time), together with literature data on nanoparticle and tumor properties, to demonstrate the effects of nanoparticle particles on systemic disposition. The current commentary used a 4-scale model (cell membrane, intracellular organelles, spatial location, time) together with literature data on the intracellular processing of membrane receptors and transporters to demonstrate disruption of transporter homeostasis can lead to drug-drug interaction (DDI) between victim drug (VD) and perpetrator drug (PD), including changes in the area-under-concentration-time-curve of VD in cells that are considered significant by the US Food and Drug Administration (FDA). The model comprised 3 computational components: (a) intracellular transporter homeostasis, (b) pharmacokinetics of extracellular and intracellular VD/PD concentrations, and (c) pharmacodynamics of PD-induced stimulation or inhibition of an intracellular kinetic process. Model-based simulations showed that (a) among the five major endocytic processes, perturbation of transporter internalization or recycling led to the highest incidence and most extensive DDI, with minor DDI for perturbing transporter synthesis and early-to-late endosome and no DDI for perturbing transporter degradation and (b) three experimental conditions (spatial transporter distribution in cells, VD/PD co-incubation time, extracellular PD concentrations) were determinants of DDI detection. We propose modeling is a useful tool for hypothesis generation and for designing experiments to identify potential DDI; its application further aligns with the model-informed drug development paradigm advocated by FDA.
Collapse
Affiliation(s)
- Roberto A Abbiati
- Institute of Quantitative Systems Pharmacology, Carlsbad, California, 92008, USA
- Department of Pharmaceutical Sciences, University of Oklahoma, Oklahoma City, Oklahoma, 73117, USA
| | - M Guillaume Wientjes
- Institute of Quantitative Systems Pharmacology, Carlsbad, California, 92008, USA
- Optimum Therapeutics LLC, 1815 Aston Ave, Suite 107, Carlsbad, California, 92008, USA
| | - Jessie L-S Au
- Institute of Quantitative Systems Pharmacology, Carlsbad, California, 92008, USA.
- Department of Pharmaceutical Sciences, University of Oklahoma, Oklahoma City, Oklahoma, 73117, USA.
- Optimum Therapeutics LLC, 1815 Aston Ave, Suite 107, Carlsbad, California, 92008, USA.
- Taipei Medical University, Taipei, Taiwan, Republic of China.
| |
Collapse
|
17
|
Zhang M, Gao S, Yang D, Fang Y, Lin X, Jin X, Liu Y, Liu X, Su K, Shi K. Influencing factors and strategies of enhancing nanoparticles into tumors in vivo. Acta Pharm Sin B 2021; 11:2265-2285. [PMID: 34522587 PMCID: PMC8424218 DOI: 10.1016/j.apsb.2021.03.033] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/05/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023] Open
Abstract
The administration of nanoparticles (NPs) first faces the challenges of evading renal filtration and clearance of reticuloendothelial system (RES). After that, NPs infiltrate through the expanded endothelial space and penetrated the dense stroma of tumor microenvironment to tumor cells. As long as possible to prolong the time of NPs remaining in tumor tissue, NPs release active agent and induce pharmacological action. This review provides a comprehensive summary of the physical and chemical properties of NPs and the influence of various biological factors in tumor microenvironment, and discusses how to improve the final efficacy through adjusting the characteristics and structure of NPs. Perspectives and future directions are also provided.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Kai Shi
- Corresponding author. Tel./fax: +86 24 43520557.
| |
Collapse
|
18
|
Kahlhofer J, Leon S, Teis D, Schmidt O. The α-arrestin family of ubiquitin ligase adaptors links metabolism with selective endocytosis. Biol Cell 2021; 113:183-219. [PMID: 33314196 DOI: 10.1111/boc.202000137] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/03/2020] [Indexed: 12/14/2022]
Abstract
The regulation of nutrient uptake into cells is important, as it allows to either increase biomass for cell growth or to preserve homoeostasis. A key strategy to adjust cellular nutrient uptake is the reconfiguration of the nutrient transporter repertoire at the plasma membrane by the addition of nutrient transporters through the secretory pathway and by their endocytic removal. In this review, we focus on the mechanisms that regulate selective nutrient transporter endocytosis, which is mediated by the α-arrestin protein family. In the budding yeast Saccharomyces cerevisiae, 14 different α-arrestins (also named arrestin-related trafficking adaptors, ARTs) function as adaptors for the ubiquitin ligase Rsp5. They instruct Rsp5 to ubiquitinate subsets of nutrient transporters to orchestrate their endocytosis. The ART proteins are under multilevel control of the major nutrient sensing systems, including amino acid sensing by the general amino acid control and target of rapamycin pathways, and energy sensing by 5'-adenosine-monophosphate-dependent kinase. The function of the six human α-arrestins is comparably under-characterised. Here, we summarise the current knowledge about the function, regulation and substrates of yeast ARTs and human α-arrestins, and highlight emerging communalities and general principles.
Collapse
Affiliation(s)
- Jennifer Kahlhofer
- Institute for Cell Biology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Sebastien Leon
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France
| | - David Teis
- Institute for Cell Biology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Oliver Schmidt
- Institute for Cell Biology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
19
|
Maeda K, Tasaki M, Ando Y, Ohtsubo K. Galectin-lattice sustains function of cationic amino acid transporter and insulin secretion of pancreatic β cells. J Biochem 2021; 167:587-596. [PMID: 31960919 DOI: 10.1093/jb/mvaa007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/09/2020] [Indexed: 01/01/2023] Open
Abstract
Maintenance of cell surface residency and function of glycoproteins by lectins are essential for regulating cellular functions. Galectins are β-galactoside-binding lectins and form a galectin-lattice, which regulates stability, clustering, membrane sub-domain localization and endocytosis of plasmalemmal glycoproteins. We have previously reported that galectin-2 (Gal-2) forms a complex with cationic amino acid transporter 3 (CAT3) in pancreatic β cells, although the biological significance of the molecular interaction between Gal-2 and CAT3 has not been elucidated. In this study, we demonstrated that the structure of N-glycan of CAT3 was either tetra- or tri-antennary branch structure carrying β-galactosides, which works as galectin-ligands. Indeed, CAT3 bound to Gal-2 using β-galactoside epitope. Moreover, the disruption of the glycan-mediated bindings between galectins and CAT3 significantly reduced cell surface expression levels of CAT3. The reduced cell surface residency of CAT3 attenuated the cellular arginine uptake activities and subsequently reduced nitric oxide production, and thus impaired the arginine-stimulated insulin secretion of pancreatic β cells. These results indicate that galectin-lattice stabilizes CAT3 by preventing endocytosis to sustain the arginine-stimulated insulin secretion of pancreatic β cells. This provides a novel cell biological insight into the endocrinological mechanism of nutrition metabolism and homeostasis.
Collapse
Affiliation(s)
- Kento Maeda
- Department of Analytical Biochemistry;, Graduate School of Health Sciences, Kumamoto University, 4-24-1 Kuhonji, Chuo-Ku, Kumamoto 862-0976, Japan
| | - Masayoshi Tasaki
- Department of Morphological and Physiological Sciences, Graduate School of Health Sciences, Kumamoto University, 4-24-1 Kuhonji, Chuo-Ku, Kumamoto 862-0976, Japan.,Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Yukio Ando
- Depatment of Amyloidosis Research, Nagasaki International University, Nagasaki 859-3243, Japan
| | - Kazuaki Ohtsubo
- Department of Analytical Biochemistry;, Graduate School of Health Sciences, Kumamoto University, 4-24-1 Kuhonji, Chuo-Ku, Kumamoto 862-0976, Japan.,Department of Analytical Biochemistry, Faculty of Life Sciences, Kumamoto University, 4-24-1 Kuhonji, Chuo-Ku, Kumamoto 862-0976, Japan
| |
Collapse
|
20
|
Understanding fundamentals of hepatocellular carcinoma to design next-generation chitosan nano-formulations: Beyond chemotherapy stride. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
21
|
Masaki K, Hiraki Y, Onishi H, Satoh Y, Roche PA, Tanaka S, Furuta K. Ligation of MHC Class II Induces PKC-Dependent Clathrin-Mediated Endocytosis of MHC Class II. Cells 2020; 9:E1810. [PMID: 32751549 PMCID: PMC7465434 DOI: 10.3390/cells9081810] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 11/17/2022] Open
Abstract
In addition to antigen presentation to CD4+ T cells, aggregation of cell surface major histocompatibility complex class II (MHC-II) molecules induces signal transduction in antigen presenting cells that regulate cellular functions. We previously reported that crosslinking of MHC-II induced the endocytosis of MHC-II, which was associated with decreased surface expression levels in murine dendritic cells (DCs) and resulted in impaired activation of CD4+ T cells. However, the downstream signal that induces MHC-II endocytosis remains to be elucidated. In this study, we found that the crosslinking of MHC-II induced intracellular Ca2+ mobilization, which was necessary for crosslinking-induced MHC-II endocytosis. We also found that these events were suppressed by inhibitors of Syk and phospholipase C (PLC). Treatments with a phorbol ester promoted MHC-II endocytosis, whereas inhibitors of protein kinase C (PKC) suppressed crosslinking-induced endocytosis of MHC-II. These results suggest that PKC could be involved in this process. Furthermore, crosslinking-induced MHC-II endocytosis was suppressed by inhibitors of clathrin-dependent endocytosis. Our results indicate that the crosslinking of MHC-II could stimulate Ca2+ mobilization and induce the clathrin-dependent endocytosis of MHC-II in murine DCs.
Collapse
Affiliation(s)
- Kento Masaki
- Department of Immunobiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Tsushima naka 1-1-1, Kita-ku, Okayama 700-8530, Japan; (K.M.); (Y.H.); (Y.S.)
| | - Yuhji Hiraki
- Department of Immunobiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Tsushima naka 1-1-1, Kita-ku, Okayama 700-8530, Japan; (K.M.); (Y.H.); (Y.S.)
| | - Hiroka Onishi
- Department of Immunobiology, Faculty of Pharmacy and Pharmaceutical Sciences, Okayama University, Tsushima naka 1-1-1, Kita-ku, Okayama 700-8530, Japan;
| | - Yuka Satoh
- Department of Immunobiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Tsushima naka 1-1-1, Kita-ku, Okayama 700-8530, Japan; (K.M.); (Y.H.); (Y.S.)
| | - Paul A. Roche
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Satoshi Tanaka
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Misasagi Nakauchi-cho 5, Yamashina-ku, Kyoto 607-8414, Japan;
| | - Kazuyuki Furuta
- Department of Immunobiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Tsushima naka 1-1-1, Kita-ku, Okayama 700-8530, Japan; (K.M.); (Y.H.); (Y.S.)
| |
Collapse
|
22
|
Gerbino A, De Zio R, Russo D, Milella L, Milano S, Procino G, Pusch M, Svelto M, Carmosino M. Role of PKC in the Regulation of the Human Kidney Chloride Channel ClC-Ka. Sci Rep 2020; 10:10268. [PMID: 32581267 PMCID: PMC7314819 DOI: 10.1038/s41598-020-67219-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 06/02/2020] [Indexed: 12/03/2022] Open
Abstract
The physiological role of the renal ClC-Ka/ClC-K1 channels is to confer a high Cl- permeability to the thin Ascending Limb of Henle (tAL), which in turn is essential for establishing the high osmolarity of the renal medulla that drives water reabsorption from collecting ducts. Here, we investigated by whole-cell patch-clamp measurements on HEK293 cells co-expressing ClC-Ka (tagged with GFP) and the accessory subunit barttin (tagged with m-Cherry) the effect of a natural diuretic extract from roots of Dandelion (DRE), and other compounds activating PKC, such as ATP, on ClC-Ka activity and its membrane localization. Treatment with 400 µg/ml DRE significantly inhibited Cl- currents time-dependently within several minutes. Of note, the same effect on Cl- currents was obtained upon treatment with 100 µM ATP. Pretreatment of cells with either the intracellular Ca2+ chelator BAPTA-AM (30 μM) or the PKC inhibitor Calphostin C (100 nM) reduced the inhibitory effect of DRE. Conversely, 1 µM of phorbol meristate acetate (PMA), a specific PKC activator, mimicked the inhibitory effect of DRE on ClC-Ka. Finally, we found that pretreatment with 30 µM Heclin, an E3 ubiquitin ligase inhibitor, did not revert DRE-induced Cl- current inhibition. In agreement with this, live-cell confocal analysis showed that DRE treatment did not induce ClC-Ka internalization. In conclusion, we demonstrate for the first time that the activity of ClC-Ka in renal cells could be significantly inhibited by the activation of PKC elicited by classical maneuvers, such as activation of purinergic receptors, or by exposure to herbal extracts that activates a PKC-dependent pathway. Overall, we provide both new information regarding the regulation of ClC-Ka and a proof-of-concept study for the use of DRE as new diuretic.
Collapse
Affiliation(s)
- Andrea Gerbino
- National Research Council, Institute of Biomembrane and Bioenergetics, Bari, IT, Italy.,Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Roberta De Zio
- Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Daniela Russo
- Department of Sciences, University of Basilicata, Potenza, IT, Italy
| | - Luigi Milella
- Department of Sciences, University of Basilicata, Potenza, IT, Italy
| | - Serena Milano
- Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Giuseppe Procino
- Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Michael Pusch
- National Research Council, Institute of Biophysics, Genova, IT, Italy
| | - Maria Svelto
- National Research Council, Institute of Biomembrane and Bioenergetics, Bari, IT, Italy.,Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Monica Carmosino
- Department of Sciences, University of Basilicata, Potenza, IT, Italy. .,Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy.
| |
Collapse
|
23
|
Rojas S, Basualto E, Valdivia L, Vallejos N, Ceballos K, Peña E, Rivas C, Nualart F, Guzmán-Gutiérrez E, Escudero C, Toledo F, Sobrevia L, Cid M, González M. The activity of IKCa and BKCa channels contributes to insulin-mediated NO synthesis and vascular tone regulation in human umbilical vein. Nitric Oxide 2020; 99:7-16. [PMID: 32165314 DOI: 10.1016/j.niox.2020.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/23/2020] [Accepted: 03/03/2020] [Indexed: 01/16/2023]
Abstract
Insulin regulates the l-arginine/nitric oxide (NO) pathway in human umbilical vein endothelial cells (HUVECs), increasing the plasma membrane expression of the l-arginine transporter hCAT-1 and inducing vasodilation in umbilical and placental veins. Placental vascular relaxation induced by insulin is dependent of large conductance calcium-activated potassium channels (BKCa), but the role of KCa channels on l-arginine transport and NO synthesis is still unknown. The aim of this study was to determine the contribution of KCa channels in both insulin-induced l-arginine transport and NO synthesis, and its relationship with placental vascular relaxation. HUVECs, human placental vein endothelial cells (HPVECs) and placental veins were freshly isolated from umbilical cords and placenta from normal pregnancies. Cells or tissue were incubated in absence or presence of insulin and/or tetraethylammonium, 1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole, iberiotoxin or NG-nitro-l-arginine methyl ester. l-Arginine uptake, plasma membrane polarity, NO levels, hCAT-1 expression and placenta vascular reactivity were analyzed. The inhibition of intermediate-conductance KCa (IKCa) and BKCa increases l-arginine uptake, which was related with protein abundance of hCAT-1 in HUVECs. IKCa and BKCa activities contribute to NO-synthesis induced by insulin but are not directly involved in insulin-stimulated l-arginine uptake. Long term incubation (8 h) with insulin increases the plasma membrane hyperpolarization and hCAT-1 expression in HUVECs and HPVECs. Insulin-induced relaxation in placental vasculature was reversed by KCa inhibition. The results show that the activity of IKCa and BKCa channels are relevant for both physiological regulations of NO synthesis and vascular tone regulation in the human placenta, acting as a part of negative feedback mechanism for autoregulation of l-arginine transport in HUVECs.
Collapse
Affiliation(s)
- Susana Rojas
- Laboratorio de Fisiología Vascular, Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad deConcepción, Concepción, Chile
| | - Emerita Basualto
- Laboratorio de Fisiología Vascular, Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad deConcepción, Concepción, Chile
| | - Luz Valdivia
- Laboratorio de Fisiología Vascular, Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad deConcepción, Concepción, Chile
| | - Natalia Vallejos
- Laboratorio de Fisiología Vascular, Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad deConcepción, Concepción, Chile; Laboratorio de Investigación Materno-Fetal (LIMaF), Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Concepción, Concepción, Chile
| | - Karen Ceballos
- Laboratorio de Investigación Materno-Fetal (LIMaF), Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Concepción, Concepción, Chile
| | - Eduardo Peña
- Departmento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Coralia Rivas
- Departmento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Francisco Nualart
- Departamento de Biología Celular, Laboratorio de Neurobiología y Células Madres Neuro-CellTT, Centro de Microscopía Avanzada CMA BIOBIO, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Enrique Guzmán-Gutiérrez
- Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile; Group of Research and Innovation in Vascular Health (GRIVAS), Chillán, Chile
| | - Carlos Escudero
- Group of Research and Innovation in Vascular Health (GRIVAS), Chillán, Chile; Vascular Physiology Laboratory, Group of Investigation in Tumor Angiogenesis (GIANT), Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile
| | - Fernando Toledo
- Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío, Chillán, Chile; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville, Spain; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston QLD, Queensland, Australia
| | - Marcela Cid
- Departmento de Obstetricia y Puericultura, Facultad de Medicina, Universidad de Concepción, Concepción, Chile
| | - Marcelo González
- Laboratorio de Fisiología Vascular, Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad deConcepción, Concepción, Chile; Laboratorio de Investigación Materno-Fetal (LIMaF), Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Concepción, Concepción, Chile; Group of Research and Innovation in Vascular Health (GRIVAS), Chillán, Chile.
| |
Collapse
|
24
|
Barthelemy C, André B. Ubiquitylation and endocytosis of the human LAT1/SLC7A5 amino acid transporter. Sci Rep 2019; 9:16760. [PMID: 31728037 PMCID: PMC6856120 DOI: 10.1038/s41598-019-53065-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/22/2019] [Indexed: 12/11/2022] Open
Abstract
The human L-type amino acid transporter 1 (LAT1), also known as SLC7A5, catalyzes the transport of large neutral amino acids across the plasma membrane. As the main transporter of several essential amino acids, notably leucine, LAT1 plays an important role in mTORC1 activation. Furthermore, it is overexpressed in various types of cancer cells, where it contributes importantly to sustained growth. Despite the importance of LAT1 in normal and tumor cells, little is known about the mechanisms that might control its activity, for example by promoting its downregulation via endocytosis. Here we report that in HeLa cells, activation of protein kinase C by phorbol 12-myristate 13-acetate (PMA) triggers efficient endocytosis and degradation of LAT1. Under these conditions we found LAT1 downregulation to correlate with increased LAT1 ubiquitylation. This modification was considerably reduced in cells depleted of the Nedd4-2 ubiquitin ligase. By systematically mutagenizing the residues of the LAT1 cytosolic tails, we identified a group of three close lysines (K19, K25, K30) in the N-terminal tail that are important for PMA-induced ubiquitylation and downregulation. Our study thus unravels a mechanism of induced endocytosis of LAT1 elicited by Nedd4-2-mediated ubiquitylation of the transporter's N-terminal tail.
Collapse
Affiliation(s)
- Céline Barthelemy
- Molecular Physiology of the Cell, Université libre de Bruxelles (ULB), IBMM (Biopark), Gosselies, Belgium
| | - Bruno André
- Molecular Physiology of the Cell, Université libre de Bruxelles (ULB), IBMM (Biopark), Gosselies, Belgium.
| |
Collapse
|
25
|
Pengnam S, Patrojanasophon P, Rojanarata T, Ngawhirunpat T, Yingyongnarongkul BE, Radchatawedchakoon W, Opanasopit P. A novel plier-like gemini cationic niosome for nucleic acid delivery. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.04.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
26
|
Xia L, Dai L, Yang Q. Transmissible gastroenteritis virus infection decreases arginine uptake by downregulating CAT-1 expression. Vet Res 2018; 49:95. [PMID: 30236161 PMCID: PMC6148772 DOI: 10.1186/s13567-018-0591-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 09/10/2018] [Indexed: 12/14/2022] Open
Abstract
Transmissible gastroenteritis virus (TGEV) is a coronavirus that causes severe diarrhea in suckling piglets. TGEV primarily targets and infects porcine intestinal epithelial cells, which play an important role in nutrient absorption. However, the effects of TGEV infection on nutrient absorption in swine have not yet been investigated. In this study, we evaluated the impact of TGEV infection on arginine uptake using the porcine small intestinal epithelial cell line IPEC-J2 as a model system. High performance liquid chromatography (HPLC) analyses showed that TGEV infection leads to reduced arginine uptake at 48 hours post-infection (hpi). Expression of cationic amino acid transporter 1 (CAT-1) was attenuated as well. TGEV infection induced activation of phospho-protein kinase C α (p-PKC α), phospho-epidermal growth factor receptor (p-EGFR), and enhanced the expression of caveolin-1, all of which appear to be involved in down-regulating arginine uptake and CAT-1 expression. These results illuminate the relationship between TGEV infection and nutrient absorption, and further our understanding of the mechanisms of TGEV infection.
Collapse
Affiliation(s)
- Lu Xia
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, China
| | - Lei Dai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, China
| | - Qian Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, China.
| |
Collapse
|
27
|
The effects of reduced dietary protein level on amino acid transporters and mTOR signaling pathway in pigs. Biochem Biophys Res Commun 2017; 485:319-327. [DOI: 10.1016/j.bbrc.2017.02.084] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 02/16/2017] [Indexed: 12/17/2022]
|
28
|
Martínez-Mármol R, Styrczewska K, Pérez-Verdaguer M, Vallejo-Gracia A, Comes N, Sorkin A, Felipe A. Ubiquitination mediates Kv1.3 endocytosis as a mechanism for protein kinase C-dependent modulation. Sci Rep 2017; 7:42395. [PMID: 28186199 PMCID: PMC5301257 DOI: 10.1038/srep42395] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 01/09/2017] [Indexed: 12/29/2022] Open
Abstract
The voltage-dependent potassium channel Kv1.3 plays essential physiological functions in the immune system. Kv1.3, regulating the membrane potential, facilitates downstream Ca2+ -dependent pathways and becomes concentrated in specific membrane microdomains that serve as signaling platforms. Increased and/or delocalized expression of the channel is observed at the onset of several autoimmune diseases. In this work, we show that adenosine (ADO), which is a potent endogenous modulator, stimulates PKC, thereby causing immunosuppression. PKC activation triggers down-regulation of Kv1.3 by inducing a clathrin-mediated endocytic event that targets the channel to lysosomal-degradative compartments. Therefore, the abundance of Kv1.3 at the cell surface decreases, which is clearly compatible with an effective anti-inflammatory response. This mechanism requires ubiquitination of Kv1.3, catalyzed by the E3 ubiquitin-ligase Nedd4-2. Postsynaptic density protein 95 (PSD-95), a member of the MAGUK family, recruits Kv1.3 into lipid-raft microdomains and protects the channel against ubiquitination and endocytosis. Therefore, the Kv1.3/PSD-95 association fine-tunes the anti-inflammatory response in leukocytes. Because Kv1.3 is a promising multi-therapeutic target against human pathologies, our results have physiological relevance. In addition, this work elucidates the ADO-dependent PKC-mediated molecular mechanism that triggers immunomodulation by targeting Kv1.3 in leukocytes.
Collapse
Affiliation(s)
- Ramón Martínez-Mármol
- Molecular Physiology laboratory, Departament de Bioquímica i Biomedicna Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain.,Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Katarzyna Styrczewska
- Molecular Physiology laboratory, Departament de Bioquímica i Biomedicna Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Mireia Pérez-Verdaguer
- Molecular Physiology laboratory, Departament de Bioquímica i Biomedicna Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Albert Vallejo-Gracia
- Molecular Physiology laboratory, Departament de Bioquímica i Biomedicna Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Núria Comes
- Molecular Physiology laboratory, Departament de Bioquímica i Biomedicna Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain.,Laboratory of Neurophysiology, Universitat de Barcelona and Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Alexander Sorkin
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Antonio Felipe
- Molecular Physiology laboratory, Departament de Bioquímica i Biomedicna Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
29
|
Gu F, Hu C, Tai Z, Yao C, Tian J, Zhang L, Xia Q, Gong C, Gao Y, Gao S. Tumour microenvironment-responsive lipoic acid nanoparticles for targeted delivery of docetaxel to lung cancer. Sci Rep 2016; 6:36281. [PMID: 27805051 PMCID: PMC5090365 DOI: 10.1038/srep36281] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 10/13/2016] [Indexed: 11/29/2022] Open
Abstract
In the present study, we developed a novel type of reduction-sensitive nanoparticles (NPs) for docetaxel (DTX) delivery based on cross-linked lipoic acid NPs (LANPs). The physicochemical properties, cellular uptake and in vitro cytotoxicity of DTX loaded LANPs (DTX-LANPs) on A549 cells were investigated. Furthermore, the in vivo distribution and in vivo efficacy of DTX-LANPs was evaluated. The results showed that DTX-LANPs had a particle size of 110 nm and a negative zeta potential of −35 mv with excellent colloidal stability. LANPs efficiently encapsulated DTX with a high drug loading of 4.51% ± 0.49% and showed remarkable reduction-sensitive drug release in vitro. Cellular uptake experiments demonstrated that LANPs significantly increased intracellular DTX uptake by about 10 fold as compared with free DTX. The cytotoxicity of DTX-LANPs showed significantly higher potency in inhibiting A549 cell growth than free DTX, while blank LANPs had a good biocompatibility. In addition, in vivo experiments demonstrated that DTX-LANPs could enhance tumour targeting and anti-tumour efficacy with low systemic toxicity. In conclusion, LANPs may prove to be a potential tumour microenvironment-responsive delivery system for cancer treatment, with the potential for commercialization due to the simple component, controllable synthesis, stability and economy.
Collapse
Affiliation(s)
- Fenfen Gu
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Chuling Hu
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Zhongguang Tai
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Chong Yao
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Jing Tian
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Lijuan Zhang
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Qingming Xia
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Chunai Gong
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Yuan Gao
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China.,Department of Clinical Pharmacy and Pharmaceutical Management, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Shen Gao
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| |
Collapse
|
30
|
Xu D, Wang H, You G. Posttranslational Regulation of Organic Anion Transporters by Ubiquitination: Known and Novel. Med Res Rev 2016; 36:964-79. [PMID: 27291023 DOI: 10.1002/med.21397] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 04/27/2016] [Accepted: 04/27/2016] [Indexed: 12/25/2022]
Abstract
Organic anion transporters (OATs) encoded by solute carrier 22 family are localized in the epithelia of multiple organs, where they mediate the absorption, distribution, and excretion of a diverse array of negatively charged environmental toxins and clinically important drugs. Alterations in the expression and function of OATs play important roles in intra- and interindividual variability of the therapeutic efficacy and the toxicity of many drugs. As a result, the activity of OATs must be under tight regulation so as to carry out their normal functions. The regulation of OAT transport activity in response to various stimuli can occur at several levels such as transcription, translation, and posttranslational modification. Posttranslational regulation is of particular interest, because it usually happens within a very short period of time (minutes to hours) when the body has to deal with rapidly changing amounts of substances as a consequence of variable intake of drugs, fluids, or meals as well as metabolic activity. This review article highlights the recent advances from our laboratory in uncovering several posttranslational mechanisms underlying OAT regulation. These advances offer the promise of identifying targets for novel strategies that will maximize therapeutic efficacy in drug development.
Collapse
Affiliation(s)
- Da Xu
- Department of Pharmaceutics, Rutgers University, Piscataway, New Jersey, 08854
| | - Haoxun Wang
- Department of Pharmaceutics, Rutgers University, Piscataway, New Jersey, 08854
| | - Guofeng You
- Department of Pharmaceutics, Rutgers University, Piscataway, New Jersey, 08854
| |
Collapse
|
31
|
Wang H, Xu D, Toh MF, Pao AC, You G. Serum- and glucocorticoid-inducible kinase SGK2 regulates human organic anion transporters 4 via ubiquitin ligase Nedd4-2. Biochem Pharmacol 2016; 102:120-129. [PMID: 26740304 PMCID: PMC5166719 DOI: 10.1016/j.bcp.2015.11.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 11/24/2015] [Indexed: 01/08/2023]
Abstract
Human organic anion transporter 4 (hOAT4) belongs to a family of organic anion transporters that play critical roles in the body disposition of clinically important drugs, including anti-viral therapeutics, anti-cancer drugs, antibiotics, antihypertensives, and anti-inflammatories. hOAT4 is abundantly expressed in the kidney and placenta. In the current study, we examined the regulation of hOAT4 by serum- and glucocorticoid-inducible kinase 2 (sgk2) in the kidney COS-7 cells. We showed that sgk2 stimulated hOAT4 transport activity. Such stimulation mainly resulted from an increased cell surface expression of the transporter, kinetically revealed as an increased maximal transport velocity Vmax without significant change in substrate-binding affinity Km. We further showed that regulation of hOAT4 activity by sgk2 was mediated by ubiquitin ligase Nedd4-2. Overexpression of Nedd4-2 enhanced hOAT4 ubiquitination, and inhibited hOAT4 transport activity, whereas overexpression of ubiquitin ligase-dead mutant Nedd4-2/C821A or siRNA knockdown of endogenous Nedd4-2 had opposite effects on hOAT4. Our co-immunoprecipitation experiment revealed that sgk2 weakened the association between hOAT4 and Nedd4-2. In conclusion, our study demonstrated for the first time that sgk2 stimulated hOAT4 transport activity by abrogating the inhibitory effect of Nedd4-2 on the transporter.
Collapse
Affiliation(s)
- Haoxun Wang
- Department of Pharmaceutics, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - Da Xu
- Department of Pharmaceutics, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - May Fern Toh
- Department of Pharmaceutics, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - Alan C Pao
- Department of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Guofeng You
- Department of Pharmaceutics, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA.
| |
Collapse
|
32
|
Xu D, Wang H, Zhang Q, You G. Nedd4-2 but not Nedd4-1 is critical for protein kinase C-regulated ubiquitination, expression, and transport activity of human organic anion transporter 1. Am J Physiol Renal Physiol 2016; 310:F821-31. [PMID: 26823285 DOI: 10.1152/ajprenal.00522.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 01/20/2016] [Indexed: 11/22/2022] Open
Abstract
Human organic anion transporter 1 (hOAT1) expressed at the membrane of the kidney proximal tubule cells mediates the body disposition of a diverse array of clinically important drugs, including anti-HIV therapeutics, antitumor drugs, antibiotics, antihypertensives, and antiinflammatories. Therefore, understanding the regulation of hOAT1 will provide significant insights into kidney function and dysfunction. We previously established that hOAT1 transport activity is inhibited by activation of protein kinase C (PKC) through accelerating hOAT1 internalization from cell surface into intracellular endosomes and subsequent degradation. We further established that PKC-induced hOAT1 ubiquitination is an important step preceding hOAT1 internalization. In the current study, we identified two closely related E3 ubiquitin ligases, neural precursor cell expressed, developmentally downregulated 4-1 and 4-2 (Nedd4-1 and Nedd4-2), as important regulators for hOAT1: overexpression of Nedd4-1 or Nedd4-2 enhanced hOAT1 ubiquitination, reduced the hOAT1 amount at the cell surface, and suppressed hOAT1 transport activity. In further exploring the relationship among PKC, Nedd4-1, and Nedd4-2, we discovered that PKC-dependent changes in hOAT1 ubiquitination, expression, and transport activity were significantly blocked in cells transfected with the ligase-dead mutant of Nedd4-2 (Nedd4-2/C821A) or with Nedd4-2-specific siRNA to knockdown endogenous Nedd4-2 but not in cells transfected with the ligase-dead mutant of Nedd4-1 (Nedd4-1/C867S) or with Nedd4-1-specific siRNA to knockdown endogenous Nedd4-1. In conclusion, this is the first demonstration that both Nedd4-1 and Nedd4-2 are important regulators for hOAT1 ubiquitination, expression, and function. Yet they play distinct roles, as Nedd4-2 but not Nedd4-1 is a critical mediator for PKC-regulated hOAT1 ubiquitination, expression, and transport activity.
Collapse
Affiliation(s)
- Da Xu
- Department of Pharmaceutics, Rutgers University, Piscataway, New Jersey
| | - Haoxun Wang
- Department of Pharmaceutics, Rutgers University, Piscataway, New Jersey
| | - Qiang Zhang
- Department of Pharmaceutics, Rutgers University, Piscataway, New Jersey
| | - Guofeng You
- Department of Pharmaceutics, Rutgers University, Piscataway, New Jersey
| |
Collapse
|
33
|
Veyhl-Wichmann M, Friedrich A, Vernaleken A, Singh S, Kipp H, Gorboulev V, Keller T, Chintalapati C, Pipkorn R, Pastor-Anglada M, Groll J, Koepsell H. Phosphorylation of RS1 (RSC1A1) Steers Inhibition of Different Exocytotic Pathways for Glucose Transporter SGLT1 and Nucleoside Transporter CNT1, and an RS1-Derived Peptide Inhibits Glucose Absorption. Mol Pharmacol 2016; 89:118-32. [PMID: 26464324 DOI: 10.1124/mol.115.101162] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/09/2015] [Indexed: 02/14/2025] Open
Abstract
Cellular uptake adapts rapidly to physiologic demands by changing transporter abundance in the plasma membrane. The human gene RSC1A1 codes for a 67-kDa protein named RS1 that has been shown to induce downregulation of the sodium-D-glucose cotransporter 1 (SGLT1) and of the concentrative nucleoside transporter 1 (CNT1) in the plasma membrane by blocking exocytosis at the Golgi. Injecting RS1 fragments into Xenopus laevis oocytes expressing SGLT1 or CNT1 and measuring the expressed uptake of α-methylglucoside or uridine 1 hour later, we identified a RS1 domain (RS1-Reg) containing multiple predicted phosphorylation sites that is responsible for this post-translational downregulation of SGLT1 and CNT1. Dependent on phosphorylation, RS1-Reg blocks the release of SGLT1-containing vesicles from the Golgi in a glucose-dependent manner or glucose-independent release of CNT1-containing vesicles. We showed that upregulation of SGLT1 in the small intestine after glucose ingestion is promoted by glucose-dependent disinhibition of the RS1-Reg-blocked exocytotic pathway of SGLT1 between meals. Mimicking phosphorylation of RS1-Reg, we obtained a RS1-Reg variant that downregulates SGLT1 in the brush-border membrane at high luminal glucose concentration. Because RS1 mediates short-term regulation of various transporters, we propose that the RS1-Reg-navigated transporter release from Golgi represents a basic regulatory mechanism of general importance, which implies the existence of receptor proteins that recognize different phosphorylated forms of RS1-Reg and of complex transporter-specific sorting in the trans-Golgi. RS1-Reg-derived peptides that downregulate SGLT1 at high intracellular glucose concentrations may be used for downregulation of glucose absorption in small intestine, which has been proposed as strategy for treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Maike Veyhl-Wichmann
- Institute of Anatomy and Cell Biology, University Würzburg, Germany (M.V., A.V., H.Ki, V.G., C.C., H.Ko.); Department of Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Germany (S.S., J.G.); Leibnitz Institute for Interactive Materials, Rheinisch-Westfälische Technische Hochschule, University Aachen, Germany (S.S.); German Cancer Research Center, Heidelberg, Germany (R.P.); Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University Barcelona & CIBER EHD, Barcelona, Spain (M.P.); and Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University Würzburg, Germany (A.F., T.K., H.Ko)
| | - Alexandra Friedrich
- Institute of Anatomy and Cell Biology, University Würzburg, Germany (M.V., A.V., H.Ki, V.G., C.C., H.Ko.); Department of Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Germany (S.S., J.G.); Leibnitz Institute for Interactive Materials, Rheinisch-Westfälische Technische Hochschule, University Aachen, Germany (S.S.); German Cancer Research Center, Heidelberg, Germany (R.P.); Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University Barcelona & CIBER EHD, Barcelona, Spain (M.P.); and Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University Würzburg, Germany (A.F., T.K., H.Ko)
| | - Alexandra Vernaleken
- Institute of Anatomy and Cell Biology, University Würzburg, Germany (M.V., A.V., H.Ki, V.G., C.C., H.Ko.); Department of Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Germany (S.S., J.G.); Leibnitz Institute for Interactive Materials, Rheinisch-Westfälische Technische Hochschule, University Aachen, Germany (S.S.); German Cancer Research Center, Heidelberg, Germany (R.P.); Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University Barcelona & CIBER EHD, Barcelona, Spain (M.P.); and Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University Würzburg, Germany (A.F., T.K., H.Ko)
| | - Smriti Singh
- Institute of Anatomy and Cell Biology, University Würzburg, Germany (M.V., A.V., H.Ki, V.G., C.C., H.Ko.); Department of Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Germany (S.S., J.G.); Leibnitz Institute for Interactive Materials, Rheinisch-Westfälische Technische Hochschule, University Aachen, Germany (S.S.); German Cancer Research Center, Heidelberg, Germany (R.P.); Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University Barcelona & CIBER EHD, Barcelona, Spain (M.P.); and Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University Würzburg, Germany (A.F., T.K., H.Ko)
| | - Helmut Kipp
- Institute of Anatomy and Cell Biology, University Würzburg, Germany (M.V., A.V., H.Ki, V.G., C.C., H.Ko.); Department of Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Germany (S.S., J.G.); Leibnitz Institute for Interactive Materials, Rheinisch-Westfälische Technische Hochschule, University Aachen, Germany (S.S.); German Cancer Research Center, Heidelberg, Germany (R.P.); Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University Barcelona & CIBER EHD, Barcelona, Spain (M.P.); and Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University Würzburg, Germany (A.F., T.K., H.Ko)
| | - Valentin Gorboulev
- Institute of Anatomy and Cell Biology, University Würzburg, Germany (M.V., A.V., H.Ki, V.G., C.C., H.Ko.); Department of Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Germany (S.S., J.G.); Leibnitz Institute for Interactive Materials, Rheinisch-Westfälische Technische Hochschule, University Aachen, Germany (S.S.); German Cancer Research Center, Heidelberg, Germany (R.P.); Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University Barcelona & CIBER EHD, Barcelona, Spain (M.P.); and Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University Würzburg, Germany (A.F., T.K., H.Ko)
| | - Thorsten Keller
- Institute of Anatomy and Cell Biology, University Würzburg, Germany (M.V., A.V., H.Ki, V.G., C.C., H.Ko.); Department of Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Germany (S.S., J.G.); Leibnitz Institute for Interactive Materials, Rheinisch-Westfälische Technische Hochschule, University Aachen, Germany (S.S.); German Cancer Research Center, Heidelberg, Germany (R.P.); Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University Barcelona & CIBER EHD, Barcelona, Spain (M.P.); and Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University Würzburg, Germany (A.F., T.K., H.Ko)
| | - Chakravarthi Chintalapati
- Institute of Anatomy and Cell Biology, University Würzburg, Germany (M.V., A.V., H.Ki, V.G., C.C., H.Ko.); Department of Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Germany (S.S., J.G.); Leibnitz Institute for Interactive Materials, Rheinisch-Westfälische Technische Hochschule, University Aachen, Germany (S.S.); German Cancer Research Center, Heidelberg, Germany (R.P.); Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University Barcelona & CIBER EHD, Barcelona, Spain (M.P.); and Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University Würzburg, Germany (A.F., T.K., H.Ko)
| | - Rüdiger Pipkorn
- Institute of Anatomy and Cell Biology, University Würzburg, Germany (M.V., A.V., H.Ki, V.G., C.C., H.Ko.); Department of Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Germany (S.S., J.G.); Leibnitz Institute for Interactive Materials, Rheinisch-Westfälische Technische Hochschule, University Aachen, Germany (S.S.); German Cancer Research Center, Heidelberg, Germany (R.P.); Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University Barcelona & CIBER EHD, Barcelona, Spain (M.P.); and Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University Würzburg, Germany (A.F., T.K., H.Ko)
| | - Marçal Pastor-Anglada
- Institute of Anatomy and Cell Biology, University Würzburg, Germany (M.V., A.V., H.Ki, V.G., C.C., H.Ko.); Department of Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Germany (S.S., J.G.); Leibnitz Institute for Interactive Materials, Rheinisch-Westfälische Technische Hochschule, University Aachen, Germany (S.S.); German Cancer Research Center, Heidelberg, Germany (R.P.); Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University Barcelona & CIBER EHD, Barcelona, Spain (M.P.); and Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University Würzburg, Germany (A.F., T.K., H.Ko)
| | - Jürgen Groll
- Institute of Anatomy and Cell Biology, University Würzburg, Germany (M.V., A.V., H.Ki, V.G., C.C., H.Ko.); Department of Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Germany (S.S., J.G.); Leibnitz Institute for Interactive Materials, Rheinisch-Westfälische Technische Hochschule, University Aachen, Germany (S.S.); German Cancer Research Center, Heidelberg, Germany (R.P.); Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University Barcelona & CIBER EHD, Barcelona, Spain (M.P.); and Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University Würzburg, Germany (A.F., T.K., H.Ko)
| | - Hermann Koepsell
- Institute of Anatomy and Cell Biology, University Würzburg, Germany (M.V., A.V., H.Ki, V.G., C.C., H.Ko.); Department of Functional Materials in Medicine and Dentistry, University Hospital Würzburg, Germany (S.S., J.G.); Leibnitz Institute for Interactive Materials, Rheinisch-Westfälische Technische Hochschule, University Aachen, Germany (S.S.); German Cancer Research Center, Heidelberg, Germany (R.P.); Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University Barcelona & CIBER EHD, Barcelona, Spain (M.P.); and Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University Würzburg, Germany (A.F., T.K., H.Ko)
| |
Collapse
|
34
|
Xu D, Wang H, You G. An Essential Role of Nedd4-2 in the Ubiquitination, Expression, and Function of Organic Anion Transporter-3. Mol Pharm 2015; 13:621-30. [PMID: 26651153 DOI: 10.1021/acs.molpharmaceut.5b00839] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Organic anion transporter-3 (OAT3) is a member of the organic anion transporter family that mediates the body disposition of a diverse array of clinically important drugs. We previously demonstrated that activation of protein kinase C (PKC) inhibits OAT3 transport activity by accelerating OAT3 internalization from cell surface into intracellular compartments. In the current study, we established that PKC-induced inhibition of OAT3 transport activity occurred through an enhanced OAT3 ubiquitination, a process catalyzed by an E3 ubiquitin-protein ligase Nedd4-2 (neural precursor cell expressed, developmentally downregulated 4-2). Overexpression of Nedd4-2 enhanced OAT3 ubiquitination, decreased OAT3 expression at the cell surface, and inhibited OAT3 transport activity. In contrast, overexpression of the ubiquitin ligase-dead mutant Nedd4-2/C821A or siRNA knockdown of endogenous Nedd4-2 had opposite effects on OAT3. Furthermore, immunoprecipitation experiments conducted both in culture cells and with rat kidney slices showed that there was a physical interaction between OAT3 and Nedd4-2. In conclusion, our results provided the first evidence that Nedd4-2 is an important regulator for OAT3 ubiquitination, expression, and transport activity.
Collapse
Affiliation(s)
- Da Xu
- Department of Pharmaceutics, Rutgers University , Piscataway, New Jersey 08854, United States
| | - Haoxun Wang
- Department of Pharmaceutics, Rutgers University , Piscataway, New Jersey 08854, United States
| | - Guofeng You
- Department of Pharmaceutics, Rutgers University , Piscataway, New Jersey 08854, United States
| |
Collapse
|
35
|
Fortian A, Dionne LK, Hong SH, Kim W, Gygi SP, Watkins SC, Sorkin A. Endocytosis of Ubiquitylation-Deficient EGFR Mutants via Clathrin-Coated Pits is Mediated by Ubiquitylation. Traffic 2015; 16:1137-54. [PMID: 26251007 DOI: 10.1111/tra.12314] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 08/03/2015] [Accepted: 08/03/2015] [Indexed: 12/11/2022]
Abstract
Signaling by epidermal growth factor receptor (EGFR) is controlled by endocytosis. However, mechanisms of EGFR endocytosis remain poorly understood. Here, we found that the EGFR mutant lacking known ubiquitylation, acetylation and clathrin adaptor AP-2-binding sites (21KRΔAP2) was internalized at relatively high rates via the clathrin-dependent pathway in human duodenal adenocarcinoma HuTu-80 cells. RNA interference analysis revealed that this residual internalization is strongly inhibited by depletion of Grb2 and the E2 ubiquitin-conjugating enzyme UbcH5b/c, and partially affected by depletion of the E3 ubiquitin ligase Cbl and ubiquitin-binding adaptors, indicating that an ubiquitylation process is involved. Several new ubiquitin conjugation sites were identified by mass spectrometry in the 21KRΔAP2 mutant, suggesting that cryptic ubiquitylation may mediate endocytosis of this mutant. Total internal reflection fluorescence microscopy imaging of HuTu-80 cells transfected with labeled ubiquitin adaptor epsin1 demonstrated that the ubiquitylation-deficient EGFR mutant was endocytosed through a limited population of epsin-enriched clathrin-coated pits (CCPs), although with a prolonged CCP lifetime. Native EGFR was recruited with the same efficiency into CCPs containing either AP-2 or epsin1 that were tagged with fluorescent proteins by genome editing of MDA-MD-231 cells. We propose that two redundant mechanisms, ubiquitylation and interaction with AP-2, contribute to EGFR endocytosis via CCPs in a stochastic fashion.
Collapse
Affiliation(s)
- Arola Fortian
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA,, USA
| | - Lai K Dionne
- Department of Pharmacology, University of Colorado Anschutz Medical Center, Aurora, CO,, USA
| | - Sun H Hong
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA,, USA
| | - Woong Kim
- Department of Cell Biology, University of Harvard School of Medicine, Boston, MA,, USA
| | - Steven P Gygi
- Department of Cell Biology, University of Harvard School of Medicine, Boston, MA,, USA
| | - Simon C Watkins
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA,, USA
| | - Alexander Sorkin
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA,, USA
| |
Collapse
|
36
|
Glushko AA, Voronkov AV, Chernikov MV. [Molecular targets for searching of endothelial-protective substances]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2015; 40:515-27. [PMID: 25895347 DOI: 10.1134/s1068162014050069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Endothelial dysfunction underlies the development of many cardiovascular diseases. Thus endothelium becomes an independent therapeutic target, and the search of new substances with endothelial-protective action to date is one of the promising tasks for pharmacotherapy and medicinal chemistry. Molecular modeling is an effective tool for solving this problem. Computer chemistry methods use is only possible in combination with detailed information on three dimensional structure and functions of molecular targets: receptors and enzymes, involved in signal transduction inside and outside of endothelial cells. Information on structure and function of various macromolecules involved in vascular tone regulation is collected in the review. The structure of endothelial NO-synthase (EC 1.14.13.39) (eNOS)--enzyme, responsible for the nitric oxide synthesis and involved in vascular tone regulation process is reviewed. The importance of eNOS substrate--L-arginine is underlined in the review in terms of this enzyme activity, regulation, the information on structure and functions of L-arginine transport system is provided. Also different ways of eNOS activity regulation are reviewed, among which are enzyme activation and concurrent inhibition by substances interaction with active center of enzyme, inhibition by caveoline binding with oxigenase domain, and also regulation by phosphorylation of certain amino acids of eNOS by proteinkinase and dephoshphorylation of them by phosphatases. The importance of membrane receptors of endothelial cells as targets for endothelial-protective substances is underlined. Among them are receptors of endothelin, platelet activation factor, prostanoids, bradykinin, histamine, serotonin and protease activated receptors. The important role of potassium and calcium ion channels of vascular cells in endothelial-protective activity is underlined. Macromolecules presented in the review finally are considered as targets for searching for medicinal substances with endothelial-protective activity using proposed ways and methods of molecular modeling.
Collapse
|
37
|
Goel P, Manning JA, Kumar S. NEDD4-2 (NEDD4L): the ubiquitin ligase for multiple membrane proteins. Gene 2014; 557:1-10. [PMID: 25433090 DOI: 10.1016/j.gene.2014.11.051] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/14/2014] [Accepted: 11/21/2014] [Indexed: 12/20/2022]
Abstract
NEDD4-2 (also known as NEDD4L, neural precursor cell expressed developmentally down-regulated 4-like) is a ubiquitin protein ligase of the Nedd4 family which is known to bind and regulate a number of membrane proteins to aid in their internalization and turnover. Several of the NEDD4-2 substrates include ion channels, such as the epithelial and voltage-gated sodium channels. Given the critical function of NEDD4-2 in regulating membrane proteins, this ligase is essential for the maintenance of cellular homeostasis. In this article we review the biology and function of this important ubiquitin-protein ligase and discuss its pathophysiological significance.
Collapse
Affiliation(s)
- Pranay Goel
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5001, Australia; Department of Medicine, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Jantina A Manning
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5001, Australia
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5001, Australia; Department of Medicine, The University of Adelaide, Adelaide, SA 5005, Australia.
| |
Collapse
|
38
|
Erpapazoglou Z, Walker O, Haguenauer-Tsapis R. Versatile roles of k63-linked ubiquitin chains in trafficking. Cells 2014; 3:1027-88. [PMID: 25396681 PMCID: PMC4276913 DOI: 10.3390/cells3041027] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/14/2014] [Accepted: 10/21/2014] [Indexed: 12/11/2022] Open
Abstract
Modification by Lys63-linked ubiquitin (UbK63) chains is the second most abundant form of ubiquitylation. In addition to their role in DNA repair or kinase activation, UbK63 chains interfere with multiple steps of intracellular trafficking. UbK63 chains decorate many plasma membrane proteins, providing a signal that is often, but not always, required for their internalization. In yeast, plants, worms and mammals, this same modification appears to be critical for efficient sorting to multivesicular bodies and subsequent lysosomal degradation. UbK63 chains are also one of the modifications involved in various forms of autophagy (mitophagy, xenophagy, or aggrephagy). Here, in the context of trafficking, we report recent structural studies investigating UbK63 chains assembly by various E2/E3 pairs, disassembly by deubiquitylases, and specifically recognition as sorting signals by receptors carrying Ub-binding domains, often acting in tandem. In addition, we address emerging and unanticipated roles of UbK63 chains in various recycling pathways that function by activating nucleators required for actin polymerization, as well as in the transient recruitment of signaling molecules at the plasma or ER membrane. In this review, we describe recent advances that converge to elucidate the mechanisms underlying the wealth of trafficking functions of UbK63 chains.
Collapse
Affiliation(s)
- Zoi Erpapazoglou
- Institut Jacques Monod-CNRS, UMR 7592, Université-Paris Diderot, Sorbonne Paris Cité, F-75205 Paris, France.
| | - Olivier Walker
- Institut des Sciences Analytiques, UMR5280, Université de Lyon/Université Lyon 1, 69100 Villeurbanne, France.
| | - Rosine Haguenauer-Tsapis
- Institut Jacques Monod-CNRS, UMR 7592, Université-Paris Diderot, Sorbonne Paris Cité, F-75205 Paris, France.
| |
Collapse
|
39
|
Becuwe M, Léon S. Integrated control of transporter endocytosis and recycling by the arrestin-related protein Rod1 and the ubiquitin ligase Rsp5. eLife 2014; 3. [PMID: 25380227 PMCID: PMC4244573 DOI: 10.7554/elife.03307] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 11/04/2014] [Indexed: 01/04/2023] Open
Abstract
After endocytosis, membrane proteins can recycle to the cell membrane or be degraded in lysosomes. Cargo ubiquitylation favors their lysosomal targeting and can be regulated by external signals, but the mechanism is ill-defined. Here, we studied the post-endocytic trafficking of Jen1, a yeast monocarboxylate transporter, using microfluidics-assisted live-cell imaging. We show that the ubiquitin ligase Rsp5 and the glucose-regulated arrestin-related trafficking adaptors (ART) protein Rod1, involved in the glucose-induced internalization of Jen1, are also required for the post-endocytic sorting of Jen1 to the yeast lysosome. This new step takes place at the trans-Golgi network (TGN), where Rod1 localizes dynamically upon triggering endocytosis. Indeed, transporter trafficking to the TGN after internalization is required for their degradation. Glucose removal promotes Rod1 relocalization to the cytosol and Jen1 deubiquitylation, allowing transporter recycling when the signal is only transient. Therefore, nutrient availability regulates transporter fate through the localization of the ART/Rsp5 ubiquitylation complex at the TGN. DOI:http://dx.doi.org/10.7554/eLife.03307.001 The plasma membrane that surrounds cells contains many different proteins that perform tasks such as detecting signals sent to the cell, and transporting molecules into or out of the cell. To adapt to changing conditions, cells remodel their membrane to change how much of each type of protein is present. A process called endocytosis—where part of the plasma membrane and the proteins it contains buds off into the cell—plays an important role in this remodeling. The fate of a membrane protein after endocytosis can depend on whether a protein ‘tag’ called ubiquitin has been added to it. Ubiquitin-marked proteins bud off into the cell and are then sent to cell structures called lysosomes to be degraded, whereas unmarked proteins are recycled back to the plasma membrane. Yeast cell membranes contain a protein called Jen1 that transports certain molecules, including one called lactate that can be used as fuel for growth. However, glucose is a preferred nutrient for yeast, so when glucose is available, another protein called Rod1 becomes activated and promotes the addition of ubiquitin to Jen1, and hence its degradation. This means that the cells can no longer use lactate as a source of energy. However, it was not known where in the cell the Rod1 protein does this. Becuwe and Léon labeled proteins involved in endocytosis with fluorescent tags and used microscopy to observe their fate in live yeast cells exposed to glucose. This revealed two roles for Rod1. At the plasma membrane, Rod1 helps Jen1 to be taken into the cell in the early stages of endocytosis. But unexpectedly, Rod1 is also found at a cellular structure called the trans-Golgi network, small membrane sacs that are typically responsible for packaging proteins so they can be transported to a new destination, in particular the plasma membrane. This suggests that Rod1 can also act at this location in the cell. When the proteins responsible for maintaining transport to the trans-Golgi network are inhibited, Jen1 is no longer degraded, even when glucose is present; instead, Jen1 is recycled back to the plasma membrane. Becuwe and Léon therefore propose that a second level of control of the degradation of plasma membrane proteins occurs in the trans-Golgi network, and so this compartment has an essential role in sorting proteins for degradation or recycling. The group of proteins that Rod1 belongs to, named arrestins, has been suggested to play important roles in several diseases, including diabetes and cancer. As many of the features of the endocytic pathway are conserved in a broad range of species, arrestins may also be important for controlling the fate of membrane proteins at multiple places in mammalian cells. However, further work is required to confirm this. DOI:http://dx.doi.org/10.7554/eLife.03307.002
Collapse
Affiliation(s)
- Michel Becuwe
- Department of Cell Biology, Institut Jacques Monod, Université Paris-Diderot, CNRS, Paris, France
| | - Sébastien Léon
- Department of Cell Biology, Institut Jacques Monod, Université Paris-Diderot, CNRS, Paris, France
| |
Collapse
|
40
|
Pratelli R, Pilot G. Regulation of amino acid metabolic enzymes and transporters in plants. JOURNAL OF EXPERIMENTAL BOTANY 2014; 65:5535-56. [PMID: 25114014 DOI: 10.1093/jxb/eru320] [Citation(s) in RCA: 218] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Amino acids play several critical roles in plants, from providing the building blocks of proteins to being essential metabolites interacting with many branches of metabolism. They are also important molecules that shuttle organic nitrogen through the plant. Because of this central role in nitrogen metabolism, amino acid biosynthesis, degradation, and transport are tightly regulated to meet demand in response to nitrogen and carbon availability. While much is known about the feedback regulation of the branched biosynthesis pathways by the amino acids themselves, the regulation mechanisms at the transcriptional, post-transcriptional, and protein levels remain to be identified. This review focuses mainly on the current state of our understanding of the regulation of the enzymes and transporters at the transcript level. Current results describing the effect of transcription factors and protein modifications lead to a fragmental picture that hints at multiple, complex levels of regulation that control and coordinate transport and enzyme activities. It also appears that amino acid metabolism, amino acid transport, and stress signal integration can influence each other in a so-far unpredictable fashion.
Collapse
Affiliation(s)
- Réjane Pratelli
- Plant Pathology, Physiology and Weed Science, Virginia Tech, Blacksburg, VA 24060, USA
| | - Guillaume Pilot
- Plant Pathology, Physiology and Weed Science, Virginia Tech, Blacksburg, VA 24060, USA
| |
Collapse
|
41
|
Kornilova ES. Receptor-mediated endocytosis and cytoskeleton. BIOCHEMISTRY (MOSCOW) 2014; 79:865-78. [DOI: 10.1134/s0006297914090041] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
42
|
Moeller HB, Aroankins TS, Slengerik-Hansen J, Pisitkun T, Fenton RA. Phosphorylation and ubiquitylation are opposing processes that regulate endocytosis of the water channel aquaporin-2. J Cell Sci 2014; 127:3174-83. [PMID: 24876223 DOI: 10.1242/jcs.150680] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The post-translational modifications (PTMs) phosphorylation and ubiquitylation regulate plasma membrane protein function. Here, we examine the interplay between phosphorylation and ubiquitylation of the membrane protein aquaporin-2 (AQP2) and demonstrate that phosphorylation can override the previously suggested dominant endocytic signal of K63-linked polyubiquitylation. In polarized epithelial cells, although S256 is an important phosphorylation site for AQP2 membrane localization, the rate of AQP2 endocytosis was reduced by prolonging phosphorylation specifically at S269. Despite their close proximity, AQP2 phosphorylation at S269 and ubiquitylation at K270 can occur in parallel, with increased S269 phosphorylation and decreased AQP2 endocytosis occurring when K270 polyubiquitylation levels are maximal. In vivo studies support this data, with maximal levels of AQP2 ubiquitylation occurring in parallel to maximal S269 phosphorylation and enhanced AQP2 plasma membrane localization. In conclusion, we demonstrate for the first time that although K63-linked polyubiquitylation marks AQP2 for endocytosis, site-specific phosphorylation can counteract polyubiquitylation to determine its final localization. Similar mechanisms might exist for other plasma membrane proteins.
Collapse
Affiliation(s)
- Hanne B Moeller
- Department of Biomedicine, Center for Interactions of Proteins in Epithelial Transport (InterPrET), Aarhus University, Aarhus DK-8000, Denmark
| | - Takwa Shaiman Aroankins
- Department of Biomedicine, Center for Interactions of Proteins in Epithelial Transport (InterPrET), Aarhus University, Aarhus DK-8000, Denmark
| | - Joachim Slengerik-Hansen
- Department of Biomedicine, Center for Interactions of Proteins in Epithelial Transport (InterPrET), Aarhus University, Aarhus DK-8000, Denmark
| | - Trairak Pisitkun
- Department of Biomedicine, Center for Interactions of Proteins in Epithelial Transport (InterPrET), Aarhus University, Aarhus DK-8000, Denmark Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Robert A Fenton
- Department of Biomedicine, Center for Interactions of Proteins in Epithelial Transport (InterPrET), Aarhus University, Aarhus DK-8000, Denmark
| |
Collapse
|
43
|
Yeast nutrient transceptors provide novel insight in the functionality of membrane transporters. Curr Genet 2013; 59:197-206. [PMID: 24114446 PMCID: PMC3824880 DOI: 10.1007/s00294-013-0413-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 09/17/2013] [Accepted: 09/30/2013] [Indexed: 11/21/2022]
Abstract
In the yeast Saccharomyces cerevisiae several nutrient transporters have been identified that possess an additional function as nutrient receptor. These transporters are induced when yeast cells are starved for their substrate, which triggers entry into stationary phase and acquirement of a low protein kinase A (PKA) phenotype. Re-addition of the lacking nutrient triggers exit from stationary phase and sudden activation of the PKA pathway, the latter being mediated by the nutrient transceptors. At the same time, the transceptors are ubiquitinated, endocytosed and sorted to the vacuole for breakdown. Investigation of the signaling function of the transceptors has provided a new read-out and new tools for gaining insight into the functionality of transporters. Identification of amino acid residues that bind co-transported ions in symporters has been challenging because the inactivation of transport by site-directed mutagenesis is not conclusive with respect to the cause of the inactivation. The discovery of nontransported agonists of the signaling function in transceptors has shown that transport is not required for signaling. Inactivation of transport with maintenance of signaling in transceptors supports that a true proton-binding residue was mutagenised. Determining the relationship between transport and induction of endocytosis has also been challenging, since inactivation of transport by mutagenesis easily causes loss of all affinity for the substrate. The use of analogues with different combinations of transport and signaling capacities has revealed that transport, ubiquitination and endocytosis can be uncoupled in several unexpected ways. The results obtained are consistent with transporters undergoing multiple substrate-induced conformational changes, which allow interaction with different accessory proteins to trigger specific downstream events.
Collapse
|
44
|
The SLC3 and SLC7 families of amino acid transporters. Mol Aspects Med 2013; 34:139-58. [PMID: 23506863 DOI: 10.1016/j.mam.2012.10.007] [Citation(s) in RCA: 503] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 08/15/2012] [Indexed: 01/18/2023]
Abstract
Amino acids are necessary for all living cells and organisms. Specialized transporters mediate the transfer of amino acids across plasma membranes. Malfunction of these proteins can affect whole-body homoeostasis giving raise to diverse human diseases. Here, we review the main features of the SLC3 and SLC7 families of amino acid transporters. The SLC7 family is divided into two subfamilies, the cationic amino acid transporters (CATs), and the L-type amino acid transporters (LATs). The latter are the light or catalytic subunits of the heteromeric amino acid transporters (HATs), which are associated by a disulfide bridge with the heavy subunits 4F2hc or rBAT. These two subunits are glycoproteins and form the SLC3 family. Most CAT subfamily members were functionally characterized and shown to function as facilitated diffusers mediating the entry and efflux of cationic amino acids. In certain cells, CATs play an important role in the delivery of L-arginine for the synthesis of nitric oxide. HATs are mostly exchangers with a broad spectrum of substrates and are crucial in renal and intestinal re-absorption and cell redox balance. Furthermore, the role of the HAT 4F2hc/LAT1 in tumor growth and the application of LAT1 inhibitors and PET tracers for reduction of tumor progression and imaging of tumors are discussed. Finally, we describe the link between specific mutations in HATs and the primary inherited aminoacidurias, cystinuria and lysinuric protein intolerance.
Collapse
|
45
|
Dutta D, Chakraborty S, Bandyopadhyay C, Valiya Veettil M, Ansari MA, Singh VV, Chandran B. EphrinA2 regulates clathrin mediated KSHV endocytosis in fibroblast cells by coordinating integrin-associated signaling and c-Cbl directed polyubiquitination. PLoS Pathog 2013; 9:e1003510. [PMID: 23874206 PMCID: PMC3715429 DOI: 10.1371/journal.ppat.1003510] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 06/06/2013] [Indexed: 12/22/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) interacts with human dermal endothelial cell surface tyrosine kinase EphrinA2 (EphA2) and integrins (α3β1 and αVβ3) in the lipid raft (LR) region, and EphA2 regulates macropinocytic virus entry by coordinating integrin-c-Cbl associated signaling. In contrast, KSHV enters human foreskin fibroblast (HFF) cells by LR-independent clathrin mediated endocytosis. The present studies conducted to identify the key molecules regulating KSHV entry in HFF cells showed that KSHV induces association with integrins (αVβ5, αVβ3 and α3β1) and EphA2 in non-LR regions early during infection and activates EphA2, which in turn associates with phosphorylated c-Cbl, myosin IIA, FAK, Src, and PI3-K, as well as clathrin and its adaptor AP2 and effector Epsin-15 proteins. EphA2 knockdown significantly reduced these signal inductions, virus internalization and gene expression. c-Cbl knockdown ablated the c-Cbl mediated K63 type polyubiquitination of EphA2 and clathrin association with EphA2 and KSHV. Mutations in EphA2's tyrosine kinase domain (TKD) or sterile alpha motif (SAM) abolished its interaction with c-Cbl. Mutations in tyrosine kinase binding (TKB) or RING finger (RF) domains of c-Cbl resulted in very poor association of c-Cbl with EphA2 and decreased EphA2 polyubiquitination. These studies demonstrated the contributions of these domains in EphA2 and c-Cbl association, EphA2 polyubiquitination and virus-EphA2 internalization. Collectively, these results revealed for the first time that EphA2 influences the tyrosine phosphorylation of clathrin, the role of EphA2 in clathrin mediated endocytosis of a virus, and c-Cbl mediated EphA2 polyubiquitination directing KSHV entry in HFF cells via coordinated signal induction and progression of endocytic events, all of which suggest that targeting EphA2 and c-Cbl could block KSHV entry and infection.
Collapse
Affiliation(s)
- Dipanjan Dutta
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Sayan Chakraborty
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Chirosree Bandyopadhyay
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Mohanan Valiya Veettil
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Mairaj Ahmed Ansari
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Vivek Vikram Singh
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Bala Chandran
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
46
|
O'Donnell AF, Huang L, Thorner J, Cyert MS. A calcineurin-dependent switch controls the trafficking function of α-arrestin Aly1/Art6. J Biol Chem 2013; 288:24063-80. [PMID: 23824189 DOI: 10.1074/jbc.m113.478511] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Proper regulation of plasma membrane protein endocytosis by external stimuli is required for cell growth and survival. In yeast, excess levels of certain nutrients induce endocytosis of the cognate permeases to prevent toxic accumulation of metabolites. The α-arrestins, a family of trafficking adaptors, stimulate ubiquitin-dependent and clathrin-mediated endocytosis by interacting with both a client permease and the ubiquitin ligase Rsp5. However, the molecular mechanisms that control α-arrestin function are not well understood. Here, we show that α-arrestin Aly1/Art6 is a phosphoprotein that specifically interacts with and is dephosphorylated by the Ca(2+)- and calmodulin-dependent phosphoprotein phosphatase calcineurin/PP2B. Dephosphorylation of Aly1 by calcineurin at a subset of phospho-sites is required for Aly1-mediated trafficking of the aspartic acid and glutamic acid transporter Dip5 to the vacuole, but it does not alter Rsp5 binding, ubiquitinylation, or stability of Aly1. In addition, dephosphorylation of Aly1 by calcineurin does not regulate the ability of Aly1 to promote the intracellular sorting of the general amino acid permease Gap1. These results suggest that phosphorylation of Aly1 inhibits its vacuolar trafficking function and, conversely, that dephosphorylation of Aly1 by calcineurin serves as a regulatory switch to promote Aly1-mediated trafficking to the vacuole.
Collapse
Affiliation(s)
- Allyson F O'Donnell
- Department of Biology, Stanford University, Stanford, California 94305-5020, USA.
| | | | | | | |
Collapse
|
47
|
Sidoryk-Wegrzynowicz M, Aschner M. Manganese toxicity in the central nervous system: the glutamine/glutamate-γ-aminobutyric acid cycle. J Intern Med 2013; 273:466-77. [PMID: 23360507 PMCID: PMC3633698 DOI: 10.1111/joim.12040] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Manganese (Mn) is an essential trace element that is required for maintaining proper function and regulation of numerous biochemical and cellular reactions. Despite its essentiality, at excessive levels Mn is toxic to the central nervous system (CNS). Increased accumulation of Mn in specific brain regions, such as the substantia nigra, globus pallidus and striatum, triggers neurotoxicity resulting in a neurological brain disorder, termed manganism. Mn has been also implicated in the pathophysiology of several other neurodegenerative diseases. Its toxicity is associated with disruption of the glutamine (Gln)/glutamate (Glu)-γ-aminobutyric acid (GABA) cycle (GGC) between astrocytes and neurons, thus leading to changes in Glu-ergic and/or GABAergic transmission and Gln metabolism. Here we discuss the common mechanisms underlying Mn-induced neurotoxicity and their relationship to CNS pathology and GGC impairment.
Collapse
|
48
|
Sorkina T, Caltagarone J, Sorkin A. Flotillins regulate membrane mobility of the dopamine transporter but are not required for its protein kinase C dependent endocytosis. Traffic 2013; 14:709-24. [PMID: 23418867 DOI: 10.1111/tra.12059] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 02/13/2013] [Accepted: 02/18/2013] [Indexed: 12/15/2022]
Abstract
Flotillins were proposed to mediate clathrin-independent endocytosis, and recently, flotillin-1 was implicated in the protein kinase C (PKC)-triggered endocytosis of the dopamine transporter (DAT). Since endocytosis of DAT was previously shown to be clathrin-mediated, we re-examined the role of clathrin coat proteins and flotillin in DAT endocytosis using DAT tagged with the hemagglutinin epitope (HA) in the extracellular loop and a quantitative HA antibody uptake assay. Depletion of flotillin-1, flotillin-2 or both flotillins together by small interfering RNAs (siRNAs) did not inhibit PKC-dependent internalization and degradation of HA-DAT. In contrast, siRNAs to clathrin heavy chain and μ2 subunit of clathrin adaptor complex AP-2 as well as a dynamin inhibitor Dyngo-4A significantly decreased PKC-dependent endocytosis of HA-DAT. Similarly, endocytosis and degradation of DAT that is not epitope-tagged were highly sensitive to the clathrin siRNAs and dynamin inhibition but were not affected by flotillin knockdown. Very little co-localization of DAT with flotillins was observed in cells ectopically expressing DAT and in cultured mouse dopaminergic neurons. Depletion of flotillins increased diffusion rates of HA-DAT in the plasma membrane, suggesting that flotillin-organized microdomains may regulate the lateral mobility of DAT. We propose that clathrin-mediated endocytosis is the major pathway of PKC-dependent internalization of DAT, and that flotillins may modulate functional association of DAT with plasma membrane rafts rather than mediate DAT endocytosis.
Collapse
Affiliation(s)
- Tatiana Sorkina
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | | | | |
Collapse
|
49
|
Kou L, Sun J, Zhai Y, He Z. The endocytosis and intracellular fate of nanomedicines: Implication for rational design. Asian J Pharm Sci 2013. [DOI: 10.1016/j.ajps.2013.07.001] [Citation(s) in RCA: 392] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
50
|
Su H, Carter CB, Laur O, Sands JM, Chen G. Forskolin stimulation promotes urea transporter UT-A1 ubiquitination, endocytosis, and degradation in MDCK cells. Am J Physiol Renal Physiol 2012; 303:F1325-32. [PMID: 22914781 PMCID: PMC3518190 DOI: 10.1152/ajprenal.00248.2012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 08/21/2012] [Indexed: 11/22/2022] Open
Abstract
The adenylyl cyclase stimulator forskolin (FSK) stimulates UT-A1 phosphorylation, membrane trafficking, and urea transport activity. Here, we found that FSK stimulation induces UT-A1 ubiquitination in UT-A1 Madin-Darby canine kidney (MDCK) cells. This suggests that phosphorylation by FSK also triggers the protein degradation machinery for UT-A1. UT-A1-MDCK cells were treated with 100 μg/ml cycloheximide to inhibit protein synthesis, with or without 10 μM FSK. Total UT-A1 protein abundance was significantly reduced after FSK treatment, concomitantly ubiquitinated UT-A1 was increased. We then specifically investigated the effect of FSK on UT-A1 expressed on the cell plasma membrane. FSK treatment accelerated UT-A1 removal from the cell plasma membrane by increasing UT-A1 endocytosis as judged by biotinylation/MesNa treatment and confocal microscopy. We further found that inhibition of the clathrin-mediated endocytic pathway, but not the caveolin-mediated endocytic pathway, significantly blocks FSK-stimulated UT-A1 endocytosis. The PKA inhibitor H89 and the proteasome inhibitors MG132 and lactacystin reduced FSK-induced membrane UT-A1 reduction. Our study shows that FSK activates the UT-A1 urea transporter and the activation/phosphorylation subsequently triggers the downregulation of UT-A1, which represents an important mechanism for the cell to return to the basal conditions after vasopressin stimulation.
Collapse
Affiliation(s)
- Hua Su
- Department of Medicine, Renal Division, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | |
Collapse
|